Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 20:30:51
Clarke’s Analysis of Drugs and P...
4279 downloads
11427 Views
44MB Size
Report
This content was uploaded by our users and we assume good faith they have the permission to share this book. If you own the copyright to this book and it is wrongfully on our website, we offer a simple DMCA procedure to remove your content from our site. Start by pressing the button below!
Report copyright / DMCA form
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 20:30:51
Clarke’s Analysis of Drugs and Poisons
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 20:30:51
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 20:30:51
Clarke’s Analysis of Drugs and Poisons in pharmaceuticals, body fluids and postmortem material FOURTH EDITION
Consulting Editors
Anthony C Moffat M David Osselton Brian Widdop Executive Development Editor
Jo Watts
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 20:30:51
Published by Pharmaceutical Press 1 Lambeth High Street, London SE1 7JN, UK 1559 St Paul Avenue, Gurnee, IL 60031, USA # Pharmaceutical Press 2011 Chapter 13: Figures 13.1–13.21, 13.25–13.32 # TICTAC Communications is a trade mark of Pharmaceutical Press Pharmaceutical Press is the publishing division of the Royal Pharmaceutical Society First edition, edited by EGC Clarke, published 1969 (Vol. 1) and 1975 (Vol. 2) Second edition (in one volume) published 1986 Third edition published 2004 Fourth edition published 2011 Typeset by Thomson Digital, Noida, India Printed in Italy by LEGO S.p.A. ISBN 978 0 85369 711 4 All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, without the prior written permission of the copyright holder. The publisher makes no representation, express or implied, with regard to the accuracy of the information contained in this book and cannot accept any legal responsibility or liability for any errors or omissions that may be made. A catalogue record for this book is available from the British Library.
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 20:30:51
Contents
VOLUME 1 Editorial Advisory Board vii Editorial and Production Staff Contributors ix About the Editors xi Foreword xii Preface xiii General Notices xv Abbreviations xix
viii
Part One: Chapters 1 Hospital Toxicology 3 DRA Uges 2 Therapeutic Drug Monitoring 59 M Hallworth 3 Workplace Drug Testing 73 A Verstraete M Peat 4 Driving Under the Influence of Alcohol 87 AW Jones 5 Driving Under the Influence of Drugs 115 BK Logan MD Osselton 6 Drug Testing in Human Sport 127 DA Cowan 7 Drug Testing in Animal Sport 138 P Teale 8 Drug-facilitated Sexual Assault 147 MD Osselton 9 Forensic Toxicology 160 MD Osselton AC Moffat B Widdop
15 Natural Toxins 243 JF de Wolff FA de Wolff 16 Pesticides 258 M Kała 17 Metals and Anions 288 R Braithwaite 18 Drugs in Saliva 308 V Spiehler 19 Hair Analysis 323 P Kintz 20 Method Development and Validation 334 FT Peters 21 Quality Control in the Pharmaceutical Industry 350 P Graham 22 Quality Control and Accreditation in the Toxicology Laboratory 361 AC Moffat 23 Measuring and Reporting Uncertainty 371 MA LeBeau 24 Pharmacokinetics and Metabolism 388 OH Drummer 25 Pharmacogenomics 401 SHY Wong 26 Interpretation of Toxicological Data 417 OH Drummer S Karch 27 Paediatric Toxicology 429 D Reith 28 Sampling, Storage and Stability 445 S Kerrigan
10 Postmortem Toxicology 176 G Jones
29 Extraction 458 T Stimpfl
11 Drugs of Abuse 190 SD McDermott
30 Colour Tests 471 B Widdop
12 Medicinal Products 208 AC Moffat AG Davidson
31 Immunoassays 496 RS Niedbala JM Gonzalez
13 Solid Dosage Form Identification 219 J Ramsey
32 Ultraviolet, Visible and Fluorescence Spectrophotometry 507 J Cordonnier J Schaep
14 Volatile Substances 230 RJ Flanagan
33 Infrared Spectroscopy 521 RD Jee
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 20:30:51
vi
Contents
34 Near-infrared Spectroscopy 538 RD Jee
44 Emerging Techniques 787 D Rudd
35 Raman Spectroscopy 553 DE Bugay PA Martoglio Smith FC Thorley
Subject Index Ii
36 Nuclear Magnetic Resonance Spectroscopy 564 JC Lindon JK Nicholson 37 Mass Spectrometry 577 D Watson 38 Liquid Chromatography–Mass Spectrometry 594 HH Maurer 39 Thin-layer Chromatography 600 CF Poole 40 Gas Chromatography 636 S Dawling 41 High Performance Liquid Chromatography 718 T Kupiec P Kemp 42 Capillary Electrophoresis 758 F Tagliaro A Fanigliulo J Pascali F Bortolotti 43 Atomic Absorption Spectroscopy, Inductively Coupled Plasma–Mass Spectrometry and Other Techniques for Measuring the Concentrations of Metals 773 A Taylor
VOLUME 2 Part Two: Monographs 807 Part Three: Indexes of Analytical Data CAS Numbers 2263 Molecular Formulae 2278 Functional Classes: Therapeutic 2305 Functional Classes: Pesticides 2315 Functional Classes: Other Substances 2317 Molecular Weights 2328 Melting Points 2342 Colour Tests 2353 Thin-layer Chromatographic Data 2358 Gas Chromatographic Data 2392 High Performance Liquid Chromatographic Data 2410 Ultraviolet Absorption Data 2427 Infrared Peaks 2442 Mass Spectral Data of Drugs 2451 Mass Spectral Data of Pesticides 2460 Reagents 2461 Pharmacological Terms 2463
Subject Index Ii
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 10:1:25
Editorial Advisory Board
Dr Craig Chatterton
Dr Jan-Piet Franke
Dr Christine Moore
CNC Forensic Toxicology Services, c/o 7 Sawley Close, Darwen, Lancashire BB3 3QY, UK
Department of Pharmaceutical Analysis, University of Groningen, A Deusinglaan 1, 9713 AV Groningen, The Netherlands
Immunalysis Corporation, 829 Towne Centre Drive, Pomona, CA 91767, USA
Dr Hee-Sun Chung
National Forensic Service (formally NISI), 331-1 Sinwol 7-Dong, Yang Chun-Ku, Seoul 158-097, Korea Dr Gail Cooper
Forensic Medicine and Science, University of Glasgow, Scotland G12 8QQ, UK Mr Simon Cosbey
5A Carnalea Aveue, Bangor BT19 1HF, Northern Ireland Dr Simon Elliott
(ROAR) Forensics, Ltd, Malvern Hills Science Park, Malvern, Worcestershire, WR14 3SZ, UK
Dr Sue Paterson Professor Bruce Goldberger
University of Florida – College of Medicine, Department of Pathology and Laboratory Medicine, 4800 SW 35th Drive, Gainesville, FL 32608, USA
Toxicology Unit, Imperial College, St Dunstan’s Road, London W6 8RP, UK Mr Chip H Walls
Forensic Toxicology Laboratory, 12500 SW 152Nd Street, Bldg B, Miami, FL 33177, USA
Dr Rodney G Gullberg
Washington State Toxicology Laboratory, 2203 Airport Way S, Suite 360, Seattle, WA 98134, USA Dr Dan S Isenschmid
Wayne County Medical Examiner’s Office, 1300 E Warren Avenue, Detroit, MI 48207, USA
vii
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 10:1:25
Editorial and Production Staff
Emma Burrows Helen Carter Tamsin Cousins Amy Cruse Simon Dunton Marian Fenton Kelly Davey Rebecca Garner Austin Gibbons David Granger Jo Humm
viii
Jean Macpherson Julie McGlashan Louise McIndoe Ithar Malik Jason Norman Karl Parsons The Prescribers, The School of Pharmacy, London, UK Jo Watts Lucy White John Wilson
Freelance Staff Irene Chiwele Millie Davis Laurent Y Galichet Poppy McLaughlin Eva Reichardt A team of dedicated copyeditors, proofreaders and indexers also helped in the preparation of this publication.
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 21:0:29
Contributors
Dr Federica Bortolotti
Mr Mike Hallworth
Dr Sean D McDermott
Department of Public Health and Community Medicine, Section of Forensic Medicine, University of Verona, Verona, Italy
Department of Clinical Biochemistry, Royal Shrewsbury Hospital, Shrewsbury, UK
Drugs Intelligence Unit, Forensic Science Laboratory, Dublin, Ireland
Dr Roger D Jee
Professor Anthony C Moffat
Dr Robin A Braithwaite
Regional Toxicology Laboratory, City Hospital NHS Trust, Birmingham, UK Dr David E Bugay
SSCI Inc., West Lafayette, USA Dr Jan Cordonnier
The School of Pharmacy, University of London, UK Dr Graham Jones
Office of Chief Medical Examiner, Edmonton, Canada Dr A Wayne Jones
Formerly, Royal Pharmaceutical Society of Great Britain and, The School of Pharmacy, University of London, London, UK Professor Jeremy K Nicholson
Department of Surgery and Cancer, Imperial College of Science, Technology and Medicine, London, UK
Dorpsstaat 106, B-8340 Sysele-Damme, Belgium
National Board of Forensic Medicine, Department of Forensic Chemistry, Linkoping, Sweden
Professor David A Cowan
Dr Maria Kała
Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania, USA
Department of Forensic Toxicology, Institute of Forensic Research, Cracow, Poland
Professor M David Osselton
Drug Control Centre, King’s College London, UK
Dr R Sam Niedbala
University of Bournemouth, Dorset, UK Dr Alastair G Davidson
Department of Pharmaceutical Sciences, University of Strathclyde, Glasgow, UK Dr Sheila Dawling
Vanderbilt University Medical Centre, Diagnostic Labs – TVC, Nashville, Tennessee, USA Professor Frederik A de Wolff
Toxicology Laboratory, Leiden University Medical Center, Leiden, The Netherlands
Dr Steven Karch
PO Box 5139, Berkeley, CA 94705, USA
Dr Jennifer Pascali
Dr Phil Kemp
Department of Public Health and Community Medicine, Section of Forensic Medicine, University of Verona, Verona, Italy
Analytical Research Laboratories, Oklahoma City, USA Dr Sarah Kerrigan
SHSU Regional Crime Laboratory, The Woodlands, Texas, USA
Dr Jacob F de Wolff
University College London Hospitals NHS Foundation Trust, London, UK
Dr Thomas Kupiec
Victorian Inst. of Forensic Medicine, Southbank, Victoria, Australia Dr Ameriga Fanigliulo
Department of Public Health and Community Medicine, Section of Forensic Medicine, University of Verona, Verona, Italy Dr Robert J Flanagan
Analytical Research Laboratories, Oklahoma City, USA
Institut f€ ur Rechtsmedizin, Universit€atsklinikum Jena, Jena, Germany Professor Colin F Poole
Department of Chemistry, Wayne State University, Detroit, Michigan, USA
Dr Marc LeBeau
Dr John Ramsey
Chemistry Unit, FBI Laboratory, Quantico, Virginia, USA
Division of Cardiological Sciences, St George’s Hospital Medical School, London, UK
Professor John C Lindon
Division of Biomedical Sciences, Imperial College of Science, Technology and Medicine, London, UK
Medical Toxicology Unit, Guy’s and St Thomas’ Hospital Trust, London, UK
Dr Barry K Logan
Dr Jesus M Gonzalez
Dr Pamela A Martoglio Smith
Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania, USA
Quest Diagnostics, Houston, Texas, USA Dr Frank T Peters
Dr Pascal Kintz
Laboratoire Chemtox, Illkirch-Graffenstaden, France
Dr Olaf H Drummer
Dr Michael A Peat
NMS Labs, Willow Grove, Pennsylvania, USA SSCI Inc., West Lafayette, USA
Dr David Reith
Dunedin School of Medicine, University of Otago, New Zealand Dr Dave Rudd
GlaxoSmithKline Manufacturing, Ware, Hertfordshire, UK Dr Johan Schaep
Chemiphar n.v., Brugge, Belgium Professor Hans H Maurer
Dr Paul Graham
Walker Graham Pharma Consulting Ltd, Ashington, Northumberland, UK
Department of Experimental and Clinical Toxicology, University of Saarland, Homburg/ Saar, Germany
Dr Vina Spiehler
422 Tustin Avenue, Newport Beach, CA 92663, USA ix
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 21:0:29
x
Contributors
Dr Thomas Stimpfl
Dr Fiona C Thorley
Dr Jo Watts
Department of Forensic Medicine, Wien, Austria
SSCI, A Division of Aptuit, Abingdon, Oxfordshire, UK
Pharmaceutical Press, Royal Pharmaceutical Society of Great Britain, London, UK
Professor Franco Tagliaro
Professor Donald RA Uges
Dr Brian Widdop
Department of Medicine and Public Health, University of Verona, University Hospital, Verona, Italy
Laboratory for Clinical and Forensic Toxicology and Drug Analysis, University Hospital Groningen and University Centre of Pharmacy, Groningen, The Netherlands
Formerly, Medical Toxicology Unit, Guy’s and St Thomas’ Hospital Trust, London, UK
Dr Andrew Taylor
Dr Alain Verstraete
Royal Surrey County Hospital, Guildford, Surrey, UK
Klinische Biologie, Universitair Ziekenhuis, Ghent, Belgium
Dr Phil Teale
Dr David Watson
Medication and Doping Control, HFL Sport Science Quotient Bioresearch, UK
Department of Pharmaceutical Sciences, University of Strathclyde, Glasgow, UK
Dr Steve HY Wong
Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 10:24:0
About the Editors
Professor Anthony C Moffat BPharm, PhD, DSc, CChem, FRSC, FRPharmS, FFIP, FFSSoc
Professor Anthony C Moffat is Emeritus Professor of Pharmaceutical Analysis at The School of Pharmacy, University of London, where he was previously Head of the Centre for Pharmaceutical Analysis. He was also Chief Scientist at the Royal Pharmaceutical Society. He has over 350 publications as well as the co-authorship of eight books. Previously he worked for the Forensic Science Service for 23 years as Research Co-ordinator (Birmingham Laboratory), Resources Manager (Huntingdon Laboratory), Head of Quality Management (HQ, London), Assistant Director (Huntingdon Laboratory), and Head of the Drugs and Toxicology Division at the Home Office Central Research Establishment, Aldermaston. He has also been a Superintendent Pharmacist in a community pharmacy, Assistant Professor of Biochemistry, Baylor College of Medicine, Houston, Texas, and Chief Pharmacist, St Leonard’s Hospital, London. An active member of many professional and learned societies, his fellowships include the Royal Pharmaceutical Society, Royal Society of Chemistry, Forensic Science Society, International Pharmaceutical Federation and the American Association of Pharmaceutical Scientists as well as the membership of the International Association of Forensic Toxicologists. Professor M David Osselton BSc, PhD, CSci, CChem, FRSC, MEWI
Professor M David Osselton started his forensic toxicology career in 1974 when he went to work with Dr Alan Curry at the Home Office Central Research Establishment, Aldermaston. He gained casework experience as Senior Toxicologist working at the Home Office Forensic Science laboratories in Nottingham and Huntingdon before returning to Aldermaston in 1984 to succeed Dr Anthony Moffat as Head of Research in Alcohol, Drugs and Toxicology. In 1991, he was appointed Head of Toxicology for the Forensic Science Service. In 2007, Professor Osselton went to Bournemouth University as Head of the Centre for Forensic Sciences. He has wide experience in toxicology casework and has been involved in numerous high profile cases working for the defence and prosecution both in the UK and overseas. He is
internationally known for his research interests in toxicology and lectures widely at conferences as a plenary and keynote speaker. Between 2003 and 2009 he was Lead Assessor (Toxicology) for the UK Council for the Registration of Forensic Practitioners (CRFP) and was Visiting Professor to the Department of Forensic Science and Drug Monitoring at Kings College, University of London (2004–2007). He is a Fellow/Member of a number of professional and learned bodies including the Royal Society of Chemistry, Royal Society of Medicine, Expert Witness Institute, International Association of Forensic Toxicologists (TIAFT), Society of Forensic Toxicologists (SOFT), LTG (formally the London Toxicology Group), UK Workplace Drug Testing Forum and is chair of the United Kingdom and Ireland Association of Forensic Toxicologists. Dr Brian Widdop BSc, PhD, SRCS, CChem, FRCS, FRCPath
Dr Brian Widdop was Director of the Medical Toxicology Unit Laboratory at Guy’s Hospital, London, from 1970 until 2002. Dr Widdop has been a Speciality Assessor for the Council for the Registration of Forensic Practitioners, a member of the WHO IPCS Working Group on Analytical Toxicology, Chief Advisor to the South East Asia Regional Office of the WHO on analytical toxicology and was a Director of the Board of the UK Horse-racing Forensic Laboratory from 1991 to 2002. From 1997 to 2001, he was joint co-ordinator of the European proficiency Testing Scheme for Drugs of Abuse. Dr Widdop has published over 80 papers on various aspects of clinical and analytical toxicology and has spoken at many international meetings. He is also a member of the editorial board of the Journal of Analytical Toxicology. Dr Widdop belongs to several international scientific societies and was a founder member of the London Toxicology Group. He has been a member of The International Association of Forensic Toxicologists for 42 years and was the recipient of the Alan Curry Award in 2002. Dr Jo Watts BSc, PhD
Dr Jo Watts attained her degree in pharmacology and toxicology followed by a PhD in neuropharmacology, both at The School of Pharmacy, University of London. She is a member of TIAFT and the LTG.
xi
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 10:27:22
Foreword
As one of the past presidents of the International Association of Forensic Toxicologists (TIAFT) it is an honour as well as a pleasure for me to write a foreword for the fourth edition of this prestigious publication. Indeed, when a publication is prepared by an impressive number of leading toxicologists working in world-famous institutions, as editors or as former or new authors – all outstanding specialists in their respective fields of activity – we as toxicologists can only expect to have another great database in our hands with which to do research or our daily work. In addition to the monographs revised from the previous editions, dealing with physicochemical and pharmacotoxicological properties of drugs and poisons, and the indexes of analytical data, review chapters have been included on various basic subjects of toxicology, such as hospital and forensic toxicology; immunoassays; analysis of alternative matrices; doping; driving under the influence of drugs and alcohol; therapeutic drug monitoring; workplace testing; quality assurance; pharmacokinetics; pesticides; volatile organic substances; natural toxins; different separation technologies; and spectrometric methods. New chapters by renowned experts have been added that deal with method development and validation; sampling, storage and stability; extraction; more recently developed analytical technologies such as liquid chromatography coupled to mass spectrometry; atomic absorption spectrometry, or inductively coupled mass spectrometry for the determination of inorganic poisons; pharmacogenomics; measuring and reporting uncertainty; drug facilitated sexual assaults; and paediatric forensic toxicology. These topics enable our younger and less young colleagues alike to familiarise themselves with these developments or to improve their knowledge. Especially at a time when shortcuts are made for budgetary reasons in healthcare and forensic systems, we need to document our performances of reliable analytical results followed by correct interpretations of these results to proof our usefulness to decision makers. Therefore, this fourth edition is published at the perfect moment. I remember very well at the beginning of my career in Luxembourg the moment I held the first edition from 1969, which was recommended to me by a French colleague. It was always a bible for me and is still an important part of my personal library. Several similar publications have been produced in the past, but they have never had the same impact on
xii
toxicology as Clarke. My professional work was also influenced by the renowned British scientists who I met during a TIAFT conference in Ghent back in the early ’70s. What a lot has changed since those days! It is an important advance that a chapter on interpretation has been added, as this is the major difference between analytical toxicology and analytical chemistry. Toxicological analysis is not analytical toxicology. From my work in forensic toxicology, I know that in court we are questioned more about interpretation of our results than about the performance of the analytical methods. Our customers should be aware that we are not ‘only making measurements’. Since toxicology is a multidisciplinary science, toxicologists need not only to have comprehensive knowledge of analytical methodology, but also to be proficient in the basics of medicine, physiology, clinical chemistry, biochemistry, pharmacodynamics and pharmacokinetics in order to provide the best possible contributions to clinicians and to forensic authorities. Toxicology is a difficult and a complex issue requiring collective information exchange among toxicology specialists from all related fields. Even the publishing of routine cases that may not be routine for other colleagues should be encouraged. In spite of the fantastic efforts in recent years to establish correlations between toxicant concentrations in body tissues and pharmacodynamic action, behaviour impairment, interindividual variability, pharmacogenetics, postmortem changes or concurrent existing pathologies, there is still a lot of research required to improve our knowledge. So, I can only recommend to my colleagues: let us do it! Even now that a staggering amount of information is available via the internet, Clarke remains a reference for old and young toxicologists. It is an easily accessible tool which can be consulted either by reading the book or by browsing and searching the online version, to give us useful structured, and peer-reviewed information written by well-known experts. I wish Clarke the best success that it deserves, to equal that of the previous three editions of this publication. Robert Wennig, PhD Past President of TIAFT
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 16:42:57
Preface
Clarke’s Analysis of Drugs and Poisons aims to be the world’s leading text on the analysis of drugs and poisons. Not only does it contain chapters on the methodology and techniques of modern analytical toxicology, but the monographs include analytical data on therapeutic drugs, drugs of abuse, drugs misused in sport as well as pesticides, metals and other poisons. This fourth edition builds on the previous editions with significant updating and improvements in scope and electronic form. Clarke, as it is affectionately known, has gained a world-wide reputation as a reliable source of toxicological information. Its presence on the benches of many different types of pharmaceutical and toxicological laboratories is a testament to its usefulness. Since the third edition was published in 2004, there have been about 120 new chemical entities brought onto the market. Some of these come from completely new chemical or pharmacological groups, but most are ‘me too’ drugs. In addition, there has been a growth of drugs misused in sport and those subject to abuse; eg legal cannabinoids and Mephedrone, and their derivatives. Nearly 400 monographs have been added to the fourth edition, with priority based on the importance of the substances covered in one of the following areas: drugs of abuse, forensic toxicology, hospital emergency toxicology, doping in sport, drugs subject to therapeutic drug monitoring and environmental toxicology. It has been impossible to include all the new drugs and poisons available, but work is continuing to add further data in the future. The information in Clarke has been designed to provide methods and data to enable analysts to detect, identify, quantify and profile drugs and poisons in a wide variety of situations. In addition, information on how to interpret the analytical data is included, since this is often the most difficult part. The book has been designed for use not only in hospital and toxicology laboratories, but also in numerous other analytical establishments. This includes quality control laboratories, and clinical laboratories engaged in drug investigations for purposes such as therapeutic drug monitoring or research into pharmacokinetics and patterns of drug metabolism. In addition, there is much information that will be of use in environmental toxicology, particularly the analysis of toxic metals and pesticides. The needs of students studying analytical and forensic toxicology have not been forgotten and the chapters form an excellent basis for study. The spin-off book Clarke’s Analytical Forensic Toxicology from the third edition of Clarke is a testament to meeting the requirements of the university teaching sector. The book is in two volumes to make it easier to use. Volume 1 contains chapters comprising methodology and analytical techniques, and the subject index to both volumes; Volume 2 contains the analytical and toxicological data, indexes to the analytical data, a list of reagents and a repeat of the subject index to both volumes. Those who regularly use Clarke will be pleased to see that the original style and form of presentation of the information has been retained from the previous edition. This tried and tested format is clear, making it easy to find relevant information. Clarke is now an established publication on MedicinesComplete, which provides online access to some of the world’s leading drug and healthcare references. This includes such reference sources as Martindale: The Complete Drug Reference, British National Formulary, The Merck Index and Stockley’s Drug Interactions. The online version of Clarke has the advantage that text searches can be performed thus aiding the reader to access relevant information more rapidly, either in Clarke alone or across multiple reference sources. Another advantage of the
online version is that it can be updated online far more frequently and easily than the conventional book form.
Volume 1 Part 1: methodology and analytical chapters This part now contains 44 chapters describing methodology and analytical techniques, which is an increase of 13 chapters from the previous edition. Three of the previous chapters have each been split into two because of the increased complexity of the topics covered. Thus there are now chapters on Driving Under the Influence of Alcohol as well as Driving Under the Influence of Drugs; Drugs in Human Sport as well as Drugs in Animal Sport; and Quality Control and Assessment in the Pharmaceutical Industry as well as Quality Control and Assessment in the Toxicology Laboratory. This latter chapter recognises the increase role of accreditation in the forensic toxicology laboratory and gives guidance on how to achieve this. A new chapter on methodology in Drug-facilitated Sexual Assault has been included to recognise the rise in this type of crime and the need for good forensic toxicological analyses. In terms of the use of particular analytical techniques, Method Development and Validation is a new chapter to assist those who need to develop their own methods and demonstrate that they are fit for purpose. Also included in this area are two new chapters on Sampling, Storage and Stability, as well as Extraction, since many toxicologists have asked for information on these topics. The increased use of liquid chromatography–mass spectrometry to replace gas chromatography–mass spectrometry in the analysis of organic compounds has been covered by a new chapter on this topic. Similarly, a new chapter on Atomic Absorption Spectroscopy, Inductively Coupled Plasma–Mass Spectrometry has been added to recognise the increased use of this combination of techniques in inorganic analysis. Four new chapters have been included to assist the toxicologist to interpret analytical data and report the results in a meaningful and clear manner. The chapter on Measuring and Reporting Uncertainty is a clear exposition that all measurements are subject to error and gives guidance on how to measure and report the uncertainty. A chapter on Paediatric Forensic Toxicology recognises that children are not just small adults and need to be treated as a separate population. Similarly, the chapter on Pharmacogenomics clearly shows how we as individuals differ in our genetic makeup and how that might affect our response to drugs. Often one of the most difficult tasks a toxicologist has is to do is to interpret the results of the analyses; a new chapter on Interpretation of Results, together with the updated chapter on Pharmacokinetics, aims to assist toxicologists in this area. This backs up the information on interpretation given in each of the methodology chapters. All the other chapters have been revised to bring them fully up to date. The structure of the spectroscopic and chromatographic chapters has been retained from the previous edition to ensure that all the relevant information is given in an easy-to-read form. The chapter on emerging techniques has been completely rewritten to acknowledge the regulatory aspects of introducing new techniques and what new instrumentation might be available in the future. The chromatographic and capillary electrophoresis systems have been extensively expanded and revised to include general screening systems as well as specialised systems for particular classes of drugs and poisons. The general systems for use have all been proven as robust xiii
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 16:42:58
xiv
Preface
and reproducible over the years, and give excellent results for use in systematic toxicological analysis. Subject index The subject index covering both volumes can be found at the end of Volume 1.
The section entitled Disposition in the Body gives data on therapeutic concentration, toxicity, bioavailability, half-life, volume of distribution, clearance, distribution in blood, plasma : saliva ratio, protein binding and dose to enable analytical data to be interpreted in the context of a given case. In addition, abstracts from published clinical studies and case histories are included. Part three: indexes to analytical data
Volume 2 Part 2: analytical and toxicological data This part contains monographs for 2111 drugs and poisons, which is an increase of around 370 from the last edition. Not only have totally new monographs been introduced, but monographs from previous editions that were excluded from the third edition have been reinstated because the drugs concerned are still used in some parts of the world. The new additions have been chosen for drugs and poisons that are new and widely used prescription drugs, novel drugs of abuse or common poisons not previously included. For example, there are now 15 new monographs on metal salts. All the other monographs have been updated from the third edition. The use of the Recommended International Nonproprietary Name (rINN) for the drug name has been continued as this is now the international standard method of nomenclature. The orientation of the chemical structures has been normalised so that the structures of similar compounds may be compared more easily. In addition, new chemical and analytical data have been added to aid the toxicologist and pharmaceutical analyst. This includes information on stability of drugs in solution and biological fluids at different temperatures, 1-chlorobutane extraction data, and infrared spectra of drug salts. Analytical data for compounds on colour tests, thin-layer chromatography, gas chromatography and high performance liquid chromatography are given from which to choose systems that will separate and identify drugs, poisons and their metabolites. This is followed by full ultraviolet, infrared and mass spectral data together with listings of the major peaks to assist further in identifying compounds. A major change has been made to the Quantification section of each monograph: it has been rearranged to give details of the analysis of each biological fluid or tissue separately instead of being ordered by technique. This makes finding an analytical method to use for a particular tissue very much easier. Additional data such as a method’s limit of detection and limit of quantification have been added when available. This has meant a considerable increase in the size of each monograph and the list of the references at the end of the monograph, but it has improved the usability of the information. All the monographs have been brought up to date by the inclusion of new references and the deletion of old ones whenever possible. The references cited give further information on published methods for separating, identifying and quantifying drugs, poisons and their metabolites. Review articles are given whenever relevant to act as a further source of concise information.
This part contains indexes of analytical, chemical and therapeutic data, arranged in a similar order to how they appear in the monographs: CAS numbers, molecular formulae, therapeutic classes, molecular weights, melting points, colour tests, thin-layer chromatographic data, gas chromatographic data, high performance liquid chromatographic data, ultraviolet absorption maxima, infrared peaks, mass spectral data of drugs, and mass spectral data of pesticides. A list of reagents and proprietary test materials mentioned in the analytical procedures in Parts One and Two is also provided, as is a list of pharmacological terms. Subject index The subject index covering both volumes is repeated at the end of Volume 2.
Preparation of this edition We are grateful to the editorial and production staff at Pharmaceutical Press who have helped in this project: Emma Burrows, Helen Carter, Tamsin Cousins, Amy Cruse, Simon Dunton, Marian Fenton, Rebecca Garner, Austin Gibbons, David Granger, Jo Humm, Jean Macpherson, Julie McGlashan, Louise McIndoe, Ithar Malik, Jason Norman, Karl Parsons, The Prescribers at The School of Pharmacy (London), Lucy White and John Wilson. There were also the freelance staff who wrote and updated the monographs, to whom we owe thanks: Irene Chiwele, Mildred Davies, Laurent Y Galichet, Poppy McLaughlin, and Eva Reichardt. A team of copyeditors, proofreaders and indexers also contributed to the production of this publication. Without the enthusiasm and dedication of these people this work would not have been published. The Editorial Board members have also assisted in many ways: they authored, refereed manuscripts and monographs, and provided analytical data from their own laboratories. They and the authors have done a great job in providing up-to-date information in an easily accessible and readable manner. A C Moffat M D Osselton B Widdop J Watts January 2011
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 10:44:1
General Notices
Health and Safety This work is intended to be used by appropriately qualified and experienced scientists. Processes and tests described should be performed in suitable premises by personnel with adequate training and equipment. Care should be taken to ensure the safe handling of all chemical or biological materials, and particular attention should be given to the possible occurrence of allergy, infection, fire, explosion or poisoning (including inhalation of toxic vapours). Cautionary notes have been included in a number of monograph entries, but the possibility of danger should always be kept in mind when handling biological samples, and medicinal or other chemical substances.
Classification At the head of each monograph, an indication is given of the classification of the compound according to its therapeutic or commercial use, its pharmacological action and/or its chemical group. The substance may, of course, have other uses or actions in addition to that stated.
Nomenclature
lauril laurilsulfate meglumine mesilate metilsulfate mofetil napadisilate napsilate octil pivalate steaglate tebutate teoclate tosilate xinafoate
n-dodecyl n-dodecylsulfate N-methylglucamine methanesulfonate methylsulfate 2-morpholinoethyl 1,5-naphthalenedisulfonate 2-naphthalenesulfonate octyl trimethylacetate steroyl-glycolate tert-butylacetate 8-chlorotheophyllinate p-toluenesulfonate 1-hydroxy-2-naphthoate
IUPAC Names The nomenclature generally follows the definitive rules issued by IUPAC, 1993.
Monograph Titles
Proprietary Names and Synonyms
The main titles of the monographs are the Recommended International Non-Proprietary Names (rINNs), this includes both drugs and pesticides. For drugs of abuse, the most common chemical names or abbreviations have been used. It is worth noting that for rINNs and chemical nomenclature, it is now general policy to use ‘f’ for ‘ph’ (e.g. in sulpha), ‘t’ for ‘th’ and ‘i’ for ‘y’. For this reason, entries in alphabetical lists and indexes should be sought in alternative spellings if the expected spellings are not found. The main title of a monograph is generally that of the free acid or base as this is the form in which the compound will usually be isolated in an analysis; details of the commonly available salts are included in subsidiary paragraphs within the monograph. The following abbreviated names for radicals and groups are used in the titles.
A selection of proprietary names have been included in the monographs. These can generally be applied to the UK, USA, Japan and a selection of African, Asian and European countries. Comprehensive lists of proprietary names worldwide, can be found in Martindale: The Complete Drug Reference, 37th edn, London, Pharmaceutical Press, 2011. Only singlesubstance preparations have been included except in the case of certain major classes of drugs for which the names of some compound preparations have been added. Some proprietary names that are not in current use have been retained. Names under the heading ‘Synonyms’ include alternative names, common titles, abbreviations and drug trial numbers.
Recommended name acetonide aceturate amsonate besilate camsilate caproate cipionate closilate edetate edisilate eglumine embonate enantate erbumine esilate gluceptate hibenzate isetionate
Chemical name (isopropylidenedioxy) N-acetylglycinate 4,40 -diaminostilbene-2,20 -disulfonate benzenesulfonate camphorsulfonate hexanoate cyclopentanepropionate p-chlorbenzenesulfonate ethylenediaminetetraacetate 1,2-ethanedisulfonate N-ethylglucamine 4,40 -methylenebis (3-hydroxy-2-naphthoate) (=pamoate) heptanoate tert-butylamine ethane sulfonate glucoheptonate o-(4-hydroxybenzoyl)benzoate 2-hydroxyethanesulfonate
CAS Registry Numbers Chemical Abstract Service (CAS) registry numbers are provided, where available, in the monographs to assist readers to refer to other information databases.
Molecular Weights Molecular weights have been calculated using the table of Atomic Weights as revised in 2001 by the Commission on Atomic Weights, IUPAC General Assembly, and based on the 12C scale. Molecular weights have been corrected to one decimal place and are listed in ascending order in the index of Molecular Weights.
Physical Characteristics Dissociation Constants Numerous methods can be used for the determination of dissociation constants, and there are often differences in the various values reported in the scientific literature. The pKa values given in the monographs have been taken from published data and should be regarded only as approximate. The temperature at which the determination was made is given where known. xv
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 10:44:1
xvi
General Notices
Information on the theory, measurement and evaluation of dissociation constants is given in The Pharmaceutical Codex, 12th edn, London, Pharmaceutical Press, 1994.
of the phase volume ratios needed for a successful extraction from 1chlorobutane. Colour Tests
Melting Points The melting points recorded in the individual monographs are listed in ascending order in the index of Melting Points. Partition Coefficients
Where colour tests are given in the monographs, these names refer to the tests described in the Colour Tests chapter, where complete tables of colours are provided. Reference should be made to this chapter for an explanation of the system used for describing the colours. The reagents used for the colour tests are also listed within the list of reagents and additional colour reaction data for approximately 250 compounds is also presented. Colour tests applicable to biological fluids are described under the Hospital Toxicology chapter.
Values for log P are given in a number of monographs. Where the pH of the aqueous phase is stated, the values given are apparent coefficients at that pH (not ion-corrected). Where no pH is stated for the aqueous phase, it can be assumed that log P is for the neutral form of the substance even though it is potentially ionisable. The values given are approximates only but they serve to indicate the characteristics of the substance when it is submitted to an extraction process. For a comprehensive collection of partition coefficients for drugs see C. Hansch et al., Exploring QSAR: Hydrophobic, Electronic and Steric Constants, Washington, American Chemical Society, 1995. Information on the theory of partition coefficients can also be found in J. Sangster, Octanol-Water Partition Coefficients: Fundamentals and Physical Chemistry, New York, John Wiley, 1997.
The thin-layer chromatographic systems referred to in the monographs are described in the TLC chapter on together with lists of data for drugs in important chemical and pharmacological classifications. General screening systems (systems TA to TF and systems TL, TAD, TAE, TAF, TAJ, TAK and TAL), which include over 1500 drugs and metabolites, are provided (see Chapter 39 for system details and references). In order to clarify the presentation of values, the data are expressed in terms of Rf100 (hRf ). Complete lists of data, in ascending order, are given in the index of Thin-layer Chromatographic Data.
Solubility
Gas Chromatography
The solubilities given in the monographs, unless otherwise stated, apply at ordinary room temperature. They have been obtained from various sources and should not be regarded as precise because of variations depending on the method and condition of determination. In general, approximate values are given when a substance is soluble in less than1000 parts of solvent. Where no figure is given, the usual solubility terms have been adopted:
The gas chromatographic systems referred to in the monographs are described in the GC chapter, together with lists of retention data for drugs in important chemical and pharmacological classifications. A general screening system (system GA), which includes over 1500 drugs and metabolites, is provided. An alternative screening system (system GB) is also included (see Chapter 40 for system details and references). For most of the systems, the data are given in terms of Retention Index. Retention times or relative retention times are used in a few systems. Complete lists of retention data, in ascending order, are given in the indexes of Gas Chromatographic Data.
Very soluble Freely soluble Soluble Sparingly soluble Slightly soluble Very slightly soluble Practically insoluble or insoluble
1 part in less than 1 1 part in 1–10 1 part in 10–30 1 part in 30–100 1 part in 100–1000 1 part in 1000–10000 1 part in more than 10000
Thin-layer Chromatography
High Performance Liquid Chromatography
Temperatures are expressed throughout the text in degrees Celsius (centigrade).
The HPLC systems referred to in the monographs are described in the HPLC chapter, together with lists of retention data for drugs in important chemical and pharmacological classifications. Six general screening systems (systems HA, HX, HY, HZ, HAA and HBK) covering between400 and 1600 drugs are provided (please note that values for system HBK have not been included within monographs and can only be found in the index) (see Chapter 41 for system details and references). The data are given in terms of Retention Index, retention time, relative retention time and column capacity ratio k (see Chapter 41). Complete lists of retention data, in ascending order, are given in the indexes of High Performance Liquid Chromatographic Data.
Analytical Data
Ultraviolet Absorption
All analytical data in the monographs apply to the form of the substance described in the main title of the monograph, unless otherwise specified. In all lists or indexes of chromatographic data, a dash indicates that the value is not known, not that the substance does not elute.
The wavelengths of principal and subsidiary peaks are recorded in each monograph for acid, alkaline and neutral solution, where available. These are generally listed from 230 nm. Values in neutral solution are given for compounds for which values in acid or alkaline solution are not available or when the values in neutral solution differ significantly from those in acid or alkaline solution. In many monographs, the ultraviolet spectrum is reproduced. In these spectra, the following notation is used:
In the solubility statements, the word ‘water’ refers to purified water, the word ‘ether’ refers to diethyl ether and the word ‘ethanol’, without qualification, refers to ethanol (95%). Temperature
Extraction It has not been possible to give direct information on the best method for extracting individual substances from various biological samples. However, useful information can be gained from the data on solubility, dissociation constant and partition coefficient. The best solvent can be chosen by reference to solubility, the pH for extraction is indicated by the pKa value, and the partition coefficient gives a quantitative measure
————————— ...................................... -----------------
acid solution alkaline solution neutral solution
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 10:44:1
General Notices Where more than one curve is shown, they do not necessarily relate to the same concentration and, consequently, points where the curves cross cannot be taken as true isosbestic points. The wavelengths of peaks in a few of the spectra may differ very slightly from those stated in the text. Where there is doubt, the values given in the text should be used. In monographs where the spectrum is reproduced, the A11 value for each peak is stated, if available. The A values apply to the form of the substance described in the main title of the monograph, unless otherwise stated. The A11 values are divided into 3 categories in order to provide an indication of reliability: n
n n
The letter ‘a’ after a figure indicates that the value is a mean value based on several reported figures, all of which lie within a range of 10% of the mean. The letter ‘b’ after a figure indicates that the value is a single reported value of unknown reliability. The letter ‘c’ after a figure indicates that the value is a mean value based on several reported figures, some of which lie outside 10% of the mean.
The phrase ‘no significant absorption’ indicates that no peaks are found at the concentrations normally used. The A11 values quoted in the monographs may be useful in identification, and may help in determining the strength of a solution which is required to obtain a curve within the instrumental range of absorption. They may also be useful to give an approximate indication of the amount of drug in a solution. However, because of instrumental differences and the possible effect of solvent and pH, A11 values are subject to considerable variation and the values quoted should not be used when an accurate assay is required. In this case, a reference specimen should be examined at the same time as the sample. The wavelengths of main peaks are listed for acid, alkaline and neutral solution from 230 nm in the index of Ultraviolet Absorption Data.
xvii
Unless otherwise stated, solutions of solids in liquids are expressed as percentage w/v, and solutions of liquids in liquids as percentage v/v. When acids of various strengths are specified, e.g. 50% sulfuric acid, this implies the appropriate dilution by volume of the strong acid in water.
Disposition in the Body Many of the monographs contain a section with the heading ‘Disposition in the Body’. The information in these statements has been obtained from a detailed survey of published papers and other reference sources. Certain monographs have a single reference at the end of the statement, and this indicates that all the disposition information has been obtained from that source. Wherever possible, information is included on absorption, distribution, metabolism, excretion, therapeutic concentration, toxicity and pharmacokinetic parameters. Entry to the literature is provided by the inclusion of abstracts of published papers on clinical studies or case histories. These abstracts include details of drug concentrations in plasma or other body fluids or tissues; in these data a dash means that the particular value was not determined, and ND or 0 means that the substance was not detected. Concentrations in body fluids or tissues are expressed in mg/L or mg/g. In some monographs, the information is incomplete, the amount of detail being dependent upon that available in the literature searched. It should not be assumed that the statements presented reflect the only significant factors in the disposition of the drug concerned. Therapeutic Concentration This is the concentration range usually observed after therapeutic doses, as reported in clinical studies and other research projects. It should not be interpreted as the concentration range required for optimum therapeutic effects.
Infrared Absorption The wavenumbers of the 6 major peaks in the range 2000–650 cm1 (5– 15 mm), in descending order or amplitude, are recorded in the monographs. In many cases, the infrared spectrum is also reproduced. When selecting the 6 principal peaks, those which are in the region where Nujol absorbs (1490–1320 cm1, 6.7–7.6 mm) have been omitted. Corrections for calibration errors have been applied where these are known. The 6 principal peaks, in ascending order of the main peak, are listed in the index of Infrared Peaks. Mass Spectrum The m/z values of the 8 most abundant ions, in descending order of intensity, are included in many monographs. Where dashes occur in the listing, this indicates that less than eight ions have been observed. The 8 principal ions, in ascending order of the main peak, are listed in the index of Mass Spectral Data of Drugs. A separate index for pesticides can also be found. The full mass spectra for the majority of the listed compounds are displayed within the monographs.
Toxicity This statement may include drug concentrations in blood or other body fluids or tissues, which have been reported to be associated with toxic or lethal effects. Because of inter-subject variations or other variable factors, the reported toxic or lethal concentrations may occasionally lie close to or within the therapeutic range. In some monographs, the toxic or lethal blood concentrations are stated in the form 60–89–150 mg/L. These figures have been obtained from a survey of a number of reported cases and represent the maximum concentrations found in 10, 50 and 90% of the subjects, respectively. Maximum permitted concentrations in air (8–h exposure limit) are those recommended by the Health and Safety Executive in Occupational Exposure Limits 2002, Guidance Note EH40/2002 Supplement, London, HMSO, 2003. Volume of Distribution This relates to plasma concentrations after IV administration, unless otherwise stated. Values are based in a body-weight of 70 kg.
Quantification The methods referred to in the references quoted under the heading ‘Quantification’ in the monographs are not intended to be recommended methods. These references are intended to be used as a guide to the literature on the particular subject. Reagents Reagents required for specific tests or methods are generally described fully in the appropriate place in the text. However, certain common reagents that are used throughout the book are described in the list of Reagents and Proprietary Test Materials. Reagent solutions are made in purified water unless otherwise specified. When ethanol, without qualification, is stated to be used, this refers to ethanol (95%).
Clearance This usually refers to the total plasma clearance (or total whole blood clearance) after IV administration. In some instances, the total clearance after an oral dose has been included if the drug is known to be well absorbed and is not subject to significant first-pass metabolism. Numerous factors and inter-subject variations may affect the absorption, distribution, metabolism and excretion of drugs. These include age, sex and disease states such as renal impairment. In addition, results of analyses may be subject to unavoidable analytical inaccuracies. Consequently, there may be considerable variations in the observed drug concentrations and in values for pharmacokinetic parameters in individual cases. Hence, the values given in the monographs should be used only as a guide and should not be taken as absolute values.
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 10:44:1
xviii
General Notices
Dose The dose recorded under this heading in the monographs indicates the usual daily dose (oral unless otherwise stated) that may be administered for therapeutic purposes. It is intended solely as a guide in deciding whether the amount taken by an individual falls within the normal dosage range and should not be taken as a recommendation for treatment. More detailed information on doses in different conditions and age groups may be found in Martindale: The Complete Drug Reference, 37th edn, London, Pharmaceutical Press, 2011; the British National Formulary, latest edition; or in the manufacturers’ data sheets for the products.
Comments This edition of Clarke could not have been completed without the comments on the second and the first editions, and the contribution of analytical data from many scientists involved in the analysis of drugs. In order to assist in the preparation of the next edition, the reader is invited to send any constructive comments and relevant new data concerning the analysis of drugs in biological materials to the Editor, Clarke’s Analysis of Drugs and Poisons, Royal Pharmaceutical Society of Great Britain, 1 Lambeth High Street, London SE1 7JN, UK. In this way, future editions will be improved to the benefit of all of those who use it.
Deletions The following substances which were included in Volumes 1 and 2* of the 1st and 2nd editions are now included in this edition: Acetyldihydrocodeine Adrenalone Allantoin Allylprodine Alphameprodine Alphamethadol Aminometradine Aminopentamide Amisometradine Amolanone Amopyroquine *Amotriphene Amprotropine Amydricaine Amylocaine Apoatropine Azacosterol Azamethonium Bromide Benzalkonium Bromide Benzamine Benzathine Penicillin Benzethidine Betameprodine Betaprodine *Brocresine
Butallylonal Butethamine Butoxamine Cetoxime Chlorisondamine Chloride Citronella Oil Clamoxyquin Clonitazene *Cloponone Codeine N-Oxide Cyclamic Acid Cyprenorphine Demecolcine Demeton-O Desomorphine Diampromide Dibutoline Sulphate *Diethylaminoethyl Diphenylpropionate Dimenoxadole *Dimethocaine Dimethylthiambutene *Dimophebumine Dioxaphetyl Butyrate Dioxathion
*Dioxyamidopyrine *Diphenazoline Dithiazanine Iodide Embramine Erythrityl Tetranitrate *Ethylisobutrazine Ethylmethylthiambutene *Ethylpiperidyl Benzilate Etonitazene Etoxeridine Etymide Fenimide *Fenmetramide Furethidine Hydromorphinol Hydroxypethidine *Imidocarb *Iminodimethylphenylthiazolidine *Iopydol *Iopydone Isobutyl Aminobenzoate *Isometamidium Isomethadone Laudexium Methylsulphate Leucinocaine Levomethorphan Levomoramide Levophenacylmorphan Lucanthone Metabutethamine Metabutoxycaine Metazocine *Methadone Intermediate Methaphenilene Methoxypromazine Methylaminoheptane Methyldesorphine Methyldihydromorphine Methylhexaneamine Methyridine Metofoline Metopon *Moramide Intermediate Morpheridine Morphine N-Oxide Mustine Myrophine Naepaine *Naftazone Narcobarbital Nicocodine Nicomorphine *Nifuroxime Noracymethadol Norbutrine *Nordefrin
*Norgestrel Norlevorphanol *Octacaine Octaverine Orthocaine Pamaquin *Panidazole *Paromomycin Pentaquin *Pethidine Intermediate A Phenadoxone Phenamidine Phenampromide *Phenatine Phenisonone Phenomorphan Phenoxypropazine Phenylpropylmethylamine *Phthivazid *Picloxydine Pipamazine Piperoxan Pipethanate Plasmocide *Proadifen Probarbital Proheptazine Properidine Pulegium Oil *Pyrrocaine Quinapyramine Chloride Racemethorphan Racemoramide *Resorantel *Rifamide *Rolicypram Stilbamidine Sulphasomizole Sulphonal *Taurolin Teclothiazide *Terodiline *Tetracosactrin *Tetraethylammonium Bromide *Thozalinone Thurfyl Nicotinate *Tiletamine Tolonium Chloride Tolycaine Triclobisonium Chloridc Tropacocaine Tropine Tymazoline Viomycin Xenysalate
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 21:7:16
Abbreviations
A11 AAFS AAS 4-ABA ABFT ABP ABV AC 2-ACB 2-ACDP ACDP ACE ACFP ACh AChE ACNB ACPO AD ADC ADCB ADH ADHD ADI AED AEME AES AFID AFM AFMAB AFNB AFS agg. AGP AIDS ALDH ALL ALS ALT 6-AM AMPA AMPK AMT amu ANB AO AOAC AORC APB APC APCI APDC APEI
1% Specific absorbance (abbreviation of A1cm ) American Academy of Forensic Sciences Anabolic/androgenic steroids; atomic absorption spectrometry 4-Aminobenzoyl-b-alanine American Board of Forensic Toxicology 2-(2-Amino-5-bromobenzoyl)pyridine Alcohol percentage by volume Acetylated 2-Amino-5-nitrobenzophenone 2-Amino-5-chlorodiphenylamine 2-Amino-20 -chloro-5-nitrobenzophenone; 2Amino-5,20 -dichlorobenzophenone Angiotensin-converting enzyme 2-Amino-5-chloro-20 -fluorobenzophenone Acetylcholine Acetylcholinesterase 2-Amino-20 -chloro-5-nitrobenzophenone Association of Chief Police Officers Alzheimer’s disease Analogue-to-digital converter 2-Amino-5,20 -dichlorobenzophenone Alcohol dehydrogenase Attention deficit hyperactivity disorder Acceptable daily intake Atomic emission detector Anhydroecgonine methylester Atomic emission spectrometry Alkali flame ionisation detection Atomic force microscopy/microscope 5-Amino-20 -fluoro-2methylaminobenzophenone 2-Amino-20 -fluoro-5-nitrobenzophenone Atomic fluorescence spectrometry aggregate (in botanical names), including two or more species which resemble each other closely a1-Acid glycoprotein acquired immunodeficiency syndrome Aldehyde dehydrogenase Acute lymphoblastic leukemia Amyloid lateral sclerosis Alanine transaminase (alanine aminotransferase) 6-Acetylmorphine a-Amino-3-hydroxy-5-methyl-4isoxazolepropionic acid AMP-activated protein kinase a-Methyltryptamine Atomic mass units 2-Amino-5-nitrobenzophenone Aldehyde oxidase Association of Analytical Chemists Association of Official Racing Chemists 3-Amino-1-phenylbutane 7-Ethyl-10-[4[N-(5aminopentanoic acid)-1piperidino]-carbonyloxycamphothecin Atmospheric Pressure Chemical Ionisation Ammonium pyrrolidine dithiocarbamate Atmospheric pressure electrospray ionisation
API APL APT AR Art 5-ASA ASL ASP AsPEX AST ASTM ASV ATD ATR AUC AUFS AV BAC BBA BBR BC BCRP 1,4-BD BDB BDMPEA BE BEN BGE bid BMAA BMC BMI BNCT BOAA BP Bp B.P. BPH BrAC BRP BSA BSH BSTFA BuChE BUN BZP CA CAM CAP CAS 2-CB CBD CBN CBQCA
Atmospheric pressure ionisation; active pharmaceutical ingredients Acute promyelocytic leukaemia Attached proton test Analytical reagent artefact 5-Aminosalicyclic acid Average signal level Amnestic Shellfish Poisoning Allele-specific primer extension Aspartate transminase (aspartate aminotransferase) American Society for Testing and Materials Anodic stripping voltametry Automated thermal desorption Attenuated total reflectance Area under the curve Absorbance units full scale Atrioventricular Blood alcohol concentration Butyl boronic acid Blood-to-breath ratio Background correction Breast cancer resistance protein 1,4-Butanediol 3,4-Benzodioxazol butanamine 4-Bromo-2,5-dimethoxyphenethylamine Benzoylecgonine Balkan endemic neuropathy Background electrolyte Twice daily b-N-Methylamino-L-alanine 4-Bromomethyl-7-methoxycoumarin Body mass index Boron neutron capture therapy b-I-Oxalylamino-L-alanine Blood pressure; Bristish Pharmacopoeia; butyrylated; benzophenone Boiling point British Pharmacopoeia Benign prostatic hyperplasia Breath alcohol concentration Biological reference preparation Bovine serum albumin; body surface area Mercaptoundecahydrododecaborate Bis(trimethylsilyl)trifluoroacetamide Butyrylcholinesterase Blood urea nitrogen N-Benzylpiperazine Carbonic anyhdrase Base-modified PEG College of American Pathologists Chemical Abstracts Service 4-Bromo-2,5-dimethoxyphenethylamine Cannabidiol Cannabinol 3-(4-Carboxy-benzoyl)-2-quinoline carboxaldehyde xix
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 21:7:16
xx
Abbreviations
CCD CD 2C-D CDT CE CEC 2C-E CEDIA CFP CFTB CG CGE cGMP CHE ChE CHF -CHNO CI 2C-I CIA CID CIEF CIn CIRMS CITP CL Cl CLCR Cmax CMC CNS -CO2 COHb COMT COPD COSY COX CPMACB CRA CRS CSEI CSF CSP CT 2C-T-2 2C-T-7 CTAB CTFEAB CTX CV CVAA CVAO CVVHDF CYP CZE 2,4-D DA DAB DACB DAD DAFB DART DBD DBQ
Charge-coupled device Circular dichroism 2,5-Dimethoxy-4-methyl-b-phenethylamine Carbohydrate-deficient transferrin Capilary electrophoresis Capillary electrophoresis; collision energy 2,5-Dimethoxy-4-ethyl-b-phenethylamine Cloned enzyme donor immunoassay Ciguatera fish poisoning 5-Chloro-20 -fluoro-2-(2,2,2-trifluroethylamino)benzophenone Chorionic gonadotrophin Capillary gel electrophoresis Cyclic GMP; current good (pharmaceutical) manufacturing practice Cholinesterase Cholinesterase Congestive heart failure Descarbamoyl artefact Chemical ionisation 2,5-Dimethoxy-4-iodo-b-phenethylamine Chemiluminescent immunoassay, capillary ion analysis Collision induced dissociation Capillary isoelectric focusing Colour index Combustion isotope ratio MS Capillary isotachophoresis Clearance Clearance Creatinine clearance Mean maximum plasma concentration Critical micelle concentration Central nervous system Artefact formed by decarboxylation Carboxyhaemoglobin Catechol-O-methyltransferase Chronic obstructive pulmonary disease Correlation spectroscopy Cyclooxygenase 2-Cyclopropylmethylamino-5chlorobenzophenone Controlled Substances Act Chemical reference substance Cation selective exhaustive injection Cerebrospinal fluid Chiral stationary phase Computed tomography 2,5-Dimethoxy-4-ethylthio-b-phenethylamine 2,5-Dimethoxy-4-propylthio b-phenethylamine Cetyl trimethyl ammonium bromide 5-Chloro-2-(2,2,2-trifluoro)ethylaminobenzophenone Ciguatoxin Coefficient of variation 2-Chlorovinylarsenous acid 2-Chlorovinyl arsenous oxide Continuous veno-venous haemodiafiltration Cytochrome P450 Capillary zone electrophoresis 2,4-Dichlorophenoxyacetic acid Dialkylated 2,5-Diaminobenzophenone 2,5-Diamino-20 -chlorobenzophenone Diode array detection/detector 2,5-Diamino-20 -fluorobenzophenone Direct analysis in real time 3,4-Benzodioxazol butanamine 2,6-Dibromoquinone-4-chlorimide
DBZ DC DCCA DCMAB DDD DDE DDS DEA DEACFB DECP DEG dEPO DEPT DESI DFA DFSA DHEA DHHS dH2O DHPLC DIPT DLLME DLS DMA p-DMAB DME DMES DMF DMS DMSA DMSO DNOC DNS-Cl DOB DOD DOM DON DOT DPA DPASV DPI DPV DQ DRESS DRIFT DSHEA DSP DTAB DUI DUIA DUID DVT DWI EA EAAS EC ECD ECG ECM ECT ED EDDP EDT
Dibenzosuberamine Direct current (3(2,2-Dichlorovinyl)-2,2dimethylcyclopropane-carboxylic acid 20 ,5-Dichloro-2-(methylamino) benzophenone Dichlorodiphenyldichloroethane Dichlorodiphenyldichloroethylene Drug detection system Drug Enforcement Agency 2-Diethylaminoethylamino-5-chloro-20 fluorobenzophenone Drug Evaluation and Classification Program Diethylene glycol Darbepoietin Distortionless enhancement by polarisation transfer Desorption electrospray ionisation Drug-facilitated assault Drug-facilitated sexual assault Dehydroepiandrosterone Department of Health and Human Services Distilled water Denaturing HPLC Diisopropyltryptamine Dispersive liquid–liquid microextraction Dynamic light scattering 2,5-Dimethoxyamfetamine p-Dimethylaminobenzaldehyde Dimethyl ether Dimethylethylsilyl Dimethylformamide Differential mobility spectrometry Dimercaptosuccinic acid Dimethylsulfoxide Dinitro-o-cresol Dansyl chloride 4-Bromo-2,5-dimethoxyamfetamine (US) Department of Defense 2,5-Dimethoxy-4-methylamfetamine Deoxynivalenol (US) Department of Transport Diphenylamine Differential pulse anodic stripping voltametry Dry powder inhalation/inhaler Differential pulse voltametry Design qualification Drug rash with eosinophilia and systemic symptoms Diffuse reflectance IR Fourier transform spectroscopy Dietary Supplement and Health Education Act Diarrhetic shellfish poisoning Dodecyl trimethyl ammonium bromide Driving under the influence Driving under the influence of alcohol Driving under the influence of drugs Deep vein thrombosis Driving while intoxicated/impaired Enzyme acceptor Electrothermal AAS Electrochemical Electron capture detection Electrocardiogram Enteric coated microcapsules Electrical capacitance tomography Erectile dysfunction; enzyme donor 2-Ethylidene-1,5-dimethyl-3,3diphenylpyrrolidine 1,2-Ethanedithiol
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 21:7:16
Abbreviations EDTA EDXRF EEG EI EIA ELCD ELF ELISA ELS EMC EMCDDA EME EMEA EMIT EMPA ENFSI EOF EPBRP EPI EPO ESA EQA ESI ET ETAAS EtG EtS ETV EU eV EWDTS FAAS FAB FAEE FAIMS FAME FASS FDA FEI FFAP FFT fg FIA FID FISH Fp FPBA FPD FPIA FPLC FPN ft FSH FT FTD FTIR FTIRD GABA GBL GC GC-HRMS GC-MS(-MS) GCS G-CSF
Ethylene diamine tetra-acetate Energy-dispersive XRF Electroencephalogram Electron Impact Enzyme immunoassay Electrolytic conductivity detection Epithelial lining fluid Enzyme-linked immunosorbent assay Evaporative light-scattering Erythromycylamine European Monitoring Centre for Drugs and Drug Addiction Ecgonine methyl ester European Agency for the Evaluation of Medicinal Products Enzyme-multiplied immunoassay technique Ethyl methylphosphonic acid European Network of Forensic Science Institutes Electroosmotic flow European Pharmacopoeia biological reference preparations Enhanced product ion Erythropoietin Electrostatic analyser External quality assurance/assessment Electrospray ionisation Ethylated Electrothermal atomic absorption spectrometry Ethyl glucuronide Ethyl sulfate Electrothermal vaporisation European Union Electron volts European Workplace Drug Testing Society Flame atomic absorption spectrometry Fast atom bombardment Fatty acid ethyl esters Field asymmetric waveform ion mobility spectrometry Fatty acid methyl ester Field-amplification sample stacking Food and Drug Administration Federation Equestre Internationale Acid-modified PEG Fast Fourier transform Femtograms Flow injection analysis; fluorescent immunoassay Flame ionisation detection; free-induction decay (NMR) Fluorescence in-situ hybridisation Freezing point 4-Fluoro-3-phenoxybenzoic acid Flame photometric detector Fluorescence polarisation immunoassay Fast protein liquid chromatography Ferric(III) chloride-perchloric acid-nitric acid Foot (feet) Follicle stimulating hormone Fourier transform Flame thermionic detection Fourier transform infrared Fourier transform infrared detector g-Aminobutyric acid g-Butyrolactone Gas chromatography High resolution mass spectrometry Tandem GC-MS Glasgow Coma Scale Granulocyte colony-stimulating factor
GFAAS GFR GH GHB GI GLC GLP GMND GMP G6PDH GPS GRM GSR GTX h HBV HCC hCG HCL -HCl -HCN HCV HD HDO HDO2 HEACFB HEPES HERG HFB HFBA HGN HHD HIV HLA HMBC HMMC HMQC HMT -H2O HOM HPLC HR HR-MS HS HS-GC HSQC -HY I IA IBS ICADTS ICH ICP ICR ICRAV ID i.d. IDA IDLH IEC
xxi
Graphite furnace atomic absorption spectrometry; electrothermal atomic absorption spectrometry Glomerular filtration rate Growth hormone g-Hydroxybutyric acid Gastrointestinal Gas-liquid chromatography Good laboratory practice Guamanian motor neuron disease Good manufacturing practice Glucose-6-phosphate dehydrogenase Genomic prescribing system Gastric release microcapsules Gunshot residue Gonyautoxins Hour(s) Hepatitis B virus Hepatocellular carcinoma Human chorionic gonadotrophin Hollow cathode lamp Artefact formed by the elimination of hydrochloric acid Artefact formed by the elimination of hydrogen cyanide Hepatitis C virus 3b-Hydrosteroid dehydrogenase Mustard sulfoxide Mustard sulfone 2-Hydroxyethylamino-5-chloro-20 fluorobenzophenone N-(2-Hydroxyethyl)-piperazine-N0 -2ethanesulfonic acid Human ether-a-go-go-related gene Heptaflurobutyrate Heptaflurobutyric anhydride Horizontal gaze nystagmus 2-Chloro-2-hydroxyethyl sulfoxide Human immunodeficiency virus Human leukocyte antigen Heteronuclear multiple bond correlation 4-Hydroxy-3-methoxymethcathinone Heteronuclear multiple quantum coherence Hexamethylenetetramine Artefact formed by dehydration of an alcohol or by rearrangement of an amino oxo compound Humic organic matter High performance liquid chromatography Heart rate High resolution mass spectrometry Headspace Headspace gas chromatography Heteronuclear single quantum coherence Acid-hydrolysed/acid hydrolysis Spin quantum number Immunoassay Irritable bowel syndrome International Council on Alcohol, Drugs and Traffic Safety International Conference on Harmonisation Inductively coupled plasma Ion cyclotron resonance International Conference of Racing Analysts and Veterinarians Isotope dilution Internal diameter Information dependent acquisition Immediately dangerous to life Ion exchange chromatography
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 21:7:16
xxii
Abbreviations
IFHA Ig IGF-1 ILAC IM IMPA IMS INAA INR IOC IP IQC IR IRMA IRMS IS ISE IT IU IUPAC IV J JRES k K-EDTA KIMS LA LAAM LAL l LAMPA LC LC-MS(-MS) LCTF LD50 LFA LH LIF LLE LLOQ ln LOCI LOD log LOQ LPG LSD LTFS M M (COOH-) M (nor-) M (OH-) M (ring) MACB MACDP MALDI 6-MAM MANFB MAO MAOI MAS mAU MBA MBDB 2,3-MBDB
International Federation of Horseracing Authorities Immunoglobulin Insulin-like growth factor-1 International Laboratory Accreditation Co-operation Intramuscular Isopropylmethylphosphonic acid Ion mobility spectrometry Instrumental neutron activation analysis International normalised ratio International Olympic Committee Identification points/intraperitoneal Internal quality control Infrared Immunoradiometric assay Isotope ratio mass spectrometry Internal standard Ion selective electrode Ion trap International unit International Union of Pure and Applied Chemistry Intravenous Indirect spin coupling J resolved experiment Column capacity ratio Potassium ethylenediamine tetraacetic acid Kinetic interaction of microparticles in solution Laser ablation Levomethadyl acetate Limulus amoebocyte lysate test Wavelength Lysergic acid N-(methylpropyl) amide Liquid chromatography Tandem LC-MS Liquid crystal tuneable filter Lethal dose to 50% of a population Lateral flow assay Luteinising hormone Laser or light induced fluorescence Liquid–liquid extraction Lower limit of quantification Logarithm to the base e (natural logarithm) Luminescent oxygen channeling immunoassay Limit of detection Logarithm to the base 10 Limit of quantification Liquified petroleum gas Lysergic acid diethylamide; lysergide Low temperature fluorescence spectroscopy Molar (moles per L) Carboxy metabolite N-Desmethyl metabolite Hydroxy metabolite Ring compound as metabolite 2-Methylamino-5-chlorobenzophenone 2-Methylamino-5-chlorodiphenylamine Matrix assisted laser desorption and ionisation 6-Monoacetyl morphine 2-Methylamino-5-nitro-20 -fluorobenzophenone Monoamine oxidase Monoamine oxidase inhibitor Magic-angle-spinning Milli-absorbance units Methyl boronic acid N-Methyl-1-(1,3-benzodioxol-5-yl)-2butanamine Methyl-2,3-benzodioxazol butanamine
MBTFA MCF MCPA MCPA-CoA MCPP mCPP MDA MDE MDEA MDI MDMA MDP2P MDPPP Me MECC (or MEKC) MECK MEKC (or MECC) MEL mEq mg mm MFD MGF mM MHRA MIBK MID Min MLR MLS MMA MMDA MMDBB 6-MNA MND mol 8-MOP MOPPP MO/TMS Mp MPA MPHP MPPP MQL Mr MR MRI MRL MRM MRO MRPL MRS MS MSC MSTFA MTA MTSS m/z NA NAA NACE NAD NAPA NAPQI
N-Methylbis(trifluoroacetamide) (1R,2S,5R)-(–)-Menthylchloroformate Methylchlorophenoxy acetic acid Methylenecyclopropylacetyl-coenzyme-A 2-(2-Methyl-4-chlorophenoxy)propionic acid 1-(-3-Chlorophenyl)piperazine Methylenedioxyamfetamine Methylenedioxyethamfetamine Methylenedioxyethylamfetamine Metered-dose inhalers 3,4-Methylenedioxymetamfetamine 1-(3,4-Methylenedioxyphenyl)-2-propanone 3,4-Methylenedioxya-pyrrolidinopropiophenone Methyl Micellar electrokinetic capillary chromatography Micellar electrokinetic chromatography Micellar electrokinetic capillary chromatography Maximum exposure limit Milliequivalent(s) Microgram(s) Micrometer(s) Mass fragmentographic detection Mechano growth factor Micrometre(s) Medicines and Healthcare Products Regulatory Agency Methyl isobutyl ketone Multiple ion detector Minute Multiwavelength linear regression Multi-angle light scattering Multi-angle light scattering; 2-methoxymetamfetamine 3,4-Methylenedioxy-5-methoxyamfetamine 2,3-Dimethylbenzodioxazolbutanamine 6-Methoxy-2-naphthyl acetic acid Motor neurone disease, mono-Ndealkyldisopyramide Mole 8-Methoxypsoralen 4-Methoxy-a-pyrrolidinopropiophenone Methoxime/trimethylsilyl Melting point Methylphosphonic acid; N,N-dimethyl-pphenylenediamine hydrochloride 40 Methyl-a-pyrrolidinohexanophenone 4-Methyl-a-pyrrolidinopropiophenone Minimal quantifiable limit Relative molecular mass Metabolic ratio Magnetic resonance imaging Maximum residue limits Multiple reaction monitoring Medical Review Officer Minimum required performance level Magnetic resonance spectroscopy Mass spectrometry Multiplicative scatter correction N-Methyltrimethylsilyltrifluoroacetamide 4-Methylthioamfetamine Merck tox screening system Mass to charge ratio Numerical aperture Neutron activation analysis Non-aqueous capillary electrophoresis Nicotinamide–adenine dinucleotide Acecainide; N-acetylprocainamide N-Acetyl-p-benzoquinoneimine
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 21:7:16
Abbreviations NAT2 NBD-F NBP NCE NCI NC-SPE ND NDPX ng -NH3 NHTSA NIAPCI NICI NIDA NIR NIST NLCP nm NMDA NMR NNRTI NOE NOESY NPC NPD NRC NRG-1 NSAI NSAID NSD NSP OAB OATPT2 OC ODS OECD OES OF OOS OTA P PAD PAGE PBMS PC PCA PCB PCC PCEEA PCEPA PCMEA PCP PCPR PCR PD PDA PDHID PDT PEEK PEG PEL PFB PFK PFP
N-Acetyltransferase 2 4-Fluoro-7-nitro-2,1,3-benzoxadiazole 4-(4-Nitrobenzyl)pyridine New chemical entity Negative chemical ionisation Non-conditioned SPE Nordiazepam Norpropoxyphene Nanogram(s) Artefact formed by elimination of ammonia (US) National Highway Traffic Safety Administration Negative ion atmospheric pressure chemical ionisation Negative ion chemical ionisation National Institute for Drug Abuse Near-infrared imaging National Institute of Standards and Technology National Laboratory Certification Program Nanometer(s) N-Methyl-D-aspartate Nuclear magnetic resonance Non-nucleoside reverse transcriptase inhibitor Nuclear Overhauser enhancement Nuclear Overhauser enhancement spectroscopy Normal phase chromatography Nitrogen phosphorus detection Nuclear Regulatory Commission Naphthylpyrovalerone (naphyrone) Non-steroidal anti-inflammatory Nonsteroidal antiinflammatory drug Nitrogen specific detector Neurotoxic shellfish poisoning Overactive bladder Organic anion transporting polypeptide 2 Oesophageal cancer Octadecylsilane Organization for Economic Development and Cooperation Occupational exposure standard Oral fluid Out-of-specification Ochratoxin-A Apparent partition coefficient Peripheral arterial disease Polyacrylamide gel electrophoresis Particle beam MS Precipitation chromatography; principle component Principal component analysis Polychlorinated biphenyl Pyridinium chlorochromate N-(1-Phenylcyclohexyl)-2-ethoxyethenamine N-(1-Phenylcyclohexyl)-3-ethoxypropanamine N-(1-Phenylcyclohexyl)-2-methoxyethenamine Phencyclidine N-(1-Phenylcyclohexyl)-propanamine Polymerase chain reaction; principal component regression Pulsed discharge Photodiode array Pulsed discharge helium ionisation detector 1,3-Propanedithiol Polyether etherketone Polyethylene glycol Permissible exposure limit Pentafluorobenzoyl Perfluorokerosene Pentafluoropropionate; puffer fish poisoning
-PFP PFPA PFTBA pg pGp PGRN PGx PH PHA PhAsO Ph. Eur. Ph. Int PI PIAPCI PICI PID PIFAB PIS PJ pKa PLA PLOT PLS PLSR PM PMA PMEA PMMA PMN PMPA PN PO p.o. P-III-P PPARd ppb PPC PPD ppm PPP PQ PRP PSI PSP PSX PtE PTFE PTV PVP QA QC qPCR QQQ QTOF RCI r.d. rDNA rf RFLP rhEPO rhGH RI RIA RMTC RNA RPC RRT
xxiii
pentafluoropropionylated Pentafluropropionic anhydride Perfluorotributylamine Picogram(s) p-Glycoprotein Pharmacogenetics Research Network Pharmacogenomics Permethylated hydroxypropyl 4-Hydroxyamfetamine Phenylarsine oxide European Pharmacopoeia International Pharmacopoeia PH of the isoelectric point of a protein Positive ion atmospheric pressure chemical ionisation Positive ion chemical ionisation Photoionisation detection Positive ion fast atom bombardment Product ion spectrum Personalised justice Negative logarithm of the dissociation constant Phospholipase A Porous layer open tubular Partial least-squares Partial least-squares regression Permethylated; personalised medicine 4-Methoxyamfetamine 4-Methoxyethylamfetamine 4-Methoxy-methamfetamine Polymorphonuclear leukocytes Pinacoylmethylphosphonic acid Propionylated per os (oral) Per oral Procollagen type III Peroxisome proliferator activated receptor d Part(s) per billion 4-Phenyl-4 piperidinocyclohexanol p-Phenylenediamine Part(s) per million a-Pyrrolidinopropiophenone Performance qualification Polyribosylribitol phosphate Pre-column separating inlet Paralytic shellfish poisoning Polysiloxane Phosphatidylethanol Polytetrafluoroethylene Temperature-programmed sample inlet, programmable temperature vaporising Poly(vinylpyrrolidone) Quality assurance Quality control Quantitative PCR Triple quadrupoles Quadrupole TOF Racing Commissioners International Relative density Recombinant DNA Radio frequency Restriction fragment length polymorphism Recombinant human erythropoietin Recombinant human growth hormone Retention index Radioimmunoassay Racing Medication and Testing Consortium Ribonucleic acid Reversed-phase chromatography Relative retention time
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 21:7:16
xxiv
Abbreviations
RSD RSS RT s SAMHSA SARM SBW SC SCF SCFC SCOT SDS SDS-PAGE SEC SEP SERM SFE SFST SHGB SI SID SIM SIMCA SIR SMAP SNAP-25 SNARE SNP SNPA SNR SNV SOFT -SO2NH SOP SORS sp. sp.gr. SPE SPME SPR SRM SSI SSNMR SSRI STA STIP STOCSY STP STR STX SVT 2,4,5-T t1/2 T1 T2 TBA TBAF TBAH TBDM TBPE TBSA
Relative standard deviation Root sum square Retention time second(s) Substance Abuse and Mental Health Services Administration Selective androgen receptor modulator Spectral band width Subcutaneous Supercritical fluid Supercritical fluid chromatography Support-coated open tubular Sodium dodecyl sulfate; standard deviation Sodium dodecyl sulfate polyacrylamide gel electrophoresis Standard error of calibration; size exclusion chromatography Standard error of prediction Selective estrogen receptor modulator Supercritical fluid extraction Standardised field sobriety test Sex hormone binding globulin Systeme international d’unites Surface ionisation detection Selected ion monitoring Soft independent modelling of class analogies Selected ion recording 2-Sulfamoylacetylphenol Synaptosome-associated protein of 25,000 daltons Acronym derived from "soluble NSF attachment receptor" Single nucleotide polymorphism N-Succinimidyl-p-nitrophenylacetate Signal-to-noise ratio Standard normal variate Society of Forensic Toxicologists Artefact formed by elimination of the sulfonamide group Standard operating procedure Spatially offset Raman spectroscopy Species (plural spp.) Specific gravity Solid-phase extraction Solid phase microextraction Surface plasmon resonance Selected reaction monitoring; standard reference materials Sonic spray ionisation Solid-state NMR Selective serotonin reuptake inhibitor Systematic toxicological analysis Systematic toxicological identification procedure Statistical TOCSY 2,5-Dimethoxy-4-methylamfetamine; short tandem repeat Short tandem repeat Saxitoxin Supraventricular tachycardia 2,4,5-Trichlorophenoxyacetic acid Half-life Spin-lattice or longitudinal relaxation time Spin–spin or transverse relaxation time Tetrabutyl ammonium hydrogen sulfate Tetrabutyl ammonium fluoride Tetrabutylammonium hydroxide Tert-butyldimethylsilyl Tetrabromophenolphthalein ethyl ester Total body surface area
TBW TCA TCD TCM TCP TCRC TDGO TDGO2 TDI TDM TdP TEA Tf -TFA TFAA TFMPP TFPI TGS THA THC THCA THC-COOH THEED THF TIAFT TIC TID TIS TLC 2,3,5-TMA 3,4,5-TMA TMAH tmax TMCS TMMA TMS TMSI TMSTFA TNF TOC TOCSY TOF TPAH TPI TPMT TRXRF TSD TSP TTX TVAC UAC UDP UGT UHPLC UK ULOQ UN UPLC USA USP UV V VAMP var. VD Vet. VGDS
Total body water Tricyclic antidepressant Thermal conductivity detector Traditional Chinese medicine 3,5,6-Trichloro-2-pyridinol Time-coupled time-resolved chromatography Thiodiglycol sulfoxide Thiodyglycol sulfone Tolerable daily intake Therapeutic drug monitoring torsades des pointes Triethylamine Transferrin Trifluoroacetylated Trifluroacetic anhydride 1-(3-Trifluoromethylphenyl)piperazine Tissue factor pathway inhibitor Triglycine sulfate Tetrahexylammonium hydrogensulfate Tetrahydrocannabinol 11-Carboxytetrahydrocannabinol Tetrahydrocannabinol-11-oic-acid Tetrahydroxyethylene diamine Tetrahydrofuran The International Association of Forensic Toxicologists Total ion current Thermionic detection Turbo ion spray Thin-layer chromatography 2,3,5-Trimethoxyamfetamine 3,4,5-Trimethoxyamfetamine Tetramethylammonium hydroxide Time to maximum plasma concentration Trimethylchlorosilane 2,3,5-Trimethoxymethamfetamine Trimethylsilyl Iodotrimethylsilane Trimethylsilyltrifluoroacetyl Tumour necrosis factor Total organic carbon Total correlation spectroscopy Time of flight Tetrapentylammonium hydroxide Terahertz pulsed imaging Thiopurine methyltransferase Total reflection XRF Thermionic specific detection Trimethylsilyl [2,2,3,3-2H4]-proprionic acid sodium salt Tetrodotoxin Total viable aerobic count Urine alcohol concentration Uridine diphosphate UDP-glucuronosyltransferase Ultra-high pressure LC United Kingdom Upper limit of quantification United Nations Ultra performance liquid chromatography United States of America United States Pharmacopeia Ultraviolet Volt(s) Vesicle associated membrane protein Variety Volume of distribution Veterinary Voluntary genomics data submission
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 21:7:16
Abbreviations Vol VSA v/v WADA WCOT WDXRF WHO
Volume(s) Volatile substance abuse Volume in volume World Anti-Doping Agency Wall-coated open tubular Wavelength dispersive XRF World Health Organization
Wt w/v w/w XRD XRF XRPD
Weight Weight in volume Weight in weight X-ray diffraction X-ray fluorescence X-ray powder diffraction
xxv
Clarke's Analysis of Drugs and Poisons Chapter No. Dated: 18/3/2011 At Time: 21:7:16
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 17/3/2011 At Time: 19:13:48
PART ONE
Chapters Methodology and analytical techniques
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 17/3/2011 At Time: 19:13:48
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:39:58
CHAPTER
1
Hospital Toxicology DRA Uges
Hospital toxicology is concerned with individuals admitted to the hospital with suspected poisoning and its prime aim is to assist in the treatment of the patient. The range of substances that may be encountered is huge and ideally the hospital laboratory will have the capability to identify and, if required, quantify pharmaceutical agents, illicit drugs, gases, solvents, pesticides, toxic metals and a host of other industrial and environmental poisons in biological fluids. In practice, few laboratories can offer such a comprehensive menu and resources are concentrated on those compounds most often involved in poisoning and for which toxicological investigations are particularly useful to the clinical services. In developed countries, hospital clinical chemistry laboratories are geared to provide these basic services and rely on support from central specialised toxicology laboratories for the rarer cases. Fortunately, in the vast majority of cases the diagnosis can be made on circumstantial and clinical evidence; there is no need for urgent analyses and these can be carried out as a routine exercise. However, when the patient’s condition is severe and the diagnosis is not clear, toxicological tests may be crucial and the analytical results must be furnished quickly (usually within 1–2 h of the patient’s arrival) if they are to have any bearing on diagnosis and treatment. Ideally, the toxic substance can be both identified and quantified within this time frame. When this is not possible, a qualitative result still has considerable value if the symptoms are consistent with the identified toxin and should be communicated to the clinician without delay. These time constraints entail an inevitable compromise between speed and analytical accuracy and precision. Consequently, the quantitative methods used may fall short of the standards required, for example, for pharmacokinetic investigations. However, they must be of sufficient quality to allow an appropriate clinical decision to be made (Peters, Maurer 2002). In this area, close liaison between the laboratory personnel and the clinician who manages the patient is essential and can save hours of fruitless effort. An attempt must be made to obtain as much information about the patient as possible. This should include not only the clinical picture, but also any previous medical history of poisoning, details of drugs or other substances to which the patient may have had access and, in cases of accidental poisoning, substances to which the patient may have been exposed. This sort of dialogue between the clinician and an experienced analytical toxicologist can often yield clues as to what the cause of toxicity might be and therefore suggest which tests should be performed as a priority. Close communication must continue if the initial tests prove negative, so that the search can be widened, or if the clinician requires advice on the interpretation of positive results. Laboratories that provide analytical toxicology analyses to assist with cases of acute and chronic poisoning often offer additional services in the area of drug abuse. An increasing number of central laboratories started with providing blood spot services. Dried whole spots on printed paper are sent to these laboratories for analysis, e.g. tacrolimus, anticonvulsants, antibiotics (tuberculosis, cystic fibrosis). This can range from diagnostic tests to uncover the covert misuse of laxatives and diuretics through to routine screening of urine samples from patients assigned to treatment and rehabilitation programmes. For the latter, the requirement is to establish the drug-taking patterns of new patients and to monitor their subsequent compliance with the prescribed treatment regime. Details of techniques suitable for these services are given in separate sections.
Causes of hospital admissions for poisoning Social and economic stresses or mental disorders often result in suicide attempts, particularly through drug overdose, one of the most common causes of emergency hospital admissions. Homicidal poisoning is relatively rare, but surviving victims of this practice are often investigated initially in the hospital environment. Individuals who have been administered substances without their knowledge to facilitate robbery or sexual abuse may also be admitted to hospital. Although in the latter scenario the victims tend to contact the medical services several days after the incident, if teenagers or young adults arrive in hospital semiconscious or disorientated, the administration of so-called date-rape drugs such as alcohol, gamma-hydroxybutyric acid (GHB), flunitrazepam or ketamine must always be considered. Poisoning in children is mainly accidental, but deliberate poisoning by parents, guardians or siblings does occur. Accidental poisoning usually takes place in the domestic environment, with young children and the elderly particularly at risk. Children may gain access to pharmaceutical products, cleaning agents (bleach, disinfectants), pesticides, alcoholic drinks and cosmetics. The confused elderly may misjudge their intake of medications or be poisoned by inappropriate handling of toxic household products. Both are susceptible to acute or chronic poisoning with carbon monoxide emitted by faulty domestic heating appliances. The workplace is another environment in which accidental poisoning occurs and the analytical results from the hospital laboratory can be important not only in medical diagnosis but also in any subsequent legal investigations that involve insurance claims. Iatrogenic intoxications occur through inappropriate medical or paramedical treatment. Neonates require intravenous dosing and the need to work out doses per kilogram of body mass or per square metre (m2) of body area introduces the risk that the total amount and volume of medicine to be administered may be miscalculated. Other causes of iatrogenic poisoning include drug interactions, use of the wrong route of administration and failure to take note of impaired liver or renal function, which reduces the patient’s ability to eliminate the drug. A common example is the accumulation of digoxin in elderly patients with reduced renal function.
Qualitative screening or quantitative analysis? Laboratories adopt different approaches to hospital toxicology. To a large extent, the range of equipment available and the skills and knowledge of the staff govern the policy adopted. Where resources are scarce, only a limited screen for common drugs and poisons may be carried out, with the main effort directed towards quantitative analyses for toxins indicated by circumstantial evidence and the patient’s clinical signs. Specialised toxicology laboratories may pursue a systematic and comprehensive toxicological screen in every case, on the grounds that the clinical and circumstantial indicators are seldom reliable, and then proceed to quantify any substances detected. While the latter approach is more likely to yield useful information, it is expensive and timeconsuming. As stated above, close liaison with the clinicians to obtain a comprehensive case history and a full clinical picture can often help to focus the resources on the qualitative and quantitative tests that are most relevant. The guidelines given in Table 1.1 are useful in this context. 3
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:39:58
4
Hospital Toxicology
Table 1.1 Guidelines to help focus resources on the most relevant qualitative and quantitative tests Indications for qualitative screening
Indications for quantitative analyses
To distinguish between apparent intoxication and poisoning
When the type and duration of treatment depends on the concentration (e.g. antidotes for paracetamol and thallium)
When information about the patient is lacking (no medical history)
When the prognosis is gauged by the plasma concentration (e.g. paraquat)
When the clinical picture is ambiguous (e.g. seizures)
To distinguish between therapeutic and toxic ingestion of drugs
Where the clinical picture may be caused by a pharmacological group of drugs rather than one particular substance (e.g. laxatives, diuretics)
Mixed intoxications (e.g. methanol and ethanol)
Cases of mixed intoxication (drugs of abuse, alcohol)
Toxicological monitoring (e.g. aluminium, Munchausen's syndrome)
Poisoning with no immediately evident clinical picture (e.g. paracetamol)
Toxicokinetic calculations
Where no reliable or selective quantitative method is available (e.g. herbal preparations)
Research (e.g. efficacy of treatment), education, prevention, etc.
Diagnosis of brain death A patient with brain death may be a potential donor of organs. In such cases, the patient should have a deep coma of known origin with no indication of a central infection and normal metabolic parameters. When the primary cause of coma is drug overdose, it is important to ensure that the drug has been eliminated prior to confirming the diagnosis of brain death. This also applies to drugs that may have been given in therapy. For example, thiopental is often given in the treatment of brain oedema and during neurosurgery. The half-lives of thiopental and its metabolite, pentobarbital, increase if cardiac function is diminished or the patient is hypothermic, and therefore plasma concentrations of both compounds must always be measured. Midazolam and diazepam are also administered frequently in treating cases of brain damage, and the continued presence of active concentrations of these drugs and their metabolites should also be excluded using specific and sensitive procedures, such as high performance liquid chromatography (HPLC) and LC-MS. Even if benzodiazepines or their metabolites cannot be detected, there remains the possibility that some may still be present; for instance, active concentrations of hydroxymidazolam glucuronide may be present, since the half-life may be increased considerably with end-stage organ failure. This may suggest a provocation test with the specific benzodiazepine antagonist flumazenil. Similarly, the presence of active levels of anticonvulsants (phenobarbital, carbamazepine, phenytoin and valproate), which are also given in the treatment of brain damage, must be excluded. Again, the use of sensitive and specific chromatographic methods is essential.
For forensic reasons At the special request of the clinician For purposes of statistics, research, education, prevention, etc.
In larger clinical laboratories, the use of various liquid chromatography–mass spectrometry (LC-MS) techniques for therapeutic drug monitoring and toxicological assays has increased considerably and the introduction of liquid chromatography linked to triple quadrupole mass spectrometry (LC-MS(-MS)) has brought about an enormous increase in reliability and sensitivity both in this application (Boermans et al. 2006) and in forensic toxicology (Roman et al. 2008). Although LC-MS(-MS) is not a comprehensive screening method, if sufficient information on the likely cause of poisoning in a drugs overdose case is available, it is possible to obtain both qualitative and quantitative data for a selection of up to 15 drugs and their metabolites within 40 minutes.
Applications Confirmation of diagnosis Most patients who reach hospital in time respond well to measures designed to support the vital processes of respiratory and cardiovascular function and, as mentioned above, toxicological investigations are of only historical value. However, it is still useful to have objective evidence of self-poisoning as this usually instigates psychiatric treatment and follow-up. Differential diagnosis of coma When circumstantial evidence is lacking, a diagnosis of poisoning may be difficult to sustain simply on the basis of clinical examination, since coma induced by drugs is not readily differentiated from that caused by disease processes. Apparent poisonings can be caused by hypoglycaemic coma, a cerebrovascular accident, exhaustion (after seizures), brain damage, meningitis, withdrawal symptoms, idiosyncratic reactions (e.g. to theophylline and caffeine), allergic reactions (shock), viral infections or unexpected symptoms of a disease (e.g. Lyme disease). In these situations, toxicological analyses serve either to confirm poisoning as the cause of coma or to rule it out in favour of an organic disorder that requires alternative medical and pathological investigations.
Influence on active therapy Although supportive therapy remains the cornerstone of the management of acute poisoning, specific antidotes are available for metals (chelation agents), anticholinesterase inhibitors (atropine), methanol and ethylene glycol (ethyl alcohol, formepizole, 4-methylpyrazole), paracetamol/acetaminophen (N-acetylcysteine), digoxin (antibody fragments), calcium blockers (calcium salt), cumarines (phytonadione) and opioids (naloxone). Given a clear diagnosis, a clinician usually administers the antidote without waiting for laboratory confirmation, but subsequent analyses may help to decide whether to continue with the therapy. For example, both parenteral and oral therapy with desferrioxamine in cases of iron poisoning is indicated if patients deteriorate and the serum iron concentration is extremely high. Measurements of cholinesterase activity in serum or red cells are useful in a situation of high-dose infusions of atropine into patients exposed to organophosphate insecticides or thiocarbamates. Measures designed to reduce the absorption of poisons from the gut, such as the use of emetics, purgatives, gastric lavage and irrigation, are now considered to be of limited value and unwarranted in most cases of poisoning. The efficacy of whole-bowel irrigation is also questionable, although some advocate its use to remove sustained-release or enteric-coated preparations of, for example, iron salts and other potentially lethal poisons that have passed into the small bowel, and in the decontamination of body packers. A single oral dose of activated charcoal has largely replaced other means of reducing absorption, although it is generally useful only when given within 1 h of ingestion and fails to absorb inorganic ions, alcohols, strong acids or alkalis, or organic solvents. Techniques to increase the rate of elimination of poisons, such as diuresis, adjusting the urinary pH, haemodialysis and peritoneal dialysis, venous–venous haemofiltration and charcoal haemoperfusion, are now rarely used. Forced diuresis is now frowned upon; it is probably beneficial only in cases of poisoning with thallium and, when coupled with alkalisation of urine, chlorophenoxy herbicides. Alkalisation of urine effectively increases the elimination of salicylates, phenobarbital and chlorophenoxy herbicides. Acidification of the urine has little merit in increasing the elimination of weakly basic substances, such as amfetamines and phencyclidine. New insights provide the indication of highdose Intralipid after a severe overdose of a wide variety of drugs, e.g. lidocaine, antidepressants (see www.lipidrescue.org). Haemofiltration also has a role in this context. ‘Gut dialysis’, or the use of multiple oral doses of activated charcoal, is thought to operate by creating a drug
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:39:58
Quality management concentration gradient across the gut wall that leads to movement of the drug from the blood in the superficial vessels of the gut mucosa into the lumen. So far, its efficacy has been demonstrated for carbamazepine, dapsone, phenobarbital, quinine and theophylline, and there is evidence for its application in poisoning with calcium antagonists. Most of these procedures carry inherent risks to the patient and, as pointed out already, their applications are limited to only a handful of poisons. Toxicological analyses to identify and quantify the poison should be used to ensure that they are used appropriately and at the same time to prevent overtreatment of patients who would recover without such interventions.
Table 1.2 Disturbance of clinical features and indications of possible causes Clinical feature
Disturbances and poisons indicated
General appearance
Restlessness or agitation (amfetamines, cocaine, lysergide (LSD), opiate withdrawal), apathy, drowsiness, coma (hypnotics, organic solvents, lithium)
Neurological disturbances
Electroencephalogram (EEG) (central depressants), motor functions (alcohol, benzodiazepines, GHB), speech (alcohol, drugs of abuse), movement disorders (hallucinogens, amfetamines, butyrophenones, carbamazepine, lithium, cocaine, ethylene glycol), reflexes, seizures (most centrally active substances in overdose or withdrawal), ataxia
Medicolegal aspects of hospital toxicology The primary role of the hospital toxicologist is to assist clinicians in the treatment of poisoned patients, irrespective of any other aspects that surround the case. However, some cases may have a criminal element. These can range from iatrogenic poisoning, in which a patient or relative sues a health authority and its staff for neglect, through to the malicious administration of drugs or poisons by a third party. The latter category includes victims of drug-facilitated sexual assault who have been administered drugs such as flunitrazepam or GHB to induce confusion and amnesia, and non-accidental poisoning in children. Mothers are the most frequent perpetrators of child poisoning and do so to attract sympathy and attention as a consequence of the child’s illness (Munchausen’s syndrome by proxy). When these situations arise, the hospital toxicologist is obliged to take special precautions to conserve all residual samples (human matrices as well as medicines) and documentation that may feature subsequently as part of a forensic investigation (see Chapter 9).
Vital signs Mental status
Psychosis (illicit drugs), disorientation, stupor
Blood pressure
Hypotension (phenothiazines, beta-blockers, nifedipine, nitroprusside and other vasodilators) Hypertension (corticosteroids, cocaine, phenylpropanolamines, anticholinergics)
Heart
Pulse, electrocardiogram (ECG) elevation of QT-time (tricyclic antidepressants, orphenadrine, calcium blockers, class III antiarrhythmics, fluoroquinolones, macrolide antibiotics, antipsychotics, antimycotics, lithium and many drug–drug interactions) Irregularities, torsades de pointes (phenothiazines, procainamide, amiodarone, lidocaine), heart block (calcium blockers, beta-blockers, digoxin, cocaine, tricyclic antidepressants)
Temperature
Clinical manifestations and biomedical tests Specific acute clinical manifestations and vital signs of the patient that can be important in suggesting the cause of poisoning are set out in Table 1.2. Biochemical tests that gauge the physiological status of the patient are more important in terms of the immediate management of the condition and some of the abnormalities found can also be diagnostic of the type of agent involved (see Table 1.6). These, together with the clinical manifestations and history, provide the basis for the order in which the toxicological tests are carried out.
Hyperthermia (LSD, cocaine, methylenedioxymetamfetamine (MDMA), selective serotonin reuptake inhibitors (SSRIs), dinitro-o-cresol (DNOC)) Hypothermia (alcohol, benzodiazepines)
Respiration
Depressed (opiates, barbiturates, benzodiazepines) Hypoventilation (salicylates)
Muscles
Spasm and cramp (strychnine, crimidine, botulism)
Skin
Dry (parasympatholytics, tricyclic antidepressants) Perspiration (parasympathomimetics, cocaine) Gooseflesh (strychnine, LSD, opiate withdrawal)
Other indicative features
Needle marks (parenteral injections: drugs of abuse, insulin),
Some poisons have characteristic odours that may be discerned on the patient’s body or on clothes, or in breath and samples of vomit, as listed in Table 1.3. Colours of the skin and of urine samples can also be useful indicators (Tables 1.4 and 1.5). However, these clues should be interpreted with caution and are not a substitute for proper clinical and toxicological evaluation. The results of biomedical tests are usually available before any toxicological tests have been completed; Table 1.6 highlights their potential diagnostic value.
Colour (red, carboxyhaemoglobin; blue, cyanosis, e.g. with ergotamine; yellow, DNOC)
Assays required on an emergency basis Table 1.7 lists the toxicological assays (mainly in serum, plasma or blood) that should be performed as soon as possible after admission and highlights those that should preferably be provided by all acute hospital laboratories. Emergency requests for the analysis of rarer poisons may be referred to a specialised centre. Such lists vary according to the pattern of poisoning prevalent in different countries or regions, and Table 1.7 is therefore presented only as a guideline. Notes that indicate the relevance of the assays are also included.
Quality management It is essential that the whole laboratory process be controlled strictly and subjected to regular internal and external assessments. All administrative and analytical activities should be described in detailed standard
5
Blisters (paraquat, barbiturates) Eyes
Pinpoint (opiates, cholinesterase inhibitors, quetiapine) Dilated pupils (atropine, amfetamines, cocaine) Reddish (cannabis) Reflex, movements, lacrimation, nystagmus (phenytoin, alcohol)
Nose
Nasal septum complications (cocaine)
Chest
Radiography (bronchoconstriction, metals, aspiration)
Abdomen
Diarrhoea (laxatives, organophosphates) Obstruction (opiates, sympatholytics such as atropine) Radiography (lead, thallium, condoms packed with illicit drugs)
Smell
Sweat, mouth, clothes, vomit (see Table 1.3)
operating procedures (SOPs), which should be reviewed and, if necessary, updated at regular intervals. The laboratory should have in place a system of internal quality controls and also participate in external proficiency-testing schemes. Particular attention should be given to the storage of raw analytical data, results and residual samples, and no unauthorised person should have access to patient information.
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:39:59
6
Hospital Toxicology
Table 1.3 Odours associated with poisoned patients
Table 1.5 Urine colours associated with various poisons
Odour
Potential agents or situation
Colour of urine
Poison or drug
Acetone/nail polish remover
Acetone, propan-2-ol, metabolic acidosis
Red/pink
(Aeroplane) Glue
Toluene, aromatic hydrocarbon sniffing
Ampicillin, aniline, blackberries, desferrioxamine, ibuprofen, lead, mercury, phenytoin, quinine, rifampicin
Alcohol
Ethanol (not with vodka), cleaners
Orange
Warfarin, rifampicin, paprika
Ammonia
Ammonia, uraemia
Brown/rust
Chloroquine, nitrofurantoin
Bitter almonds, silver polish
Cyanide
Bleach, chlorine
Hypochlorite, chlorine
Disinfectant
Creosote, phenol, tar
Formaldehyde
Formaldehyde, methanol
Foul
Bromides, lithium
Hemp, burnt rope
Cannabis
Garlic
Arsenic, dimethyl sulfoxide (DMSO), malathion, parathion, yellow phosphorus, selenium, zinc phosphide
Table 1.6 Biochemical and haematological abnormalities in poisoning Abnormality
Indication
Acid–base disturbances Metabolic acidosis
Ethylene glycol, salicylate, methanol, cyanide, iron, amfetamines, MDMA
Metabolic alkalosis
Chronic use of diuretics or laxatives
Camphor, naphthalene, p-dichlorobenzene
Respiratory acidosis
Opiates
Nicotine, carbon monoxide
Respiratory alkalosis
Salicylates, amfetamines, theophylline
Organic solvents
Diethyl ether, chloroform, dichloromethane
Increased anion gap
Ethylene glycol
Peanuts
Rodenticide
Increased osmolar gap
Alcohols, glycols, valproate
Pears
Chloral hydrate, paraldehyde
Electrolyte disturbances
Plants with special odours
For example Taxus, Convallaria
Hypocalcaemia
Rotten eggs
Disulfiram, hydrogen sulfide, hepatic failure, mercaptans (additive to natural gas), acetylcysteine
Ethylene glycol, oxalates, phosphates, diuretics, laxatives
Hyperkalaemia
Digoxin, potassium salts
Hypokalaemia
Theophyllline, insulin, oral antidiabetic drugs, diuretics, chloroquine
Hypernatraemia
Sodium chloride, sodium bicarbonate
Hyponatraemia
MDMA, diuretics
Mothballs Smoke
Shoe polish
Nitrobenzene
Turpentine
Turpentine, wax, solvent of parathion, polish
Glucose Table 1.4 Typical colours of the skin with poisoning
Hypoglycaemia
Colour of skin
Poison or situation
Blue, cyanosis
Hypoxia, methaemoglobinaemia, sulfhaemoglobin
Liver enzymes
Blue, pigment
Dye (amitriptyline or chloral hydrate tablets), paint
Raised transaminases
Yellow (jaundice)
Liver damage (alcohol, borate, nitrites, scombroid fish, rifampicin, mushrooms, metals, paracetamol, phosphorus, solvents)
Haematological
Insulin, oral antidiabetic drugs, ethanol (children), paracetamol (with liver failure) Paracetamol, amfetamines, MDMA, iron, Amanita phalloides, strychnine
DNOC
Anaemia, raised zinc protoporphyrin, basophilic stippling
Reddish
Carbon monoxide
Carboxyhaemoglobin
Carbon monoxide
Black, necrosis
Sodium or potassium hydroxide, sulfuric acid, burning, intra-arterial injection
Methaemoglobinaemia
Chlorates, nitrites
Raised prothrombin time
Paracetamol, coumarin anticoagulants
Yellow
Where possible, the laboratory should seek accreditation by an external authority (see Chapter 22). Request forms A specially designed request form for toxicological analyses is a useful way not only to obtain essential demographic information on the patients and the analyses required but also to gather details of symptoms, drugs prescribed, biochemical abnormalities and previous medical history. This supplements the oral information provided by the clinician. On completion of the analyses, a copy of the form with the results and interpretation entered can be returned to the clinician. An example of a request form is shown in Fig. 1.1. Collection and choice of samples Blood, serum or plasma
Blood is usually easy to obtain and the analytical results can be related to the patient’s condition and also be used in pharmacokinetic or toxicokinetic calculations. A 10 mL sample of anticoagulated blood (sodium
Lead
edetate) and 10 mL of clotted blood should be collected from adults on admission (proportionately smaller volumes from young children). Most quantitative assays are carried out on the plasma, but anticoagulated whole blood is essential if the poison is associated mainly with the red cells (e.g. carbon monoxide, cyanide, lead, mercury). Serum from coagulated blood can also be used, although the levels are almost always the same as those in plasma. Serum has the advantage that there is no potential interference from any additive. The disadvantage is that clotting takes time and occurs only at room temperature, which creates problems with the analysis of unstable analytes that require samples to be cooled immediately by immersion in ice. It is advisable in addition to collect a 2 mL blood sample into a fluoride/oxalate tube if ethanol ingestion is suspected. However, since most of the fluoride tubes used in hospitals do not contain enough sodium fluoride to completely inhibit microbial production of alcohol (the minimum fluoride concentration required in blood is 1.5% w/v), these samples are not acceptable for forensic purposes. There are conflicting reports of the dangers of contamination of samples collected after the use of disinfectant swabs containing ethanol or 2-propanol and then analysed for ethanol content. Volunteer studies (Malingre et al. 2005) have suggested that this is not a
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:39:59
Quality management
7
Table 1.7 Emergency toxicological assays Assay(s)
Intervention
Comments
Anticholinesterase inhibitors(a)
Atropine (since 2008 the use of pralidoxime or obidoxime is contraindicated)
Measure serum (or preferably red cell) cholinesterase activity
Antiepileptics (carbamazepine, phenytoin)
Multiple-dose activated charcoal
—
Benzodiazepines
Flumazenil antidote only in severe cases
Consider presence of active metabolites; withdrawal seizures
Beta-blockers
Glucagon, isoprenaline
—
Calcium antagonists
Calcium salt infusions, Intralipid
Verapamil: severe prognosis
Carboxyhaemoglobin(a)
Hyperbaric oxygen
No value after administration of oxygen
Chloroquine
High doses of diazepam
Monitor serum K+
Cocaine
Diazepam, haloperidol
—
Digoxin(a)
Potassium salts, Fab antidote
Monitor serum K+, measure serum digoxin prior to giving Fab fragments
Ecstasy group (methylenedioxyamfetamine (MDA), MDMA)
Single-dose activated charcoal, diazepam, dantrolene
Check for metabolic acidosis and hyponatraemia, hyperthermia
Ethanol(a)
Haemodialysis, vitamin B
Monitor blood glucose in children
Iron(a)
Desferrioxamine, IV þ PO
Measure unbound iron; colorimetric assays for serum iron unreliable in presence of desferrioxamine
Isoniazid
Pyridoxine
—
Lithium(a)
Haemodialysis, vitamin B
Measure serum level 6 h after ingestion
Methaemoglobin(a)
Methylene blue
Methaemoglobinaemia caused by nitrites, chlorates, dapsone, aniline
Methanol, ethylene glycol plus other alcohols
Methylpyrazole or ethanol and haemodialysis
Monitor serum ethanol levels to ensure optimum antidote administration
Methotrexate
Folinate, venous–venous haemofiltration
Measure plasma methotrexate level 4–6 h after ingestion
Nifedipine: acidosis
Opiates
Naloxone
—
Osmolality
—
Increased by alcohol, glycols, severe valproate overdose
Paracetamol(a)
N-Acetylcysteine, methionine
Measure serum level at least 4 h after ingestion; prothrombin time and international normalised ratio (INR) are useful prognostic indicators
Paraquat (qualitative urine test)(a)
Activated charcoal
Urine test diagnostic; plasma levels useful in predicting outcome
Salicylate(a)
HCO3 infusion, haemodialysis
Repeat serum salicylate assays may be needed because of continued absorption of the drug
Strychnine
Diazepam
—
Thallium
Prussian (Berlin) blue orally
Treatment continued until urine thallium levels 9 L 13–96
sum 3 L sum 5
P 5–15 0.2 L 0.5–1(d) (for addicts 1–10) Amphotericin B
924.1
S
T 0.025–1
T (3–) 5–10
P 1.5–3.5 Ampicillin
349.4
S
T 0.02–1
Amprenavir
505.6
Pl S
P (2 h) 4-6; (4–6 h) 1.5–4.5
Amrinone
187.2
Pl
1–2 (4)
Amsacrine
393.5
Pl add 1 drop lactic acid then 48 h at 20 add
T 0.03
352.5
S
0.15
0.14 (d)
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:42:8
Interpretation and advice
35
Table 1.28 continued Compound
Atenolol
Relative molecular mass 266.3
Material(a)
S
Reference concentration (mg/L) Therapeutic(b)
Toxic(c)
0.1–0.6 (1)
2 L 27(d)
Atovaquone
366.8
Pl
(10) 15–30 (50)
Atracurium
929.2
S
0.1–1 (5)
Atropine
289.4
S
0.002–0.025
0.03–0 L 0.2
Azapropazone
336.4
Pl
30–90
Azathioprine
277.3
S Pl
P 0.05–0.3
152.2
S
0.04–0.3
Azelastine
6–Mercaptopurine
383
Pl
0.002–0.003 (0.01)
1–2
Azithromycin
749.0
Pl
0.04–1
Aztreonam
435.4
S
T 1–10
Baclofen
213.7
S
0.08–0.6
1.1–3.5
Barbital
184.2
Pl
5–30
20
Tissue >2 mg/g P 50–250 L 6–9.6 L 90 Barbiturates Intermediate acting
S
1–5
10–30
Long acting
S
10–40
40–60
Short acting
S
1–5
7–10
Pl
0.001
L >30 L >80 L >10–15 Barium
137.3
Bendroflumethiazide
421.4
Pl
0.05–0.1
Benoxaprofen
301.7
Pl
Peak 0.5) 0.8–1.3
Child 0.2–0.4
L3
312.8
S
0.1–0.6
0.7
58.9
B
0.0001–0.0022
Cocaine
303.4
S
0.05–0.3
0.25–5
Codeine
299.4
S
T 0.01–0.05
0.3–1
P 0.05–0.250
L 1.6
285.4
S
399.4
S
Desmethylclozapine Cobalt
L 1–20
Morphine Colchicine Colistin
1170
S
0.15 0.0003–0.0024
0.005
P 0.003
L 0.024(d)
1–5 -(10) Cystic fibrosis 10–350 (d)
Copper
63.6
Pl
106 units
0.6–1.5
2 L5 50
Cresol
108.1
Pl
Cromoglicate sodium
512.3
Pl
0.01
26.0
B
0.001–0.006
L 120 Cyanide Cyclizine
0.5
Smoker 0.005–0.012 (–0.15)
L (1) 4–5 0.75–1
266.4
S
0.1–0.25 (0.03–0.3)
252.5
S
0.004–0.025
Cyclobarbital
236.3
S
2–10
10–15
Cyclobenzaprine
275.4
S
0.003–0.036
0.4
Cyclophosphamide
279.1
Pl
10–25
42.1
Pl
80–180
L 15 Norcyclizine
L 20
Cyclopropane Cyproheptadine
287.4
Pl
0.05
Cytarabine (Ara C)
243.2
Pl add 1 drop 1 mol/L tetrahydrouridine for stabilisation
0.05–0.5
Danazol
337.5
Pl
0.2 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:43:17
40
Hospital Toxicology
Table 1.28 continued Compound
Relative molecular mass
Material(a)
Reference concentration (mg/L) Therapeutic(b)
Dantrolene
314.3
S
Toxic(c)
0.4–1.5 T 0.3–1.4 P 1–3
Dapsone
248.3
S
0.5–5
10–20 L 18(d)
Deptropine Desferrioxamine Ferrioxamine Dexamethasone
333.5
S
406.8
S
1082.5
S
0.5–3
Pl
0.05–0.265
392.5
0.015 3–15
Dexfenfluramine
231.2
S
0.03–0.06
0.15–0.25
Dextromethorphan
271.4
S
0.01–0.04
0.1
Dextromoramide
392.5
S
0.075–0.15
0.2
Dextropropoxyphene
339.5
Pl
0.05–0.75
Pl
sum 0.75–3
sum 3
Pl
Anxiolytic 0.125–0.25
1.5
Antiepileptic 0.25–0.5
L5
L3 L 0.9 1 L2 Nordextropropoxyphen Diazepam
284.7
Eclampsia, tetanus, strychnine poisoning 1–1.5 Nordazepam Diazinon
270.7
S
Approximately the same as diazepam 0.2–1.8
304.3
S
0.05–0.1 (0.5)
Diazoxide
230.7
S
10–50
Dibenzepin
295.4
S
T 0.025–0.15
50–100
P 0.1–0.5 Desmethyldibenzepin
S
sum 0.2–0.4
354.5
S
0–0.013
sum 3 L 18
Dichlorodiphenyltrichloroethane Dichloromethane
84.9
S
L 280
2,4–Dichlorophenoxyacetic acid
221.0
S
100
Dichlorvos
221.0
B
Diclofenac
296.2
S
Dicoumarol
336.3
S
8–30 (50)
50–70
Dicycloverine
309.5
Pl
0–0.1
0.2
Didanosine
236.2
Pl
0.5–2.9
Dieldrin
380.9
S
0–0.0015
L 200 L 29(d) T 0.05–0.5
50–60(d)
P 0.1–2.2
L 0.5
Diethylcarbamazine
199.3
Pl
0–0.2
Diethylpropion
205.3
Pl
0.007–0.2
0.15–0.3 2 L 5.4(d)
Difenacoum
444.0
Pl
Diflunisal
250.2
S
0.5 (9) 40–100 (200)
300–500 L 600
Digitoxin
764.9
S
0.01–0.03
0.03
Digoxin
780.9
S
T 0.0005–0.001 (was 0.002)
T (0.0014) 0.0025–0.007
L 0.04–0.1 L (0.0015) 0.01–0.03 depending on potassium level
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:43:35
Interpretation and advice
41
Table 1.28 continued Compound
Relative molecular mass
Material(a)
Reference concentration (mg/L) Therapeutic(b)
Toxic(c) 0.5–1
Dihydrocodeine
301.4
S
0.03–0.25
Dihydroergotamine
583.7
Pl
0.001–0.01
Diltiazem
414.5
S
0.05–0.4
0.8
Dimethadione
129.1
S
500–1000
1000
L2
L 2–6 Dimethyltryptamine
188.3
S
0.001–0.1
Dimetindene
292.4
S
P 0.01–0.05
Dinitro–o–cresol (DNOC)
198.1
S
1–5
30–60
Diphenhydramine
225.4
Pl
0.1–1
1
Diphenoxylate
452.6
S
0.01
Dipipanone
349.5
Pl
0.05
0.2
Dipyridamole
504.6
S
1–2
4
Diquat
184.2
SU
Disopyramide
339.5
Pl
2–7
8
297.5
S
20% active stronger anticholinergic
sum 8–10
296.5
S
0.05–0.4
0.5–5
171.3
S
0.3–1.4
70.1
B
427.6
B
L75–100 L5
T 0.1–1
Nordisopyramide Disulfiram
0.1–0.4
L8 Diethyldithiocarbamate Divinyl oxide Dixyrazine
L 700 0.3
L 5.5(d), 9.4(d)
Domperidone
425.9
S
0.005–0.025 (0.04)
Donepezil
379.5
Pl
0.03–0.075
(d)
Dosulepin (dothiepin)
295.4
S
0.02–0.15 (0.4)
0.8
Desmethyldosulepin
S
0.1–0.2
0.75
Dosulepin S–oxide
S
0.04–0.4
0.65–2.2
Active metabolite 6-Odesmethyldonepezil L (1) 5–19
Doxacurium chloride Doxapram
1106.1 378.5
Pl
0.01–0.3
S
(1.5) 2–5.2
9 (doxapram and keto– doxapram
Doxazosin
451.5
S
0.01–0.05
Doxepin
279.4
Pl
sum 0.05–0.35
265.4
S
sum 0.2–0.35
Doxorubicin
543.5
S
0.006–0.02
Doxycycline
444.5
S
(1-) 5–10
30
Doxylamine
270.4
S
0.05–0.2
1–2
Dronabinol (D9–tetrahydrocannabinol, THC)
314.5
Pl
0.01–0.2
0.1 L 1–18
Nordoxepin
0.5–1 L 2–4
L5
Droperidol
379.4
Pl
0.05
Dyphylline
254.3
S
6.5–14 (–20)
Edrophonium
165.2
S
0.15–0.2
0.15
Efavirenz
315.7
Pl
1.0–4.0
4–6
Emetine
480.6
Pl
-0.1
0.5
Enalapril
376.5 S
0.01–0.05 (0.1)
Desethylenalapril
40
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:43:44
42
Hospital Toxicology
Table 1.28 continued Compound
Relative molecular mass
Material(a)
Reference concentration (mg/L) Therapeutic(b)
Encainide
352.2
S
3-Methoxy-O-desmethylencainide (MODE)
368.5
S
O–Desmethylencainide (ODE)
Toxic(c)
0.06–0.28
338.5
S
0.1–0.3
0.3
Endrin
380.9
S
0–0.003
0.01–0.03
Enoximone
248.3
Pl
0.2
Entacapone
305.3
Pl
0.4–1.0 (–7.0)
Ephedrine
165.2
S
0.02–0.2
1 L 5 (d)
Epirubicin
543.5
S
0.01–0.05
Eprosartan
520.6
S
0.01–0.04
Erythromycin
733.9
S
0.5–6
12–15
T 0.5–1 P 4–12 Esmolol
295.4
Pl
0.15–2
Estazolam
294.8
S
0.055–0.2
Etacrynic acid
303.1
S
0.05–0.1
Ethambutol
204.3
S
0.5–6.5
6–10
46.1
B
0–25
1000–4500 L (2250) 4000–6000
Ethchlorvynol
144.6
S
0.5–8
20 L 50
Ethinamate
167.2
S
5–10
50–100
Ethosuximide
141.2
S
40–100
Ethyl ether
74.1
S
500–1500
Ethylene glycol
62.1
S
276.4
S
Ethanol
L 100 (200 (d)) (100) 150–200 L 250 L (1400) –1900 200–500 L 2000 Etidocaine
0.5–1.5
Etilefrine
181.2
S
P 5–15
Etodolac
287.4
S
20–50
Etomidate
244.3
Pl
0.1–0.5 (–1)
Etoposide
588.6
S
T 2–6
Everolimus
958.2
B
T 0.002–0.006 (0.01)
1.6–2
P 8–14 0.006–0.009
At 2 h 0.003–0.015 Immunoassay 30–35% higher due to metabolites Famotidine
337.4
S
0.02–0.06 (0.2)
0.42 (d)
Felbamate
238.2
Pl
30–60
70–120
Felodipine
384.3
S
0.001–0.008 (0.012)
0.01–0.015
Fenbufen
254.3
S
-60
Fenfluramine
231.3
Pl
0.05–0.15
Fenitrothion
277.3
Fenofibrate
360.8
0.5–0.7 L6 1
Pl
5–30
Fenoldopam
305.8
Pl
0.003–0.06
Fenoprofen
242.3
S
15–65
Fenoterol
303.4
Pl
(0.001) 0.01–0.04
Fentanyl
336.5
S
0.001–0.002
Fexofenadine
501.7
Pl
0.3–0.6
Finasteride
372.6
Pl
0.008–0.01
0.002–0.02
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:43:58
Interpretation and advice
43
Table 1.28 continued Compound
Flecainide
Relative molecular mass 414.4
Material(a)
S
Reference concentration (mg/L) Therapeutic(b)
Toxic(c)
T 0.45–0.9
1.5–3
P 0.75–1.25
L 2.6(d),13(d)
Flucloxacillin
453.9
Pl
3–30
Fluconazole
306.3
S
5–15 (40)
50–75
Flucytosine
129.1
S
T 25–50
100
P 50–100 Flumazenil
303.3
S
0.01–0.05
0.5
P 0.2–0.3 Flunarizine
404.5
S
0.025–0.2
0.3
Flunitrazepam
313.3
S may be reduced by microorganisms
0.005–0.015
0.05
S U not in glass container
After 24 h stop medication (equilibrium) 0.08–0.15
Fluoride
19
T 0.5–2 L3
5–Fluorouracil
130.1
S
0.05–0.3
0.4–0.6 1 neurotoxic
Fluoxetine
309.3
S
0.1–0.45
1.5–2
295.3
S
sum 0.15–0.5 (0.9)
0.4
434.5
S
0.001–0.015
Norfluoxetine
L 0.9–5.0 Flupentixol Fluphenazine
437.5
S
(0.0002–) 0.001–0.017
0.05–0.1
Flupirtine
304.3
Pl
0.5–1.5
3–4
Flurazepam
387.9
S
0.0005–0.03
0.15–0.2
Sedation 0.007
L 0.5–17
0.04–0.15
sum 0.2–0.5
N–Desalkylflurazepam
288.9
S
Flurbiprofen
244.3
5–15
Fluvoxamine
318.3
S
0.05–0.25
Furosemide
330.8
S
2–5 (10)
25–30
Gabapentin
171.2
Pl
2–20
25
0.65
Galantamine
287.4
Pl
0.03–0.14
Gallopamil
484.6
Pl
0.02–0.1
L 8(d)
255.2
S
0.5–5
T 3–5
T 0.2–1
P 20
Gamma-hydroxybutyric acid (GHB) see Hydroxybutyrate Ganciclovir
P 5–12.5 Gemcitabine
263.2
Pl
3–6
Gemfibrozil
250.3
Pl
-25
Gentamicin
449–477
S
P 4–15
T2
T 0.05–2 Glibenclamide
494.0
S
0.03–0.35
0.6
Glipizide
445.5
Pl
0.1–1.0 (2.5)
2
Glutethimide
217.3
S
2–12
12–20
Gold
197.0
S
3–8
10–15
L 30 Granisetron
312.4
Pl
0.009–0.017
Griseofulvin
352.8
Pl
0.3–1.3 (2.5)
Guaifenesin
198.2
B
0.3–1.4
Guanethidine
198.3
S
0.01
Haloperidol
375.9
S
0.005–0.015 (0.04)
0.05–0.1
Halothane
197.4
B
22–84
L 200
Heptabarb(ital)
250.3
Pl
1–4
8–15
L 0.5
L 20 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:44:6
44
Hospital Toxicology
Table 1.28 continued Compound
Relative molecular mass
Material(a)
Reference concentration (mg/L) Therapeutic(b)
Toxic(c)
0.2–1(–1.5)
Heptaminol
145.2
Pl
Heptobarbital
218.2
S
50–100
125–150
Hexachlorophene
406.9
S
0.003–0.65 (1)
L 35
0.01
n–Hexane
86.2
Pl
Hexapropymate
181.2
S
2–5
10–20
Hexobarbital
236.3
S
1–5
8 (10–20)
Hydralazine
160.2
S
(0.05) 0.2–0.9
L 50 Hydrochlorothiazide
297.7
S
0.07–0.45
Hydrocodone
299.4
S
0.002–0.024 (0.05)
Hydrogen sulfide
34.1
S
Hydromorphone
285.3
S
0.008–0.032
L >0.1
4–Hydroxybutyrate (GHB)
104.1
Pl
50–120
80 (abuse)
0.1 L 0.1 (0.2) L 0.92
L 250–280 (abuse) Hydroxychloroquine Hydroxyzine
335.9 374.9
S S
T 0.1–0.4
0.5–0.8
P 0.5–2.0
L4
P 0.05–0.09
0.1 L 39(d)
Ibuprofen
206.3
S
15–30 (5–50)
Idebenone
338.4
S
0.05–0.2
Imipenem
317.4
S
T 0.5–5
100
P 0.65–0.85 P 20–75 Imipramine Desipramine
280.4
S
0.045–0.15
266.4
S
0.075–0.25 sum 0.15–0.3
sum 0.5 L sum (0.8–) 4.5–13
Indinavir
613.8
Pl S
P (1–3 h) 7–12; (4–7 h) 3–7
10
T (10–12 h) Pl neg 0.1–0.5; resistant >0.75 Indometacin
357.8
S
0.5–3
Indoramin
347.5
Pl
0.025–0.1
Iproniazid
179.2
Pl
5
Iron
35.8
S P non-haemolytic
0.5–2
Iron
35.8
B
380–625
137.1
S
T 0.2–1
4–6
6 child 2–8 L 17
Isoniazid
P 3–10 Isopropanol
60.1
B
Acetone
58.1
B
20 L (30–) 100 200–400 L 1000
5–20
200–400 L 550
Isosorbide dinitrate
236.1
S
0.003–0.018
isosorbide mononitrate
191.1
S
0.2–0.5
Isotretinoin
300.4
Pl
T 0.4–1.8; oral 0.1–0.5
Isoxicam
335.3
S
5–25
Isradipine
371.4
Pl
0.0005–0.002 (–0.01)
Itraconazole
705.6
S
T >0.25
Hydroxyitraconazole Kanamycin
484.5
S
sum 1–4
sum 6
S
T 1–4
T 5–10
P 15–25
P 25–30
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:44:19
Interpretation and advice
45
Table 1.28 continued Compound
Relative molecular mass
Material(a)
Reference concentration (mg/L) Therapeutic(b)
Toxic(c) 7 (abuse)
Ketamine
237.7
S
0.5–6.5
Ketanserin
395.4
S
0.015–0.2
Ketazolam
368.8
S
0.001–0.02
P 0.08–1 Nordazepam
270.7
S
0.2–0.6
Ketobemidone
247.3
Pl
0.025–0.030
1–2
Ketoconazole
531.4
S
T 0.3–0.5
Ketoprofen
254.3
S
1–5
Ketorolac
255.3
S
0.22–0.35
5 (plasma)
Ketotifen
309.4
Pl
0.001–0.004
0.02
P 3–10 (20) P 5–15 (–20)
L 1.2 (d) Labetalol
328.4
S
0.025–0.2
Lacidipine
455.5
Pl
0.003–0.006
Lamotrigine
256.1
S
2–15
0.5–1 15 L 50(d)
Lead
207.2
B Heparinised
30 (4)
L >900 Methapyrilene
L 2–380(d) Methaqualone
250.3
Pl
0.4–5.0
>2 L >8
Methazolamide
236.3
S
Methimazole (thiamazole)
114.2
Pl
40 0.5–2 (–3)
Methocarbamol
241.2
S
25–40 (–50)
Methohexital
262.3
Pl
(0.5–) 1–6
Methotrexate
454.4
S
Active > 0.005
T >4.5 (24 h after dose); >0.45 (48 h after dose)
Methoxsalen (8-methoxypsoralene)
216.2
S
0.1–0.2
1
250
T 0.025–0.1 P 0.1–0.4 Methoxyflurane
165.0
B
30–200
2-Methyl-4-chlorophenoxyacetic acid (MCPA)
200.6
Pl
100 (500)
2-Methylchlorophenoxypropionic acid (MCPP)
214.6
Methyldopa
211.2
S
1–5
Methylenedioxyamfetamine (MDA, love drug))
179.2
S
0.4
Methylenedioxyethylamfetamine (MDEA, Eve)
207.3
Pl
0.2
L1
3,4Methylenedioxymethylamfetamine (MDMA, XTC)
193.2
S
0.1–0.35
0.35–0.5
Methylenedioxyamfetamine
L 180 100 (500) L 669 (d) 7–10 9(d) 1 (1.5) L2
179.2
S
Methylfentanyl
350.5
S
L 0.4–0.8
Methylphenidate
233.3
S
Methylthioamphetamine (4–MTA, pMTA)
181.3
Pl
Methyprylon
183.2
Pl
10–20
S
0.01–0.06
L 0.002–0.011 0.005–0.06
(0.5) 0.8 L 2.3 1 2(d),4.2(d) 12–75 (–128) L 50 (–100)
Metiamide Metildigoxin
795.0
Pl
0.0005–0.0008 (–0.003)
Metipranolol as desacetylmetipranolol
267.3
Pl
0.02–0.08g
Metoclopramide
299.8
S
0.04–0.15
0.0025–0.003 L 0.005
0.1–0.2 L 4.4(d)
Metoprolol
267.4
S
0.1–0.6
(0.65(d)) 1
T 0.02–0.34
L (4.7(d)) 12–18
Metronidazole
171.2
S
(3–)10–30
150 (200(d))
Mexiletine
179.3
S
0.5–2
2–4
Mianserin
264.4
S
0.015–0.07 (0.14–)
0.5–5 sum 0.3–0.5 L
L 35(d) 250.3
S
sum 0.04–0.125
Miconazole
Desmethylmianserin
416.1
Pl
1 (2–9)
Midazolam
325.8
S
0.08–0.25 (postoperative awake 0.1–0.04)
1–1.5 (glucuronide) -metabolites also active table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:44:39
48
Hospital Toxicology
Table 1.28 continued Compound
Relative molecular mass
Material(a)
Reference concentration (mg/L) Therapeutic(b)
Toxic(c)
Mifepristone
429.6
Pl
1–2
Milrinone
211.2
S
0.15–0.25
0.3
Minoxidil
209.3
Pl
0.04–0.25 (oral)
1.4(d),3.1(d) L 2.7(d)
Mirtazepine
265.4
S
0.02–0.1 (–0.3)
251.4
S
sum 0.05–0.3
500 (less with alcohol ingestion) 100–125
SB
0–30
157.2
S
1.1–5
275.2
Pl
>0.005
Paraquat
186.3
SU
0.05
Parathion
291.3
Pl
Paraoxon
L 2 (4 h); 0.1 (24 h) Paraoxon
0.01–0.05 0.05–0.08
275.2
Paroxetine
329.4
S
(0.01) 0.07–0.15 (0.25)
0.3
Pefloxacin
333.4
S
T 0.1–6
25
P 5–10 Pemoline
176.2
Pl
1–7
Penbutolol
291.4
S
(0.01–) 0.3–0.7 (1.0)
Penciclovir
253.3
S
T 0.1–0.3
Penfluridol
524.0
S
0.004–0.025
Penicillin (benzyl)
334.4
S
1–10
P 1.75–2 (oral); 10–20 (IV)
( D-)Penicillamine
149.2
Pl
1.7–5.6 (–11)
Pentachlorophenol
266.3
S
0–0.1 (–0.2 dietary)
Pentamidine
340.4
Pl
0.3–0.5
Pentazocine
285.4
S
0.01–0.2 (0.5)
1–2
Pentobarbital
226.3
Pl
1–10 (25–40)
(5–) 8–10
Pentoxifylline (oxpentifylline)
278.3
Pl
0.5–2
Perazine
339.5
S
0.02–0.35
325.5
S
0.04–0.55
30 L >45
L >1 (3) L (8–) 15–25
Desmethylperazine
0.5
Pericyazine (periciazine)
365.5
S
0.005–0.03
Perindopril
368.5
Pl
(0.05–) 0.08–0.15
0.1
Perphenazine
404.0
S
0.0004–0.03
0.05
Pethidine (meperidine)
247.3
S
0.1–0.8
(1–) 2
233.3
S
0.3
0.5
Phenacetin
179.2
S
5–20
50
Phenazone (antipyrine)
188.2
S
5–25
50–100
L >5 Norpethidine
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:44:58
Interpretation and advice
51
Table 1.28 continued Compound
Relative molecular mass
Material(a)
Reference concentration (mg/L) Therapeutic(b)
Toxic(c)
Phencyclidine (PCP)
243.4
S
0.007–0.24
Phendimetrazine
191.3
S
0.02–0.24 (–0.3)
Phenelzine
136.2
S
0.001–0.002 (–0.2)
0.5
Phenformin
205.3
Pl
0.03–0.1 (–0.3)
0.6
Pheniramine
240.3
Pl
0.01–0.27
L 1.9 (d)
Phenmetrazine
177.2
S
0.02–0.25
0.5 L 4
Phenobarbital
232.2
S
2–30 (–40)
30–40
94.1
S
Phenprocoumon
280.3
S
1–3
5
Phensuximide
189.2
S
4–10
80
Phentermine
149.2
S
0.03–0.1
Phenylbutazone
308.4
S
50–100
L (0.3–) 1–5
L >1.5 L3
L 45–120 Phenol
50 L 90
P 10–20 L 7.6(d) 120–200 L 400–500 Phenylephrine
167.2
S
0.03–0.1 (–0.3)
Phenylpropanolamine (norephedrine)
151.2
S
0.05–0.5
2
Phenytoin
252.3
S
8–20; baby 6–14
25; baby 15
L 48 L 70
Free fraction Physostigmine
275.3
S
0.2–2
S
8
Selenium
79.0
Pl
0.045–0.13
0.4
Sertraline
306.2
S
0.05–0.25 (–0.5)
0.29(d);1.6(d)
Sildenafil
474.6
Pl
0.025–0.25 (0.5)
Depending on cardiac function
Silver
107.9
B
0–0.005
T (10–12 h) 0.1–0.4 10 L 30 L (4–) 10–50
with silver sulfadiazine ointment for burns 0.06–0.6 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:45:6
54
Hospital Toxicology
Table 1.28 continued Compound
Sirolimus
Relative molecular mass 914.2
Material(a)
Pl
Reference concentration (mg/L) Therapeutic(b)
Toxic(c)
0.004–0.015
0.020
single therapy 0.012–0.020 Sotalol
272.4
S
0.5–3 (5)
5–10 L 40(d)
Sparteine
234.4
Pl
0.5–1
Spironolactone
416.6
S
0.1–0.5
340.5
S
0.05–0.25 (–0.5)
Stiripentol
Canrenone
234.3
Pl
4–20
20
Streptomycin
581.6
S
T 1–5
T5
P 15–40
P 40–50
Strontium
87.6
S
0.03
Strychnine
334.4
S
0.075–0.1
Sufentanil
386.6
S
L 0.2–2 0.0005–0.005
L 0.001–0.007(d)
P 0.01–0.02 Sulfaguanidine
214.2
S
30–50
Sulfamethoxazole
253.3
S
30–60 (100–200)
Sulfanilamide
172.2
S
100–150
Sulfasalazine
398.4
Pl
5–30 (–70)
Sulfinpyrazone
404.5
Pl
6–17 (–21)
Sulfonamides
SU
T 35–75
200–400
200
P 80–150 Sulindac
356.4
S
0.5–5
340.4
S
sum (including sulfone) 1–5
Sulpiride
341.4
S
0.04–0.6
Sultiam (sulthiame)
290.4
S
Sulindac sulfide
L 3.8 (d)
P 0.15–0.75 0.5–12.5
12–15 L 20–25
Sumatriptan Suramin Tacrine
295.4
Pl
0.018–0.06
1407.2
Pl
150–250
300
198.3
Pl
0.007–0.03
0.02
Tacrolimus
804.0
B
T 0.003–0.01
T 0.003–0.01
Talinolol
363.5
Pl
0.04–0.15
L 5(d)
Talipexole
282.2
Pl
Continuous infusion 0.003–0.01
Tamoxifen
371.5
S
0.05–0.5
Teicoplanin
189.4
S
10–40
200
Temazepam
300.7
S
0.3–0.9
1 L 8.2(d)
Tenoxicam
337.4
Pl
5–10
Terazosin
387.4
Pl
0.02–0.08 (–0.1)
Terbinafine
291.4
S
T 0.01–0.03
277.4
S
P 0.4–0.8
Terbutaline
225.3
S
0.001–0.006 (–0.01)
L 0.04
Terfenadine
471.7
Pl
0.0015–0.0045
0.06
T 0.02–0.15
P 0.5–3 Norterbinafine
L 0.4(d) Tetrachloroethylene
165.8
S
Tetracycline
444.4
S
5–10
L 4–5
Tetrahydrocannabinol
314.5
Pl (unstable)
P 0.05–0.125
Tetrazepam
288.8
Pl
0.05–0.6 (–1)
30
T 1–5 0.001–0.01
0.004–0.0015
Clarke's Analysis of Drugs and Poisons Chapter No. 1 Dated: 15/3/2011 At Time: 17:45:8
Interpretation and advice
55
Table 1.28 continued Compound
Relative molecular mass
Material(a)
Reference concentration (mg/L) Therapeutic(b)
Toxic(c)
Thalidomide
258.2
Pl
0.5–1.5 (–8)
Thallium
204.4
B
20
P 5–15 Thiazinamium Thiocyanate
299.5
S
0.05–0.15
58.0
S
Non-smokers 1–4
0.3 35–50
Smokers 3–12
L 200
As metabolite of nitroprusside 6–30 Thiopental
242.3
S
1–5 (flat EEG: 25–40)
10 (40–50)
Pentobarbital
226.3
S
5–10
10–15
Thioproperazine
466.6
S
0.001–0.02
0.1
Thioridazine
370.6
S
0.2–1
2 (5)
386.6
S
0.3 (0.2–1.6)
L 10–100
L 3–10 Mesoridazine
402.6
S
25 þþ and 20 mg/100 mL) and a first-order kinetic model at lower concentrations. Graph (a) is plotted on Cartesian coordinates and illustrates the hockey-stick shape of the curve; graph (b) shows the corresponding semi-logarithmic plot.
Clarke's Analysis of Drugs and Poisons Chapter No. 4 Dated: 10/3/2011 At Time: 16:47:58
106
Driving Under the Influence of Alcohol
Absorption and elimination kinetics Much less is known about the absorption kinetics of ethanol than about the elimination kinetics. The mathematical modelling of the absorption phase is complicated because uptake from the gut is sometimes so fast that insufficient C–t data points are available for curve fitting. Furthermore, ethanol is a drug absorbed from both the stomach and the upper part of the small intestines. Indeed, the rate of uptake of alcohol after passing through the pyloric sphincter is much faster than from the stomach. The speed of absorption and time of occurrence of Cmax therefore depend to a large extent on factors that influence gastric emptying, such as fed or fasting state before drinking. Unlike many prescription drugs, the dose of ethanol is not swallowed immediately, but is instead ingested in portions often over several hours. In most experiments dealing with the pharmacokinetics of ethanol, a single moderate dose is consumed in 5–15 min as a bolus. In the real world, people more often consume spirits, beer and/or wine repetitively in divided doses, which makes it much more difficult to model the shape of the BAC curve. Unlike most drugs, ethanol is absorbed to some extent through the gastric mucosa and 20% of the dose enters the bloodstream in this way, although empirical support for this is not easy to find in the literature. A mathematical model of the entire BAC curve is possible if it is assumed that absorption occurs according to first-order kinetics and that elimination, which occurs simultaneously, is a zero-order process. Under these assumptions, the entire concentration–time profile of ethanol can be fitted to the equation: Ct ¼ C0 ð1 e kt Þ k0 t where Ct ¼ BAC at some time t after administration; C0 ¼ initial backextrapolated BAC at the time of starting to drink; k ¼ first-order absorption rate constant; k0 ¼ zero-order elimination rate constant; and t ¼ time after drinking. Factors that influence gastric emptying, such as food in the stomach, will delay the rate of absorption resulting in a lower Cmax and delayed tmax. With a rapid absorption, the Cmax might coincide with time of last drink or occur before the first blood sample is taken. With delayed absorption, as might occur under some circumstances, a plateau in the C–t profile is likely, which implies that the rates of absorption and elimination are the same. Other evidence exists that, even with delayed absorption, the BAC reaches about 80% of the final Cmax within 15– 30 min of the end of drinking (Jones, Neri 1991). Because ethanol exhibits saturation-type kinetics, the peak BAC and the area under the curve (AUC) increase more than proportionally with increase in the dose administered, which is particularly evident after very small doses (Wagner et al. 1985). When the rate of delivery of ethanol to the liver is slow, the AUC is smaller for a given dose and vice
versa (Levitt, Levitt 1994). The systemic availability (bioavailability) of ethanol must be considered when blood alcohol calculations are made for legal purposes. If some part of the dose of ethanol fails to reach the systemic circulation, owing to first-pass metabolism or other reason, then the results of calculations of BAC are obviously incorrect. If only the post-absorptive phase of the C–t profile is considered, then the equation widely used in forensic science practice is: Ct ¼ C 0 bt where Ct is the blood alcohol concentration, C0 is the extrapolated concentration of alcohol in blood at the time of starting to drink (yintercept) and b is the zero-order slope of the declining phase, sometimes denoted as k0. Disappearance rate of alcohol from blood The slope of the pseudo-linear declining phase of the blood alcohol curve represents the rate at which ethanol is eliminated from the bloodstream. People who drink excessively over long periods of time develop a higher capacity to metabolise ethanol compared with moderate or occasional drinkers. This phenomenon, known as metabolic tolerance, is reflected in a steeper slope in the C–t plot during the post-absorptive phase and is often seen in alcoholics during detoxification after a period of binge drinking. Elimination rates of alcohol up to 35 mg/100 mL per h have been reported in these individuals. However, after a few days of abstinence, the rate returns to the expected 15 mg/100 mL per h (range 10–20 mg/100 mL per h) seen in moderate drinkers. The faster rate of metabolism is considered to reflect induction of the microsomal enzyme CYP2E1 as discussed earlier, and this occurs after heavy drinking lasting several days or weeks without an alcohol-free period. The quantities of alcohol necessary and the duration of drinking required to cause induction of the microsomal enzymes have not been determined in humans. The information in Table 4.9 gives a likely physiological range of elimination rates of ethanol from blood in humans and the circumstances under which such rates might be encountered (Jones 2010b). Accordingly, for the vast majority of people, the rate of ethanol elimination is likely to vary three-to-four fold or from 10 per 35 mg/100 mL per h. In blood alcohol calculations for research and legal purposes, back-extrapolation with an alcohol burn-off rate of 10–25 mg/100 mL per h is appropriate (mean 15 mg/100 mL per h). In apprehended drunken drivers, because of the many alcoholics in this population, the average elimination rate of alcohol from blood is 19 mg/100 mL per h, being slightly faster in women than in men (Jones, Andersson 1996a). The information about elimination rate of ethanol in impaired drivers has been determined by taking two blood samples about 1 h apart in over 1000 offenders. Under the assumptions of zero-order
Table 4.9 Rates of ethanol elimination from blood that might physiologically be observed under various conditions or circumstances Elimination rate Very slow
Expected values (mg/ 100 mL per h)(a) 8–10
Conditions or circumstances in which such rates might be expected Malnourished individuals or people eating low-protein diets. Medical conditions such as advanced liver cirrhosis or portal hypertension and reduced hepatic blood flow. Administration of a drug (e.g. 4methylpyrazole or fomepizole) that blocks the action of alcohol dehydrogenase
Slow
10–13
Healthy individuals after overnight (10 h) fast and a single bolus dose of ethanol (1.0 g/L) of 75.5% and 75.1% for the walk-and-turn and one-leg-stand, respectively (Burns, Moskowitz 1977).
HGN is not a divided-attention test but rather takes advantage of the CNS-depressant properties of alcohol and other drugs, which produce among other things an observable lateral jerking of the eye, or nystagmus, that can be observed by asking the subject to track a stimulus such as a fingertip or penlight across the field of vision while keeping the head still. While pronounced nystagmus can result from other causes (brain damage, inner-ear disturbances, thermal imbalance in the inner ear, rotation, etc.), it is produced reliably by alcohol at blood concentrations known to impair driving ability. Further, other CNS-depressant drugs can also elicit HGN (e.g. depressants, phencyclidine (PCP) and volatile solvents), which makes the test a useful indicator of use of some other classes of drugs in addition to alcohol. Only limited evaluation of these field sobriety tests has been done for different categories of drug use. In investigations of suspected drugimpaired driving, the value of these and similar tests is to demonstrate an inability on the part of the subject to integrate simple psychomotor and cognitive skills, and as such to demonstrate evidence of impairment. The Drug Evaluation and Classification Program (DECP) that has been established in some jurisdictions in the USA, and modified for use in other countries such as Germany and Australia, seeks to classify subjects as under the influence of a specific class or classes of drugs based on certain psychomotor and physiological measurements. Table 5.3 shows a matrix that relates these effects to the classes of drugs recognised in the DECP. In some European countries, such as the UK and Norway, impairment assessment is done by a physician. On-site chemical tests Of increasing interest to the law-enforcement and traffic safety community is the use of ‘point of contact’ or ‘on-site’ drug testing. The use and acceptability of such tests vary in different countries. Urine was initially used for this purpose in some countries, and lateral-flow immunoassaybased dipstick devices or test cups were commercially produced and have since proliferated. The devices test for common drugs of abuse including cocaine, marijuana and opiates and are generally sensitive and specific enough for this purpose, although there are quality differences between manufactured devices. Four such devices showed sensitivity ranging from 82.9% to 100% for cannabinoids and from 82.5% to 100% for cocaine, and all showed 100% for opiates. Accuracy ranged from 94.0% to 98.3% for marijuana, from 97.4% to 98.0% for cocaine, and from 99.7% to 100% for opiates (Buchan et al. 1998). The benefit of the on-site urine test was the ability to confirm the officer’s suspicions about drug use at the time of the arrest, and the devices had great potential for use in enforcement checkpoints or during random screening in jurisdictions where ‘per se’ laws were in effect. The limitations of the devices included the limited scope of drugs that could be detected (typically five to seven drug classes on any given device, with variable cross-reactivity to compounds within the class) and, on a practical level, the need to transport the subject to a private location to provide the urine sample. Often observed collection was not possible owing to the unavailability of a same-sex observer, creating the potential for substitution or adulteration. While urine is a good specimen for screening, it is essentially ‘out of the body’ and drugs may be detected in urine after the effects on driving ability have worn off; hence this is not a good specimen for interpretative purposes. More recently, on-site oral fluid drug testing has gained in popularity. OF testing offers the same benefits of an immediate result to corroborate the officer’s opinion regarding drug use, but has the advantages over urine of being capable of being conducted at the roadside, eliminating the cost and time of transporting the subject. It also offers the opportunity of obtaining a sample proximate to the time of driving, it provides a non-invasive, clean sample, and collection can be directly observed irrespective of the sex of the suspect or the arresting officer. OF also has an advantage over blood of not requiring a licensed collector or phlebotomist, and the associated cost of that procedure. Following the result of a field test, OF can also be collected and sent to a laboratory for the confirmatory test needed for forensic purposes. OF test devices currently have a limited scope compared with a laboratory test but they can cover the major drug
Clarke's Analysis of Drugs and Poisons Chapter No. 5 Dated: 10/3/2011 At Time: 16:56:54
Introduction
119
Table 5.3 Symptomatology chart for drug effects (IACP, DECP Program) Categories
Depressants
Stimulants
Hallucinogens
PCP
Narcotic analgesic
Inhalants
Cannabis
HGN
Present
None
None
Present
None
Present
None
Vertical nystagmus
Present (high dose)
None
None
Present
None
Present (high dose)
Present
Lack of convergence
Present
None
None
Present
None
Present
Present
Pupil size(a)
Normal
Dilated
Dilated
Normal
Constricted
Normal
Dilated
Reaction to light
Slow
Slow
Normal
Normal
Little or nonvisible
Slow
Normal
Pulse rate(b)
Down
Up
Up
Up
Down
Up
Up
Blood pressure(c)
Down
Up
Up
Up
Down
Up/down or Normal, DOS
Normal
Body temperature
Normal
Up
Up
Up
Down
Muscle tone
Flaccid
Possibly rigid
Possibly rigid
Rigid
Normal or flaccid
Flaccid
Normal
General indicators
Uncoordinated Disoriented Sluggish Thick slurred speech Drowsiness Drunk-like behaviour Fumbling Impaired vision Droopy eyes Body tremors will be evident with methaqualone
Restlessness Body tremors Excited Talkative Exaggerated reflexes Anxiety Grinding of teeth Insomnia Dry mouth Irritability Runny nose Redness to nasal area
Dazed appearance
Perspiring Warm to touch Repetitive speech Cyclic behaviour Speech difficulty Incomplete verbal responses Increased pain threshold Possibly violent
On the nod Dry mouth Facial itching Droopy eyelids Low raspy slow speech Track marks Euphoria Nausea Fresh puncture wounds
Odour of substance Traces on clothes or face Bloodshot watery eyes Confused Disoriented Lack of muscle control Nausea Flushed face Headaches
Odour of cannabis Cannabis debris in mouth Eyelid tremors Reddening of conjunctiva Body tremors Disoriented Relaxed inhibitions Possible paranoia
Body tremors Synaesthesia Paranoia Hallucinations Disoriented Memory loss Flashbacks Piloerection Perspiring Uncoordinated Speech difficulty
(a)
Pupil size normal range, 3.0–6.5 mm. Pulse normal range, 60–90 beats/s. Blood pressure normal range, 120–140 mmHg systolic, 70–90 mmHg diastolic.
(b) (c)
classes encountered in the driving population. OF on-site tests have been deployed in a large-scale roadside random testing programme in Australia, with some success (Boorman, Owens, 2009), and on-site OF testing has recently been trialled by police in the UK. Studies conducted as part of the Driving under the Influence of Drugs and Alcohol (DRUID) project in the European Union have evaluated available on-site devices and found them to be significantly improved over their earlier versions (DRUID 2009). The merits of OF as a sample for confirmatory testing are discussed below. Any on-site drug test needs to take account of the fact that not all drugs that can cause impairment will be detected in an on-site test and that the primary consideration in whether to release a driver would be his or her appearance of sobriety and fitness to drive. Choice of specimen for drug testing in impaired driving cases The three options for sample collection for DUI enforcement purposes are blood, OF and urine. Each has its advantages and disadvantages. Urine has the advantage of being easily collected (privacy issues around observed collection have been discussed above). The concentration of drugs in urine is dependent on the volume of liquid consumed, the degree of hydration of the subject and the time of ingestion, as it is on the amount of drug ingested. Urine drug concentrations are often several orders of magnitude higher than blood drug concentrations, making urine an ideal sample for preliminary screening. Depending on the drug, it or its metabolites may be detected for days or occasionally weeks after administration. Shorter-acting drugs may be detected in the urine even after the effect of the drug has disappeared. This is true, for example, of some of the short-acting benzodiazepines, g-hydroxybutyric acid (GHB) and zaleplon.
Urine drug concentrations can be normalised for creatinine concentration, which corrects for dilution caused by the ingestion of large volumes of water, but such a correction is of limited value in a single urine specimen, and does not add to an interpretation of the degree of intoxication. Urine is, therefore, an excellent specimen for answering the question ‘Did the donor at some time prior to providing the specimen use or ingest this drug?’, but it is of little use in determining whether the subject was impaired or intoxicated at the time that the sample was collected. The investigator must consider the result of the urine drug test in the light of objective evidence of impairment (slurred speech, staggering gait, inappropriate conduct or response, poor coordination, glassy stare, agitation, restlessness, etc.) to reach any conclusion about the subject’s fitness to drive. The toxicologist can then use his or her specialised knowledge to assess whether the observed effects are consistent with the known properties of the drugs present. Another limitation of urine is that, following drug use, it may take some time before a drug is metabolised and excreted. If ingestion had been very recent, the urine may test negative for the presence of the drug, even though the drug is present in the body and exerting an effect on the subject. In spite of these limitations, urine has been a popular specimen for drug testing in impaired driving cases because of the ease with which it can be collected, the essentially unlimited volume for testing, and the ease with which urine can be screened in the laboratory by immunoassay on automated instruments. Blood has emerged as the preferred specimen for collection and analysis in impaired-driving investigations. The presence of drugs in the circulating blood, in equilibrium with the brain, provides the most direct measure of a person’s likelihood of intoxication. From an interpretive standpoint, blood drug concentrations can usually be related to some degree to published concentrations associated with known doses
Clarke's Analysis of Drugs and Poisons Chapter No. 5 Dated: 10/3/2011 At Time: 16:56:57
120
Driving Under the Influence of Drugs
or patterns of therapeutic or recreational use. Blood drug concentrations are a function of the dose, time since last use, and whether the use is acute or chronic. They can be useful in distinguishing drug use from abuse and assessing the likelihood of drug interactions, and can be related to behavioural studies of effects on critical abilities related to driving, although relationships between blood drug concentration and any specific degree of impairment or effect on driving have not been established for many drug substances. Apart from alcohol, the exception to this may be other members of the depressant drug class, which have been studied to a greater degree (Bramness et al. 2003, 2004). Hysteresis or varying early phase and late phase effects for a given concentration as a result of acute tolerance or withdrawal make this relationship more complex for stimulant drugs, and issues of chronic tolerance can impact on the interpretation of opiate concentrations. Consequently, the interpretation of blood drug concentrations still needs to be treated with care and the full range of possible ingestion patterns, and likely effects, must be considered. Blood must be collected by a medical professional, which can add expense and time to the collection process. The time factor is a consideration for drugs such as zolpidem, tetrahydrocannabinol (THC) and some of the synthetic cannabinoids (JWH-018 and JWH-250), which are distributed and metabolised out of the blood rapidly. Consequently the concentrations in the blood at the time of collection may not reflect the concentrations present at the time impairment is first observed or during driving. Typical volumes of blood available for testing are around 10 mL which is usually adequate for a sensitive broad-spectrum screen, confirmation and quantification. It can be difficult to collect an adequate volume from some individuals with collapsed veins. Drugs are typically quite stable in blood provided that it is collected over an enzyme inhibitor such as sodium fluoride. Collection with an anticoagulant such as heparin, citrate or oxalate is important in order to prevent clotting and inhomogeneity in sampling. As noted above there is increased interest in OF testing for drugs in driving investigations. OF (a mixture of salivary, parotid and crevicular ultrafiltrates and mucus) has gained popularity as an alternative to blood because of the ease and low cost of collection proximate to the time of driving, and the increasing reliability of on-site test devices. In the USA, 16 states now allow OF collection in their DUI statutes, and others are following suit. In some European countries, OF is also accepted in court for traffic enforcement cases subject to appropriate laboratory confirmation. Several large-scale surveys of drivers have also recently assessed the utility of OF for traffic investigation, and have found generally good correlation between the presence of drugs in OF and the presence in a subsequently collected blood sample (Diplock, Plecas, 2009). This meets the requirement in ‘per se’ jurisdictions where the mere presence of a proscribed drug in a body fluid constitutes the
offence. OF drug concentrations are, however, more difficult to interpret quantitatively than blood concentrations. Partition of drugs into OF is a function of many factors: salivary flow, oral pH, drug polarity and pKa, contamination of the oral cavity with drug (especially an issue for smoked drugs such as cocaine or marijuana), and the degree of protein binding of the drug in blood (only non-bound drug partitions into the OF). Consequently, it is inadvisable to try to predict blood drug concentrations based on OF concentrations, and there are currently limited data available to determine whether impairment can be predicted based on quantitative OF test results (Wille et al. 2009). Analytically, the target compounds in OF can be different from whole blood, as the less polar parent drug is often the major analyte present in OF as is the case with THC and cocaine. More polar highly bound drugs, such as the benzodiazepines, can be present at much lower concentrations in OF. In summary, blood remains the specimen of choice in DUID investigations, but OF is emerging as a valuable alternative within the correct statutory framework.
Laboratory approaches to drug testing in DUID cases At the heart of every DUID investigation is a forensically defensible drug test. This generally means a test with appropriate scope to identify the range of drugs that are known to affect driving and are prevalent in the impaired driving population at large, supplemented with other drugs that may be an issue locally. Prescription drugs, over-the-counter drugs and drugs of abuse should all be included. Two sets of guidelines with largely overlapping recommendations for scope and limit of detection in blood and urine, for both screening and confirmation, are shown in Table 5.4 (Farrell et al. 2007; Walsh et al. 2008). Most laboratories begin the analytical process with a test for volatiles to establish any role for alcohol in the subject’s impairment. This can also disclose the presence of volatiles including anaesthetic gases, and solvents subject to abuse such as toluene, xylene or difluoroethane. The volatiles screen is usually followed by an immunoassay screen. The immunoassay provides a valuable role in screening for the presence of certain drug classes, for example identifying the presence of amfetamines or benzodiazepines but not the identity of the specific amfetamine or benzodiazepine. The variable cross-reactivity of the assay to members within a class must be kept in mind. For example, many benzodiazepine immunoassay tests do not cross-react well with lorazepam (Ativan), so a chromatographic screen will be needed to exclude those drugs with poor cross-reactivity. Appropriate cut-offs need to be established depending on the matrix being tested (see Table 5.4). It is not appropriate to apply most urine drug screening cut-offs to blood analysis, since urine drug concentrations may exceed those in the blood by several orders of magnitude, and the presence of some drugs might be subsequently missed.
Table 5.4 Recommended scope and analytical cut-offs of toxicological analysis in DUID investigations Blood (ng/mL) Target analyte
Screen
Urine (ng/mL) Confirmation
Screen
Confirmation
DRE category – cannabis THC
–(a)
2
–(a)
2
Carboxy-THC
10
5
20(b)
5
11-OH-THC
–(a)
2
–(a)
2
Metamfetamine
20
20
200
50
Amfetamine
20
20
200
50
MDMA
20
20
200
50
MDA
20
20
200
50
Cocaine
–(a)
10
–(a)
20
Benzoylecgonine
50
50
300
50
Cocaethylene
–(a)
10
–(a)
20
DRE category – CNS stimulants
Clarke's Analysis of Drugs and Poisons Chapter No. 5 Dated: 10/3/2011 At Time: 16:56:57
Laboratory approaches to drug testing in DUID cases
121
Table 5.4 continued Blood (ng/mL) Target analyte
Screen
Urine (ng/mL) Confirmation
Screen
Confirmation
DRE category – CNS depressants Alprazolam
–(c)
Chlordiazepoxide
–
(c)
Clonazepam
–
(c)
7-Aminoclonazepam
–
(c)
Diazepam
–
(c)
Nordiazepam
50
10
–(c)
50 total(b)
50
–
(c)
50 total(b)
10
–
(c)
50 total(b)
10
–
(c)
50 total(b)
20
–
(c)
50 total(b)
20
100
50 total(b)
Lorazepam
–
(c)
10
–
(c)
50 total(b)
Oxazepam
50
50
100
50 total(b)
Temazepam
–
50 total(b)
50
–
(c)
(d)
25
–
(d)
50
Amitriptyline
–(d)
25
–(d)
50
Nortriptyiline
–(d)
25
–(d)
50
Diphenhydramine
–(d)
25
–(d)
50
Carisoprodol
–(d)
500
–(d)
500
Meprobamate
–(d)
500
Trazodone
(c)
–
500
–(d)
Zolpidem
–
(d)
20
–(d)
20
Butalbital
–(a)
100
–(a)
100
Phenobarbital
–(a)
100
–(a)
100
100
100
200
100
Phenytoin
–(d)
500
–(d)
5000
Carbamazepine
–
(d)
500
–(d)
5000
Topiramate
–(d)
1000
–(d)
1000
g-Hydroxybutyrate
–(d)
5000
–(d)
10 000
Codeine
–(e)
10
–(e)
50
6-Acetylmorphine
–(e)
10
–(e)
10
Hydrocodone
–
(e)
10
–
50
Hydromorphone
–(e)
10
–(e)
50
Methadone
50
10
300
50
Morphine
20 free(f)
50 total(b)
Secobarbital
DRE category – narcotic analgesics
(e)
10
200
Oxycodone
–
(e)
10
–
(e)
50
Propoxyphene
50
50
300
50
Tramadol
–(d)
20
–(d)
20
Dextromethorphan
–(d)
20
–(d)
50
Phencyclidine
10
10
25
10
DRE category – dissociative drugs
THC, D -tetrahydrocannabinol; carboxy-THC, 11-nor-9-carboxy-D -tetrahydrocannabinol; 11-OH-THC, 11-hydroxy-D -tetrahydrocannabinol; MDMA, 3,4methylenedioxymetamfetamine; CNS, central nervous system. (a) Immunoassay screening not targeted to this analyte. (b) Combination of free and conjugated analyte. (c) Immunoassay screening targeted to nordiazepam, oxazepam or both; not an effective tool for screening all drugs in this class. (d) Not routinely screened for by immunoassay. (e) Immunoassay screening targeted to morphine; not an effective tool for screening all drugs in this class. (f) Free drug, not conjugated. 9
9
Preliminary drug screening Many different types of immunoassay screening tests are available commercially. The techniques in most widespread use include enzyme multiplied immunoassay technique (EMIT), ELISA, kinetic interaction of microparticles in solution (KIMS) and fluorescence polarisation immunoassay (FPIA; see Chapters 3, 18 and 32). When applied appropriately, with the correct calibrators and controls, any of these techniques is suitable for drug screening in DUID cases. ELISA is in most widespread use, and has the advantage as a heterogeneous assay with a separation step
9
of being readily applied to blood in addition to other matrices. Homogeneous systems such as EMIT typically require some pretreatment (protein precipitation or extraction) prior to analysis of samples other than urine or serum. An immunoassay panel for blood or urine typically includes cocaine metabolites, opiates, amfetamines, methylenedioxyamfetamines, cannabis metabolites, methadone, PCP, propoxyphene, barbiturates and benzodiazepines. For OF, the same general targets would be included, with the exception of using a THC-specific assay for cannabinoids, and a cocaine-specific assay rather than a metabolite assay. Immunoassay is only a presumptive technique, and should not be
Clarke's Analysis of Drugs and Poisons Chapter No. 5 Dated: 10/3/2011 At Time: 16:56:57
122
Driving Under the Influence of Drugs
considered proof of the identification of a compound without a complementary confirmatory analysis. Immunoassay screening covers only a partial range of drugs, and typically would not detect other potentially impairing compounds such as novel analgesics, tramadol and tapentadol, or anticonvulsant drugs such as gabapentin, topiramate or leveteracitam, or partial g-aminobutyric acid (GABA) agonists such as zolpidem or zopiclone. All these drugs may play a role in driver impairment and underscore the need for a comprehensive chromatographic screen (GCMS or LC-MS) in addition to the immunoassay.
Chromatographic screening The ‘gold standard’ for confirmatory identification of drugs in biological samples is GC-MS or LC-MS. Typically, two fractions, one for basic drugs and the second for weakly acidic or neutral drugs, are obtained. Basic drugs Drug extraction
The procedure described below efficiently extracts basic drugs, including those commonly encountered in DUID cases such as those indicated in Table 5.4. The procedure described below includes a back -extraction into acid, which produces a very clean extract and reduces the amount of time required to analyse the GC-MS data, and the time spent on system maintenance. The back extraction, however, eliminates weak bases or amphoteric compounds, notably some benzodiazepines, which must be analysed for using a separate confirmatory procedure that does not include the back-extraction step. 1. Add 1 mL of the sample to a 20-mL disposable glass culture tube. 2. To each tube add 50 mL internal standard. 3. Add 1 mL pH 9.0 borate buffer to each tube and vortex mix for approximately 10 seconds. 4. Add 3 mL n-butyl chloride to each tube, cap, and rotate on a tube rotator for a minimum of 5 minutes. 5. Centrifuge until the organic and biological matrix layers are separated. 6. Transfer the n-butyl chloride organic layer to a clean, labelled, 10mL, conical, disposable centrifuge tube. 7. Back extract by adding 200 mL 3 mol/L HCl to the n-butyl chloride. 8. Cap and rotate for at least 5 minutes. 9. Centrifuge until separated; aspirate and discard the (upper) n-butyl chloride layer. 10. Remove any remaining butyl chloride by blowing air over the lower layer. 11. Add 100 mL concentrated ammonium hydroxide and 100 mL saturated ammonium carbonate to each tube to make the extract alkaline. 12. Vortex mix for 15–30 seconds. 13. Add 150 mL of chloroform and vortex mix for 15 seconds. 14. Centrifuge until the organic and aqueous layers are separated. 15. Using a serological disposable pipette, transfer approximately 50–75 mL of the lower organic layer into two labelled autosampler vials for GC-MS analysis. GC-MS analysis
Many different conditions of flow rate, columns and temperature programming can be used for the GC-MS analysis of basic drugs to emphasise or optimise certain separations for frequently encountered compounds that might co-elute or interfere. The conditions for optimum separation have to be balanced against the run time so as to effectively use the laboratory’s resources and keep up with casework. The following method is an example of such a system that allows the chromatography of approximately 250 commonly encountered basic drugs and metabolites. Simultaneous analysis is performed on a dual-column gas chromatograph with a nitrogen phosphorus detector (NPD) on column 1 and a mass-selective detector (MSD) on column 2. Use of identical columns and simultaneous injection permits the comparison of the mass- and nitrogen-specific chromatograms, which assists with data interpretation and peak identification. In this example, both columns are 5%
phenylmethylsiloxane column (DB-5 or equivalent) 30 m 0.25 mm i. d., 0.25 mm film thickness. The temperature programme starts at 90 C, rises by 15 C/min for 12 minutes (to 180 C), then rises by 8 C/min for 15 minutes (to 300 C) and holds for 4 minutes. The total run time is 31 minutes. Weakly acidic and neutral drugs Extraction
The procedure described here isolates drugs with acidic and neutral character, such as non-steroidal anti-inflammatory compounds and paracetamol, carbamates (such as carisoprodol and meprobamate), barbiturates and anticonvulsants (such as carbamazepine and phenytoin; Logan et al. 2000). 1. Add approximately 1 g XAD resin (washed in ethyl acetate) to disposable glass culture tubes (20 mL). 2. Add 5 mL deionised water and 50 mL internal standard to each tube. 3. Add 1 mL of sample to the appropriately labelled tubes. 4. Thoroughly vortex mix each tube for approximately 60 seconds and allow to stand for at least 60 seconds to allow settling. 5. Aspirate the blood–water layer from each tube, leaving the XAD in the tube. 6. Add 6 mL ethyl acetate to each tube and vortex thoroughly for 60 seconds. 7. Centrifuge until separated, and transfer the ethyl acetate to an appropriately labelled, clean, conical, disposable tube. 8. Evaporate to dryness at approximately 50 C under air until very dry (approximately 20 minutes). 9. Reconstitute using 75 mL acetonitrile and wash with 500 mL heptane. 10. Vortex mix for about 15 seconds and centrifuge until separated. 11. Aspirate the heptane layer (top), and transfer the acetonitrile layer to labelled autosampler vials for GC-MS analysis. GC analysis
As with basic drugs, many GC protocols are suitable for this screen. Since the number of compounds being tested for is considerably smaller (30), the run time is correspondingly shorter. GC is performed on a gas chromatograph equipped with a flame ionisation detector (FID, for screening) or a: MSD (for confirmation). Helium is used as the carrier gas. The column is a 5% phenylmethylsilicone column (DB-5 or equivalent), 30 m 0.32 mm i.d., 0.25 mm film thickness. The temperature programme starts at 60 C, rises by 10 /min for 16 minutes, then rises by 20 C/min to 295 C and holds for 1 minute. The total run time is 21 minutes.
Determining an appropriate protocol for your laboratory While attention should be paid to the opinion of the arresting officer about what drugs are suspected in any given case, the toxicologist should apply a good broad-spectrum drug screen to be able to exclude the possibility of other drugs with similar effects. For example, as discussed later, an individual withdrawing from intravenous amfetamine use may appear drowsy and have constricted pupils, poor psychomotor performance and injection marks. This constellation of symptoms is very similar to those caused by a narcotic analgesic, and consequently a test only for opiates would not disclose the actual drug responsible for the person’s intoxication. The class of CNS-depressant drugs is probably the most challenging analytically, since it includes some chemically very diverse drugs, from solvents and inhalants, to long- and short-acting benzodiazepines, antidepressants, antihistamines and novel recreational agents such as GHB and butane-1,4-diol. Practically speaking, an escalation approach is the most economical and effective, beginning with the commonly encountered drugs and expanding and adding assays for drugs that could account for the symptoms observed on the basis of review of the preliminary data. It is critical that laboratories performing this work observe accepted standards for forensic analysis, including: maintaining security and chain of custody, maintaining written protocols for commonly performed
Clarke's Analysis of Drugs and Poisons Chapter No. 5 Dated: 10/3/2011 At Time: 16:56:58
Determining an appropriate protocol for your laboratory
123
Table 5.5 CNS-depressant drugs associated with impaired driving Antidepressants
Anxiolytics
Sedative hypnotics
Analgesics
Antipsychotics
Anticonvulsants
Muscle relaxants
Antihistamines
Amitriptyline Nortriptyline Amoxapine Clomipramine Imipramine Desipramine Doxepin Meprobamate Trazodone Trimipramine
Alprazolam Clonazepam Diazepam Oxazepam Triazolam
g-Hydroxybutyric acid (GHB) Flunitrazepam Lorazepam Temazepam Zolpidem Zopiclone Zaleplon
Tramadol Tapentadol Codeine Morphine Hydrocodone Dextropropoxyphene Pentazocine Fentanyl Methadone Pethidine Hydromorphone Oxycodone
Chlorpromazine Mesoridazine Thioridazine Thiothixene Loxapine
Phenobarbital Carbamazepine Chlordiazepoxide Phenytoin Clonazepam Topiramate
Butalbital Carisoprodol Diazepam Cyclobenzaprine
Diphenhydramine Chlorphenamine (Chlorpheniramine) Brompheniramine
procedures, validating method accuracy through use of appropriate standards and controls, verifying the identity of any drugs reported (using MS wherever possible), implementing the scientific review of data, participating in proficiency testing programmes and ensuring that staff have appropriately documented qualifications and training. Priority drugs associated with driving impairment CNS depressants
Many drugs have CNS-depressant properties either as a targeted effect or as a side-effect of the drug. While not intended to be a comprehensive list, Table 5.5 presents some common drugs with CNS-depressant activity that have been implicated in impaired driving cases. Most of these drugs exert their effects through the GABA pathways, with the exception of the opiates which cause sedation through the opioid m and k receptors. Alcohol is the best-known example of a CNS-depressant, and the general presentation of alcohol impairment is seen with these drugs, as discussed below. Performance in divided-attention tests, such as field impairment tests, is affected similarly by many CNS-acting drugs and observable nystagmus can be seen using the HGN test. Driving has been affected demonstrably in driving simulators, in on-road driving studies and from epidemiological and anecdotal reports for many drugs, and monographs concerning the benzodiazepines, GHB and carisoprodol are included in two special issues of Forensic Science Reviews (Vol. 14, 2002, and Vol. 15, 2003). By virtue of their common effect, combining alcohol with prescription or recreational drugs causes an additive or synergistic effect. Drugs with CNS-depressant properties are frequently prescribed in combination for the legitimate treatment of a constellation of effects associated with illness. One of the most commonly encountered examples in DUID casework is that of chronic pain patients. A patient who suffers from chronic debilitating back pain might be prescribed one or two centrally acting muscle relaxants such as carisoprodol or cyclobenzaprine, a barbiturate such as butalbital, an opioid analgesic such as propoxyphene, hydrocodone or oxycodone, a sleeping aid such as zolpidem, and often an antidepressant drug to treat the depression that typically accompanies chronic pain. The resultant combined effect makes some decrement in driving performance almost inevitable, and the patient, pharmacist and physician have an obligation to monitor the effectiveness of the treatment, select drugs with the least likelihood for impairment, use minimum effective doses, advise the patient of the risks to driving, and take care when adjusting the dose, changing the dosing schedule or adding new medications. The patient needs to be similarly advised that fatigue, alcohol and his or her illness itself can lead to unexpected changes in the risk of impairment. Whether the impairment is a result of legitimate, compliant, prescription use or not is immaterial from a public safety standpoint, and a driver impaired by use of prescription drugs must be subject to the same removal from the road as a recreational drug user. As noted earlier, there is currently insufficient information to relate blood drug concentrations to a specific degree of impairment; however, CNS-depressant drugs do display a qualitative dose–response relationship between concentration and effect, with higher concentrations being
associated with more obvious symptoms, progressing from mild impairment to unconsciousness, even in subjects with some tolerance. Effects on driving can occur with the therapeutic use of a single drug. Blood drug concentration data can help to establish whether a subject is following his or her prescribed course of medication. A subject under the influence of a CNS-depressant will typically display signs that are familiar from impairment through alcohol. Consistently noted are problems with fine or coarse motor control, staggering gait (ataxia), loss of balance, impairment in divided-attention tests, poor concentration and slurred speech. Subjects often have a sleepy or dazed appearance, may have difficulty understanding questions or responding appropriately, and may be disoriented to time and place. Readily observable HGN is a consistent and common feature of CNS-depressant intoxication. Driving behaviours commonly noted include weaving within or between lanes, failure to notice and obey traffic signals, failure to obey posted speed limits, wide turns, rear-end collisions, no use of turn signals and driving at night without lights. Marijuana
Marijuana (cannabis) is the most popular recreational drug after alcohol in most jurisdictions. Obtained from the plant Cannabis sativa, the leaves and buds contain a variety of cannabinoids that posses psychoactive effects, the predominant one being D9-THC. Cannabinoids have significant behavioural and physiological effects that contribute to changes in a person’s ability to drive safely. The drug is popular for its relaxation-promoting and euphoric properties, accompanied by sedation and changes in perception. Accompanying effects include altered time and distance perception, poor concentration, impaired learning and recall, increased appetite and mood changes. Associated physiological effects include increased pulse and blood pressure, and bloodshot conjunctivae. A loss of convergent vision is also reported. Beginning in 2008, synthetic cannabinoid agonists developed as investigational drugs started to appear in the recreational drug market. Sold as ‘Spice’, ‘Pep-spice’, ‘K2’ and by many other names, they are marketed as incense products or ‘legal highs’, and often labelled not for human consumption. A very diverse range of structural families has been identified with significant binding and agonist effects at the cannabinoid CB1 receptor, responsible for the intoxicating effects of THC. Preliminary data suggest that these compounds have very similar effects to marijuana, and will probably become important to consider in DUID investigations. The compounds most frequently reported to date are JWH-018, JWH-019, JWH-073, JWH-250, CP47,497, HU-210, WIN55212, RCS-4 among many others. These drugs generally do not cross-react with cannabinoid immunoassays, and will probably require detection levels of less than 1 ng/mL in blood, necessitating LCMS(-MS) or other high-sensitivity mass-spectrometric techniques. The predominant effects of concern with respect to the effects of cannabinoids on driving are sedation, vigilance, and the effects on concentration, divided attention, perception, and temporal and spatial orientation. The sedative effects can be similar to those of CNS depressants, and the associated driving behaviours are similar also, resulting in weaving, slowed responses and inattentiveness, frequently resulting in collisions.
Clarke's Analysis of Drugs and Poisons Chapter No. 5 Dated: 10/3/2011 At Time: 16:56:58
124
Driving Under the Influence of Drugs
The epidemiological literature with respect to cannabis has been reviewed extensively by Huestis (2001). Many of the underlying studies, however, suffer from a lack of appropriate control groups, and confounding factors (time, dose, duration, last use, combined use of cannabis with alcohol or other drugs, laboratory test cut-offs, etc.) make comparisons between these studies difficult. The ability to compare and compile study data is another reason for the international efforts towards standardised methods for drug testing in DUID research (Walsh et al. 2008). In concert, however, the behavioural, epidemiological and toxicological studies that have been undertaken to date point towards a general picture of an overrepresentation of cannabinoid-positive blood, OF or urine samples (indicating use) in drivers involved in accidents or arrested for impaired driving, when compared with the general population. This has been further demonstrated in the recent roadside surveys discussed earlier (Bierness et al. 2010). The second source of evidence for a relationship between cannabis use and driving impairment comes from laboratory-based tests of psychomotor performance. These studies often suffer from limitations on what dose of drug can ethically be administered to subjects and which subjects can be included. Studies may also be subject to observation effects in which the participants can exert greater effort because of the motivating factor of being under observation in the study. The value of the studies is that they can isolate and demonstrate decrements in specific measures of psychomotor and cognitive performance (e.g. tracking, reaction time, error rate, vigilance, learning, recall, divided attention). By extension, the evidence for impairment in the driving task after cannabis use, which incorporates many of these components, is verified. Laboratory-based studies suffer from a practical limit to which subjects can be dosed with recreational drugs; consequently, they probably underestimate the potential effect in chronic or frequent recreational users. Finally, and perhaps most convincingly, a limited number of studies have been performed in which subjects have been administered cannabis and then tasked with driving in electronic driving simulators, or actually operating vehicles on open or closed driving courses. These studies have also been reviewed by Huestis (2001), and reveal evidence of significant mild-to-moderate decrements in driving performance at the doses given. In an early study, Klonoff (1974) tested the effects of low doses of THC (5–8.5 mg) on subjects who were then asked to undertake closed-course and on-street driving in an urban setting in Vancouver, Canada. The authors reported impairment of attentiveness (increased distractibility), problems with braking, bad judgement and difficulty in concentrating. These effects were most pronounced on city streets when the demands of the driving were greatest. Illustrating some of the contradictions of these studies, however, some drivers actually improved in performance after the cannabis dose. This highlights one of the mitigating effects of cannabis intoxication, whereby some subjects have demonstrated an ability to focus concentration on tasks for short periods of time and perform quite well. This has led to the conclusion that sustained attention, or vigilance, rather than attention in general is most affected by cannabis. This finding can explain why it may be difficult to demonstrate impairment in field sobriety tests, which by their nature are of short duration. A research group in Australia has studied the effects of marijuana smoking on standardised field sobriety tests (Papafotiou et al. 2005a). They found that there was a dose-dependent increase in errors in the field sobriety tests, with between 30% and 40% of subjects showing clear indicators of impairment. The same doses of marijuana produced impairment in driving performance (Papafotiou et al. 2005b). On-road driving studies performed in the Netherlands (Robbe, O’Hanlon 1993, 1999) have produced other evidence of impairment. Volunteers smoked cannabis in doses of 100, 200 and 300 mg/kg (the latter being consistent with a ‘user-preferred dose’) and were assessed in an on-road driving task 40 and 100 minutes later. Drivers showed evidence of decrements in maintaining lateral vehicle position (weaving) at all doses, equivalent to blood alcohol levels of between 0.3 and 0.7 g/L. Importantly, no correlation was found between the blood THC concentrations and the degree of effect. Using similar methodology, the authors showed that, when cannabis was combined with alcohol, the effects of both drugs on driving were increased.
In summary, cannabis is a psychoactive drug that can influence mood, concentration and judgement, in addition to its sedating properties, all of which contribute in a dose-related fashion to impairment of vehicle operation. Compared with many other drugs, the level of impairment after mild-to-moderate single-dose recreational use is low, equivalent to a BAC in the range 0.3–0.7 g/L. Combining alcohol and cannabis produces a greater impairment than either alone, and cannabis use should be considered in determining the likely impairment in subjects with low BACs. The blood THC and metabolite concentrations are not well correlated with effect, although some authors have explored the possibility of relating these concentrations to time since last use (Huestis et al. 2005). CNS stimulants
In contrast to the sedative drugs discussed above, stimulants generally increase neural activity, which, in moderation, may not be entirely detrimental. Caffeine can revive the drowsy driver, and patients with attention deficit hyperactivity disorder (ADHD) or narcolepsy can benefit from appropriate doses of methylphenidate or amfetamine. Just as these drugs can act to restore some balance in these individuals, they can upset the delicate neural homeostasis in a healthy individual. Stimulants, principally cocaine and the amfetamines, are used recreationally for their excitatory, euphorigenic properties. In this context they produce intense, overwhelming and distracting effects. After acute administration, users report feeling elated or powerful, having superior intellect and insight, and sexual arousal and stimulation. Time may appear to pass more quickly, speech becomes faster and less coherent, and users can become impatient and agitated, sometimes to the point of violence. These perceptual changes are accompanied by increases in pulse and blood pressure, pupillary dilatation, sweating and psychomotor restlessness, manifested as pacing, fidgeting and scratching. Simple reaction time may be improved under the influence of stimulants, but this is only one component of driving; in fact, complex reaction time, which demands impulse control, intelligent decision-making and appropriate response, may be affected adversely. The intensity of these effects depends on the dose, on the route of administration, and to some extent on the user’s experience with, and tolerance of, the effects of the drug. Inevitably, these effects, when combined, are detrimental to complex task performance and make drivers less attentive, while the psychomotor excitation demands greater focus on muscle control and vehicle operation. These opposing effects result in poorer driving. As with any other drugs, the effects are likely to become more apparent when demands are high, such as when driving in bad weather, in heavy traffic, in an unfamiliar environment or in a defective vehicle. In addition to these acute effects, humans display both acute and chronic tolerance of the effects of stimulants. With cocaine and the amfetamines, the initial excitatory, euphoric phase can be followed by a withdrawal phase, the intensity of which depends on the duration and intensity of drug use. Binges of metamfetamine use are generally longer, of the order of days, than those for cocaine use, for which binges typically run only to hours, and amfetamine withdrawal is often more severe. This is in part a reflection of the different half-lives of the drugs, with cocaine having a 1–2 h half-life, while for amfetamines it is of the order of 7–15 h. During excitation, there is some downregulation of the dopamine receptors, reduction in dopamine transporter activity and reduction in concentrations of the enzyme tyrosine hydroxylase, responsible for dopamine synthesis. This decline in dopamine activity has the opposite effect to that experienced during excitation. Subjects are typically fatigued, lack energy, can be irritable and depressed, and are anhedonic and dysphoric. Sometimes delusions and pseudohallucinations can occur, and the subject can become psychotic. Often this is called the ‘crash’ phase, during which the subjects sleep a fitful, restless sleep, sometimes for days in the case of metamfetamine withdrawal. During this phase, subjects can appear as if under the influence of a CNS-depressant or opiate, and a poorer complex task performance is expected, such as in driving a vehicle. Drivers have been reported as being lethargic, sleepy, with very poor lane travel (weaving), and frequently drive at high speed, drive off the road or drive into oncoming traffic.
Clarke's Analysis of Drugs and Poisons Chapter No. 5 Dated: 10/3/2011 At Time: 16:56:58
References Isenschmid (2001) and Logan (2001) reviewed the available epidemiological, psychomotor performance and anecdotal evidence for impairment in driving after stimulant use, from the effects of both the acute excitation and later withdrawal phases, and concluded that these effects are real and significant. Blood cocaine and amfetamine drug concentrations must be interpreted with caution, since a single blood drug measurement does not predict what phase of intoxication the subjects may be experiencing, and consequently what pattern of effects are likely to predominate. Nevertheless, blood drug concentrations can show whether drug use is consistent with recreational doses in the case of cocaine, and may help distinguish between legitimate dosing of amfetamines in the treatment of narcolepsy, ADHD or eating disorders, and recreational use. The doses of stimulant drugs required to produce the sought-after euphoric effects are accompanied inevitably by deterioration in driving performance. Hallucinogens
Drugs with hallucinogenic properties have an obvious deleterious effect on driving. Inability to distinguish illusion from reality results in poor decision-making and consequently poorer driving. Drugs such as psylocibe mushrooms, mescaline, lysergide (LSD), ketamine and PCP can produce fully formed hallucinations, seeing objects, shapes or individuals that are not present, and synaesthesia or blending of sensory information such as ‘seeing’ sounds, or ‘hearing’ colours. Ketamine and its psychomotor effects on driving have been evaluated (Mozayani 2001). Many other drugs can produce milder hallucinations, including, as noted earlier, cannabis and stimulants. Methylenedioxy-substituted amfetamines, such as MDMA (ecstasy), methylenedioxyamfetamine (MDA) or methylenedioxyethamfetamine (MDEA; Eve) can also produce hallucinations, particularly tactile ones, that enhance sensitivity to touch. However, the predominant impairing effects of that class of compounds appear to be more related to their excitatory and stimulant properties (Logan, Couper 2001). Interpretation of results In many jurisdictions, the law requires that the presence of drugs in blood should be quantified and reported to the court. This then poses the question ‘If you have a quantitative measurement, what does it mean?’. Unlike alcohol, where a large body of information is available that enables the toxicologist to offer a generalised statement about the effects that might be expected when an individual has a known BAC, it is not possible to relate blood drug concentrations to drug effects or degrees of intoxication. Drugs affect different people in different ways and it is unlikely that anybody who has used a particular psychoactive drug will exhibit all of the signs and symptoms listed in the sections above to a noticeable degree. One approach to using quantitative results is to employ population profiles or cumulative frequency plots (Figure 5.1) to allow the results of a particular analysis to be compared with those of a population of drivers who have previously been classified as unfit to drive by qualified medical personnel. Examination of data 100 90
Percentage of cases
80 70 60
Tempazepam
50
ND
40
Diazepam
30 20 10 0
0
1
2
3
4
5
6
7
8
9
Concentration (µg/mL) Figure 5.1 Cumulative frequency profiles for diazepam, nordiazepam (ND) and temazepam in 354 cases where all three drugs were detected.
125
plotted in this way allows the toxicologist to compare an analytical result against the frequency with which it has been encountered in a casework population. Although impairment and symptoms cannot be correlated directly with blood drug concentrations, cumulative data plots enable the toxicologist to assist the court by comparing specific case results against a population of drivers who have previously been certified as unfit to drive. For example, Figure 5.1 shows population profiles for diazepam, nordiazepam and temazepam in 354 cases where all three drugs were detected. According to this figure, a subject with a blood diazepam concentration of 2 mg/mL would have a higher concentration than 75% of the 354 cases previously reviewed. This provides the court with some context for understanding the drug concentration reported in any individual case.
References Beirness DJ, Beasley EE (2010). A roadside survey of alcohol and drug use among drivers in British Columbia. Traffic Inj Prev 11: 215–221. Boorman M, Owens K (2009). The Victorian legislative framework for the random testing drivers at the roadside for the presence of illicit drugs: an evaluation of the characteristics of drivers detected from 2004 to 2006. Traffic Inj Prev 10: 16–22. Bramness JG et al. (2003). Testing for benzodiazepine inebriation--relationship between benzodiazepine concentration and simple clinical tests for impairment in a sample of drugged drivers. Eur J Clin Pharmacol 59: 593–601. Bramness JG et al. (2004). Impairment due to intake of carisoprodol. Drug Alcohol Depend 74: 311–318. Buchan BJ et al. (1998). Evaluation of the accuracy of on-site multi-analyte drug testing devices in the determination of the prevalence of illicit drugs in drivers. J Forensic Sci 43: 395–399. Burns M, Moskowitz H (1977). Psychophysical Tests for DWI Arrest, DOT-HS-501242. Washington DC: National Highway Traffic Safety Administration, US Department of Transportation. DRUID (2009). Evaluation of Oral Fluid Screening Devices by TISPOL to Harmonise European Police Requirements (ESTHER). Project No. TREN-05FP6TR-S07.61320-518404-DRUID. Available at: www.bast.de/nn_107548/ Druid/EN/deliverales-list/downloads/Deliverable__3__1__1,templateId¼raw, property¼publicationFile.pdf/Deliverable_3_1_1.pdf (accessed 16 November 2010). Diplock J, Plecas D (2009). Clearing the Smoke on Cannabis: Respiratory effects of cannabis smoking. Ottawa, Canada: Canadian Centre on Substance Abuse. Dubowski KM (1994). Quality assurance in breath-alcohol. J Forensic Sci 35: 1414–1423. Farrell LJ et al. (2007). Recommendations for toxicological investigation of drug impaired driving. J Forensic Sci 52: 1214–1218 [Erratum 2007; 53: 239]. Huestis M (2001). Cannabis (marijuana) – effects on human performance and behaviour. Forensic Sci Int 14: 15–61. Huestis MA et al. (1992). Blood cannabinoids II. Models for the prediction of time of marijuana exposure from plasma concentrations of delta-9-tetrahydrocannabinol (THC), and 11-nor-9-carboxy-deltahydrocannabinol (THCCOOH). J Anal Toxicol 16: 283–286. Huestis MA et al. (2005). Estimating the time of last cannabis use from plasma delta9-tetrahydrocannabinol and 11-nor-9-carboxy-delta9-tetrahydrocannabinol concentrations. Clin Chem 51: 2289–2295. ICADTS Working Group (2007).Categorization system for medicinal drugs affecting driving performance. Ann Arbor, MI: The International Council on Alcohol, Drugs & Traffic Safety. Available at: www.icadts.nl/reports/medicinaldrugs1.pdf (accessed 20 October 2010). ICADTS Drug List (July 2007). Ann Arbor, MI: The International Council on Alcohol, Drugs & Traffic Safety. Available at: www.icadts.nl/reports/medicinaldrugs2.pdf (accessed 20 October 2010). Isenschmid DS (2001). Cocaine – effects on human performance and behaviour. Forensic Sci Rev 14: 61–101. Klonoff H (1974). Marijuana and driving in real life situations. Science 186: 317–324. Logan BK (2001). Methamphetamine – effects on human performance and behavior. Forensic Sci Rev 14: 133–151. Logan BK, Couper FJ (2001). Methylenedioxymethamfetamine (MDMA, ecstasy) and driving impairment. J Forensic Sci 46: 1426–1433. Logan BK et al. (2000). Carisoprodol, meprobamate and driving impairment. J Forensic Sci 45: 619–623. Longo MC et al. (2000). The prevalence of alcohol, cannabinoids, benzodiazepines and stimulants amongst injured drivers and their role in driver culpability: part II: the relationship between drug prevalence and drug concentration, and driver culpability. Accid Anal Prev 32: 623–632. Mozayani A (2001). Ketamine – effects on human performance and behaviour. Forensic Sci Rev 14: 123–132. Papafotiou K et al. (2005). An evaluation of the sensitivity of the Standardised Field Sobriety Tests (SFSTs) to detect impairment due to marijuana intoxication. Psychopharmacology (Berl) 180: 107–114.
Clarke's Analysis of Drugs and Poisons Chapter No. 5 Dated: 10/3/2011 At Time: 16:56:58
126
Driving Under the Influence of Drugs
Papafotiou K et al. (2005). The relationship between performance on the standardised field sobriety tests, driving performance and the level of delta9-tetrahydrocannabinol (THC) in blood. Forensic Sci Int 155: 172–178. Robbe WJ, O’Hanlon JF (1993). Marijuana and Actual Driving Performance, DOT HS 808 078. Washington DC: US Department of Transport. Robbe HWJ, O’Hanlon JF (1999). Marijuana, Alcohol and Actual Driving Performance, DOT HS 808 939. Washington DC: US Department of Transport. Schwilke EW et al. (2006). Changing patterns of drug and alcohol use in fatally injured drivers in Washington State. J Forensic Sci 51: 1191–1198.
Senna MC et al. (2010). First nationwide study on driving under the influence of drugs in Switzerland. Forensic Sci Int 20 198: 11–16. Walsh JM et al. (2002). Driving under the influence of drugs (DUID) Legislation in the United States. Bethesda MD: The Walsh Group. Available at: www. walshgroup.org/MANUAL%20FINAL.pdf (accessed 16 November 2010). Walsh JM et al. (2008). Guidelines for research on drugged driving. Addiction 103: 1258–1268. Wille SM et al. (2009). Relationship between oral fluid and blood concentrations of drugs of abuse in drivers suspected of driving under the influence of drugs. Ther Drug Monit 31: 511–519.
Clarke's Analysis of Drugs and Poisons Chapter No. 6 Dated: 15/3/2011 At Time: 11:51:37
CHAPTER
6
Drug Testing in Human Sport DA Cowan
Introduction Drug abuse in sport is often called doping, the international word ‘dope’ being used both as a noun and as a verb. This is thought to originate from the Dutch word ‘doop’ which means ‘Christian baptism’. It seems likely that the religious fervour associated with this ceremony resulted in the use of the same word, in a somewhat cynical and contemptuous way, to describe the state of intoxication and euphoria induced by certain drugs. The word does not appear in this context before the twentieth century despite the practice of horse ‘nobbling’, which was known well before this time and is described separately in Chapter 7. The word appears to have come into use early in the twentieth century, and it is probably associated with the rise of the pharmaceutical industry. The abuse of drugs in an attempt to enhance performance in human sporting competitions is not new. For example, the Greek authors Phylostratos and Galen commented on the ethics of competitors in the Olympics who would take any preparation to improve their performance. Roman gladiators were often drugged to make their fights more lusty and bloody as demanded by the spectators. The effect of drugs on performance is often extremely difficult to determine, and there is little definitive published work for any species. The results that have been published are often conflicting; some workers suggest an increase in the competitor’s performance and others suggest no improvement. The test system may not adequately relate to the appropriate sporting performance, such as increase in muscle strength and sprint running. Changes in speed of less than 1% cannot be demonstrated with statistical significance because of the many variable and uncontrollable factors, yet an improvement of only 1% represents a lead of 1 metre in a 100-metre sprint or 17.6 yards over 1 mile, which is an enormous lead in either race. Furthermore, athletes may take far larger amounts of drugs than would be ethically acceptable in most human experiments. The toxic side-effects of drugs are less difficult to ascertain, but the conclusions drawn from the available data are often circumstantial (e.g. the possible links between taking anabolic steroids and liver cancer). Nevertheless, there is sufficient evidence of the harmful effects when certain drugs are misused to justify their prohibition from sports competitions.
minimum of interference with the normal therapeutic use of drugs. Thus, a competitor who is undergoing a legitimate course of treatment is not disqualified. However, this creates many problems in deciding which drugs should or should not be prohibited, and whether or not a competitor who requires a particular drug treatment should be allowed to compete. WADA has recently published the new World Anti-Doping Code (WADA 2009h). In this document doping is defined as ‘the occurrence of one or more of the anti-doping rule violations set forth in Article 2.1 through Article 2.8 of the Code’. WADA has published criteria that it uses when deciding what should be prohibited. Among these is Article 4.3.1.2 of the Code, which gives as one of the criteria for prohibiting a substance as ‘Medical or other scientific evidence, pharmacological effect or experience that the use of the substance or method represents an actual or potential health risk to the athlete’ (note that the italicised words are WADA-defined terms). The IOC and now WADA list examples of prohibited drugs according to their pharmacological classification (Table 6.1), but the entries are often far from explicit, using the words ‘and other substances with similar chemical structure or similar biological effect(s)’. For most substances, the mere presence of the substance or a diagnostic metabolite in the biological fluid sampled constitutes an offence, but for some substances (Table 6.2) there is a reporting threshold. Furthermore, certain methods are prohibited (Table 6.3); one category of which is to attempt to prevent detection of doping. WADA states that ‘The use of any drug should be limited to medically justified indications’. It has a system in place (WADA 2009d) whereby athletes being treated with certain medications such as b2-agonists may obtain a ‘Therapeutic Use Exemption’ certificate that will allow them to use that medication without contravening the WADA Code. WADA maintains a comprehensive set of guidance documents to help with the exemption process to permit needed medical treatment. Also WADA permits the use of certain substances (Table 6.4) other than during competitions. Finally, some sports prohibit additional substances (Table 6.5) and WADA publishes a comprehensive Prohibited List (WADA 2010b) each year.
Reported analytical findings Rules In human sports, the main controlling body is the International Olympic Committee (IOC). However, since 2004 doping issues have been taken over by the World Anti-Doping Agency (WADA; originally called the International Anti-Doping Agency). The IOC and WADA jointly issued the Olympic Movement Anti-Doping Code, which defined doping as: The use of an expedient (substance or method) that is potentially harmful to athletes’ health and/or capable of enhancing their performance, or The presence in the athlete’s body of a prohibited substance or evidence of the use thereof or evidence of the use of a prohibited method. The stated intention of the Medical Commission of the IOC was to ban those drugs that are likely to be harmful when misused, with the
Data for human sports have been available since 1987 and are presented for the years 1987, 1990, 1995, 2000 and 2005 in Table 6.6. Figure 6.1 shows the proportion of samples analysed by WADA-accredited laboratories in the years 1988 to 2008 reported for the three most commonly found prohibited substances. Note the marked increase in the reporting of testosterone in recent years following the reduction of the reporting threshold by WADA. Note also the decrease in the reporting of salbutamol in the last 2 years following the increase in the reporting threshold by WADA.
Sampling Sample collecting procedures must take into consideration both scientific and legal aspects: n n
The health of the individual being sampled must be safeguarded. Incorrect labelling, contamination or sample switching must be avoided at the time of sample collection and subsequently. 127
Clarke's Analysis of Drugs and Poisons Chapter No. 6 Dated: 15/3/2011 At Time: 11:51:37
128
Drug Testing in Human Sport
Table 6.1 Substances prohibited by the World Anti-Doping Agency at all times Category Anabolic agents
Substance 1. Anabolic androgenic steroids (AAS) (a) Exogenous(a) AAS, including: 1-androstendiol (5a-androst-1-ene-3b,17b-diol); 1-androstendione (5a-androst-1-ene-3,17-dione); bolandiol (19-norandrostenediol); bolasterone; boldenone; boldione (androsta-1,4-diene-3,17-dione); calusterone; clostebol; danazol (17a-ethynyl-17b-hydroxyandrost-4-eno[2,3-d]isoxazole); dehydrochlormethyltestosterone (4-chloro-17b-hydroxy-17a-methylandrosta-1,4-dien-3-one); desoxymethyltestosterone (17a-methyl-5a-androst-2en-17b-ol); drostanolone; ethylestrenol (19-nor-17a-pregn-4-en-17-ol); fluoxymesterone; formebolone; furazabol (17b-hydroxy-17a-methyl-5a-androstano[2,3-c]-furazan); gestrinone; 4-hydroxytestosterone (4,17b-dihydroxyandrost-4-en-3-one); mestanolone; mesterolone; metenolone; methandienone (17b-hydroxy-17a-methylandrosta-1,4-dien-3-one); methandriol; methasterone (2a, 17a-dimethyl-5a-androstane-3one-17b-ol); methyldienolone (17b-hydroxy-17a-methylestra-4,9-dien-3-one); methyl-1-testosterone (17b-hydroxy-17a-methyl-5a-androst-1-en-3-one); methylnortestosterone (17b-hydroxy-17a-methylestr-4-en-3-one); methyltrienolone (17b-hydroxy-17a-methylestra-4,9,11-trien-3-one); methyltestosterone; mibolerone; nandrolone; 19-norandrostenedione (estr-4-ene-3,17-dione); norboletone; norclostebol; norethandrolone; oxabolone; oxandrolone; oxymesterone; oxymetholone; prostanozol (17b-hydroxy-5a-androstano[3,2-c] pyrazole); quinbolone; stanozolol; stenbolone; 1-testosterone (17b-hydroxy-5a-androst-1-en-3-one); tetrahydrogestrinone (18a-homo-pregna-4,9,11-trien-17b-ol-3-one); trenbolone and other substances with a similar chemical structure or similar biological effect(s) (b) Endogenous(b) AAS when administered exogenously: androstenediol (androst-5-ene-3b,17b-diol); androstenedione (androst-4-ene-3,17-dione); dihydrotestosterone (17b-hydroxy-5a-androstan-3-one); prasterone (dehydroepiandrosterone, DHEA); testosterone and the following metabolites and isomers: 5a-androstane-3a,17a-diol; 5a-androstane-3a,17b-diol; 5a-androstane-3b,17a-diol; 5a-androstane-3b,17bdiol; androst-4-ene-3a,17a-diol; androst-4-ene-3a,17b-diol; androst-4-ene-3b,17a-diol; androst-5-ene-3a,17adiol; androst-5-ene-3a,17b-diol; androst-5-ene-3b,17a-diol; 4-androstenediol (androst-4-ene-3b,17b-diol); 5-androstenedione (androst-5-ene-3,17-dione); epi-dihydrotestosterone; epitestosterone; 3a-hydroxy-5aandrostan-17-one; 3b-hydroxy-5a-androstan-17-one; 19-norandrosterone; 19-noretiocholanolone 2. Other anabolic agents, including but not limited to: Clenbuterol, selective androgen receptor modulators (SARMs), tibolone, zeranol, zilpaterol
Hormones and related substances
The following substances and their releasing factors are prohibited:
b2-Agonists
All b2-agonists including their D- and L-isomers are prohibited.
1. 2. 3. 4. 5.
Erythropoiesis-stimulating agents (e.g. erythropoietin (EPO), darbepoietin (dEPO), haematide) Growth hormone (GH), insulin-like growth factors (e.g. IGF-1), mechano growth factors (MGFs) Chorionic gonadotrophin (CG) and luteinizing hormone (LH) in males Insulins Corticotrophins
and other substances with similar chemical structure or similar biological effect(s) Therefore, formoterol, salbutamol, salmeterol and terbutaline when administered by inhalation also require a Therapeutic Use Exemption in accordance with the relevant section of the International Standard for Therapeutic Use Exemptions Despite the granting of a Therapeutic Use Exemption, the presence of salbutamol in urine in excess of 1000 mg/L will be considered as an Adverse Analytical Finding unless the Athlete proves, through a controlled pharmacokinetic study, that the abnormal result was the consequence of the use of a therapeutic dose of inhaled salbutamol Hormone antagonists and modulators
Diuretics and other masking agents
(a) (b)
The following classes are prohibited: 1. Aromatase inhibitors including, but not limited to: anastrozole, letrozole, aminoglutethimide, exemestane, formestane, testolactone 2. Selective oestrogen receptor modulators (SERMs) including, but not limited to: raloxifene, tamoxifen, toremifene 3. Other anti-oestrogenic substances including, but not limited to: clomiphene, cyclofenil, fulvestrant 4. Agents modifying myostatin function(s) including but not limited to: myostatin inhibitors Masking agents are prohibited. They include: Diuretics, probenecid, plasma expanders (e.g. IV administration of albumin, dextran, hydroxyethyl starch and mannitol) and other substances with similar biological effect(s) Diuretics include: acetazolamide, amiloride, bumetanide, canrenone, chlorthalidone, etacrynic acid, furosemide, indapamide, metolazone, spironolactone, thiazides (e.g. bendroflumethiazide, chlorothiazide, hydrochlorothiazide), triamterene, and other substances with a similar chemical structure or similar biological effect(s) (except drosperinone and topical dorzolamide and brinzolamide, which are not prohibited)
'Exogenous' refers to a substance that is not ordinarily capable of being produced by the body naturally. 'Endogenous' refers to a substance that is capable of being produced by the body naturally.
Clarke's Analysis of Drugs and Poisons Chapter No. 6 Dated: 15/3/2011 At Time: 11:51:37
Sampling Table 6.2 WADA Code. Summary of urinary concentrations above which WADA-accredited laboratories must report findings for specific substances (WADA 2009f) Substance
Urinary concentration to be reported
Carboxy-THC(a)
>15 mg/L
Cathine(b)
>5 mg/L
Ephedrine
>10 mg/L
Epitestosterone(c)
>200 mg/L
Methylephedrine
>10 mg/L
Morphine(d)(e)
>1 mg/L
19-Norandrosterone(c)
>2 mg/L
Salbutamol(d)(f)
>1 mg/L
testosterone/epitestosterone (T/E) ratio(g)
>4
Table 6.4 Substances prohibited by the World Anti-Doping Agency only in-competition Stimulants
(a) Non-specified stimulants: Adrafinil; amfepramone; amiphenazole; amfetamine; amfetaminil; benzphetamine; benzylpiperazine; bromantan; clobenzorex; cocaine; cropropamide; crotetamide; dimethylamfetamine; etilamfetamine; famprofazone; fencamine; fenetylline; fenfluramine; fenproporex; furfenorex; mefenorex; mephentermine; mesocarb; methamfetamine (D-); methylenedioxyamfetamine; methylenedioxymethamfetamine; pmethylamfetamine; modafinil; norfenfluramine; phendimetrazine; phenmetrazine; phentermine; 4phenylpiracetam (carphedon); prolintane
11-Nor-delta 9-tetrahydrocannabinol-9-carboxylic acid. Unless it may be as a metabolite of a permitted substance such as pseudoephedrine. Threshold adjusted if specific gravity >1.020. (d) Sum of glucuronide conjugate and free drug concentrations. (e) Unless it may be as a metabolite of a permitted substance such as codeine. (f) Concentrations greater than 500 mg/L and less than 1 mg/L should be reported as consistent with the use of a b2-agonist. (g) Testosterone/epitestosterone ratio. Although a report must be issued for samples with a T/E ratio greater than 4, samples with lower ratios must also be reported if there is evidence of an exogenous origin of testosterone. (b) (c)
A stimulant not expressly listed in this section is a Specified Substance (b) Specified stimulants (examples): Adrenaline(b); cathine(c); ephedrine(d); etamivan; etilefrine; fenbutrazate; fencamfamin; heptaminol; isometheptene; levmetamfetamine; meclofenoxate; methylephedrine(d); methylphenidate; nikethamide; norfenefrine; octopamine; oxilofrine; p-hydroxyamfetamine; pemoline; pentetrazol; phenpromethamine; propylhexedrine; selegiline; sibutramine; strychnine; tuaminoheptane and other substances with a similar chemical structure or similar biological effect(s)
Table 6.3 Methods prohibited by the World Anti-Doping Agency at all times Enhancement of oxygen transfer The following are prohibited:
Narcotics
Chemical and physical manipulation 1. Tampering, or attempting to tamper, in order to alter the integrity and validity of Samples collected during Doping Controls is prohibited. These include but are not limited to catheterisation, urine substitution and/or alteration 2. Intravenous infusions are prohibited except in the management of surgical procedures, medical emergencies or clinical investigations Gene doping The transfer of cells or genetic elements or the use of cells, genetic elements or pharmacological agents to modulate expression of endogenous genes having the capacity to enhance athletic performance is prohibited Peroxisome proliferator activated receptor d (PPARd) agonists (e.g. GW 1516) and PPARd-AMP-activated protein kinase (AMPK) axis agonists (e.g. AICAR) are prohibited
n
The rights of the individual or team must be safeguarded against error by the analyst.
Samples in human sport are now usually collected by agencies certified in accordance with the ISO 9001 standard (www.iso.org/iso/ iso_catalogue/management_standards/iso_9000_iso_14000.htm) and using doping control officers who have been appropriately trained to collect the samples. Apart from its Code (WADA 2009h), WADA publishes an International Standard for Testing (WADA 2009c), which incorporates elements from ISO/PAS 18873 (which was withdrawn in 2005) and the ISO 9000 series of quality management system standards. WADA and most international federations always provide a second portion of the sample for defence use. This is to be opened only after the first sample has been found to contain a banned drug, and after the competitor has been notified and invited to attend the second analysis, with his or her own expert if the competitor so wishes.
All stimulants (including both their D- and Loptical isomers where relevant) are prohibited, except imidazole derivatives for topical use and those stimulants included in the 2009 Monitoring Program(a) Stimulants include:
(a)
1. Blood doping, including the use of autologous, homologous or heterologous blood or red blood cell products of any origin 2. Artificially enhancing the uptake, transport or delivery of oxygen, including but not limited to perfluorochemicals, efaproxiral (RSR13) and modified haemoglobin products (e.g. haemoglobin-based blood substitutes, microencapsulated haemoglobin products)
129
The following narcotics are prohibited: Buprenorphine, dextromoramide, diamorphine (heroin), fentanyl and its derivatives, hydromorphone, methadone, morphine, oxycodone, oxymorphone, pentazocine, pethidine
Cannabinoids
Cannabinoids (e.g. hashish, marijuana) are prohibited
Glucocorticosteroids*
All glucocorticosteroids are prohibited when administered by oral, intravenous, intramuscular or rectal routes In accordance with the International Standard for Therapeutic Use Exemptions, a declaration of use must be completed by the Athlete for glucocorticosteroids administered by intra-articular, periarticular, peritendinous, epidural, intradermal and inhalation routes, except as noted below Topical preparations when used for auricular, buccal, dermatological (including iontophoresis/phonophoresis), gingival, nasal, ophthalmic and perianal disorders are not prohibited and require neither a Therapeutic Use Exemption nor a declaration of use
(a) The following substances included in the 2009 Monitoring Program (bupropion, caffeine, phenylephrine, phenylpropanolamine, pipradol, pseudoephedrine, synephrine) are not considered as Prohibited Substances. (b) Adrenaline associated with local anaesthetic agents or by local administration (e.g. nasal, ophthalmological) is not prohibited. (c) Cathine is prohibited when its concentration in urine is greater than 5 mg/L. (d) Each of ephedrine and methylephedrine is prohibited when its concentration in urine is greater than 10 mg/L. (e) WADA use the term 'glucocorticosteroid' for what is more conventionally known as a corticosteroid.
Clarke's Analysis of Drugs and Poisons Chapter No. 6 Dated: 15/3/2011 At Time: 11:51:39
130
Drug Testing in Human Sport
Table 6.5 Substances prohibited by particular sports and only incompetition Alcohol Alcohol (ethanol) is prohibited in-competition only, in the following sports. Detection will be conducted by analysis of breath and/or blood. The doping violation threshold (haematological values) is 0.10 g/L. n Aeronautic (FAI) n Archery (FITA, IPC) n Automobile (FIA) n Boules (IPC bowls) n Karate (WKF) n Modern Pentathlon (UIPM) for disciplines involving shooting n Motorcycling (FIM) n Ninepin and Tenpin Bowling (FIQ) n Powerboating (UIM) Beta-blockers
Unless otherwise specified, beta-blockers are prohibited in-competition only, in the following sports. n Aeronautic (FAI) n Archery (FITA, IPC) (also prohibited Out-ofCompetition) n Automobile (FIA) n Billiards and Snooker (WCBS) n Bobsleigh (FIBT) n Boules (CMSB, IPC bowls) n Bridge (FMB) n Curling (WCF) n Golf (IGF) n Gymnastics (FIG) n Motorcycling (FIM) n Modern Pentathlon (UIPM) for disciplines involving shooting n Ninepin and Tenpin Bowling (FIQ) n Powerboating (UIM) n Sailing (ISAF) for match race helms only n Shooting (ISSF, IPC) (also prohibited Out-ofCompetition) n Skiing/Snowboarding (FIS) in ski jumping, freestyle aerials/halfpipe and snowboard halfpipe/big air n Wrestling (FILA)
Beta-blockers include, but are not limited to, the following: acebutolol, alprenolol, atenolol, betaxolol, bisoprolol, bunolol, carteolol, carvedilol, celiprolol, esmolol, labetalol, levobunolol, metipranolol, metoprolol, nadolol, oxprenolol, pindolol, propranolol, sotalol, timolol
Urine Urine is the preferred body fluid. Its collection is non-invasive, it is generally available in sufficient quantity, and the drugs or their metabolites tend to be present in relatively high concentrations. The disadvantages are that a drug may be present as its metabolites or in a conjugated form, and the parent drug may be present only in a relatively low concentration. Furthermore, the relationship with the concentration in blood is very imprecise. Substitution of samples is clearly a possibility that must be avoided and particular care is required during the period of waiting before a sample is obtained to balance this risk against the desire for privacy on the part of a person. It has been reported that racing cyclists have carried a rubber bladder of (negative) urine under their arm, connected by a rubber tube to the appropriate discharge point. Blood The principal advantage of a blood sample is that its integrity is easier to safeguard because it is usually collected by a doctor or phlebotomist experienced in the procedure. In addition, drug concentrations in blood are interpreted more easily than those in urine and certain drugs that are not excreted in urine in significant quantities (e.g. human growth hormone) can be detected in blood. Since the 2000 Olympic Games in Sydney, blood has been collected routinely by some federations (e.g. the International Cycling Union) as a ‘health check’. Any competitor whose haematocrit is above 50% is not permitted to compete. This test is intended to limit the use of erythropoietin (EPO) to stimulate red cell production. However, this haematocrit test is readily circumvented and depends on too many factors; the use of haemoglobin concentration is preferred. Furthermore, blood samples may also be collected for more sophisticated tests to indicate the use of EPO. In addition, the administration of small doses (so-called microdosing) of EPO has been shown (Ashenden et al. 2006) to reduce the chance of detection of EPO use and yet still raise haemoglobin concentrations, hence the need to collect blood samples. Recently, tests for recombinant human growth hormone (rhGH) administration and for blood transfusion have been developed (see later) that rely on the use of blood samples.
Other matrices At the present time, WADA does not permit the use of alternative biological matrices such as oral fluid or hair to counter an analytical finding obtained from either a urine or blood sample. Nevertheless, Kintz has reviewed hair testing and doping control in human sport (Kintz 1998) and an interesting case report of its use in detecting a variety of prohibited substances in body builders has been published (Dumestre-Toulet et al. 2002) (see Chapter 19).
Table 6.6 Prohibited substances most commonly reported by WADA-accredited laboratories, in order of frequency(a) Number of reports Substance
1987
Substance
1990
Substance
1995
Substance
2000
Substance
2005
Nandrolone
262
Nandrolone
192
Testosterone
293
Salbutamol
367
Testosterone
1132
Pseudoephedrine
100
Testosterone
83
Testosterone
171
Cannabis
224
Nandrolone
325
Cannabis
503
Pseudoephedrine
123
Nandrolone
212
Testosterone
306
Salbutamol
357
Ephedrine
58
Stanozolol
79
Methandienone
132
Cannabis
295
Nandrolone
298
Phenylpropanolamine
57
Phenylpropanolamine
64
Salbutamol
132
Pseudoephedrine
136
Stanozolol
233
Methenolone
42
Ephedrine
43
Pseudoephedrine
102
Ephedrine
129
Amfetamine
194
Stanozolol
37
Codeine
32
Ephedrine
78
Stanozolol
116
Terbutaline
171
110
Methandienone
27
Methenolone
25
Stanozolol
78
Terbutaline
hCG
143
Codeine
26
Amfetamine
24
Methenolone
39
Methandienone
75
Budesonide
116
Amfetamine
24
Methandienone
23
Clenbuterol
31
Lidocaine
64
Ephedrine
(a)
1987 was first year of available data.
93
Clarke's Analysis of Drugs and Poisons Chapter No. 6 Dated: 15/3/2011 At Time: 11:51:39
Analytical approach
131
0.90 0.80 0.70
%
0.60 0.50 0.40
Nandrolone
Testosterone
08 20
06 20
04 20
02 20
00 20
98 19
96 19
94 19
92 19
90 19
19
88
0.30 0.20 0.10 0.00
Salbutamol
Figure 6.1 The proportion of human sports samples analysed by WADA-accredited laboratories in the years 1988 to 2008 reported for the three prohibited substances most commonly found.
Analytical approach
Table 6.7 Minimum required performance levels
With the exception of anabolic steroids, prohibited substances are generally administered at or near the therapeutic dose, which results in relatively low concentrations in biological fluids. The laboratory is provided with a coded sample to preserve the anonymity of the athlete. They may also be given a declaration of any drug that has recently been taken by the athlete but, apart from that, there is usually no evidence whether or not a drug has been administered, or what sort of drug it might be. As with equine testing, any drug used in human treatment or in veterinary practice may be found. Thus, screening procedures are designed to be both sensitive and of wide coverage. The material analysed is usually in a fairly fresh condition. The analyst thus has a clearer picture of a normal sample than does the forensic or hospital chemist, who may be required to examine a wide variety of materials in various states of decomposition. Any sample that fails a screening test is invariably submitted to rigorous confirmatory testing (see below) before an adverse report is issued. The WADA-accredited laboratory must reliably be able to detect and confirm the presence of prohibited substances or their metabolites at least down to a minimum required performance level (MRPL; see Table 6.7) (WADA 2009f). However, WADA states that ‘for non-threshold substances prohibited in-competition only, it is not recommended that laboratories report below 10% of the MRPL’. This refers to stimulants, narcotics and betablockers only (the latter being prohibited only by particular sports) since, for glucocorticoids, WADA states that ‘laboratories are not to report below the MRPL’. Although, with some exceptions, the parent drug is the entity that appears in the Prohibited List, screening procedures rely upon the detection of either the unchanged drug or its metabolites. The identification of the corresponding metabolites is often useful supplementary evidence to support the identification of the parent drug, and indeed WADA expects the laboratory to identify as many of the presumptive analytical findings from the screening procedures as possible. In addition, the presence of metabolites in the appropriate concentrations relative to the parent drug helps to support the conclusion that a drug has been administered. Conversely, the absence of any expected metabolites is possible evidence that a sample has been contaminated; this should be refutable by a proper chain of custody. Occasionally, the parent drug is not excreted in urine at a detectable concentration and a knowledge of the metabolic pathways is thus essential. An example of this is the identification of 19-norandrosterone and 19-noretiocholanolone (Fig. 6.2) in the urine of humans as evidence of the administration of the anabolic steroid nandrolone or a 19norsteroid precursor. Drugs can be used either to improve or to impair athletic performance, though in human sport the latter category of drug is
Prohibited class
Specific examples/ exceptions
Concentration
Stimulants
0.5 mg/L 0.2 mg/L
Strychnine Narcotics
0.2 mg/L 10 mg/L
Buprenorphine
10 mg/L
Anabolic agents
Hormone antagonists and modulators
Clenbuterol
2 mg/L
Methandienone
2 mg/L
Methyltestosterone
2 mg/L
Stanozolol
2 mg/L
Epitestosterone
2 mg/L
Aromatase inhibitors, SERMs and other anti-oestrogenic substances
50 mg/L
Beta2-Agonists
100 mg/L
Beta-blockers
0.5 mg/L
Diuretics
0.25 mg/L 30 mg/L
Glucocorticosteroids Peptide hormones
hCG
5 IU/L
OH H H
H
O
O
O H H
H HO
HO 19-Norandrosterone
H 19-Etiocholanolone
Figure 6.2 Main metabolites of nandrolone (19-norandrosterone).
Clarke's Analysis of Drugs and Poisons Chapter No. 6 Dated: 15/3/2011 At Time: 11:51:39
132
Drug Testing in Human Sport
unlikely to be used. No single analytical scheme will suffice to cover so many different types of compound; various approaches have evolved in sports drug testing laboratories to address this challenge. Some drugs are notable for being excreted in urine almost entirely in conjugated form as, for instance, most anabolic steroids. When the presence of these drugs is suspected, hydrolysis before extraction is essential, although liquid chromatography–tandem mass spectrometry (LC-MS(-MS)) of intact conjugates is starting to be used. Liquid–liquid extraction may be used, for example with alkalinisation of the urine, to prepare samples for the detection of basic drugs by gas chromatography (GC) with nitrogen-selective detection. Alternatively, drugs have been extracted on the styrene–divinylbenzene copolymer XAD-2 resin. The development of solid-phase extraction (SPE) in the cartridge format in the late 1970s (Shackleton, Whitney 1980) and the rapid advances made in the technology associated with the technique have provided an attractive alternative to liquid–liquid extraction in many drug-screening programmes. Immunochemical methods that covered anabolic steroids were first introduced into human drug screening programmes in the 1970s (Brooks et al. 1979). However, in the late 1980s, these were largely replaced by GC-MS methods. Unlike many horseracing drug testing laboratories, human sports drug testing laboratories have not employed thin-layer chromatography (TLC) and do not use enzyme-linked immunosorbent assay (ELISA) extensively, but have used XAD-2 resin in columns for sample extraction and now, more commonly C8 and C18 SPE. Instrumental methods based on GC-MS, GC–high resolution mass spectrometry (GC-HRMS), GC-MS(-MS) and LC-MS(-MS) are preferred by most laboratories.
some polar compounds, such as ephedrine, are readily extracted in this procedure, it is best suited to less polar compounds. Anabolic steroids and corticosteroids
1. Condition C18 or C8 SPE cartridges with methanol and then water. Add 4 mL of the urine sample. 2. Wash with an equal volume of water and then elute the adsorbed steroids with an equal volume of methanol. 3. Remove the methanol under nitrogen, dissolve the residue in 1 mL of 0.1 mol/L phosphate buffer (pH 6.2) containing b-glucuronidase from Escherichia coli (approximately 1.4 units using 4-nitrophenylb-D-glucuronide as the substrate at 37 C). 4. Incubate for either a minimum of 2 h at 50 C or overnight at 37 C. 5. Cool and add approximately 100 mg potassium carbonate and extract the steroids with 5 mL distilled diethyl ether. 6. Divide the ether into two portions. 7. Evaporate each portion under nitrogen, dry the residue in a vacuum desiccator over phosphorus pentoxide–potassium hydroxide. 8. To one portion, add 200 mL of LC mobile phase, transfer to an autosampler vial and immediately cap the vial. 9. Examine the solution by LC-MS(-MS) for corticosteroids (and anabolic steroids such as tetrahydrogestrinone that are not amenable to trimethylsilylation for GC-MS analysis). 10. Derivatise the other portion by the addition of 100 mL N-methyl-Ntrimethylsilyl-trifluoroacetamide–ammonium iodide–ethanethiol (1000 : 2:6) and heating at 60 C for at least 15 min. 11. Examine the solution by capillary column GC-MS. Mass spectral and chromatographic data for many anabolic steroids and their metabolites have been published by Ayotte et al. (1996) and are given as system GAI (see Chapter 40): n
Solvent extraction In general, the choice of solvent is dictated by the wide range of drugs to be covered, or the need to extract a specific drug as in confirmatory analysis procedures. In many sports drug testing laboratories throughout the world, SPE has replaced liquid–liquid extraction for the isolation of drugs from both urine and plasma. Based upon the studies of Shackleton and Whitney (1980), the use of C18 or C8 bonded-phase cartridges for the isolation of anabolic steroids and their metabolites is the most common approach for sample extraction.
Sample preparation Extraction method for drugs and metabolites Note the initial pH and specific gravity of each sample. Basic drugs
1. Take an aliquot (4.0 mL) of each urine sample in a 10 mL centrifuge tube. 2. Add 0.5 mL of 5 mol/L potassium hydroxide solution, approximately 3 g of sodium chloride, 100 mL of diphenylamine as a reference standard solution (80 mg/L, final concentration 2 mg/L) and 1.6 mL of tbutyl methyl ether. 3. Vortex thoroughly and then mix for at least 10 min on a rotary mixer. 4. Centrifuge to separate the phases at 800g for 5 min. 5. Remove about 1 mL of the ethereal layer and transfer to an autosampler vial containing approximately 20 mg of anhydrous sodium sulfate to dry the extract (take care that none of the lower aqueous layer is transferred). 6. Examine 2 mL of this extract by GC using a cross-linked 5% phenylmethylsilicone-fused silica column (12.5 m 0.32 mm i.d., 0.52 mm) using nitrogen–phosphorus detection. The use of sodium chloride to increase the ionic strength of the aqueous phase increases the extraction of many of the compounds into the ether, which obviates the need for solvent evaporation. Although
n n
Column: methylsilicone fused silica (25 m 0.2 mm i.d., 0.11 mm), connected to a mass spectrometer. Temperature programme: 180 C for 1 min to 280 C at 8 /min. Carrier gas: He.
Diuretics
1. Take an aliquot (2.0 mL) of each urine sample in a 10 mL centrifuge tube. 2. Add 100 mL mefruside internal standard (10 mg/mL) and 2 mL 0.1 mol/L acetate buffer (pH 5.2). 3. Vortex thoroughly for 30 s. 4. Decant the urine onto a cross-linked polymeric sorbent cartridge with hydrophilic and lipophilic moieties (such as Abselut Nexus) and allow it to pass through the column. Do not let the bed dry out. 5. Add 1 mL of purified water onto each cartridge and allow it to pass through the column. Do not let the bed dry out. 6. Add 1 mL of methanol–water (20 : 80) onto each cartridge and allow it to pass through the column. Apply vacuum to the column to dry the bed. 7. Release the vacuum and place a labelled 10 mL glass centrifuge tube under the cartridge to collect the sample eluent. 8. Elute the adsorbed compounds with 3 mL methanol under gravity and then with the aid of a vacuum for a minimum of 30 s to dry the bed and achieve maximum recovery. 9. Evaporate the methanolic solution to dryness using oxygen-free nitrogen at 60 C. 10. Reconstitute the samples by adding 200 mL of LC mobile phase. 11. Vortex; transfer the reconstituted extract to a 0.2 mL tapered autosampler vial and cap the vial securely. 12. Examine 10 mL of the solution by LC-MS(-MS) for diuretics.
Gas chromatography and gas chromatography–mass spectrometry The GC procedures referred to above detect a wide range of compounds in urine samples, at concentrations in the order of 0.1 mg/L. They depend on the fact that all the compounds of interest contain at least one nitrogen atom and produce a signal in an alkali flame-ionisation
Clarke's Analysis of Drugs and Poisons Chapter No. 6 Dated: 15/3/2011 At Time: 11:51:40
Methods detector. The methods of extraction and the selectivity of the detector ensure minimal interference from other compounds that do not contain nitrogen, although certain plasticisers that contain phosphorus, such as tributyl phosphate, may produce signals. In addition, other nitrogencontaining compounds that are not prohibited in human sport (e.g. antihistamines) produce interfering peaks. Identification is based on the retention index; alternatively, retention time (relative to a standard) may be used. Details of retention indices or relative retention times of compounds in the systems described below are given in Chapter 40 and in the index of Gas Chromatographic Data. The identity of a substance should be confirmed using derivative formation and GC-MS, comparing the data obtained with reference material analysed contemporaneously (see Criteria for identification below).
Liquid chromatography–mass spectrometry With the development of atmospheric-pressure ionisation (API) techniques, LC-MS has found increasing application in doping control for both the qualitative and quantitative analysis of drugs. Screening methods have been developed for corticosteroids (Deventer, Delbeke 2003; Mazzarino, Botre 2006) and diuretics (Deventer et al. 2002; Ventura et al. 2008). In human sports drug testing, LC-MS(-MS) has been used more frequently over the last 10 years. For example, Barro´n et al. (1996) developed a direct method to determine anabolic steroids in human urine by on-line SPE LC-MS with a particle beam interface. Bean and Henion showed that it was possible to determine intact, i.e. conjugated, anabolic steroids using LC-electrospray ionisation (ESI)-MS(-MS) (Bean, Henion 1997). Thevis and colleagues used LC-MS(-MS) for the rapid screening of samples for beta-blockers (Thevis et al. 2001). Diuretics have been screened by LC-MS techniques (Ventura et al. 1991) and by LC-MS(-MS) (Thieme et al. 2001). Recently, the use of sub-2 mm porous particles to enable faster separations and greater separation power has become possible (Mazzeo et al. 2005). High-pressure pumping systems (greater than the conventional 400 bar [40 MPa] limit) needed to obtain the linear velocities desired are now readily available. Although the frictional heating caused by the velocity of the mobile phase through the column can limit the benefit that can be obtained using small particles, the use of 2.1 mm and narrower internal diameter columns has minimised this potential difficulty. Th€ orngren and colleagues have used ultra-performance liquid chromatography (UPLC)-MS(-MS) to screen 130 different substances (diuretics, masking agents, central nervous system stimulants and opiates) in urine (Th€ orngren et al. 2008) in approximately 6 min.
Isotope ratio mass spectrometry Combustion isotope ratio MS (CIRMS) is now used routinely by several WADA-accredited laboratories as an additional tool to help distinguish an individual whose testosterone : epitestosterone ratio may be naturally beyond the normal range from one who was administered testosterone. This technique relies on the fact that synthetic testosterone has a different proportion of 13C to the more abundant 12 C than the normal endogenous steroid (de la Torre et al. 2001). The extracted steroids are separated by GC and then converted into CO2 and the relative amounts of 12C to 13C as CO2 is determined for each eluting steroid in turn. Typically, the testosterone metabolites androsterone and etiocholanolone or androstanediols are monitored (Aguilera et al. 2000), or the metabolites 5a-androstanediol and 5bandrostanediol (Aguilera et al. 2001; Shackleton et al. 1997b), often comparing the results with pregnanediol or pregnanetriol as endogenous internal standards (Aguilera et al. 1999; Shackleton et al. 1997a). Flenker and colleagues and Cawley and colleagues have published reference isotope ratios for endogenous steroids (Cawley et al. 2009; Flenker et al. 2008) and Cawley has also published a method for the direct analysis of testosterone rather than its metabolites (Cawley
133
et al. 2009). Grosse and colleagues have shown that, in some urine samples, 19-norsteroids may be produced in small quantities from endogenous steroids (Grosse et al. 2005). Hebestreit and co-workers have published a method to distinguish this or the minute amounts that may be produced, especially in females, from the administration of nandrolone (Hebestreit et al. 2006). Buisson and colleagues (2009) have also used GC-CIRMS to detect exogenous hydrocortisone administration. Cawley and Flenker have published a tutorial article that reviews the use of CIRMS in doping control (Cawley, Flenker 2008).
Methods In human sports drug testing, MS is essential for the definite identification of a prohibited substance, with the exception of peptide hormones and glycoproteins, such as human chorionic gonadotrophin (hCG) for which a validated immunoassay is required for detection and quantification. For confirmation of hCG, a second different immunoassay is required. Specific techniques and methodologies for other peptide hormones and glycoproteins such as EPO have been described by WADA (WADA 2009a) and one for recombinant hCG (rhGH) is currently being considered. Criteria for identification WADA requires laboratories to be accredited to ISO 17025 to be eligible for WADA accreditation. ISO/IEC 17025 : 2005 requires traceability of measurements and for sports drug testing this is considered to be met by WADA, when identifying a prohibited substance (WADA 2003), by the direct comparison with a reference material or reference collection analysed in parallel or in series with the test sample. A reference material is generally accepted as a homogeneous, stable chemical with a wellestablished structure. The material may be characterised structurally within the laboratory using appropriate techniques or validated against a certified reference material or by comparison with uncontroversial published data. WADA permits the use of a reference collection ‘obtained from a verified administration study in which scientific documentation of the identity of metabolite(s) can be demonstrated’ (WADA 2009b). Certified reference materials issued by organisations accredited for compliance with ISO Guide 34 : 2000 are often not available and hence there is the need to use authenticated administrations for comparison purposes. Many of the most common substances are now available with certificates of analysis, and thereby fully meet the generic traceability requirements of ISO/IEC 17025 : 2005. This is particularly important for quantitative analysis, where it is usually extremely difficult for an individual laboratory to determine the purity of a non-certified material to a sufficient standard to be able to establish the measurement uncertainty. WADA generally expects a chromatographic retention time match between the analyte and a reference collection sample (see above) or reference material analysed using the same procedure in the same assay. This retention time difference must be not more than 1% or 0.2 min, whichever is the smaller, and three diagnostic ions in the electron impact (EI) and chemical ionisation (CI) mass spectra must also match to within 20% of the relative abundance of each ion (Table 6.8). Figure 6.3 illustrates the relative abundance criteria that, paradoxically, show a step change for the data obtained using CI, LC or tandem MS. These standards generally meet or exceed those required by the Substance Abuse and Mental Health Services Administration (SAMHSA) for the US federal employment drug testing programmes (see also Chapter 3). Although library data may be useful in the early phase of substance identification, especially in generic screening procedures, they are not considered sufficiently reliable for the final identification. Similarly, published data are used more to assure reliability than directly for substance identification. The required documentation for the analytical certificate is clearly set out in ISO/IEC 17025 : 2005 but merely requires a statement as to the substance found. However, WADA also sets out a
Clarke's Analysis of Drugs and Poisons Chapter No. 6 Dated: 15/3/2011 At Time: 11:51:40
134
Drug Testing in Human Sport
Table 6.8 Maximum relative ion intensity tolerances for substance identification using MS Relative abundance (% of base peak)
EI-GC-MS
CI-GC-MS; GC-MSn; LC-MS; LC-MSn 15% (absolute)
>50%
10% (absolute)
25–50%
20% (relative)
25% (relative)
20 000) and accurate mass LC-MS instrumentation is considered by many to offer the potential to address many of these issues. Screening based upon full-scan accurate mass LC-MS is potentially both broad ranging and highly selective and could replace more limited targeted screens such as those based upon triple-quadrupole systems. Inclusion of additional analytes is straightforward and, at least for the purpose of gathering information on previous use, retrospective. The use of chemometric tools either to directly detect novel agents or to identify metabolite patterns indicative of abuse may also offer a solution to designer drugs. A potentially more serious threat is posed by the switch to biological drugs and therapies emerging from the pharmaceutical industrial and medical research laboratories. Development of methods to detect the misuse of GH and EPO presented both the analytical laboratories and the sporting authorities with significant challenges. Rumours of the abuse of more exotic protein-based agents such as snake and cone snail venoms have circulated around the world, although seized materials claiming to contain these materials have often proved to be counterfeit. While these challenges are far from trivial, developments in protein MS, transcriptomics and metabolomics provide the promise that detection methods will become available, either directly or indirectly via biomarkers (Teale et al. 2009). It should also be noted that, for the most part, animal sport testing laboratories have the advantage of dealing with protein therapeutics that are clearly endogenous. For example, all the pharmaceutical preparations of EPO currently available are based upon human EPO, the primary structure of which is significantly different from equine or canine EPO.
Clarke's Analysis of Drugs and Poisons Chapter No. 7 Dated: 10/3/2011 At Time: 17:7:16
References A more difficult area to address is likely to be the use of therapies that do not directly involve administration of a drug. Examples of these include shock wave therapy and gene doping. Direct detection of these therapies may prove difficult, although significant research into the detection of gene doping has been undertaken and various targets for detection have been identified. A highly attractive possibility is the use of biomarkers. If successful, appropriate biomarkers could provide evidence of misuse of a wide range of agents and possibly replace traditional means of screening.
References Bailly-Chouriberry L et al. (2008). Detection of secondary biomarker of met-eGH as a strategy to screen for somatotropin misuse in horseracing. Analyst 133: 270–276. Bailly-Chouriberry L et al. (2008). Identification of recombinant equine growth hormone in horse plasma by LC-MS/MS: a confirmatory analysis in doping control. Anal Chem 80: 8340–8347. Biddle STB (2005). Monitoring the use of anabolic oestrus suppressants in the racing greyhound bitch: testosterone. In: Albert PH et al., eds. Proceedings of the 15th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket), 107–115. Biddle STB (2008). Metabolism of methyltestosterone in the greyhound. Rapid Commun Mass Spectrom 2009; 23: 713–721. Birks EK (2007). Anti-rhEPO antibodies in racehorses: defining the problem. In: Houghton E et al., eds. Proceedings of the 16th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 409. Bobin S et al. (2001). Approach to the determination of insulin-like-growth-factor-I (IGF-I) concentration in plasma by high-performance liquid chromatographyion trap mass spectrometry: use of a deconvolution algorithm for the quantification of multiprotonated molecules in electrospray ionization. Analyst 126: 1996–2001. Boyer S et al. (2007). Detection of testosterone propionate administration in horse hair samples. J Chromatogr B Analyt Technol Biomed Life Sci 852: 684–688. Brockwell M et al. (1992). The identification of the metabolites of testosterone, 19nortestosterone and 1-dehydrotestosterone in greyhound urine. In: Short CR, ed. Proceedings of the 9th International Conference of Racing Analysts and Veterinarians. Baton Rouge, LA: Dupre’s Printing, Copying, 57–68. Carter WG (2001). Medication violations and penalties for RCI Class 1, 2 and 3 foreign substances: a preliminary report. In: Williams RB et al., eds. Proceedings of the 13th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 303–309. Chakraborty J et al. (1976). A radioimmunoassay method for prednisolone: comparison with the competitive protein binding method. Br. J Clin Pharmacol 3: 903–906. De Kock SS et al. (2001a). Growth hormone abuse in the horse: establishment of an insulin-like growth factor base. In: Williams RB et al., eds. Proceedings of the 13th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 94–97. De Kock SS et al. (2001b). Growth hormone abuse in the horse: preliminary assessment of a mass spectrometric procedure for IGF-1 identification and quantitation. Rapid Commun Mass Spectrom 15: 1191–1197. De Kock SS et al. (2001). Administration of bovine, porcine and equine growth hormone to the horse: effect on insulin-like growth factor-I and selected IGF binding proteins. J Endocrinol 171: 163–171. Dumasia MC et al. (1973). Production and properties of antisera to dexamethasone-protein conjugates. Biochem J 133: 401–404. Dumasia MC, Houghton E (1981). Studies related to the metabolism of anabolic steroids in the horse – the identification of some 16-oxygenated metabolites of testosterone and a study of Phase II metabolism. Xenobiotica 11: 323–331. Dumasia MC, Houghton E (1984). Studies related to the metabolism of anabolic steroids in the horse: the phase I and phase II biotransformation of 19-nortestosterone in the equine castrate. Xenobiotica 14: 647–655. Dumasia MC, Houghton E (1991). Screening and confirmatory analysis of betaagonists, beta-antagonists and their metabolites in horse urine by capillary gas chromatography–mass spectrometry. J Chromatogr 564: 503–513. Dumasia MC et al. (1983). Studies related to the metabolism of anabolic steroids in the horse: the metabolism of 1-dehydrotestosterone and the use of fast atom bombardment mass spectrometry in the identification of steroid conjugates. Biomed Mass Spectrom 10: 434–440. Dumasia MC et al. (1986). Development of a gas chromatographic-mass spectrometric method using multiple analytes for the confirmatory analysis of anabolic steroids in horse urine. I. Detection of testosterone phenylpropionate administrations to equine male castrates. J Chromatogr 377: 23–33. Dumasia MC et al. (1996). LC/MS analysis of intact steroid conjugates: a preliminary study on the quantification of testosterone sulfate in equine urine. In: Auer DE, Houghton E, eds. Proceedings of the 11th International Conference of Racing
145
Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 188–194. Eenoo PV et al. (2001). Screening for diuretics in urine by LC/MS. In: Williams RB et al., eds. Proceedings of the 13th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 214–221. Faustino-Kemp J et al. (2001). The use of IGF-1 as a marker for detecting administration of growth hormone. In: Williams RB et al., eds. Proceedings of the 13th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 321–323. Guan F et al. (2003). Sensitive liquid chromatographic/tandem mass spectrometric method for the determination of beclomethasone dipropionate and its metabolites in equine plasma and urine. J Mass Spectrom 38: 823–838. Guan F et al. (2005). Detection, quantification and confirmation of anabolic steroids in equine plasma by liquid chromatography and tandem mass spectrometry. J. Chromatogr B Analyt Technol Biomed Life Sci 829: 56–68. Guan F et al. (2007). LC-MS/MS method for confirmation of recombinant human erythropoietin and darbepoetin alpha in equine plasma. Anal Chem 79: 4627–4635. Guan F et al. (2008). Differentiation and identification of recombinant human erythropoietin and darbepoetin alfa in equine plasma by LC-MS/MS for doping control. Anal Chem 80: 3811–3817. Harkins JD et al. (1996). Determination of highest no effect dose (HNED) for local anaesthetic responses to procaine, cocaine, bupivacaine and benzocaine. Equine Vet J 28: 30–37. Henion JD, Maylin GA (1980). Qualitative and quantitative analysis of hydrochlorothiazide in equine plasma and urine by high-performance liquid chromatography. J Anal Toxicol 4: 185–191. Ho EN et al. (2005). Metabolic studies of methenolone acetate in horses. Anal Chim Acta 540: 111–119. Ho EN et al. (2004). Detection of endogenous boldenone in the entire male horses. J Chromatogr B Analyt Technol Biomed Life Sci 808: 287–294. Ho EN et al. (2006). Comprehensive screening of anabolic steroids, corticosteroids, and acidic drugs in horse urine by solid-phase extraction and liquid chromatography-mass spectrometry. J Chromatogr A 1120: 38–53. Ho EN et al. (2007a). Metabolic studies of turinabol in horses. Anal Chim Acta 586: 208–216. Ho EN et al. (2007b). Metabolic studies of mesterolone in horses. Anal Chim Acta 596: 149–155. Houghton E (1977). Studies related to the metabolism of anabolic steroids in the horse: 19-nortestosterone. Xenobiotica 7: 683–693. Houghton E, Dumasia MC (1979). Studies related to the metabolism of anabolic steroids in the horse: testosterone. Xenobiotica 9: 269–279. Houghton E, Dumasia MC (1980). Studies related to the metabolism of anabolic steroids in the horse: the identification of some 16-oxygenated metabolites of 19-nortestosterone. Xenobiotica 10: 381–390. Houghton E (1981). The use of combined high performance liquid chromatography negative ion chemical ionization mass spectrometry to confirm the administration of synthetic corticosteroids to horses. Biomed Mass Spectrom 8: 558–564. Houghton E et al. (1982). The use of capillary column gas chromatography and negative ion chemical ionization mass spectrometry to confirm the administration of synthetic corticosteroids to horses. Biomed Mass Spectrom 9: 459–465. Houghton E et al. (1984). The identification of C-18 neutral steroids in normal stallion urine. Biomed Mass Spectrom 11: 96–99. Houghton E et al. (1986). Development of a gas chromatographic-mass spectrometric method using multiple analytes for the confirmatory analysis of anabolic steroid residues in horse urine. II. Detection of administration of 19-nortestosterone phenylpropionate to equine male castrates and fillies. J Chromatogr 383: 1–8. Howitt RG et al. (2007). Routine drug screening by accurate mass using liquid chromatography/time of flight mass spectrometry. In: Houghton E et al., eds. Proceedings of the 16th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 222–230. Hudson S et al. (2007). Can multiple instrumental analyses be replaced by a single analysis?. In: Houghton E et al., eds. Proceedings of the 16th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 534–538. IFHA (2009). International Agreement on Breeding and Racing. Boulogne: IFHA. www.horseracingintfed.com/resources/2009_choose_eng.pdf. ILAC (1997). Accreditation Requirements and Operating Criteria for Horseracing Laboratories, ILAC G-7. Rhodes, NSW: International Laboratory Accreditation Cooperation. www.ilac.org/documents/ILAC_G7_06_2009.pdf. ISO (2003). ISO 17025. General Requirements for the Competence of Calibration and Testing Laboratories. Geneva: ISO. Jondorf WR (1977). 19-Nortestosterone, a model for the use of anabolic steroid conjugates in raising antibodies for radioimmunoassay. Xenobiotica 7: 671–681. Jondorf WR, Moss MS (1977). On the detectability of anabolic steroids in horse urine (proceedings). Br J Pharmacol 60: 297P–298P. Jondorf WR, Moss MS (1978). Radioimmunoassay technique for detecting urinary excretion products after administration of synthetic anabolic steroids to the horse. Xenobiotica 8: 197–206.
Clarke's Analysis of Drugs and Poisons Chapter No. 7 Dated: 10/3/2011 At Time: 17:7:16
146
Drug Testing in Animal Sport
Jouvel C et al. (2000). Detection of diazepam in horse hair samples by mass spectrometric methods. Analyst 125: 1765–1769. Kim JY et al. (2000). Measurement of 19-nortestosterone and its esters in equine plasma by high-performance liquid chromatography with tandem mass spectrometry. Rapid Commun. Mass Spectrom 14: 1835–1840. Koupai-Abyazani MR et al. (1994a). Identification of levodopa and its metabolites in equine biological fluids by liquid chromatography-atmospheric pressure ionisation mass spectrometry. In: Kallings P et al., eds. Proceedings of the 10th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 123–126. Koupai-Abyazani MR et al. (1994b). Determination of triamcinolone acetonide in equine serum and urine by liquid chromatography-atmospheric pressure ionisation mass spectrometry. In: Kallings P et al., eds. Proceedings of the 10th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 209–213. Lampinen-Salomonsson M et al. (2006). Detection of altrenogest and its metabolites in post administration horse urine using liquid chromatography tandem mass spectrometry--increased sensitivity by chemical derivatisation of the glucuronic acid conjugate. J Chromatogr B Analyt Technol Biomed Life Sci 833: 245–256. Lasne F, de Ceaurriz J (2000). Recombinant erythropoietin in urine. Nature 405: 635. Lasne F et al. (2005). Detection of recombinant epoetin and darbepoetin alpha after subcutaneous administration in the horse. J Anal Toxicol 29: 835–837. Matassa LC et al. (1992). Solid-phase extraction techniques for the determination of glycopyrrolate from equine urine by liquid chromatography-tandem mass spectrometry and gas chromatography–mass spectrometry. J Chromatogr 573: 43–48. McKeever KH et al. (1996). Erythropoietin: a new form of blood doping in horses. In: Auer DE, Houghton E, eds. Proceedings of the 11th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 79–84. McKinney AR et al. (2001). Metabolism of methandrostenolone in the horse: a gas chromatographic–mass spectrometric investigation of phase I and phase II metabolism. J Chromatogr B Biomed Sci Appl 765: 71–79. McKinney AR et al. (2004). Detection of stanozolol and its metabolites in equine urine by liquid chromatography-electrospray ionization ion trap mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 811: 75–83. McKinney AR (2009). Modern techniques for the determination of anabolic-androgenic steroid doping in the horse. Bioanalysis 1: 785–803. Noble GK, Sillence MN (2001). The potential of mediator hormones as markers of growth hormone abuse in racehorses. In: Williams RB et al., eds. Proceedings of the 13th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 88–90. Pompa G et al. (1994). Prolonged presence of isoxsuprine in equine serum after oral administration. Xenobiotica 24: 339–346. Popot MA et al. (2001a). Detection of equine recombinant growth hormone administration in the horse. In: Williams RB et al., eds. Proceedings of the 13th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 98–104. Popot MA et al. (2001b). Determination of clenbuterol in horse hair by gas chromatography–tandem mass spectrometry. Chromatographia 53: S375–S379. Popot MA et al. (2001c). High performance liquid chromatography-ion trap mass spectrometry for the determination of insulin-like growth factor-I in horse plasma. Chromatographia 54: 737–741. Popot MA et al. (2001d). IGF-I plasma concentrations in non-treated horses and horses administered with methionyl equine somatotropin. Res Vet Sci 71: 167–173. Popot MA et al. (2008). Determination of IGF-I in horse plasma by LC electrospray ionisation mass spectrometry. Anal Bioanal Chem 390: 1843–1852. Ralston JM et al. (1992). Detection of 19-nortestosterone in equine and greyhound urine. In: Short CR, ed. Proceedings of the 9th International Conference of Racing Analysts and Veterinarians. Baton Rouge, LA: Dupre’s Printing, Copying, 69–73. Roberts J et al. (2003). Evaluation of commercial ELISA kits to detect the administartion of human erythropoietin (rhuEPO) to horses. In: Hill D, Hill WT, eds. Proceedings of the 14th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 234–242. RCI (2002). Uniform Classification Guidelines for Foreign Substances and Recommended Penalties and Model Rules. Lexington, KY: RCI. Russell C, Maynard S (2001). Environmental contamination with isoxsuprine. In: Williams RB et al., eds. Proceedings of the 13th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 381–383. Ryan M et al. (1996). Detection and confirmation of clidinium bromide in equine urine using LC/MS/MS and GC/MS techniques. In: Auer DE, Houghton E, eds. Proceedings of the 11th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 488–493.
Samuels T et al. (1994). Applications of bench-top LC/MS to drug analysis in the horse: I. Development of a quantitative method for urinary hydrocortisone. In: Kallings P et al., eds. Proceedings of the 10th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 115–118. Scarth J et al. (2009). Presence and metabolism of endogenous androgenic-anabolic steroid hormones in meat-producing animals. Food Addit Contam Part A Chem Anal Control Expo, Risk Assess 26: 640–671. Schoene C et al. (1994). Preliminary study of the metabolism of 17 alpha-methyltestosterone in horses utilizing gas chromatography–mass spectrometric techniques. Analyst 119: 2537–2542. Shackleton CH, Whitney JO (1980). Use of Sep-pak cartridges for urinary steroid extraction: evaluation of the method for use prior to gas chromatographic analysis. Clin Chim Acta 107: 231–243. Smith RL (2001). The zero tolerance approach to doping control in horseracing: a fading illusion. In: Williams RB et al., eds. Proceedings of the 13th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 9–14. Stanley SM et al. (1995). Detection of flunixin in equine urine using high-performance liquid chromatography with particle beam and atmospheric pressure ionization mass spectrometry after solid-phase extraction. J Chromatogr B Biomed Appl 667: 95–103. Stanley SD et al. (2001). Unique functionalised polymeric columns for solid phase extraction. In: Williams RB et al., eds. Proceedings of the 13th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 241–244. Stevenson AJ (1995). The Canadian approach: limitations on analytical methodology In: Testing for Therapeutic Medications, Environmental and Dietary Substances in Racing Horses. Maxwell H Gluck Equine Research Center, Lexington, Kentucky. Stewart RT et al. (2009). Metabolism of stanozolol: chemical synthesis and identification of a major canine urinary metabolite by liquid chromatography– electrospray ionisation ion trap mass spectrometry. J Steroid Biochem Mol Biol 117: 152–158. Tang PW et al. (2001). Analysis of corticosteroids in equine urine by liquid chromatography-mass spectrometry. J Chromatogr B Biomed Sci Appl 754: 229–244. Tay S et al. (1996). Evaluation of ELISA tests for erythropoietin (EPO) detection. In: Auer DE, Houghton E, eds. Proceedings of the 11th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 410–414. Teale P, Houghton E (2010). Metabolism of anabolic steroids and their relevance to drug detection in horseracing. Bioanalysis (in press). Teale P, Houghton E (1991). The development of a gas chromatographic/mass spectrometric screening procedure to detect the administration of anabolic steroids to the horse. Biol Mass Spectrom 20: 109–114. Teal P et al. (2009). Biomarkars: unrealized potential in sports doping analysis. Bioanalysis 1: 1103–1118. Tobin T et al. (1999). Testing for therapeutic medications: analytical/pharmacological relationships and limitations on the sensitivity of testing for certain agents. J Vet Pharmacol Ther 22: 220–233. Whittem Tet al. (1998). Detection of morphine in mane hair of horses. Aust Vet J 76: 426–427. Williams TM et al. (2000). Characterization of urinary metabolites of testosterone, methyltestosterone, mibolerone and boldebone in greyhound dogs. J Vet Pharmacol Ther 23: 121–129. Woods WE et al. (1988). Immunoassay detection of drugs in racing horses. VI. Detection of furosemide (Lasix) in equine blood by a one step ELISA and PCFIA. Res Commun Chem Pathol Pharmacol 61: 111–128. Woodward K et al. (2003). The application of APCI LC-MS to equine sport testing. In: Hill D, Hill WT, eds. Proceedings of the 14th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 215–220. Wynne PM (2000). The application of SPE to veterinary drug abuse. In: Simpson MJK, ed. Solid Phase Extraction. New York: Marcel Dekker, 273–306. Wynne PM et al. (2001a). An improved method for the automated extraction of anabolic steroids from equine urine. In: Williams RB et al., eds. Proceedings of the 13th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 245–251. Wynne PM et al. (2001b). Reduced blocking rates through application of a new NCSPE sorbent to the extraction of equine urine. In: Williams RB et al., eds. Proceedings of the 13th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 445–452. Wynne PM et al. (2004). Approaches to the solid-phase extraction of equine urine. Chromatographia 59: S51–S60. Yiu KCH et al. (2003). Detection of basic drugs in equine urine by liquid chromatography–mass spectrometry. In: Hill D, Hill WT, eds. Proceedings of the 14th International Conference of Racing Analysts and Veterinarians. Newmarket: RW Publications (Newmarket) Ltd, 154–161.
Clarke's Analysis of Drugs and Poisons Chapter No. 8 Dated: 10/3/2011 At Time: 17:12:0
CHAPTER
8
Drug-facilitated Sexual Assault MD Osselton
Drug-facilitated assault (DFA) has emerged as a major area of forensic work during recent years as a consequence of media publicity attributed to a handful of drugs such as flunitrazepam, gamma-hydroxybutyrate (GHB) and ketamine. Although drug-facilitated sexual assault (DFSA) has attracted much attention, drug-related assault may also be encountered in cases of robbery or uncharacteristic and antisocial behaviour by an individual. DFSA has been loosely described by the popular media as ‘date rape’ and linked with drink spiking, particularly in club venues. Despite media publicity, drink-spiking cases probably make up only a small percentage of DFSA cases and the majority of cases are associated with recreational drug and alcohol use by the victim. In a typical drinkspiking scenario, a potential sexual offender surreptitiously spikes the drink of an unsuspecting person with a sedative substance for the purpose of ‘drugging’ and subsequently sexually assaults the victim while he or she is under the influence of this substance. Victims often report loss of memory during and after these incidents and may wake up in unfamiliar places, inappropriately dressed and often with the sense but not the actual recollection of having had sex. DFSA is not limited to male assaults on females but may involve female assault on males as well as homosexual assault. DFSA is not a new crime and appears to be documented in the Bible where Lot’s daughters are recorded to have used wine to drug him before having sexual intercourse with him in order to procreate. Both daughters became pregnant and subsequently produced sons Moab and Benammi (Genesis 19 : 30-38). This chapter will consider the terminology used in association with drug-related assault, the types of substances most frequently encountered, and the approaches that may be taken when investigating such cases.
Definitions ‘Assault’ may be defined as ‘an intentional act by one person that creates an apprehension in another of an imminent harmful or offensive contact’. In the UK the Sexual Offences Act 2003 came into force in May 2004 and essentially repealed most of the existing statute law relating to sexual offences. Under the auspices of the UK Sexual Offences Act 2003 a person A is guilty of a sexual assault if: (i) he/she intentionally touches another person (B); (ii) the touching is sexual; (iii) B does not consent to the touching; and (iv) A does not reasonably believe that B consents. It is up to the court to decide whether ‘belief’ is reasonable after having regard to all the circumstances, including any steps A has taken to ascertain whether B consents. This law covers any kind of intentional sexual touching of another person without their consent. It includes touching any part of their body, clothed or unclothed, either with the body or with an object (www.homeoffice.gov.uk/crime/sexualoffences/ legislation/act.html). The Act therefore makes it an offence for any male or female to intentionally touch another person sexually without his or her consent. Under the same Act, rape occurs if a male penetrates with his penis the vagina, or the anus or mouth of a female or male without their consent. The issue of consent is therefore a key issue. The Sexual Offences Act 2003 for the first time in UK law provides a clear definition of consent, making it easier for courts to decide upon evidence put before them. Section 74 of the Act defines consent as follows: ‘A person consents if he/she agrees by choice and has the freedom and capacity to make that choice.’
Date rape, also referred to as ‘acquaintance rape’, defines a sexual assault/rape or attempted assault/rape by an acquaintance who may have been previously known to the victim or whom the victim has met socially for the first time. Although the terms ‘date rape’ and ‘drug-assisted rape’ have been used interchangeably, the two should not be confused. Drug-facilitated sexual assault occurs when a person is subjected to non-consensual sexual acts while their capacity to provide consent is impaired (or they are unconscious) owing to the effect(s) of ethanol, a drug and/or other intoxicating substance, and are therefore prevented from resisting and/or unable to consent. The drug can be any substance that induces changes to the physical state of the intended victim or that exerts mind-altering properties. While UK law has been cited above as an example of legislation, the definitions of rape, sexual assault and drug-facilitated sexual assault are not dissimilar in other countries. In the USA, sexual assault may be referred to in different terms in the laws of different states (sexual battery, criminal sexual assault, rape, etc.); however, the underlying principles of law remain essentially the same.
Frequency/statistics of DFSA The extent of DFSA cases has not been officially recorded and only estimates of its prevalence are available in the literature. The extent of DFSA is difficult to gauge because some incidents are not reported to the police and others are not reported within a time frame that enables drug exposure to be identified by the analysis of blood or urine specimens. The European Monitoring Centre for Drugs and Drug Addiction stated that between 1997 and 2007 there had been a rise in the number of reports in which drugs and alcohol had been used to immobilise victims for the purposes of sexual assault (www.emcdda.europa.eu). It further reported that surveys in six European Union countries suggested that up to 20% of women had experienced some form of sexual assault in their adult lifetime. In 2001 the British Crime Survey estimated the total number of rapes of females to be between 11 000 and 39 000 (Walby, Allen 2004); however, the proportion of cases in which drugs were implicated is unknown. Figures taken from the British Crime Survey 2010 relating to male and female rape and sexual assault between 2004/ 2005 and 2009/2010 are summarised in Table 8.1, which shows an apparent decrease in the number of cases. The way the statistics have been collected has changed, however, and therefore the latest results are not directly comparable with those of earlier years. As with the earlier reported figures by Walby and Allen, there is no indication in the British Crime Survey 2010 of what proportion of cases were implicated as involving drug use. It is notable that during the same period the number of drug-related offences rose by 62% from 145 837 to 234 998. Only a small number of studies have been published in the scientific literature that provide information on the extent of alcohol and drug use in DFSA cases. For example, the number of reported sexual assaults in the USA according to the FBI Uniform Crime Report is presented in Table 8.2. Both the number and the rate of reported rapes declined throughout most of the 1990s, reaching a low in 1999. It is also well known that the number of reported rapes is significantly lower than the actual number. In 1999, for example, the Bureau of Justice Statistics estimated that there were over 141 000 cases of sexual assault, 58% more than the number actually reported to the police.
147
Clarke's Analysis of Drugs and Poisons Chapter No. 8 Dated: 10/3/2011 At Time: 17:12:0
148
Drug-facilitated Sexual Assault
Table 8.1 Rapes and sexual assaults reported between 2005 and 2010, abstracted from the British Crime Survey 2010 Year
Rape – female over 16 years
Rape – female under 16 years
Sexual assault on females over 13 years
Rape – males over 16 years
2005/2006
8786
3153
17 158
460
2006/2007
8247
2853
16 883
431
2007/2008
7731
2413
15 780
344
2008/2009
7950
2538
15 503
339
2009/2010
9102
2926
15 713
372
Elsohly and Salamone (1999) surveyed 1179 urine samples from suspected DFSA victims collected from 49 states, Puerto Rico and the District of Columbia over a 26-month period. The urine specimens were screened using immunoassay for amfetamines, barbiturates, benzodiazepines, cannabinoids, cocaine metabolite (benzoylecgonine), methaqualone, opiates, phencyclidine and propoxyphene. In addition, samples were screened for flunitrazepam metabolites and GHB by gas chromatography–mass spectrometry and for ethanol by gas chromatography using a flame ionisation detector. No drug substances were detected in 468 of the samples; 451 specimens tested positive for ethanol, 218 for cannabinoids, 97 for benzoylecgonine, 97 for benzodiazepines, 51 for amfetamines, 48 for GHB, 25 for opiates and 12 for barbiturates. Of the samples shown to contain drugs, 35% contained multiple drugs. A similar study undertaken in the UK analysed the toxicology results from 1014 cases submitted to the Forensic Science Service London laboratory between January 2000 and December 2002 (Scott-Ham, Burton 2005). Alcohol was reported to be present in 470 of all cases (46%) either alone or in combination with drugs. Drugs of abuse were detected in 344 cases (34%), with cannabis being the most commonly detected in 260 cases (26%), cocaine in 110 cases (11%), MDMA in 47 cases (5%), amfetamine in 23 cases (2%), diamorphine in 12 cases (1%) and ketamine in 3 cases (0.5%). Potentially stupefying drugs were detected in 187 cases (18%), although in the vast majority of these cases the drugs had been taken therapeutically by the victim or taken after the incident. Only 21 cases (2%) were attributed to involuntary ingestion, i.e. as a result of deliberate drink spiking. During the 3-year period of the survey no evidence for the use of flunitrazepam was observed. As in the ElSohly study, only a few cases were encountered in which a sedative drug was detected and its presence could not be attributed to voluntary use
by the complainant. Scott-Ham and Burton further analysed the blood and urine alcohol concentrations in cases of alleged DFSA (Scott-Ham, Burton 2006). Of the 1041 cases scrutinised, 391 had blood and/or urine samples collected within 12 h of an alleged incident. The authors backcalculated the alcohol concentrations in these 391 samples to estimate the possible alcohol concentration at the time of the alleged incident. Following back-calculation, 60% of cases were estimated to have a blood alcohol concentration in excess of 150 mg alcohol per 100 mL of blood at the time of the alleged incident. Hall et al. (2008) surveyed blood alcohol concentrations (BACs) determined in cases of alleged DFSA in Northern Ireland between 1999 and 2005 and reported that the estimated average BAC (218 mg/100 mL) remained broadly similar during the period of the study. Jones et al. (2008) analysed blood and urine specimens from 1806 female victims of alleged non-consensual sexual activity. No alcohol or drugs were reported to be present in 559 cases (31%) and ethanol alone was detected in 772 cases (43%). In 215 cases (12%) ethanol occurred together with at least one other drug. The mean, median and highest concentrations of ethanol in blood (N ¼ 806) were 124, 119 and 370 mg/100 mL, respectively. Amfetamine and cannabinoids were reported to be the most common illicit drugs detected. Elliott and Burgess (2005) reported on 169 clinical requests relating to surreptitious drug administration over a 2-year period between 2002 and 2004. Approximately half of the cases analysed were negative for alcohol or drugs, but in the cases that were positive alcohol and common drugs of abuse were found to be present. Neither GHB nor flunitrazepam was detected in any of the cases. These studies indicate that the patterns of drug and alcohol use in DFSA cases is similar in different countries and that alcohol is the most frequently encountered substance in these cases.
Toxicological approach to DFSA Table 8.2 Number of reported sexual assaults – FBI Uniform Crime Report Year
Number of sexual assaults
Rate per 100 000
1990
102 560
41.2
1991
106 590
42.3
1992(a)
109 060
42.8
1993
106 010
41.1
1994
102 220
39.3
1995
97 460
37.1
1996
96 252
36.3
1997
96 153
35.9
1998
96 144
34.5
(b)
1999
89 411
32.8
2000
90 178
32.0
2001
90 863
31.8
2002
95 136
33.0
2003
93 433
32.1
(a) (b)
The highest. The lowest.
Delay in collecting samples can be a critical factor for drugs that exhibit short elimination half-lives, e.g. GHB, as these may not be detectable at concentrations above endogenous levels after around 10–12 h of administration. In the UK, the Association of Chief Police Officers (ACPO) produced the report Operation MATISSE – Investigating drug facilitated sexual assault (Gee et al. 2006), which assessed the time periods between alleged incidents and the collection of urine samples. They found that, out of a total of 117 samples collected, 25.6% of samples had been collected within the first 6 h, 47.8% within 11.59 h, 84.5% within 23.59 h and 94.8% within 47.59 h. It is essential that specimens for analysis should be collected as soon as possible after the victim becomes aware that a drug-related sexual assault has taken place. This of course depends on the victim reporting the incident to the appropriate authorities and is outside the toxicologist’s control. Laboratories can, however, take a proactive role in working with their local authorities, colleges, health departments, police forces and forensic medical examiners to make them aware of the need to educate the population at large to report incidents and of the need to collect specimens as soon as possible after an alleged offence is suspected to have taken place. Urine specimens can be collected without the need for trained medical personnel to be present and without the need for observed supervised collection, thus facilitating the obtaining of an early evidence specimen while waiting for medical examiners to arrive and collect blood specimens. In order
Clarke's Analysis of Drugs and Poisons Chapter No. 8 Dated: 10/3/2011 At Time: 17:12:0
Toxicological approach to DFSA • • • • • • • • • • • • • • • • • • • • •
• • •
• • • • •
149
Name of subject Age Sex Weight Height Build (proportional/stocky or muscular/obese/slim etc.) Date of incident Time of incident Date samples taken Time samples taken Occupation of complainant Name and contact details of investigating officer Name and contact details of the doctor that examined the victim Details of the incident – full case history What led the victim to suspect that a DFSA offence had taken place? Did the victim fall unconscious? If so provide approximate times, e.g. how long was he/she unconscious? Does the victim suffer from any medical condition requiring treatment from a physician? Does the victim suffer from any medical condition, e.g. diabetes, depression? Did the victim empty his/her bladder prior to reporting the incident? Had complainant taken any alcohol, drugs or medication within 48 hours prior to the incident? If so what? How much? and when? If alcohol was consumed in the period leading up to the incident was this more than would have normally been consumed? If yes give details of times and amounts consumed. Were alcohol or any drugs consumed after the incident? If yes what, how much and when? Were any symptoms experienced, e.g. drowsiness, dizziness, nausea, vomiting, impairment of memory, thirst, sweating or shivering, unusual taste, hallucinations? Were any of the following symptoms observed by police officers or the medical examiner? Drowsiness, poor coordination, unsteadiness, shivering, sweating, nystagmus, hyperactivity, abnormal pupil size Is the complainant a known drug user? If so obtain details Is the identity of the suspect known? If so have blood or urine specimens been obtained from the suspect for analysis? What is the occupation of the suspect? What are the hobbies of the suspect, e.g. making model aircraft Have any other exhibits been collected and submitted for analysis, e.g. cups, glasses, tablets, etc.?
Figure 8.1 Information required in cases of suspected drug-facilitated sexual assault.
to avoid irretrievable loss of forensic evidence, it is recommended that a urine specimen should be collected before the commencement of any interviews with the victim. At least 20–25 mL urine should be collected as soon as possible after an incident has been reported. Ideally the urine should contain sodium fluoride as preservative at a minimum final concentration of around 1.5% w/v. There have been anecdotal reports of concerns being expressed in some areas of the USA that supervised collection should be enforced, as in workplace drug testing, to ensure that alleged victims do not add drug substances to their urine specimens in order to make malicious accusations against an innocent party. As soon as the forensic medical examiner is available, a specimen of blood should be collected. The volume of blood collected should ideally be of a minimum volume of 10 mL and should be collected into a sealed glass container with a minimum final concentration of 1.5% w/v sodium fluoride as preservative. A second urine specimen (20 mL) should also be collected into a container with sodium fluoride preservative as above. All specimens should be clearly labelled with the name of the victim as well as the time and date that the specimen was collected. Specimen containers should not be over-filled. This avoids breakage if the sample is frozen at a later time. Once the early evidence specimens have been obtained, the case history and information regarding the
events and circumstances should be recorded. Many police forces and clinics will have their own protocols for collecting details of the case circumstances. However, it is important that the case history should be as detailed and comprehensive as possible. The information that should be collected is summarised in Figure 8.1. Information can provide the toxicologist with important clues as to what, or what not, to look for and in cases where limited sample volumes have been submitted for analysis can make the difference between detection of an incapacitating agent and a negative finding. In addition collection of a detailed case history and blood, hair and urine from the alleged victim, any suspected tablets, powders, drinks, containers or residues in cups, etc. should be collected and submitted for possible analysis. While this may be impracticable if the alleged drink spiking occurred in a bar, many alleged incidents take place in the home where exhibits are less likely to have been disposed of. The examination of drinking vessels may reveal drug or tablet residues. In cases where gelatin capsules have been added to hot drinks it is not uncommon to be able to see the melted capsule adhering to the base of the cup. If the subject has vomited and vomit stains are available, these may also be considered for analysis. Alleged incidents in public bars may be recorded on security video systems and examination of these can also provide useful evidence of an alleged incident.
Clarke's Analysis of Drugs and Poisons Chapter No. 8 Dated: 10/3/2011 At Time: 17:12:1
150
Drug-facilitated Sexual Assault
Samples and sample collection Blood, urine and hair are recommended for collection wherever possible, although each has its merits and limitations. Blood is the tissue of choice for analysis because it provides potentially interpretable results and helps place drug consumption within a fairly narrow time frame (up to 24–36 h depending on the drug consumed and the amount taken). Many drugs may be detected in blood for 24–36 h after consumption, although GHB is unlikely to be present in concentrations greater than endogenous concentrations approximately 6–8 h following consumption (Andresen et al. 2010). If it is suspected that GHB has been consumed, urine should be collected in addition to blood. Blood and urine should be stored in a refrigerator prior to submission to the forensic laboratory for analysis. As well as being easy to collect, urine is usually plentiful and easy to analyse, contains higher concentrations of drugs or their metabolites than blood, and provides a wider window of detection than blood. Depending on the drugs consumed, urine may enable the detection of drugs of abuse, benzodiazepines, flunitrazepam metabolites, trichlorinated compounds and many basic drugs for up to 2–3 days after consumption and hence is particularly useful in cases where a long delay in reporting an incident has occurred. The window of detection for GHB in urine, although greater than that of blood, is still short and is unlikely to yield meaningful results for this compound if more than 12 h has elapsed between ingestion and specimen collection. Alcohol may usefully be analysed in urine and used for back-calculations up to around 24 h after consumption but after this time the results are likely to provide only limited information. The major flunitrazepam metabolite (7-aminoflunitrazepam) can be detected in urine for up to 72 h following the ingestion of a single therapeutic dose (Negrusz et al. 2000; Snyder et al. 2001). Table 8.3 summarises the approximate detection times for alcohol and some common drugs in blood and urine after single or therapeutic doses. In cases where the time elapsed between an incident and reporting exceeds 24–48 h, hair analysis should be considered. Hair grows, on average, at a rate of approximately 1 cm per month and therefore a hair specimen should be collected at the time of the first examination and the victim advised to return to provide a second hair specimen after approximately 6 weeks. The first hair specimen taken after the incident may then be used as a control and any drugs taken around the time of the incident will hopefully show up in the second sample. Protocols for hair collection and analysis are provided in Chapter 19. If it is proposed to collect hair specimens, a clear explanation should be given to the alleged
Table 8.3 Approximate detection times for alcohol and common drugs following the consumption of single or therapeutic doses
victim of the potential outcomes in terms of the results of the analysis. The subject should be advised that where he or she is a voluntary drug user but has not declared the fact, the ramifications of this being disclosed in court could be potentially damaging.
Sample analysis Beverages, cups and tablets Commercial ‘Quick Test’ kits have been marketed for use by the public to test drinks for adulteration in situ in a nightclub scenario. Beynon et al. (2006) reported on the evaluation of two such kits – ‘Drink Guard’ and ‘Drink Detective’ – and concluded that ‘whilst drink spike testing kits may be a useful public health tool, neither kit demonstrated high levels of sensitivity, specificity or utility’ under the laboratory protocols used for the evaluation. They further stated that ‘in practice this could leave the public feeling falsely reassured or overplay the magnitude of drink spiking’. Liquids submitted for analysis should be carefully examined against control samples for signs of adulteration, which might be indicated by cloudiness, opacity, colour changes (the manufacturers of flunitrazepam incorporated a blue dye into the tablets to provide an indication of drink spiking), particles floating on the surface or residues in the bottom of the container. If a liquid preparation of an adulterant such as GHB is added to a beverage there may be no easily recognisable sign of adulteration. However, if a liquid herbal preparation is added, e.g. valerian tincture, distinct colour changes may be observed. Herbal preparations such as crushed herbal sleep aid tablets often yield floating debris that is easily recognisable. The addition of coloured tablets or capsules containing oily drug formulations may be indicated by a coloured scum or immiscible oily globules floating on the surface of the drink. Most tablets comprise bulking agents in addition to the active pharmaceutical ingredient and these may sometimes be left as visible residues in the bottom of a drinking container. Figures 8.2–8.5 illustrate, respectively, a crushed tablet showing parts of coating prior to addition to a beverage, an Arthrotec tablet partially dissolved in water, a residue in a tea cup following removal of the beverage by decanting and floating waxy particles following the addition of crushed tablet with wax coating. In some cases there may be sufficient residue present in a container that it can be scraped from the bottom and analysed directly using colour tests, ultraviolet spectrophotometry (UV), Fourier transform infrared spectrometry (FTIR), thin-layer chromatography (TLC), high performance liquid chromatography (HPLC) or gas chromatography– mass spectrometry (GC-MS). In cases where only a slight residue may be indicated, the receptacle should be washed with a small quantity of ethanol, which can be retrieved, evaporated and subsequently tested by immunoassay screening or instrumental analysis such as high
Detection time (h) Drug
Blood(a)
Urine(a)
Alcohol
8–10
24
Amfetamines
12
24–48
Barbiturates
24
242–48
Benzodiazepines (including metabolites)
48
48–72
Cannabinoids
4–12(b)
12–48
Cocaine
12
12
Benzoylecgonine
12–24
24–48
Flunitrazepam
12–24
48–72
GHB
1600
29
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:16
194
Drugs of Abuse
Screening tests Colour/spot tests provide a valuable indication of the content of any particular item tested, but it must be stressed that positive results to colour tests are only presumptive indications of the possible presence of the drug. Colour tests have the advantage that they can be used as field tests by unskilled operators, with the obvious need for follow-on analysis in the laboratory. One of the most important and widely used colour tests is the Marquis reagent test. Another important screen is thin-layer chromatography (TLC). This has many advantages as an analysis/screening tool. It is quick, easy to use and has a low cost; it is relatively sensitive and can give a good degree of discrimination. Solvent systems TA and TB are suitable for many drugs (see Table 11.4 and Chapter 39). Visualisation of many of the drugs may be achieved by a variety of methods. However, spraying with acidified potassium iodoplatinate reagent is suitable for many drugs. Qualitative analysis Using a capillary column gas chromatograph and a suitable temperature programme coupled to a mass spectrometer (GC-MS; see Chapter 37), the drug components of most samples can be separated and identified. Because of the reduced capital outlay of such instruments in recent times, it is not uncommon for laboratories to have several such instruments working with automatic samplers (possibly on a 24-hour basis). The use of GC-MS has become the routine method of identification of most drugs. A general GC-MS screen method (GAK, Table 11.4) can be used to separate and/or identify most of the drugs encountered in exhibits. Figure 11.2 shows the separation achieved, by this method, of a mixture of the main drugs described in this chapter. Identification of the various components of a suspect mixture can be made by library searching against commercial libraries but it is important to run a standard of the specific drug being tested, e.g. pure heroin. This obviously will need to give a retention time and mass-spectral match. High performance liquid chromatography (HPLC) is a simple and reliable method of analysis of most drugs. It is both accurate and precise and thus lends itself to quantitative analysis. It is especially useful for compounds that are thermally labile. HPLC has some advantages over GC because of the variety and combinations of mobile phases that can be chosen. There is also a choice of detectors available for specific applications. HPLC can, however, involve significantly more method development than GC and the resolution of GC is greater. No one system is suitable for optimum separation of all the different drug types, so different systems are used to give optimum separations for specific analysis. The system that is best for the separation of heroin/ acetylcodeine/noscapine/papaverine (Huizer 1983) will not be the same as that which separates cocaine from its impurities and processing byproducts (Moore, Casale 1994). A general screening method such as HBC (Table 11.4) can be used for the separation of heroin (diamorphine), cocaine, amfetamine and metamfetamine (Fig. 11.3). Using this system, metamfetamine and MDMA co-elute. However, by changing the method to HBD, amfetamine, metamfetamine, MDMA and methylenedioxyethamfetamine (MDEA) may be separated (Fig. 11.4). This illustrates the versatility of HPLC. Separations can occur between compounds that co-elute by altering the elution system. A system such as the above could be used as a screen, but identification would necessitate some spectroscopic method such as MS or infrared (IR) spectroscopy. Most modern laboratories are now equipped with Fourier transform IR (FTIR) spectrometers (see Chapter 33). These have many advantages over traditional IR instruments. They are faster and can work with smaller samples. When they are coupled with a microscope, tiny samples can be analysed. The difficulty with IR analysis of drug samples is the presence of other material that will interfere with the spectrum. These interfering compounds could be other drugs that occur naturally in the
samples (or from the synthetic process) or could be adulterants such as caffeine and paracetamol. IR analysis can, however, give valuable information on chemicals that are not suitable for GC-MS analysis. Another technique that is popular is GC-FTIR. Because of the speed of scanning of the FTIR it can be used to obtain a spectrum of the compounds that have been separated by GC. In practice, neither spectra nor pure reference samples may be available for comparison for the more unusual substances. In this situation, nuclear magnetic resonance (NMR) spectroscopy (see Chapter 36) is the method of choice. Quantitative analysis For most controlled drugs there are no minimum quantities below which an offence does not occur. The quantitative analysis of drugs is therefore not carried out routinely on all exhibits. The main reason for determining purity/drug content of powders and tablets is to enable a court to establish a monetary value of the seizure or when sentencing structures are based on equivalent pure drug content. In some countries the death sentence can apply if one is convicted of possession/supply of greater than a specified quantity of a substance. In some situations, information on drug purity will be used for intelligence purposes such as to assess trends in the illicit drug market or for use in drug comparison and profiling. Having already identified the powder as, for example, heroin, quantitative analysis may be carried out by GC using flame ionisation detection (FID) or by HPLC. In performing quantitative analysis it is always desirable to include an internal standard in the analysis. This has the advantages of ease of use, increased accuracy, no need for volumetric glassware, no need to measure injection volume and easy determination of reproducibility, and can be used as a monitor for GC or HPLC systems. An internal standard must meet the following criteria: it must be absent from the sample; it must be readily available (and not too costly); and it must be pure, show good chromatographic behaviour, be reproducible and be soluble in the solvent used. Straight-chain hydrocarbons (for GC) fulfil all of these requirements and they elute as a homologous series, so they are a popular choice as internal standard. It has been suggested that the internal standard that one chooses should be chemically related to the compound being analysed. However, provided that it fulfils the above criteria, any compound can be chosen. A general approach to quantification by GC could be as follows. The GC conditions for the qualitative analysis can again be used. A standard curve is established by preparing up to five standard solutions of the drug being quantified. A range from 1 mg/mL to 5 mg/mL is prepared using solvent containing an internal standard. A concentration of internal standard of 0.5 or 1 mg/mL will normally be adequate. A test sample is prepared that will have a concentration between 1 and 5 mg/mL, i.e. within the range of the standard curve. If the test sample is outside the range, a second sample is prepared based on the information from the first sample. In general it is suggested that at least two samples of the powder being tested be taken for quantitative analysis and an average of these be taken as the true result. The amount of the drug in the test sample can now generally be calculated by the data-analysis function of the instrument. Both GC and HPLC are used extensively for quantitative analysis and it is useful to compare the results obtained by one method with the other, for a given drug. Profiling and comparison A more detailed analysis of drug samples can be used to provide ‘collective’ information. This is generally called profiling when it involves the chemical analysis of powders, or is known as characterisation when the physical properties of tablets and other dosage forms are measured. Chemical profiling has been the technique most widely used and is often based on the chromatographic separation of impurities and precursors (as in the case of amfetamine and metamfetamine) or other naturally occurring components and adulterants (e.g. heroin, cocaine,
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:17
Analysis of seized drugs
195
Table 11.4 GC, HPLC and TLC conditions Condition code
Source
Details
SD McDermott (unpublished)
Column: HP Ultra-1 cross-linked methylsiloxane (12.5 m 0.2 mm i.d., 0.33 mm)
Gas chromatography GAK
Carrier gas: He, 1 mL/min, 50 : 1 split ratio Temperature programme: 60 C for 2 min to 180 C at 15 C/min to 290 C at 25 C/min for 3 min MS conditions: low mass 40, high mass 550 with a solvent delay of 1.5 min GAL
Clarke (1989)
Column: OV-1 (12 m 0.32 mm i.d., 0.52 mm) Carrier gas: He, 1.5 mL/min, 25 : 1 split ratio Temperature: 260 C
GAM
McDermott (unpublished)
Column: HP Ultra-1 (12 m 0.2 mm i.d., 0.33 mm), split ratio 50 : 1 Temperature programme: 150 C (no hold) to 295 C at 15 C/min
GAN
Lee (1995)
Column: HP-1 (12 m 0.22 mm i.d., 0.33 mm) Temperature programme: 100 C for 2 min to 300 C at 15 C/min for 5 min
GAO
Blackledge, Miller (1991)
Column: HP-1 (25 m 0.2 mm i.d., 0.33 mm) Temperature programme (lactone): 70 C for 1 min to 300 C at 20 C/min for 3 min Temperature programme (BSTFA derivative): 100 C for 1 min to 300 C at 20 C/min for 7 min
GAP
SD McDermott (unpublished)
Column: HP Ultra-1 cross-linked methyl siloxane (12.5 m 0.2 mm i.d., 0.33 mm) Carrier gas: He, 1 mL/min, 50 : 1 split ratio Temperature programme: 70 C for 2 min to 210 C at 20 C/min for 1 min MS conditions: low mass 40, high mass 550 with a solvent delay of 1.5 min
GAQ
SD McDermott (unpublished)
Column: HP 101 cross-linked methylsiloxane (12 m 0.2 mm i.d., 0.33 mm) Carrier gas: He, 1 mL/min, 50 : 1 split ratio Temperature programme: 200 C for 1 min to 270 C at 5 C/min to 295 C at 20 C/min for 3 min
High performance liquid chromatography HBC
McDermott (unpublished)
Column: Spherisorb 5 ODS-1 (150 4.6 mm i.d.) at 30 C Mobile phase: acetonitrile: TEAP(a) (50 : 50), flow rate 1.5 mL/min Detector: DAD (l = 254 nm)
HBD
McDermott (unpublished)
Column: Spherisorb 5 ODS-1 (150 4.6 mm i.d.) at 30 C Mobile phase: acetonitrile: TEAP(a) (20 : 80), flow rate 1.0 mL/min Detector: DAD (l = 254 nm)
HBE
Clarke (1989)
Column: Supelcosil 5 LC-18 (250 4.6 mm i.d.) Mobile phase: phosphate buffer(b) : methanol (60 : 40) Detector: UV (l = 309 nm)
HBF
Borner, Brenneisen (1987)
Column: Spherisorb ODS-1 (250 4.6 mm i.d., 10 mm) Mobile phase: 0.3 mol/L ammonium acetate in water (buffered to pH 8 with ammonia): 0.3 mol/L ammonium acetate in methanol (100 : 0 for 2 min to 5 : 95 at 14 min) Detector: UV (l = 269 nm)
HBG
Mesmer, Satzger (1998)
Column: Bondapak C18 (300 3.9 mm i.d., 10 mm) Mobile phase: phosphate buffer(c) : methanol (70 : 30), flow rate 1 mL/min Detector: UV (l = 215 nm)
HBH
Japp et al. (1988)
Column: ODS (250 5 mm i.d., 5 mm) Mobile phase: methanol : water : phosphate buffer(d) (pH 7.25, 55 : 25 : 20), flow rate 1 mL/min Detector: UV (l = 240 nm)
HBI
Japp et al. (1988)
Column: ODS (250 5 mm i.d., 5 mm) Mobile phase: methanol : water : phosphate buffer(d) (pH 7.67, 70 : 10 : 20), flow rate 1 mL/min Detector: UV (l = 240 nm)
HBJ
CND Analytical (1989)
Column: Bondex C18 (300 3.9 mm) Mobile phase: methanol : water (70 : 30), flow rate 1 mL/min Detector: UV (l = 254 nm)
HL
Baker et al. (1980)
Column: Spherisorb ODS silica (250 4.6 mm i.d., 5 mm) Mobile phase: 0.01 mol/L sulfuric acid : methanol : acetonitrile (7 : 8 : 9) table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:17
196
Drugs of Abuse
Table 11.4 continued Condition code
Source
Details
Thin-layer chromatography TAH
Silica gel plates with hexane : diethyl ether (4 : 1)
TAI
Silica gel plates with acetone
TAJ
Silica gel plates with n-butanol : acetic acid : water (2 : 1 : 1)
(a)
TEAP, triethylammonium phosphate buffer made up by preparing a 1.0 mol/L phosphoric acid (65 mL of 85% phosphoric acid to 1 L of water) titrated to pH 2.5 with triethylamine (approximately 10 mL triethylamine per 100 mL 1.0 mol/L phosphoric acid). A 10 mL quantity of this solution is made up to 1 L of water to give the working solution of 10 mmol TEAP. (b) Phosphate buffer is prepared by adding 10 mL of phosphoric acid to 1 L of water, followed by the addition of sufficient 2 mol/L sodium hydroxide to raise the pH to 6.5. (c) Phosphate buffer is 10 mmol potassium dihydrogen phosphate, adjusted to pH 3 with phosphoric acid. (d) 0.1 mol//L phosphate buffer is prepared by dissolving 14.35 g sodium dihydrogen phosphate dihydrate and 1.14 g disodium hydrogen phosphate in 1 L of water.
Figure 11.2 Gas chromatographic separation.
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:18
Analysis of seized drugs
Figure 11.3 HPLC separation.
197
are suspected of being linked as part of a local distribution chain. This is known as comparison or tactical profiling and may be carried out as a routine requirement in forensic casework. A second stage (intelligence or strategic profiling) exists where answers to wider questions may be sought. These will depend on the drug concerned, but include estimating the number of different profile types in circulation and relating them to the number of active laboratories and how long they have been in operation; determining the extent of importation by comparing the profiles of police and customs seizures; identifying the route of synthesis and types of precursors used; creating large-scale maps of drug distribution and identifying the country or region of origin. Tablet comparisons using general physical features, gross drug content and microscopic examination of defects and punch marks (socalled ballistic analysis) can be of some value, but they suffer from the fact that at any one time a large fraction of illicit tablets in circulation may be almost identical. Such small differences as may exist could simply reflect inherent differences in the punches and dies of a multiple-stage tabletting machine. This can be illustrated by the Mitsubishi logo (Fig. 11.5), which was found on over half of all MDMA tablets seized in Europe in the late 1990s. A similar pattern was also found in the UK for amfetamine in the early 1990s, when nearly half of all samples belonged to one profile type. In these circumstances, any connection between two separate seizures of otherwise identical tablets or powders may be purely fortuitous. This, in turn, raises other problems with profiling. It is necessary to maintain a database of profiles such that the significance of any ‘match’ or ‘non-match’ can be critically assessed. However, in a situation where profiles may change with time, what constitutes a ‘current’ database is not always clear. In the case of determination of country of origin, authentic samples are required in order to provide a statistical ‘training set’, yet such samples may be difficult to obtain and their true provenance uncertain. A general approach to the analysis of unknown substances A general approach to the analysis of unknown substances has been outlined in Fig. 11.1. Different approaches will be required depending on whether the exhibit is a powder, vegetable matter, tablet/capsule or liquid. Powders
When a powder is submitted for analysis, the most likely drugs to be present include heroin, cocaine, amfetamine and metamfetamine. Although others may be present, these are the most common. Initial examination will involve describing/detailing of the packaging material. If there are multiple packs present then a subsample may be removed for analysis. The powder must be weighed before analysis. The powder is then homogenised and an aliquot is taken for analysis. A screen (colour test, TLC, HPLC, GC) will indicate the drug(s) present. Identification of the drug can be achieved using GC-MS or FTIR spectroscopy. GC-MS has an advantage over FTIR spectroscopy because of retention time and a mass-spectral comparison with a known standard.
Figure 11.4 HPLC separation.
cannabis resin). Detection may range from flame-ionisation to isotoperatio mass spectrometry. Non-separation methods, for example using IR, Raman or X-ray diffraction spectroscopy, have only limited scope. Drug profiling may be used for two quite separate purposes. In the first case, it can establish connections between a number of exhibits that
Figure 11.5 Impressions on illicit MDMA tablets.
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:18
198
Drugs of Abuse
Identification of the other components in the powder can be achieved by using a range of analytical techniques including FTIR, X-ray fluorescence (XRF), X-ray diffraction (XRD), NMR spectroscopy and others. The drug content can be quantified by GC or HPLC by preparation of a curve using a range of concentrations of the drug in question. In some circumstances comparison may be required between powders for links in a specific case or for intelligence purposes, and GC and HPLC can be employed to examine some of the minor ingredients of the powder. Vegetable material
This includes cannabis plants, cannabis resin, khat and psilocybe mushrooms. A physical examination would include a description of the material followed by a measurement of the weight of material or the height of the plant. A subsample of the population may be chosen and an aliquot taken for analysis. Homogenisation may be necessary depending on the material. The physical appearance will generally give a very good idea of the drug present, for example cannabis plant or psilocybe mushrooms. We can therefore go directly to a specific test rather than use a screening technique. Identification can be carried out by a combination of microscopic and chemical techniques. The drug content can be quantified by GC or HPLC and comparison/ profiling carried out by chemical and physical comparisons. Tablets and capsules
Tablet or capsules submitted to the laboratory include ecstasy (MDMA, MDEA, etc.), benzodiazepines, steroids, LSD squares and others. An initial examination will include a description of any markings or logos, counting of the items, measurement of the tablet/capsule and possibly a weight. An examination of a tablet/capsule identification database such as TICTAC may give an indication of the drug present (see Chapter 13). Subsampling followed by homogenisation will lead to essentially a powder sample and the procedure for analysis of powder samples may then be followed. For comparison/profiling a physical comparison of the logo or mark may be the most informative piece of information available. Liquids and other forms
These include GHB, steroid oils, cocaine liquor and others. A physical examination may give an indication of the drug likely to be present. The physical measurements to be noted would include the volume, colour, odour and general appearance of the liquid. A subsample may be taken and an aliquot removed for analysis. It may be necessary to (base) extract the drug from the liquid into an organic solvent prior to analysis. The physical examination may allow the analyst to proceed to the identification stage, otherwise a screen may be used to indicate the presence of a certain drug. Identification, quantification and comparison/profiling can be carried out along the lines outlined for powders.
only a small amount (or none) of the final product (i.e. the controlled substance) may be found. In these instances, detailed explanations of the synthetic routes may be required. An explanation of the role of each of the chemicals found at the scene could be required. The scientist must also be aware of alternative explanations for the presence of the chemicals, as this is the likely defence in such cases. In many instances the precursor chemicals themselves are controlled. The forensic investigation of clandestine laboratories has been well documented (Christian 2003) and many of the complications associated with such laboratories have been explored. In the USA, the number of clandestine metamfetamine laboratories has increased enormously in recent times. Illegal seizures increased from 7438 in 1999 to 12 484 in 2005. The hazards and contamination issues associated with such laboratories pose difficulties for those involved in the process and for public health in general (Martyny et al. 2007). Capillary electrophoresis (CE) has been employed to characterise phosphorus species used in the manufacturing process (Knops et al. 2006).
Concealment of drugs The internal concealment of illicit drugs to smuggle quantities across borders has been a phenomenon for decades (Fineschi et al. 2002; Gill, Graham 2002; Takekawa et al. 2007). The ‘body packer’ or ‘mule’ carries quantities of drugs that are generally well wrapped to withstand the biological hazards of internal concealment. Deaths related to such activities are not uncommon. However, it is not known how widespread the practice is as the detection is frequently associated with instances that have adverse medical consequences. Impregnation of items with cocaine is also a novel method of concealment. Cocaine-impregnated silicone in baseball cap parts were detected and chloroform was used to extract the cocaine (Microgram 2003). Items of clothing impregnated with cocaine were extracted with methanol (McDermott, Power 2005) and in this instance ‘moth balls’ containing camphor were added to the suitcase to deter detection by dogs. An unusual way of smuggling cocaine is the use of a cocaine–poly (methylmethacrylate) solid solution (Gostic, Klemenc 2007).
Analysis of the main drugs of abuse In this section, methods are described for the analysis of the main drugs of abuse. In all cases a number of analysis methods are described. The methods chosen will depend on the aim of the analysis, the apparatus/ equipment available, legal aspects and the number of analyses to be performed, and may depend on other details associated with the specific drugs seizure. The analytical data for the various drugs are shown in Tables 11.5 and 11.6.
Clandestine laboratories Because of the increase in abuse of synthetic drugs, clandestine laboratories have become an increased part of forensic investigations. The investigation of such sites is very interesting as they reveal (in situ) the synthetic processes, intermediates, and often notes and chemical equations describing the various reactions used. These laboratories, however, are also very hazardous sites to investigate. The use of the word ‘laboratory’ disguises the more usual scenario of a garage, shed or kitchen. Forensic scientists frequently become involved in an advisory capacity in the initial stages of a clandestine laboratory investigation. Information may come to light about certain chemicals being used at a premises and the scientist is responsible for formulating an opinion as to whether a controlled substance is being produced. The police can then act on the basis of this opinion. Many countries have specially trained police and scientists to deal with the specific problems that clandestine laboratories pose. These problems could be in the form of hazardous chemicals (acids, bases, solvents and reagents) and fire and explosion potentials. Ultimately, if the seizure results in a court case, the testimony in these cases can be technically demanding for the scientist. In many situations,
Amfetamine and metamfetamine Amfetamine (a-methylphenethylamine; 1-phenylpropan-2-amine) and metamfetamine (N-methyl-a-methylphenethylamine; N-methyl1-phenylpropan-2-amine) in free base form are both liquids. Amfetamine is normally produced as amfetamine sulfate or phosphate and is common in Europe. Metamfetamine is normally produced as metamfetamine hydrochloride and is more popular in North America and Japan. Street-level amfetamine and metamfetamine are normally submitted to the laboratory as white/off-white powders with relative low purity (e.g. 5%). Synthesis of amfetamine and metamfetamine
Many methods are available for the illicit synthesis of amfetamine, but the Leuckart reaction has been the most popular. This method is simple and rapid, gives a good yield and does not involve any particularly hazardous chemicals or procedures. It may be considered as a three-step reaction involving the condensation of phenyl-2-propanone (P-2-P) with formamide followed by a hydrolysis of the N-formylamfetamine and finally purification by steam distillation.
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:19
Analysis of the main drugs of abuse
Table 11.5 Analytical data using GC Drug
Retention time (min)
GC system
Amfetamine
4.85
G1
Methamfetamine
5.52
G1
MDMA
8.70
G1
Heroin
14.19
G1
Cocaine
199
Metamfetamine can be made by the Leuckart reaction using methylamine and formic acid or N-methylformamide in the condensation step. Analysis of amfetamine and metamfetamine
12.68
G1
LSD
7.28
G2
LAMPA
8.06
G2
Psilocin
4.88
G3
Because many of the street-level samples submitted to the laboratory are relative low in purity (5%), pre-concentration of samples may be required for the analysis to be successful. This may be achieved by base extracting the amfetamine/metamfetamine into ether and evaporating to dryness in an airflow without heat. A few drops of methanol can then be added and the methanolic solution transferred to a plastic insert prior to analysis. Colour test Marquis test gives an orange colour for both amfetamine and metamfetamine.
Psilocybin
6.90
G3
TLC
Cathine
4.06
G4
Cathinone
3.93
G4
GHB
5.60
G5
n n n
GBL
3.98
G5
Diazepam
13.51 (4.47)
G1 (G6)
Flunitrazepam
14.02 (5.24)
G1 (G6)
Flurazepam
14.59 (6.14)
G1 (G6)
Nitrazepam
14.48 (5.94)
G1 (G6)
Fluoxymesterone
9.04
G7
Nandrolone
7.16
G7
Testosterone
7.98
G7
Methyltestosterone
8.33
G7
Cannabinol
13.86
G1
Tetrahydrocannabinol
13.64
G1
Cannabidiol
13.32
G1
Table 11.6 Analytical data using HPLC Drug
Retention time (min)
HPLC system
Amfetamine
2.06 (3.68)
H1 (H2)
Methamfetamine
2.42 (4.67)
H1 (H2)
MDMA
2.42 (5.31)
H1 (H2)
Heroin
2.88
H1
Cocaine
5.21
H1
LSD
5.34
H3
LAMPA
5.66
H3
Psilocin
6.80(a)
H4
Psilocybin
3.20(a)
H4
GHB
3.5
H5
GBL
4.0
H5
Diazepam
2.29(a) (10.41)(a)
H6 (H7)
Flunitrazepam
0.86(a) (3.34)(a)
H6 (H7)
Flurazepam
3.12(a) (12.98)(a)
H6 (H7)
Nitrazepam
0.87(a) (3.22)(a)
H6 (H7)
Fluoxymesterone
5.5
H8
Nandrolone
6.0
H8
Testosterone
7.5
H8
Methyltestosterone
9.5
H8
Cathine Cathinone
Cannabinol
11.77(a)
HL
Tetrahydrocannabinol
13.35(a)
HL
7.47(a)
HL
Cannabidiol (a)
k values.
TA: amfetamine Rf ¼ 0.43; metamfetamine Rf ¼ 0.31 TB: amfetamine Rf ¼ 0.15; metamfetamine Rf ¼ 0.28 Visualisation is with acidified iodoplatinate solution.
Separation/identification
(For analytical data see Tables 11.5 and 11.6.) In addition, it is common practice with primary amines to prepare derivatives such as N-methylbis(trifluoroacetamide) (MBTFA) or trifluoroacetic anhydride (TFAA) derivatives. It is good practice to analyse both derivatised and underivatised samples since N-hydroxylamines may give the same product as the parent amines. Using a concentrated/base-extracted sample, the molecular ion peaks m/z 134 and m/z 148 for amfetamine and metamfetamine, respectively, can readily be achieved in an underivatised sample. Both amfetamine and metamfetamine have one asymmetrical carbon atom, resulting in a pair of enantiomers in each case. Depending on the synthetic route l-, d- and dl-amfetamine or metamfetamine could be encountered in samples submitted to the laboratory for analysis. These optical isomers differ in their pharmacological activity and are subject to different regulatory measures in certain countries. In those countries where the specific optical isomer needs to be identified, chiral analysis can be undertaken by derivatisation/GC, by the use of chiral columns (GC and HPLC) and by the use of CE (Anastos et al. 2005; Fanali et al. 1998; Lebelle et al. 1995; Sellers et al. 1996). Enantiomeric analysis of metamfetamine samples (Lee et al. 2007) showed that up until 1997 the vast majority of the metamfetamine encountered in the Republic of Korea was the S-(þ)-enantiomer, but from 1997 onwards the R()-enantiomer began to appear and increased continuously until 2005 when 50% of the metamfetamine samples seized contained the R-()-enantiomer. Quantitative analysis and profiling of amfetamine/ metamfetamine
Amfetamine and metamfetamine can be quantified by HPLC or GC. Normally if GC is used the samples are base extracted into an organic solvent and either run directly or derivatised and then run. Using HPLC there is no need to extract and, in many cases, this is the preferred method for quantitative analysis of amfetamine/metamfetamine. Amfetamine produced illicitly often contains impurities that are a result of the manufacturing process. The presence of these impurities can be used to compare and distinguish samples of amfetamine since material used in the same manufacturing batch would almost certainly have the same number and relative amount of identical impurities. Samples from the same illicit laboratory produced at different times may show strong similarities, whereas samples from unrelated laboratories are expected to show major qualitative and quantitative differences. Basic extracts into organic solvents are subjected to GC or GC-MS analysis. Samples are compared by visual inspection of the GC trace and by quantitative comparisons. Metamfetamine impurity profiling is also carried out by GC analysis, with the impurities also giving information on the synthetic route (Seta et al. 1994). A recent study (Lee et al. 2006) of the impurities present as a result of the synthetic process showed that the selected impurity peaks may be utilised as the indicators of synthetic conditions and analysis of their patterns can supply valuable information for understanding the
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:20
200
Drugs of Abuse
conditions of clandestine synthesis of metamfetamine. The use of thermal desorption followed by GC-MS was found to be an efficient method for impurity profiling of metamfetamine (Kuwayama et al. 2007). The purpose of the comparison/profiling is: to identify dealer–user links; to establish possible sources, i.e. the clandestine laboratory; and to build up databases to allow interpretation in comparison casework. This approach has been used in Australia to examine samples over the period 1998–2002 (Qi et al. 2006). A series of studies were undertaken to develop a harmonised method for the profiling of amfetamine synthesised by three different methods. The study covered the sample preparation, extraction procedure, optimisation of GC-MS method, selection of target compounds and numerical comparison of amfetamine impurity profiles (Aalberg et al. 2005a, b; Andersson et al. 2007a,b,c; Lock et al. 2007). This inter-laboratory study found that the variation from laboratory to laboratory was affected by such factors as homogeneity of samples and concentration effects due to dilution. Cannabis, cannabis resin and cannabinoids Herbal cannabis (marijuana) means all parts of the plant Cannabis sativa L., but excludes the seeds and mature/woody stalk material. Cannabis sativa L., which can be grown in all parts of the world, is an annual plant and attains a height of 1–5 m. When it is planted for the production of hemp fibre, the stalks are crowded and without foliage except near the top. The wild growing plant, in contrast, has numerous branches. The resin of the plant occurs mainly in the flowering area, the leaves and the stem, particularly at the top of the plant. The highest amount of resin is found in the flowering part. Up to the time of flowering, the male and female plants produce resin almost equally. After shedding their pollen the male plants soon die. The resin is found in the glandular trichomes. The leaves of Cannabis sativa L., are compound and consist of 5 to 11 separate leaflets. Each leaflet is characteristically hair covered and veined and has serrated edges (Fig. 11.6). Cannabis herbal material may be encountered in blocks of dried flowering tops and dried leaves. Cannabis resin (hashish) is a compressed solid made from the resinous parts of the plant and is usually produced in 250 g blocks. Herbal cannabis imported into Europe may originate from West Africa, the Caribbean or South East Asia, but cannabis resin derives largely from either North Africa or Afghanistan. Cannabis and cannabis resin are normally mixed with tobacco and smoked, but can be ingested. The average ‘reefer’ ‘joint’ cigarette contains around 200 mg of herbal cannabis or cannabis resin. The main psychoactive compound in cannabis and cannabis resin is D9-THC. Cannabinol (CBN) and cannabidiol (CBD) are among the other main components.
Figure 11.7 Microscopic examination of cannabis: (a) cystolith hair; (b) large glandular hair with several cells in the head and stalk; (c) head of one of the large glandular hairs; (d) calcium carbonate.
A sample of the material is extracted with petroleum ether. The petroleum ether is removed and evaporated to dryness. The addition of Duquenois reagent followed by concentrated hydrochloric acid will yield a purple colour after a few minutes. The addition of chloroform should result in a purple colour in the chloroform layer. This result can be taken positive for cannabinoids. TLC n n
TAH: THC Rf ¼ 0.50; CBD Rf ¼ 0.60; CBN Rf ¼ 0.45 Visualisation is by fast blue BB with THC showing a red colour, CBD showing an orange colour and CBN showing a purple colour.
Microscopic examination of cannabis/cannabis resin
The most characteristic botanical feature visible under the microscope is the hairs. There are two types of hair: n n
Cystolithic hairs Glandular hairs.
Colour test The presence of cannabinoids in suspect material can be
The cystolithic hairs contain a deposit of calcium carbonate at their base. These hairs are mostly single celled. The glandular hairs are most important since they contain and secrete the resin. They are short and may be unicellular or multicellular. The bigger glandular hairs have a multicellular stalk with heads containing 8–16 cells (Fig. 11.7). The microscopic test is carried out by putting a small portion of the dry material (cannabis herbal material or cannabis resin) on a microscope slide. A few drops of Duquenois reagent are added followed by a few drops of concentrated hydrochloric acid. The cystolithic hairs contain a deposit of calcium carbonate at their base and a characteristic effervescence can be observed. The heads at the end of the glandular hairs will become a red/purple colour. An alternative method is to add a few drops of chloral hydrate solution to the dry material. This is particularly useful for getting more detailed information on the structure of the plant tissue since it removes coloured materials such as chlorophyll.
indicated by the Duquenois–Levine test.
Quantitative analysis and comparison of cannabis/cannabis resin
Figure 11.6 Cannabis plant.
As already stated, cannabis resin is normally produced in 250 g blocks. Frequently these blocks carry an impression, e.g. a number or a letter or a symbol. Comparison can be made between different blocks on the basis of similar impressions, though unrelated blocks often have the same impression. The street-level deal of cannabis resin is typically a finger-sized portion (normally 1–10 g) possibly wrapped in tinfoil or plastic. It may be possible to link a smaller piece of cannabis resin to its original block by a physical fit between the smaller and larger pieces. GC or HPLC may be used to obtain a chemical profile of the cannabis/cannabis resin (see Tables 11.5 and 11.6). The THC content can be calculated and comparison can be made on that basis. It must be noted that variations can occur in the THC content of a single block of cannabis resin as the THC content decreases with age and storage conditions. The outer material in a block of cannabis resin can differ from that in the centre. The variation of distribution of cannabinoids within blocks of resin was studied and it was noted that composition of the resin changed even within a 12-month period (Lewis et al. 2005). The main chemical constituents of cannabis have been well documented (Elsohly, Slade 2005).
Analysis of cannabis/cannabis resin
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:20
Analysis of the main drugs of abuse The THC contents of the various forms of cannabis can vary enormously. For example, recent seizures of cannabis in the UK had the following THC contents: sensemilla (indoor intensively cultivated herbal cannabis), mean 16.2%, range 4.1–46%; imported herbal material, mean 8.4%, range 0.3–22%; resin (mostly from North Africa), mean 5.9%, range 1.3–28% (Hardwick, King 2008). Short tandem repeat (STR) DNA markers have been used to indicate the likely origin of a cannabis crop and STR markers can permit the identification of hydroponically propagated clonal drug lines, providing evidence to link illegal operators (Gilmore et al. 2007). Cocaine Cocaine is a naturally occurring alkaloid found in certain varieties of plants of the genus Erythroxylum. Coca cultivation is distributed throughout the central and northern Andean Ridge, with approximately 60% in Peru, 30% in Bolivia, and the remainder in Columbia, Ecuador, Venezuela, Brazil, Argentina and Panama. Cocaine production
Production of illicit natural cocaine involves three steps: 1. Extraction of crude coca paste from the coca leaf. 2. Purification of the coca paste to cocaine base. 3. Conversion of cocaine base to cocaine hydrochloride. Cocaine is normally encountered in the laboratory in paper or plastic packs of white powder and can be analysed without extraction. Analysis of cocaine Colour test Cobalt thiocyanate test or modified cobalt thiocyanate test (Scott test): a blue colour indicates the presence of cocaine. Odour test A 5% methanolic solution of sodium hydroxide added to the test sample and warmed gives a characteristic odour in the presence of cocaine. TLC n n
TA: Rf ¼ 0.65; TB: Rf ¼ 0.47 Visualisation is with acidified iodoplatinate solution.
Separation/identification
(For analytical data see Tables 11.5 and 11.6.) In addition to GC and HPLC, IR spectroscopy is routinely used in cocaine cases if a distinction is to be made between cocaine as a salt, e.g. cocaine hydrochloride, and cocaine in base form. The cocaine base is known as crack and, unlike cocaine hydrochloride, can be consumed by smoking. The differences in their IR spectra are shown in Fig. 11.8. The differences in the spectra at 1736 and 1709 cm1 for the base and 1729 and 1711 cm1 for the hydrochloride are explained (Elsherbini 1998) by the effect of the hydrochloride ion on the C¼O stretching bands. A simple laboratory test also exists for the determination of the chemical form of cocaine (Logan et al. 1989). Quantitative analysis and profiling of cocaine samples
Quantitative analysis of cocaine samples may be carried out by GC or HPLC. The general quantitation method previously described may also be used for cocaine. In some jurisdictions, preparations containing less than 0.1% cocaine base are exempt from certain controls and analytical procedures must be designed to accommodate this. Because of the unsophisticated nature of the cocaine manufacturing process, a multitude of trace-level alkaloid impurities are present in illicit cocaine. Many of these impurities are naturally occurring alkaloids that originate from the coca leaf and are carried through the manufacturing process. In addition, cocaine is also contaminated with a variety of manufacturing by-products. The relative amounts of these compounds can be used to compare cocaine samples (Moore, Casale 1998). The alkaloid impurities, which originate in the coca leaf, include cis- and trans-cinnamoylcocaine, tropacocaine, truxillines and hydroxycocaines. Manufacturing by-products found in illicit cocaine include hydrolysis products such as benzoylecgonine, ecgonine methyl ester, ecgonine and benzoic acids.
201
Oxidation by-products also arise and these include N-norcocaine and N-norecgonine methyl ester. New impurities arising from the oxidation of crude cocaine base have been reported recently (Casale et al. 2007). In addition to the above, solvent residues may be detected by NMR or headspace GC. The solvents detected include acetone, methyl ethyl ketone, benzene, toluene and diethyl ether (Cole 1998). A comparison of cocaine samples can be achieved by a combination of qualitative analysis for the presence/absence of certain trace impurities and by quantitative analysis of the cocaine and other ingredients. Isotope ratio analysis has also been used in the profiling of cocaine (Benson et al. 2006). A recent study explored the optimisation and harmonisation of a profiling method for cocaine in two separate laboratories using eight main alkaloids as the comparators (Lociciro et al. 2007). Heroin Street-level heroin (diamorphine, diacetylmorphine) is normally encountered in the laboratory in paper or plastic packs containing typically 100–200 mg of brown (and sometimes white) powder. The street-level purity varies depending on availability and other factors, but values of 40–60% are common. Production of heroin
The raw material for the production of heroin is opium. Opium is a naturally occurring product of the plant Papaver somniferum L. (opium poppy). Opium is purified to form crude morphine. The morphine is acetylated with acetic anhydride to produce diacetylmorphine, the primary constituent of illicit heroin samples. Sometimes known as ‘Chinese heroin’, heroin from south east Asia is a white powder consisting of heroin hydrochloride and minor amounts of other opiate alkaloids, but adulterants are unusual. This material is ideally suited to injection. Heroin from south west Asia is a much cruder product. Typically seen as a brown powder containing heroin base, it has variable amounts of other opiate alkaloids (e.g. monoacetylmorphine, noscapine, papaverine and acetylcodeine) as well as adulterants such as caffeine and paracetamol. It is believed that these cutting agents are added to heroin either at the time of manufacture or during transit. Brown heroin may be ‘smoked’ by heating the solid on a metal foil above a small flame and inhaling the vapour. Those intending to inject brown heroin must first solubilise it with, for example, citric acid or ascorbic acid. Analysis of heroin Colour test Marquis reagent gives a purple/violet colour.
Other opiate alkaloids (morphine, codeine, monoacetylmorphine and acetyl codeine) give the same positive reaction to the Marquis test. TLC n n
TA: Rf ¼ 0.47. TB: Rf ¼ 0.15 Visualisation is with acidified iodoplatinate solution.
Separation/identification
(For analytical data see Tables 11.5 and 11.6.) Quantification and profiling of heroin
Heroin may be quantified by either GC or HPLC. A problem associated with GC analysis is that heroin may hydrolyse to 6-O-monoacetylmorphine and/or transacetylation may occur of the common cutting agent paracetamol by heroin in the injection port of the GC column. The use of fresh samples and of chloroform as solvent can avoid these problems. By examining the amount of heroin, papaverine, noscapine and acetylcodeine in the samples it is possible to discriminate between or show a link between samples (Besacier, Chaudron-Thoxet 1999; Dufey et al. 2007; Seta et al. 1994; Stromberg et al. 2000). This method of comparison was used successfully on 500 heroin samples that were divided into nine groups (Zhang et al. 2004). It may be further possible to examine heroin samples and show potential links between samples by the presence (and amount) of adulterants such as caffeine or by the presence of less common adulterants such as diazepam, phenobarbital
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:20
202
Drugs of Abuse
Figure 11.8 Infrared spectra of (A) cocaine base and (b) cocaine hydrochloride.
or mannitol hexa-acetate (El Haj et al. 2004). In large seizures, differences may be found between various samples originating from the seizure that indicate that it includes more than one batch of heroin. Chemical profiling was used to determine the country of origin of heroin samples in Australia (Collins et al. 2006) and stable isotope analysis was used to complement the chemical profiling on the same
samples (Casale et al. 2006). Isotope ratio analysis has increasingly been used as a comparison tool for heroin (Carter et al. 2005; Zhang et al. 2005). In addition to examining the relative ratios of the main components, it is possible to analyse for solvent residues (Cole 1998; Dams et al. 2001).
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:20
Analysis of the main drugs of abuse LSD LSD is one of the most potent hallucinogenic substances known. Its properties were first discovered in the 1930s and its popularity as a drug of abuse was very high during the 1960s and 1970s when it was associated with the hippy movement. Synthesis of LSD
LSD can be produced by several different methods, the majority of which use lysergic acid as the starting material. Lysergic acid itself is also produced in clandestine laboratories using, most commonly, ergometrine or ergotamine tartrate as the starting material. Ergotamine refluxed with potassium hydroxide and hydrazine in an alcohol–water mixture produces lysergic acid. The methods used for the production of LSD yield a crude product, which is then cleaned up and converted to a more stable form, e.g. the tartrate salt. In the past, LSD was encountered in a variety of substrates including powder in gelatine capsules, gelatine squares, sugar cubes and ‘microdots’. Nowadays LSD is encountered mostly in paper dose form. The paper dosages are produced by soaking pre-printed paper in a solution of LSD. These sheets are then perforated into squares (typically 5 5 mm) with each square (‘tab’) containing approximately 50 mg of LSD. The designs on the paper can vary from one design per square to one large design covering many squares (Fig. 11.9).
203
Quantitative analysis and comparison of LSD
HPLC is the method of choice for quantitative analysis of LSD. Using the solvent mixture methanol–water (1 : 1), quantitative extraction of LSD from paper squares is normally achieved after sonication for 20 min (McDonald et al. 1984). In some instances, a comparison is requested between one square of LSD and a large sheet of perforated squares. This can be an easy matter if the design on the large sheet spreads over the whole sheet and the ‘missing’ square fits neatly into the pattern. In other instances, the design may be on each individual square (or there may be no design). In such a case it is necessary to examine the colour/design/dimensions of the squares and the perforation pattern. Chemical comparisons can also be undertaken, but squares from the same larger sheet can vary in the amount of LSD on each square. MDMA
The only analogue of LSD to receive widespread interest is lysergic acid N-(methylpropyl)amide (LAMPA) and any analytical technique should be capable of separating LAMPA from LSD. The presence of LSD may be signalled early by placing the suspect paper under long-wavelength UV light. The presence of LSD is indicated by a blue fluorescence. Colour test Van Urk reagent gives a purple colour.
MDMA is the prototypical member of a large series of phenethylamine designer drugs and has become one of the main drugs of abuse in many countries in northern Europe. Clandestine production is largely centred in Europe. A number of homologous compounds with broadly similar effects, e.g. MDA (methylenedioxyamfetamine), MDEA (methylenedioxyethamfetamine) and MBDB (N-methyl-1-[1,3-benzodioxol-5yl]-2-butanamine) have also appeared, but have proved less popular. These substances are collectively known as the ‘ecstasy’ drugs. MDMA is the most common drug encountered in ‘ecstasy’ tablets. The tablets are typically 10 mm in diameter, either flat or biconvex, and weigh approximately 300 mg. The MDMA content varies but is generally in the range 80–100 mg per tablet. The tablets normally carry a characteristic logo/imprint. These designs are not restricted to MDMA tablets, but may be found on amfetamines and other illicit products. In other words, the logo and other physical characteristics provide no reliable information on the drug content. Many hundreds of different impressions have been found and several examples are shown in Fig. 11.5.
TLC
Synthesis of MDMA
Analysis of LSD
n n n
TAI: LSD Rf ¼ 0.58; LAMPA Rf ¼ 0.49 Visualisation: observe the plate under UV light (254 and 365 nm) Spray with Van Urk reagent and heat to give a blue spot.
Separation/identification
(For analytical data see Tables 11.5 and 11.6.) Some difficulty may be encountered in obtaining an unequivocal identification of LSD is because of its low dosage (50 mg or less). However, if the sample is concentrated, a satisfactory analysis can be achieved. Place a suspect LSD square in a glass vial and cover with methanol. After soaking (or sonication) for 10–20 min, the methanol can be transferred to a plastic insert for analysis. Another method is to add concentrated ammonia (2 drops) to the methanol. In addition to chromatographic separation, LSD can be discriminated from other ergot alkaloids by its MS fragmentation pattern; for example, the presence in the LSD spectrum of a m/z 100 fragment nearly as intense as the as the m/z 111 fragment differentiates LSD from other di-substituted amides (Clarke 1989).
Figure 11.9 Examples of LSD paper squares.
Several methods of synthesis can be employed including: 1. Amine displacement method using safrole as the starting material. 2. Via the intermediate 1-(3,4-methylenedioxyphenyl)-2-propanone (MDP2P) with isosafrole or a nitrostyrene as starting material. Analysis of MDMA Colour test Marquis test gives a blue/black colour. TLC n n
TA: Rf ¼ 0.31; TB: Rf ¼ 0.23 Visualisation is with acidified iodoplatinate solution.
Separation/identification
(For analytical data see Tables 11.5 and 11.6.) Base extraction into an organic solvent and/or derivatisation prior to GC-MS analysis is common with MDMA. Quantitative analysis and profiling/comparison
In order to perform a quantitative analysis on ‘ecstasy’ tablets they must first be ground to produce a homogeneous powder and the MDMA content determined by either GC (either directly or base extracted) or HPLC. Chemical profiling of tablets containing MDMA involves the examination/quantification of the drug and main adulterants present such as caffeine, sugars and binding agents. In addition to the main ingredients, many trace-level impurities from the synthetic process can be present and these can be used for comparison (Bohn et al. 1993; Renton et al. 1993). A cross-laboratory study on organic impurity profiling of MDMA tablets based on 46 organic impurities yielded separation between population of samples from the same synthesis batch and samples from different batches (Weyermann et al. 2008). Trace metal analysis has proved to be a useful method for comparing MDMA tablets (Koper et al. 2007; Waddell et al. 2004). The metals arise in the tablets as a result of the catalyst or reducing agent in the synthesis. Isotope ratio analysis has also been used to discriminate between different tablets
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:21
204
Drugs of Abuse
containing MDMA (de Korompay et al. 2008; Palhol et al. 2004). Recent advances in impurity profiling in MDMA tablets have employed a variety of techniques, with multiple techniques being used in many situations (Waddell-Smith 2007). As already mentioned, tablet comparisons can also be made using (so-called) ballistic analysis. In this, general physical features and microscopic examination of defects and punch marks are used for comparison. The difficulty is that, at any one time, a large fraction of illicit tablets in circulation may be almost identical. Anabolic steroids Anabolic steroids may be abused by ‘body builders’ and athletes. In the UK, 48 steroids are listed specifically and generic legislation covers certain derivatives of 17-hydroxyandrostan-3-one or 17-hydroxyestran-3-one. Methandienone, nandrolone, oxymetholone, stanozolol, and testosterone and its esters account for most cases. Further nonsteroidal anabolic compounds are also controlled, i.e. human chorionic gonadotrophin (HCG), clenbuterol, non-human chorionic gonadotrophin, somatotropin, somatrem and somatropin. Certain anabolic steroids are scheduled in the US Controlled Substances Act, but these drugs are not listed in the UN Conventions. A large number of the anabolic steroids encountered in seizures have been found in counterfeited packaging and the drug content may differ qualitatively or quantitatively from what is indicated in the information on the product label. This mislabelling can be particularly frustrating to the forensic chemist trying to identify the particular steroid in the product. Formulations may be either as tablets or as steroid esters dissolved in vegetable oil suitable for injection. The oils may be extracted using a steroid : hexane : methanol ratio of 1 : 2 : 1 (Chiong et al. 1992) with the methanol layer being used for analysis. Because of the large number of steroid products available, analytical information is presented here on only four: fluoxymesterone, nandrolone, testosterone and methyltestosterone. Further information on the analysis of anabolic steroids in urine is to be found in Chapters 6 and 7 on Drugs in sport.
TLC n n n
TA: diazepam Rf ¼ 0.75; flunitrazepam Rf ¼ 0.63; nitrazepam Rf ¼ 0.62; flurazepam Rf ¼ 0.68 TB: diazepam Rf ¼ 0.23; flunitrazepam Rf ¼ 0.10; nitrazepam Rf ¼ 0.30; flurazepam Rf ¼ 0.00 Visualisation is with acidified iodoplatinate solution.
Separation/identification
(For analytical data see Tables 11.5 and 11.6.) GHB and analogues g-Hydroxybutyric acid (GHB) was originally developed as an anaesthetic drug and is still used for that purpose in some countries. It acts as a central nervous system depressant and hypnotic, and is chemically related to the brain neurotransmitter GABA (g-aminoxybutyric acid). Synonyms include sodium oxybate, g-OH, somatomax, ‘GBH’ and ‘liquid ecstasy’. The effects of GHB have been likened to those produced by alcohol and there are claims that it has anabolic properties. GHB is easily manufactured by adding aqueous sodium hydroxide to g-butyrolactone (GBL) to leave a weakly alkaline solution. Not only is the precursor GBL widely used as an industrial solvent, it can also be ingested directly to produce the same effects as GHB. Illicit GHB is normally sold in solution as a clear liquid in 30 mL opaque plastic bottles. The typical dose is around 10 mL equivalent to about 1 g or more of GHB. The sodium and potassium salts of GHB are hygroscopic. This property mean that GHB is almost never found as a powder or in tablets. Analysis of GHB
The legal distinction between GHB and GBL, coupled with the potential for GBL to undergo interconversion with GHB, raises important issues in the analytical approach to GHB analysis. The potential exists for aqueous-based GBL products to undergo conversion to GHB in the time between manufacture and consumption. Some of the factors affecting this interconversion have been explored (Ciolino et al. 2001). Colour test With 1% cobalt nitrate a pink to violet colour is indicative of GHB.
Analysis of steroids
Separation/identification
TLC
(For analytical data see Tables 11.5 and 11.6.) GC analysis of GHB samples will result in conversion of GHB to GBL, thus necessitating the need for derivatisation prior to analysis (Blackledge, Miller 1991). Test samples are taken to dryness under a stream of dry air. Samples are then derivatised with 99 : 1 N,O-bis(tri-methylsilyl)trifluoroacetamide (BSTFA) : trimethylchlorosilane (TMCS) in the presence of pyridine and incubated at 70 C for 30 min. GHB is detected as the di-trimethylsilyl (TMS) derivative and GBL does not form a silyl derivative. HPLC can be used without derivatisation (Mesmer, Satzger 1998). In a study of the reaction of GHB with alcohol it was found that an ester was formed under certain conditions and this can be separated from both GHB and GBL by HPLC and GC (Hennessy et al. 2004). Capillary electrophoresis has been used to separate GHB, GBL and 1,4butanediol (Dahlen, Vriesman 2002). Carbon isotope ratio analysis has been used to examine the discrimination between endogenous and exogenous GHB (Saudan et al. 2007).
n n n
TP: fluoxymesterone Rf ¼ 0.51; nandrolone Rf ¼ 0.87; testosterone Rf ¼ 0.60; methyltestosterone Rf ¼ 0.70 TQ: fluoxymesterone Rf ¼ 0.09; nandrolone Rf ¼ 0.48; testosterone Rf ¼ 0.07; methyltestosterone Rf ¼ 0.16 Visualisation is with ethanol–sulfuric acid or p-toluene sulfonic acid solution.
Separation/identification
(For analytical data see Tables 11.5 and 11.6.) Benzodiazepines There are 34 benzodiazepines listed in Schedule 4 of the UN 1971 Convention. Most are now rarely prescribed and abuse is largely restricted to pharmaceutical preparations containing diazepam, flunitrazepam, nitrazepam, flurazepam and temazepam. They may be used in conjunction with opiates (e.g. heroin) or in their own right. A particular problem occurred in Scotland during the mid-1990s when the contents of gel-filled temazepam capsules were injected. Abuse of temazepam declined following the withdrawal of capsules from the market and their replacement with tablets. In other countries, flunitrazepam became the most widely abused benzodiazepine. This drug also gained a certain notoriety for its association with ‘date-rape’. For these reasons, flunitrazepam was moved to Schedule 3 of the UN 1971 Convention and is therefore subject to more stringent controls. Analytical information is presented here only for diazepam, flunitrazepam, nitrazepam and flurazepam. Analysis of benzodiazepines Colour test Zimmerman’s test: reddish-purple or pink indicates the possiblility of some of the benzodiazepines.
Khat Catha edulis is a flowering evergreen shrub cultivated in East Africa and the Arabian Peninsula. The leaves and fresh shoots are commonly known as khat, qat or chat. Khat can be used by chewing the leaves or by brewing as a ‘tea’ and daily consumption can be up to several hundred grams. Khat has stimulant effects similar to those of amfetamine. Alcoholic extracts (tinctures) of khat have been noted especially in ‘herbal high’ sales outlets and at music festivals. The active components of khat, cathinone (()-1-aminopropiophenone) and cathine ((þ)-norpseudoephedrine), are usually present at around 0.3–2.0% (Lee 1995). Both substances are close chemical
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:22
Analysis of the main drugs of abuse relatives of synthetic drugs such as amfetamine and methcathinone. Khat must be used fresh as the more active cathinone begins to deteriorate rapidly after harvesting. Both cathine and cathinone are scheduled under the UN 1971 Convention, but khat itself is only specifically listed in a few jurisdictions. Analysis of khat
Approximately 5–6 g of plant material is cut into small pieces. Methanol (15–20 mL) is added and sonicated for 15 min. The green methanolic solution is filtered/decanted and condensed to near dryness. Approximately 20 mL of 0.2 mol/L sulfuric acid is added and the solution acquires a reddish hue. A chloroform extract will remove the neutral organic compounds. The aqueous layer (red layer) is basified with saturated sodium bicarbonate solution. Methylene chloride (20 mL) is added to extract the cathinone and cathine. A stream of air is used to reduce the volume to approximately 1 mL. Colour test Cathinone gives no reaction with Marquis reagent, but does produce a slow-forming yellow/orange colour with Chen’s reagent. TLC n n
TE: cathinone Rf ¼ 0.46; cathine Rf ¼ 0.25 Visualisation: UV (254 nm) and 0.5% ninhydrin with cathinone showing an orange colour and cathine a purple colour.
Separation/identification
(For analytical data see Tables 11.5 and 11.6.) Psilocybe mushrooms The hallucinogenic substances psilocin and its phosphate ester psilocybin occur in a number of fungi, particularly those of the genus Psilocybe. These are small grey mushrooms which grow wild over large areas. Although such material is not in itself controlled, and neither is its cultivation, it has been held in UK case law that the deliberate drying or processing of these mushrooms constitutes preparation of a controlled drug. Analysis of psilocybe mushrooms
A small quantity (approximately 1 g) of the dried mushrooms is sonicated with methanol (approximately 5 mL) for 10 min. The liquid is removed and reduced in volume at room temperature in an air flow. Psilocybin can be converted to psilocin by heating. This conversion can also occur if the mushrooms are not dried prior to or when they arrive into the laboratory. Colour test Ehrlich reagent: a violet colour is indicative of psilocybin and psilocin TLC n n n
TAN: psilocybin Rf ¼ 0.34; psilocin Rf ¼ 0.59 TA: psilocybin Rf ¼ 0.05 psilocin Rf ¼ 0.39 Visualisation is with Van Urk’s reagent, with both compounds showing a blue/violet colour.
205
the 1980s that the phenomenon of so-called designer drugs was fully recognised. Starting in the late 1980s, a large series of designer drugs began to appear, all of which were based on the phenethylamine nucleus. Just as with the production of the major illicit phenethylamines (e.g. MDMA), much of this synthetic activity took place in Europe. Table 11.7 lists a number of designer drugs that have appeared in Europe and the USA since the mid-1990s. This list, which may not Table 11.7 Designer drugs reported in Europe and the USA since the mid-1990s Compound/drug
Acronym
UN/UK
Ring-substituted phenethylamines 3,4-Methylenedioxyamfetamine
MDA
þ/þ
3,4-Methylenedioxymetamfetamine
MDMA
þ/þ
3,4-Methylenedioxyethylamfetamine
MDE(A)
þ/þ
4-Bromo-2,5-dimethoxyamfetamine
DOB (Bromo-STP)
þ/þ
4-Methoxyamfetamine
PMA
þ/þ
N-Hydroxy-MDA
N-OH MDA
þ/þ
3,4-Methylenedioxypropylamfetamine
MDPA
/þ
N-Methyl-1-(1,3-benzodioxol-5-yl)-2butanamine
MBDB
/þ
1-(1,3-Benzodioxol-5-yl)-2-butanamine
BDB
/þ
4-Bromo-2,5-dimethoxyphenethylamine
2C-B
/þ
3,4-Methylenedioxydimetamfetamine
MDDM
/P
2,5-Dimethoxy-4-(n)propylthiophenethylamine
2C-T-7
/þ
4-Allyloxy-3,5-dimethoxyphenethylamine
AL
/þ
3,5-Dimethoxy-4methylallyloxyphenethylamine
MAL
/þ
N-Hydroxy-MDMA
FLEA
/P
2,5-Dimethoxy-4-chloroamfetamine
DOC
/þ
4-Methylthioamfetamine
4-MTA
/P
2,5-Dimethoxy-4-ethylthiophenethylamine
2C-T-2
/þ
4-Methoxy-N-metamfetamine
Me-MA
/þ
6-Chloro-MDMA
—
/þ
N-(4-Ethylthio-2,5-dimethoxyphenethyl)hydroxylamine
HOT-2
/þ
2,5-Dimethoxy-4-iodo-phenethylamine
2C-I
/þ
4-Methoxy-N-ethylamfetamine
—
/þ
N-Substituted amfetamines without ring substitution /P
Separation/identification
N-Hydroxyamfetamine
N-OHA
(For analytical data see Tables 11.5 and 11.6.) Direct injection of psilocybe mushroom extracts onto a GC column will convert psilocybin to psilocin by thermal dephosphorylation and only psilocin will be detected. Thus prior derivatisation is necessary if psilocybin is to be detected. To eliminate sugars that may interfere with derivatisation, 1 mL of acetone is added to the methanolic solution and the mixture allowed to stand for 30 min and then filtered. The solution is taken to dryness in a stream of air. Pyridine (15 mL), TMCS (15 mL) and BSTFA (100 mL) are added and heated at 100 C for 30 min. Psilocin is converted to psilocin di-TMS and psilocybin to psilocybin tri-TMS. LC-MS and LC-MS-MS have also been used to analyse the constituents of magic mushrooms (Kamata et al. 2005).
N,N-Dimetamfetamine
—
/
N-Acetylamfetamine
—
/
Di-(1-phenylisopropyl)amine
DIPA
/
N,N-Dimethyl-5-methoxytryptamine
5-MeO-DMT
/þ
N,N-Di-(n)-propyltryptamine
DPT
/þ
4-Acetoxy-N,N-di-isopropyltryptamine
—
/
a-Methyltryptamine
a-MT
/
1-PEA
/
'Designer Drugs' Although a few ring-substituted phenethylamines (e.g. 2,5-dimethoxy4-bromoamfetamine (DOB), 4-bromo-2,5-dimethoxyamfetamine) had been subject to limited abuse since the 1960s, it was not until
Tryptamines
Other phenylalkylamines and miscellaneous 1-Phenethylamine N-Methyl-1-phenethylamine
N-Me-PEA
/
4-Methyl-1-phenethylamine
4-Me-PEA
/
1-Phenyl-3-butanamine
—
/
N-Benzylpiperazine
BZP
/
Methcathinone
—
þ/þ
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:22
206
Drugs of Abuse
necessarily be complete, shows that the phenethylamines comprise the largest group. Ring-substituted compounds were more common than N-substituted homologues without ring substitution. The substances shown in Table 11.7 have appeared both as powders and as tablets, often manufactured, packaged or marked in such a way that they may appear to the user to be amfetamine or MDMA. Considerable scope exists to develop further series of phenethylamine-related ‘designer drugs’. Thus ring-substituted analogues of cathinone and methcathinone might have MDMA-like activity. As mentioned earlier, the EU, via Europol and EMCDDA, carry out risk assessments on new synthetic drugs. Since 1997 ten risk assessments have been undertaken on the following drugs: MBDB, 4-MTA, GHB, ketamine, p-methoxymethamphetamine (PMMA), 2C-1, 2C-T-2, 2CT-7, TMA-2 and benzylpiperazine (BZP). Various studies have outlined the analytical methods available to separate and identify these and other designer drugs (Blachut et al. 2002; Furnari et al. 1998; Poortman, Lock 1999; Tsai et al. 2006) A detailed chemical identification of DOB has been described using CE, IR spectroscopy and GC-MS (da Costa et al. 2007). Raman spectroscopy has been used as a screen for tablets containing DOB (Bell et al. 2007). Piperazines have increasingly become an alternative to amfetaminederived drugs of abuse (EMCDDA 2005). Among the most prominent of these have been N-benzylpiperazine (BZP), 1-(3-trifluoromethylphenyl) piperazine (TFMPP) and 1-(3-chlorophenyl)piperazine (mCPP). The legal status of piperazines is very inconsistent and many European countries are considering control measures, while others already control mCPP (EMCDDA 2005). In the USA, BZP and TFMPP were temporarily placed into Schedule 1 of the Controlled Substances Act in 2002 followed by final placement of BZP into Schedule 1 2004. In 2003 both BZP and TFMPP were controlled in Japan. The analysis of aryl-piperazines has been carried out by HPLC and GC-MS with and without derivatisation (de Boer et al. 2001; Tsutsumi et al. 2005). Analytical profiles covering a range of techniques have been reported for a number of tryptamines (Spratley et al. 2005). The approach to the analysis of these compounds, especially the phenethylamine-related ‘designer drugs’, could be in line with the general procedure outlined for powders and with specific reference to the analytical procedures employed to analyse MDMA or amfetamine. Those substances listed in UN 1971 or which are controlled in UK by the Misuse of Drugs Act 1971 are shown by (þ) in Table 11.7. Substances pending control in the UK are shown by ‘P’. In the USA, unscheduled substances may still be deemed to be controlled by virtue of the Controlled Substances Analogue Enforcement Act 1986.
Conclusion Many other compounds are encountered in the laboratory as ‘drugs of abuse’ such as opium, phencyclidine and analogues, tryptamines, barbiturates, methadone, morphine, dihydrocodeine, ephedrine, ketamine and alkyl nitrites. However, the general approach to the analysis of an unknown substance previously outlined should pose no difficulty to the identification of any of these drugs. Analytical information and background information on many of the drugs can be found in some of the general texts in the area (CND Analytical 1994; Cole, Caddy 1995; Gough 1991; Karch 1996, 1998; Klein et al. 1989; Redda 1989; Shulgin, Shulgin 1991; Smith 2005; UN 1994; Weaver, Yeung 1995).
References Aalberg L et al. (2005). Development of a harmonized method for the profiling of amphetamines. I. Synthesis of standards and compilation of analytical data. Forensic Sci Int 149: 219–229. Aalberg L et al. (2005). Development of a harmonized method for the profiling of amphetamines. II. Stability of impurities in organic solvents. Forensic Sci Int 149: 231–241. Aitken CG (1999). Sampling – how big a sample? J Forensic Sci 44: 750–760. Anastos N et al. (2005). Capillary electrophoresis for forensic drug analysis. Talanta 67: 269–279. Andersson K et al. (2007). Development of a harmonised method for the profiling of amphetamines: III. Development of the gas chromatographic method. Forensic Sci Int 169: 50–63.
Andersson K et al. (2007). Development of a harmonised method for the profiling of amphetamines: IV. Optimisation of sample preparation. Forensic Sci. Int 169: 64–76. Andersson K et al. (2007). Development of a harmonised method for the profiling of amphetamines VI: Evaluation of methods for comparison of amphetamine. Forensic Sci Int 169: 86–99. Baker PB et al. (1980). Determination of the distribution of cannabinoids in cannabis resin using high performance liquid chromatography. J Anal Toxicol 4: 145–152. Bell SE et al. (2007). Screening tablets for DOB using surface-enhanced Raman spectroscopy. J Forensic Sci 52: 1063–1067. Benson S et al. (2006). Forensic applications of isotope ratio mass spectrometry – a review. Forensic Sci Int 157: 1–22. Besacier F, Chaudron-Thoxet H (1999). Chemical profiling of illicit heroin samples. Forensic Sci Rev 11: 105–119. Blachut D et al. (2002) Identification and synthesis of some contaminants present in 4-methoxyamphetamine (PMA) prepared by the Leuckart method. Forensic Sci Int 127: 45-62. Blackledge RD, Miller MD (1991). The identification of GHB. Microgram 24: 172–179. Bohn M et al. (1993). Synthesis markers in illegally manufactured 34-methylenedioxyamphetamine and 3,4-methylenedioxymethamphetamine. Int J Legal Med 106: 19–23. Borner S, Brenneisen R (1987). Determination of tryptamine derivatives in hallucinogenic mushrooms using high-performance liquid chromatography with photodiode array detection. J Chromatogr 408: 402–408. Carter JF et al. (2005). Isotope ratio mass spectrometry as a tool for forensic investigation (examples from recent studies). Sci Justice 45: 141–149. UN (1988). Recommended Methods for Testing Benzodiazepine Derivatives under International Control. New York: United Nations, 37–38. Casale J et al. (2006). Stable isotope analyses of heroin seized from the merchant vessel Pong Su. J Forensic Sci 51: 603–606. Casale JF et al. (2007). Four new illicit cocaine impurities from the oxidation of crude cocaine base: formation and characterization of the diastereomeric 2,3dihydroxy-3-phenylpropionylecgonine methyl esters from cis- and trans-cinnamoylcocaine. J Forensic Sci 52: 860–866. Chiong DM et al. (1992). The analysis and identification of steroids. J Forensic Sci 37: 488–502. Christian DR (2003). Forensic Investigation of Clandestine Laboratories: Boca Raton, FL: CRC Press. Ciolino LA et al. (2001). The chemical interconversion of GHB and GBL: forensic issues and implications. J Forensic Sci 46: 1315–1323. Clarke AB, Clark CC (1990). Sampling of multi-unit drug exhibits. J Forensic Sci 35: 713–719. Clarke CC (1989). The differentiation of lysergic acid diethylamide (LSD) from Nmethyl-N-propyl and N-butyl amides of lysergic acid. J Forensic Sci 34: 532–546. CND Analytical (1989). Analytical Profile of the Anabolic Steroids. Auburn, AL: CND Analytical Inc. CND Analytical (1994). Forensic and Analytical Chemistry of Clandestine Phenethylamines. Auburn, AL: CND Analytical Inc. Cole MD (1998). Occluded solvent analysis as a basis for heroin and cocaine sample differentiation. Forensic Sci Rev 10: 113–120. Cole MD, Caddy B (1995). The Analysis of Drugs of Abuse: An instruction manual. New York: Ellis Horwood. Collins M et al. (2006). Chemical profiling of heroin recovered from the North Korean merchant vessel Pong Su. J Forensic Sci 51: 597–602. Colon M et al. (1993). Representative sampling of street drug exhibits. J Forensic Sci 38: 641–648. da Costa JL et al. (2007). Chemical identification of 2,5-dimethoxy-4-bromoamphetamine (DOB). Forensic Sci Int 173: 130–136. Dahlen J, Vriesman T (2002). Simultaneous analysis of gamma-hydroxybutyric acid, gamma-butyrolactone, and 1,4-butanediol by micellar electrokinetic chromatography. Forensic Sci Int 125: 113–119. Dams R et al. (2001). Heroin impurity profiling: trends throughout a decade of experimenting. Forensic Sci Int 123: 81–88. de Boer D et al. (2001). Piperazine-like compounds: a new group of designer drugsof-abuse on the European market. Forensic Sci Int 121: 47–56. de Korompay A et al. (2008). Supported liquid-liquid extraction of the active ingredient (3,4-methylenedioxymethylamphetamine) from ecstasy tablets for isotopic analysis. J Chromatogr A 1178: 1–8. Dufey V et al. (2007). A quick and automated method for profiling heroin samples for tactical intelligence purposes. Forensic Sci Int 169: 108–117. El Haj BM et al. (2004). Heroin profiling: mannitol hexaacetate as an unusual ingredient of some illicit drug seizures. Forensic Sci Int 145: 41–46. Elsherbini SH (1998). Cocaine base identification and quantification. Forensic Sci Rev 10: 1–12. Elsohly MA, Slade D (2005). Chemical constituents of marijuana: the complex mixture of natural cannabinoids. Life Sci 78: 539–548. EMCDDA (2005). Annual report on the state of the drugs problem in the European Union. Lisbon: European Monitoring Centre for Drugs and Drug Addiction.
Clarke's Analysis of Drugs and Poisons Chapter No. 11 Dated: 10/3/2011 At Time: 17:45:22
References Fanali S et al. (1998). New strategies for chiral analysis of drugs by capillary electrophoresis. Forensic Sci Int 92: 137–155. Fineschi V et al. (2002). The cocaine “body stuffer” syndrome: a fatal case. Forensic Sci Int 126: 7–10. Frank RS et al. (1991). Representative sampling of drug seizures in multiple containers. J. Forensic Sci 36: 350–357. Furnari C et al. (1998). Identification of 3,4-methylenedioxyamphetamine analogs encountered in clandestine tablets. Forensic Sci Int 92: 49–58. Gill JR, Graham SM (2002). Ten years of “body packers” in New York City: 50 deaths. J Forensic Sci 47: 843–846. Gilmore S et al. (2007). Organelle DNA haplotypes reflect crop-use characteristics and geographic origins of Cannabis sativa. Forensic Sci Int 172: 179–190. Gostic T, Klemenc S (2007). Evidence on unusual way of cocaine smuggling: cocaine–polymethyl methacrylate (PMMA) solid solution – study of clandestine laboratory samples. Forensic Sci Int 169: 210–219. Gough TA (1991). The Analysis of Drugs of Abuse. New York: Wiley. Hardwick S, King L (2008). Home Office Cannabis Potency Study. London:Home Office. Hennessy SA et al. (2004). The reactivity of gamma-hydroxybutyric acid (GHB) and gamma-butyrolactone (GBL) in alcoholic solutions. J Forensic Sci 49: 1220–1229. Huizer H (1983). Analytical studies on illicit heroin. II. Comparison of samples. J Forensic Sci 28: 40–48. Japp M et al. (1988). Collection of analytical data for benzodiazepines and benzophenones. J Chromatogr 439: 317–339. Kamata T et al. (2005). Liquid chromatography–mass spectrometric and liquid chromatography-tandem mass spectrometric determination of hallucinogenic indoles psilocin and psilocybin in “magic mushroom” samples. J Forensic Sci 50: 336–340. Karch SB (1996). The Pathology of Drug Abuse, 2nd edn. New York: CRC Press. Karch SB (1998). Drug Abuse Handbook. London: CRC Press. Klein M et al. (1989). Clandestinely Produced Drugs, Analogues and Precursors: Problems and solutions. Washington, DC: United States Department of Justice Drug Enforcement Administration. Knops LA et al. (2006). Capillary electrophoretic analysis of phosphorus species in clandestine methamphetamine laboratory samples. J Forensic Sci 51: 82–86. Koper C et al. (2007). Elemental analysis of 3,4-methylenedioxymethamphetamine (MDMA): A tool to determine the synthesis method and trace links. Forensic Sci Int 171: 171–179. Kuwayama K et al. (2007). Contribution of thermal desorption and liquid-liquid extraction for identification and profiling of impurities in methamphetamine by gas chromatography-mass spectrometry. Forensic Sci Int 171: 9–15. Lebelle MJ et al. (1995). Chiral identification and determination of ephedrine, pseudoephedrine, methamphetamine and methcathinone by gas chromatography and nuclear magnetic resonance. Forensic Sci Int 71: 215–223. Lee JS et al. (2006). Analysis of the impurities in the methamphetamine synthesized by three different methods from ephedrine and pseudoephedrine. Forensic Sci Int 161: 209–215. Lee JS et al. (2007). Monitoring precursor chemicals of methamphetamine through enantiomer profiling. Forensic Sci Int 173: 68–72. Lee MM (1995). The identification of cathinone in Khat (Catha adulis): a time study. J Forensic Sci 40: 116–121. Lewis R et al. (2005). Distribution of the principal cannabinoids within bars of compressed cannabis resin. Anal Chim Acta 538: 399–405. Lociciro S et al. (2007). Cocaine profiling for strategic intelligence purposes, a crossborder project between France and Switzerland. Part I. Optimisation and harmonisation of the profiling method. Forensic Sci Int 167: 220–228. Lock E et al. (2007). Development of a harmonised method for the profiling of amphetamines V: Determination of the variability of the optimised method. Forensic Sci Int 169: 77–85. Logan BK et al. (1989). A simple laboratory test for the determination of the chemical form of cocaine. J Forensic Sci 34: 678–681. Magg V (2003). Decriminalisation of cannabis use in Switzerland from an international perspective- European, American and Australian experiences. Int J Drug Policy 14: 279–281. Martyny JW et al. (2007). Chemical concentrations and contaminations associated with clandestine methamphetamine laboratories. J Chem Health Safety 14: 40–52. McDermott SD, Power JD (2005). Drug smuggling using clothing impregnated with cocaine. J Forensic Sci 50: 1423–1425.
207
McDonald PA et al. (1984). An analytical study of illicit lysergide. J Forensic Sci 29: 120–130. Mesmer MZ, Satzger RD (1998). Determination of gamma-hydroxybutyrate (GHB) and gamma-butyrolactone (GBL) by HPLC/UV-VIS spectrophotometry and HPLC/thermospray mass spetrometry. J Forensic Sci 43: 489–492. Microgram (2003) Cocaine impregnated silicone in baseball cap parts in Peru. Microgram Bulletin 36: 271. Moore JM, Casale JF (1994). In-depth chromatographic analyses of illicit cocaine and its precursor, coca leaves. J Chromatogr A 674: 165–205. Moore JM, Casale JF (1998). Cocaine profiling methodology – recent advances. Forensic Sci Rev 10: 13–15. Palhol F et al. (2004). 15N/14N isotope ratio and stratistical analysis: an efficient way of linking seized ecstasy tablets. Anal Chim Acta 510: 1–8. Poortman AJ, Lock E (1999). Analytical profile of 4-methylthioamphetamine (4MTA), a new street drug. Forensic Sci Int 100: 221–233. Qi Y et al. (2006). Australian Federal Police seizures of illicit crystalline methamphetamine (‘ice’) 1998-2002: impurity analysis. Forensic Sci Int 164: 201–210. Redda KK (1989). Cocaine, Marijuana, Designer Drugs, Chemistry, Pharmacology and Behaviour. Boca Raton, FL: CRC Press. Renton RJ et al. (1993). A study of the precursors, intermediates and reaction byproducts in the synthesis of 3,4-methylenedioxymethylamphetamine and its application to forensic drug analysis. Forensic Sci Int 60: 189–202. Saudan C et al. (2007). Carbon isotopic ratio analysis by gas chromatography/ combustion/isotope ratio mass spectrometry for the detection of gammahydroxybutyric acid (GHB) administration to humans. Rapid Commun. Mass Spectrom 21: 3956–3962. Sellers JK et al. (1996). High performance liquid chromatographic analysis of enantiomeric composition of abused drugs. Forensic Sci Int 8: 91–108. Seta S et al. (1994). Impurity profiling analysis of illicit drugs. Forensic Sci Int 69 (special issue): 1–102. Shulgin A, Shulgin A (1991). PiHKAL: A chemical love story. Berkeley, CA: Transform Press. Smith FP (2005). Handbook of Forensic Drug Analysis. New York: Elsevier. Spratley TK et al. (2005). Analytical profiles for five "designer" tryptamines. Microgram 3: 54–68. Stromberg L et al. (2000). Heroin impurity profiling. A harmonization study for retrospective comparisons. Forensic Sci Int 114: 67–88. Takekawa K et al. (2007). Methamphetamine body packer: acute poisoning death due to massive leaking of methamphetamine. J Forensic Sci 52: 1219–1222. Tsai CC et al. (2006). Optimization of the separation and on-line sample concentration of phenethylamine designer drugs with capillary electrophoresis-fluorescence detection. J.Chromatogr A 1101: 319–323. Tsutsumi H et al. (2005). Development of simultaneous gas chromatography–mass spectrometric and liquid chromatography–electrospray ionization mass spectrometric determination method for the new designer drugs, N-benzylpiperazine (BZP), 1-(3-trifluoromethylphenyl)piperazine (TFMPP) and their main metabolites in urine. J Chromatogr B Analyt Technol Biomed Life Sci 819: 315–322. Tzidony D, Ravreby M (1992). A statistical approach to drug sampling: a case study. J Forensic Sci 371549. UN (1994). Rapid Testing Methods of Drugs of Abuse. New York: United Nations. UN (1998). Recommended Methods for Testing Opium, Morphine and Heroin. New York: United Nations. Waddell RJ et al. (2004). Classification of ecstasy tablets using trace metal analysis with the application of chemometric procedures and artificial neural network algorithms. Analyst 129: 235–240. Waddell-Smith RJ (2007). A review of recent advances in impurity profiling of illicit MDMA samples. J Forensic Sci 52: 1297–1304. Weaver K, Yeung E (1995). An Analyst’s Guide to the Investigation of Clandestine Laboratories. Ottawa, ON: Drug Analysis Service. Weyermann C et al. (2008). Drug intelligence based on MDMA tablets data I. Organic impurities profiling. Forensic Sci Int 177: 11–16. Zhang D et al. (2004). Component analysis of illicit heroin samples with GC/MS and its application in source identification. J Forensic Sci 49: 81–86. Zhang D et al. (2005). Origin differentiation of heroin sample and its acetylating agent with (13)C isotope ratio mass spectrometry. Eur J Mass Spectrom 11: 277–285.
Clarke's Analysis of Drugs and Poisons Chapter No. 12 Dated: 10/3/2011 At Time: 14:35:15
CHAPTER
12
Medicinal Products AC Moffat and AG Davidson
Introduction Medicinal products should be safe and efficacious. Manufacturers of medicinal products are required by law to possess marketing authorisations from government regulatory agencies in countries in which their products are marketed and to manufacture their products in compliance with current Good (Pharmaceutical) Manufacturing Practice (cGMP) standards. These requirements include conducting appropriate quality control tests to check that the product conforms to a specification that assures its safety and efficacy. Typical specifications include tests to verify the chemical composition and physical properties of the medicine and to ensure that the medicine is not contaminated by microorganisms or other substances. Although authentic medicinal products are subject to quality control testing by the manufacturer, they may have been counterfeited, adulterated or stored poorly, and therefore need to be checked for the following purposes: n n n n n n n
Determine or confirm their composition Assess their suitability for use Investigate defects Identify unknown medicinal products Identify contaminants Determine whether the products have been adulterated Establish whether the products are counterfeit.
Independent quality checks of medicinal products may be carried out by official medicines control laboratories as part of a government surveillance programme, and by hospital quality control laboratories, public analyst laboratories and forensic laboratories. This chapter describes the philosophies, strategies and methodologies for the analysis and testing of medicinal products by laboratories that may not have access to the manufacturers’ research and development data or to the manufacturers’ test methods, specifications and reference materials for the products. It focuses on the information about the medicinal product that can be derived from other sources and on the conclusions that can then be made about the quality of the product.
Submission of samples and choice of tests A wide range of tests that utilise a variety of analytical techniques are available to laboratories. Samples submitted for testing should therefore be accompanied by a clear written request about the nature of the investigation required. The provision of relevant background information about the sample, including the reason for the request, allows the laboratory to choose the most appropriate tests, analytical techniques and, where relevant, acceptance criteria (i.e. criteria that allow an objective assessment to be made about the quality of the product). In most cases that involve checking of the quality of a medicinal product, it is preferable to use the test methods in the product specification defined in the manufacturer’s marketing authorisation for the product. This is because the product specifications, including the test methods and acceptance criteria, have been assessed by the relevant government regulatory agency as being valid and justified. Some manufacturers publish their analytical methods in the scientific literature. Consequently, it may be worthwhile to check the literature or maintain a 208
library of published methods for widely used medicinal products. Alternatively, in certain circumstances, manufacturers may provide the laboratory with the test methods, acceptance criteria and reference materials. If the laboratory does not have access to the authorised finished product specification, including the test methods and acceptance criteria, pharmacopoeial monographs can be used, where applicable (see later). In the absence of a pharmacopoeial monograph, alternative approaches are required to obtain as much information as possible about the quality of the medicinal product. This requires the development of valid test methods to measure relevant quality parameters and the application of generally accepted criteria to assess the quality. However, if the laboratory uses analytical techniques different from those used by the manufacturer (e.g. a high performance liquid chromatographic (HPLC) technique instead of an ultraviolet (UV) spectrophotometric technique), the results may differ significantly from those obtained by the manufacturer, particularly if high levels of impurities are present. This is because analytical techniques differ in their accuracy, precision, selectivity, and/or specificity and sensitivity. It is therefore important to consider the findings in relation to the technique used. This chapter describes the tests that should be carried out to provide information about different aspects of medicinal products. A summary of the principal tests and techniques for checking known products and investigating unknown products is given in Table 12.1.
Counterfeit medicines The World Health Organization (WHO) describes counterfeit medicines as part of the broader phenomenon of substandard pharmaceuticals – medicines manufactured below established standards of quality and therefore dangerous to patients’ health and ineffective for the treatment of diseases. The difference is that counterfeits are deliberately and fraudulently mislabelled with respect to identity or source. Counterfeiting occurs with both branded and generic products. Counterfeit medicines may: n contain no active ingredient n contain the wrong active ingredient (e.g. a cheap antibiotic instead of an expensive antibiotic) n contain an incorrect (usually low) quantity of the active ingredient n be in low-quality packaging n be manufactured using low-quality active ingredient or excipients n be manufactured under poor standards of cGMP compliance. Counterfeit products that contain no active ingredient and those that contain the wrong active ingredient or the correct active ingredient in the wrong amount can be detected by carrying out appropriate identification and quantitative tests (see below). In situations where resources for accurate and precise quantitative testing are limited, for example in developing countries, basic tests, including semi-quantitative tests, may be sufficient to detect these types of counterfeit medicines (see below). It may also be possible to identify products as being counterfeit by their general appearance (colour, markings, etc.), particularly when the appearance differs from a genuine batch of the product. For this reason, laboratories that regularly undertake checks for counterfeit medicines maintain a stock of genuine reference products for comparison. Examination of the labelling should also be carried out to check the
Clarke's Analysis of Drugs and Poisons Chapter No. 12 Dated: 10/3/2011 At Time: 14:35:15
Direct comparison of products
209
Table 12.1 Summary of tests and techniques Purpose
Test(s)
1. General check of quality
Test to full specification
Analytical technique(s)(a)
Comment Refer to typical monographs for dosage forms in pharmacopoeias
Identity
See 2 below
Assay
See 5 below
Homogeneity
Uniformity of content
Contaminants
HPLC, GC, TLC, CE, etc.
Release of active ingredient
Dissolution test
Microbial quality
TVAC, specific microorganisms, sterility test
2. Confirm identity of product
Instrumental tests for active ingredient (s)
NIR, IR or combination of UV, HPLC, TLC, colour reaction, melting point, etc.
WHO basic tests
Colour reactions, TLC
3. Rapid identification of unknown product
Visual comparison of physical characteristics (size, colour, shape, etc.) against library
TICTAC, Identidex, etc.
See Chapter 13
Instrumental checks provide greater assurance about identity
Simple screening tests
UV, TLC, HPLC, colour tests
See Chapters 32, 39, 41 and 30
4. Unambiguous identification of unknown products
Specific identification tests
IR, NMR, LC-MS, GC-MS, CE, chiral HPLC (for enantiomeric substances), AAS (for inorganic moieties)
See relevant chapters on these techniques
5. Quantification of active ingredient(s)
Assay
HPLC, GC, UV, CE
See relevant chapters on these techniques
6. Homogeneity of active ingredient
Uniformity of content (or mass)
Assay of several (e.g. 10) dosage units or subsamples of the product
Uniformity of content preferred, particularly for unit doses with a content of active substance The OP compounds can be analysed without prior derivatisation. Screening for these compounds by GC is assisted greatly by a dual FID–NPD system. Detection by NPD gives a 10-fold increase in sensitivity for phosphorus-containing compounds compared with those detected using nitrogen only. This is very important because many OP pesticides are highly toxic and their concentrations in biological samples are low. Mass fragmentation of these compounds to typical ions (which vary widely in intensity) allows the chemical class to be determined. Molecular ions of OP compounds can be monitored using the thermospray LC-MS technique (Niessen 1999). Both protonated and ammoniated molecules are detected in the positive-ion mode without any fragmentation. Phosphates and phosphorodithioates appear to give a better response than other chemical classes. Phosphorothioates with chlorine, bromine or nitrate substitution on the ring are especially intense. The thermospray mass spectra obtained by operating in the negative-ion mode are more compound dependent. Human exposure to OP pesticides is often assessed by measuring general dialkyl phosphate metabolites of OPs in urine. Six common urinary dialkyl metabolites can be quantified by GC-MS to confirm cumulative exposure to OP pesticides (De Alwis et al. 2006). An experiment on animals demonstrated the possibility of using hair analysis by GC-NCI-MS for monitoring low-level exposure to diazinon (Tutudaki, Tsatsakis 2005). Hair was also shown to be a promising material by documenting non-fatal intoxication in two men, one exposed to alachlor a year before sampling and the other exposed to carbofuran 14 days before sampling. The concentrations of alachlor in five analysed hair segments were between 12 and 136 pg/mg. Carbofuran and its main metabolite (3-hydroxycarbofuran) were detected in the hair strand (global analysis) at concentrations of 207 and 164 pg/mg, respectively (Dulaurent et al. 2008). Blood and tissue levels Pre-treatment plasma concentrations (mg/L) in cases of non-fatal organophosphate poisoning are summarised below (Kłys et al. 1989, 1991, 1992). Chlorphenvinphos
Fenitrothion
Phosalone
n = 20
n = 15
n = 16
0.1–10.6
0.096–0.35
0.005–0.39
¼ AChE activity in international units per millilitre ðIU=mLÞ The precision of the method is 210 IU/mL. Normal values for AChE activity in whole blood range from 3500 IU/mL to 8000 IU/mL. Commercial kits for the determination of AChE activity in red blood cells, whole blood and plasma are available (EQM Research, Inc., Cincinnati, OH, USA).
Many studies have been carried out on the distribution of organophosphate in cases of fatal poisoning with this group of pesticides. Their concentrations (mg/L or mg/kg) in various body fluids and tissues can help to determine the specimens of choice in postmortem toxicological analysis. The following data have been reported: For chlorphenvinphos (Kłys et al. 1989):
Determination of organophosphates in urine Most compounds of
this chemical class are hydrolysed rapidly by plasma and tissue enzymes with the production of many metabolites. Metabolites and their conjugates are excreted in urine and are known to be unstable in stored specimens. To derive data that accurately represent the true degree of exposure, as indicated by the concentration of OP compounds, it is essential to obtain and analyse samples as soon as possible after an incident. Urine samples should be analysed within a week of obtaining the sample and kept at 20 C prior to analysis (Comer et al. 1976).
Blood
Liver
Kidney
Lung
Brain
n=7
n=7
n=5
n=7
n=6
0.30–15.0
0.32–15.94
0.16–15.0
0.80–15.94
0.1–4.3
Blood
Liver
Kidney
Colorimetric procedure In the colorimetric procedure (Namera et al.
n=6
n=5
n=2
n=5
1.9–14.8
0.8–11.5
3.9–4.5
0.04–1.15
2000), to 1 mL of urine (pH 5–8) add 0.1 mL of 45% (w/v) of 4-(4nitrobenzyl)pyridine (NBP) solution in acetone, vortex for 30 s and heat
For fenitrothion (Kłys et al. 1992): Brain
Clarke's Analysis of Drugs and Poisons Chapter No. 16 Dated: 10/3/2011 At Time: 18:13:16
280
Pesticides
Values reported in single cases are given below. For dimethoate (Tarbah et al. 2007): Blood
Urine
Brain
Liver
Lung
Myocardial muscle
Skeletal muscle Kidney
Gall bladder
Stomach contents
38
0.47
2.2
4.6
7.6
7.6
21
31
104
In this case the blood alcohol concentration was 2.85 g/L and the blood was also positive for cyclohexanone and cyclohexanol, a mixture used as a vehicle in the commercial product. For omethoate (Pavlic et al. 2002): Cardiac blood
Urine
Bile
Liver
Kidney
Stomach contents
208
225
524
341
505
48 223
For dichlorvos (Moriya et al. 1999): Vitreous Thoracic Thoracic Cerebro- Perispinal cardial humour aorta inferior fluid blood vena cava fluid blood
Bile
0.043
8.99 0.542
0.082
0.027
0.438 0.067
Spleen Stomach contents
2929
55
intensity, but together with diagnostic fragments they enable identification to be made. In LC-MS methods, the carbamates do not present a serious problem. Positive ion detection with a soft ionisation technique is the method of choice (Niessen 1999). Some compounds, e.g. carbofuran and promecarb, can be determined by a chemiluminescence method (on-line conversion into methylamine by irradiation with UV light, and then reaction with tris(2,20 -bipyridine)ruthenium(III), which was generated through the on-line photooxidation of tris-2,20 -bipyridineruthenium(II) with peroxydisulfate) (Perez-Ruiz et al. 2002). Toxicity Carbamate pesticides have a similar action to that of the OP compounds in causing a decrease in cholinesterase activity, but the binding to the active site of the cholinesterase enzyme is reversible. Consequently, although the symptoms are practically identical to those of organophosphorus poisoning, they have a shorter duration. Blood and tissue levels Blood carbofurane concentrations in 15 nonfatally poisoned patients ranged from 0.15 to 2.78 mg/L, and in seven fatal cases concentration ranges were as follows (Kłys, Bialka 1990):
In this case no dichlorvos was detected in blood from the left and right cardiac chambers, the pulmonary arteries and veins or the right femoral vein. Dichlovos was also absent from the urine, cerebrum, lung, kidney and right femoral muscle. For chlorpyrifos-methyl (Moriya et al. 1999):
Blood
Liver
Kidney
0.31–3.90
0.09–9.34
0.54–6.29
Blood Left cardiac chambers Right cardiac chambers
Pulmonary arteries
Pulmonary veins Thoracic aorta
Thoracic inferior vena cava Right femoral vein
1.01
1.71
4.15
2.83
0.99
2.24
0.62
Cerebrospinal fluid
Pericardial fluid
Vitreous humour
Lung left hilus
Myocardium
Right kidney
Right femoral muscle
0.01
0.01
0.01
8.60
0.49
0.47
0.39
Liver deep right lobe
Spleen
Cerebrum
Stomach contents
Bile
Urine
1.41
0.666
0.38
2041
ND
ND
ND – not detected.
Concentrations of other OP compound in postmortem blood have also been reported: Compound
Concentration (mg/L)
Reference
Chlorpyrifos
0.21–2.05 (n = 6)
(Park et al. 2009)
Diazinon
0.24–2.82 (n = 4)
(Park et al. 2009)
Diazinon
0.4–277 (n = 3)
(Poklis et al. 1980)
Malathion
0.35–1.32 (n = 4)
(Park et al. 2009)
Malathion
175–517 (n = 6)
(Jadhav et al. 1992)
Parathion
0.21–19.64 (n = 17)
(Park et al. 2009)
Phosalone
0.024–0.19 (n = 3)
(Kłys et al. 1991)
Carbamates Analysis These compounds can be divided into various subclasses, characterised by their different thermal stabilities. N-Methylcarbamates give thermal decomposition products, mainly substituted phenols. The molecular ions that arise from these products are more abundant in mass spectra. The compounds from other subclasses are more thermally stable. The molecular ions are of low
In a case of oral ingestion of thiodicarb (which is unstable under acidic condition and rapidly hydrolysed to methomyl), the parent compound was detected only in gastric contents at a concentration of 24.3 mg/L. In all other fluids and tissues, various concentrations of methomyl – its metabolite – were detected. Methomyl concentrations (mg/L or mg/kg) were as follows (Hoizey et al. 2008):
Stomach Peripheral Urine contents blood
Bile
Liver
Kidney Lung
Brain
Heart
19.9
2.7
0,7
1.7
9.3
3.6
0.7
8.5
1.5
Traces of methomyl were also detected in vitreous humour. Moreover, blood concentrations (mg/L) of zolpidem (2.87), bromazepam (2.39), nordazepam (4.21) and levopromazine (0.64) were above their common therapeutic ranges. In contrast to this case, blood and liver methomyl concentrations following methomyl ingestion in several published fatal case reports varied between 0.003 and 63.5 mg/L, and between ‘not detectable’ and 1.2 mg/kg, respectively (Hoizey et al. 2008). Blood methomyl levels in nine fatal cases ranged from 8 mg/L to 56 mg/L (Baselt 2004, pp. 700–701).
Clarke's Analysis of Drugs and Poisons Chapter No. 16 Dated: 10/3/2011 At Time: 18:13:17
Quantification of pesticides
281
Blood methomyl concentrations (mg/L) in a case reported by Moriya and Hashimoto (1999) were site-dependent as shown below: Blood Left cardiac chambers
Right cardiac chambers
Pulmonary arteries
Pulmonary veins
Thoracic aorta
Thoracic inferior vena cava
Right femoral vein
4.89
1.08
4.75
4.09
7.00
0.56
3.91
Cerebrospinal fluid
Pericardial fluid
Vitreous humour
Lung left hilus
Lung right hilus
Right kidney
Right femoral muscle
5.37
4.75
3.67
1.17
1.86
ND
2.61
Liver deep right lobe
Spleen
Cerebrum
Stomach contents
Bile
Urine
Myocardium
ND
ND
2.26
340
5.68
4.76
0.08
ND, not detected.
In three cases involving fatal ingestion of carbaryl, concentration ranges in blood and urine (mg/L) and tissues(mg/kg) were as follows (Baselt 2004, pp. 167–169):
In a case of endrin suicide poisoning, its concentrations (mg/L or mg/kg) in different postmortem materials were as follows (Moriya, Hashimoto 1999):
Blood
Liver
Kidney
Brain
Urine
Blood
6–27
12–29
1.9–25
4.6
31
Left cardiac chambers
Right cardiac Thoracic chambers aorta
Thoracic Right femoral inferior vena cava vein
Kidney
0.615
0.568
0.542
0.453
0.353
ND
Cerebrospinal fluid
Pericardial fluid
Vitreous humour
Lung left hilus
Lung right Myocarhilus dium
0.515
1.00
1.67
6.20
1.19
0.467
Liver deep right lobe
Urine
Cerebrum Stomach contents
Bile
Right femoral muscle
13.8
ND
1.93
2.06
2.08
In a lethal poisoning with ethiofencarb and ethanol, concentrations of the parent carbamate compound and its two major metabolites (mg/ L) in antemortem (taken on admission to hospital) and postmortem biological fluids (death occurred 3 h after admission, samples were taken at autopsy 2 days after death) were measured with the following results (Al Samarraie et al. 2009): Antemortem blood
Postmortem peripheral blood
Ethiofencarb (mg/L)
18.8
26.4
1.7
Ethiofencarb sulfoxide (mg/L)
25.6
37.9
41.2
0.5
0.9
0.4
1.2
2.6
Ethiofencarb sulfone (mg/L) Ethanol (g/kg)
Postmortem urine
1100
ND, not detected.
Pyrethrins and pyrethroids
Not analysed
Chlorinated hydrocarbons Analysis The chlorinated hydrocarbons chemical class of pesticides may
be analysed intact using chromatography with a dual FID-NPD detection system, but greater sensitivity can be achieved using an ECD. Among hyphenated techniques, GC-MS is the method of choice for this class of pesticide. The methods applied in clinical and forensic cases do not need to be highly sensitive because most compounds that belong to this class are only slightly toxic and severe symptoms of poisoning are observed only after ingestion of large quantities (several grams). Moreover, the symptoms often result from the solvents in which the chlorinated hydrocarbons are formulated. Useful references for the determination of chlorinated hydrocarbons in human or avian serum are Brock et al. (1996) and Sundberg et al. (2006) using GC after SPE, and Lo´pez et al. (2007) using GC-MS or GC-ECD. Toxicity Chlorinated hydrocarbons are neurotoxins that also damage the liver and kidneys. Major clinical features of poisoning are headache, disorientation, paraesthesia and convulsions. Blood and tissue levels A blood lindane concentration of 0.13 mg/L was measured in a patient who developed seizures after ingesting about 250 mg (Burton et al. 1991). An adult male who ingested 57 g of 1% lindane lotion and became comatose had a blood lindane concentration of 1.3 mg/L on the first day of admission to hospital and died 7 days later with a postmortem blood concentration of 0.02 mg/L (Kurt et al. 1986). In a fatal case of endosulfan poisoning in a woman, the blood endosulfan concentration was 30 mg/L (Bernardelli, Gennari 1987).
Analysis The term ‘pyrethrins’ is used collectively for the six insecticidal
constituents present in extracts of the flowers of Pyrethrum cinerariaefolium and other species. Pyrethrins comprise esters of the natural stereoisomers of chrysanthemic acid (pyrethrin I, cinerin I and jasmolin I) and the corresponding esters of pyrethric acid (pyrethrin II, cinerin II and jasmolin II). Their low photochemical stability leads to synthetic analogues (pyrethroids), which are highly toxic to insects. In recent years pyrethroids have been manufactured and used in large quantities. These contain no nitrogen and therefore both GC-FID and GC-MS are appropriate detection systems. The compounds can be analysed easily by chromatography, either without derivatisation or after methylation (Bissacot, Vassilieff 1997; Cherstniakova et al. 2006; FernandezGutierrez et al. 1998). Toxicity Pyrethrins and pyrethroids have relatively low toxicity to humans, but exposure to these compounds by inhalation can cause localised reactions to the upper and lower respiratory tract, which leads to oral and laryngeal oedema, coughing, shortness of breath and chest pain. In acutely exposed sensitised patients a serious asthmatic-type reaction can be triggered that can prove fatal within a few minutes. Nitrophenols and nitrocreosols Analysis Dinitro-o-creosol (DNOC) can be measured in blood speci-
mens by colorimetry (Smith et al. 1978). Toxicity Dinitrophenol, dinitro-o-creosol and dinoseb stimulate oxida-
tive metabolism in the mitochondria and cause profuse sweating, headache, tachycardia and fever. Blood levels Blood and urine measurements are useful as an aid to diagnosis and treatment. Symptoms of headache and malaise are noted with DNOC blood concentrations of around 40 mg/L and serious intoxication has been observed in workers with concentrations of 44–60 mg/L (Bidstrup et al. 1952). In a fatal case, the postmortem blood concentration was 75 mg/L (Bidstrup et al. 1952). In two deaths attributed to the
Clarke's Analysis of Drugs and Poisons Chapter No. 16 Dated: 10/3/2011 At Time: 18:13:18
282
Pesticides
use of dinitrophenol, its concentrations in blood were 28 and 36.1 mg/L (Miranda et al. 2006). In another case involving ingestion of dinitrophenol together with medicinal drugs, the postmortem blood contained 48.4 mg/L of dinitrophenol, 1.2 mg/L of 2-amino-4-nitrophenol, citalopram and its desmethylated metabolite (in toxic concentrations), and olanzapine, desalkylflunitrazepam and nordiazepam (in therapeutic or subtherapeutic concentrations) (Politi et al. 2007). Chlorinated phenoxy acids Analysis Substituted phenoxy acids occur in commercial products as
salts or esters. Conversion of salts by extraction and derivatisation to the corresponding methyl esters improves their chromatographic properties. The presence of isooctyl (2,2,4-trimethylpentyl) esters of chlorinated phenoxy acid herbicides can be indicated using mass chromatography with ions of m/z 41, 55, 57, 69, 71 and 85, as well as high-mass ions of significant intensity (Fysh, Whitehouse 1986). Toxicity Chlorinated phenoxy acids are corrosive chemicals that damage the skin, eyes, and respiratory and gastrointestinal tracts. Ingestion of large doses causes vomiting, abdominal pain, diarrhoea, metabolic acidosis, pulmonary oedema and coma. Some herbicide preparations of 2,4-dichlorophenoxyacetic acid (2,4-D) and 2,4,5-trichlorophenoxyacetic acid (2,4,5-T) have been shown to contain a contaminant, dioxin (2,3,7,8-tetrachlorodibenzodioxin), which is a potent teratogenic agent. Alkalinisation of the urine to increase the excretion of 2,4-D and other chlorophenoxy compounds after poisoning has proved an effective therapy. Blood and tissue levels A patient who survived after taking 7 g of 2,4-D had an initial plasma concentration of 400 mg/L (Park et al. 1977). In another non-fatal case, a maximum 2,4-D plasma level of 1031 mg/L was recorded (Rivers et al. 1970). In five fatal cases, blood 2,4-D concentrations have ranged from 58 mg/L to 826 mg/L (Baselt 2004, pp. 323–325). A patient who died 30 h after taking a mixture of 2-(2-methyl-4-chlorophenoxy)propionic acid (MCPP; 44 g), 2,4-D (39 g) and chlorpyrifos had blood concentrations of 389 mg/g MCPP and 325 mg/g 2,4-D. Postmortem tissue concentrations were also reported (Osterloh et al. 1983). Ingestion of 4-chloro-2-(methylphenoxy)acetic acid (MCPA) led to the following concentrations of MCPA and its metabolite, p-chloro-ocresol (mg/L or mg/kg) in postmortem materials (Takayasu et al. 2008):
Quaternary ammonium compounds Analysis Paraquat and diquat are not extractable by conventional LLE. A
recently published comparison study of different extraction procedures showed that a chloroform–ethanol 7 : 3 (v/v) solvent mixture was the most effective extraction solvent (Baeck et al. 2007). The diene or monoene reduction products of paraquat and diquat produced by sodium borohydride can be extracted by diethyl ether from alkaline solution for chromatography (Draffan et al. 1977). Very limited data are available for the mass spectral characterisation of these compounds (de Almeida, Yonamine 2007). Colorimetric determination of paraquat and diquat after reduction with sodium dithionite under alkaline conditions is probably the most widely used technique. Both of the bipyridylium reduction products have absorbance maxima at 396 and 379 nm. Using an ion-pairing extraction technique, a lower limit of measurement of 50 mg/L can be achieved (Jarvie, Stewart 1979). However, it is not possible to include an internal standard in this method. Radioimmunoassay (Fatori, Hunter 1980) and fluorescence polarisation immunoassay (Colbert, Coxon 1988) methods for the determination of paraquat in serum are very sensitive and require only small sample volumes, but they are not widely available. Paraquat can also be determined in serum by HPLC-UV after extraction onto a disposable cartridge of end-capped octadecyl silica (Fig. 16.4) (Croes et al. 1993) down to a concentration of 25 mg/L. Diquat may be analysed in biological specimens by most of the procedures described for paraquat. Specific HPLC procedures for paraquat and/or diquat have also been described (Ameno et al. 1995; Arys et al. 2000; Hara et al. 2007). Difenzoquat, diquat and paraquat can be quantified in human whole blood in addition to other quaternary ammonium compounds by LCMS(-MS) with positive ESI following SPE and ion-pair chromatography (Ariffin, Anderson 2006). Toxicity Ingestion of concentrated paraquat formulations causes burning of the mouth, oesophagus and stomach, and after massive absorption patients die of multiple organ failure. Absorption of smaller amounts can lead to renal damage followed by a progressive pulmonary fibrosis that causes death from respiratory failure, in some cases after 2–3 weeks of ingestion. Treatments to reduce absorption or increase elimination have not been effective. A strongly positive urine test with the dithionite test in a sample collected more than 4 h after ingestion indicates a poor prognosis. Measurement of the plasma paraquat con-
Heart blood
Peripheral blood
Urine
Brain
Left lung
Right lung
Liver
Left kidney
Right kidney
Stomach contents
MCPA
888.3
578.1
52.2
770.9
2864
1 362
1 135
867.1
755.5
10 200
p-Chloro-o-cresol
2.16
1.92
0.24
1.64
9.06
7.41
10.36
5.56
4.85
16.45
Triazines Analysis Triazines contain several nitrogen atoms and show enhanced
NPD and FID ratios. Most triazines, which are readily amenable to GC-MS, exhibit highly characteristic mass spectra of the parent compounds and yield the important degradation products hydroxy- and desalkyl triazines. By using LC-MS with APCI and ESI, and optimising the in-source parameters, the protonated triazine molecule can be seen without fragmentation (Niessen 1999). Quantification of atrazine metabolites, e.g. atrazine mercapturate, desethyl and desisopropyl atrazine, as markers of atrazine exposure, in urine has been achieved using isotope-dilution calibration of an LC-MS(-MS) method (Nguyen et al. 2007). Toxicity Ingestion of about 100 g of atrazine can lead to coma, circulatory collapse, metabolic acidosis and gastric bleeding. This may be followed by renal failure, hepatic necrosis and a disseminated intravascular coagulopathy that may prove fatal. Haemodialysis is recommended for severe cases. Blood levels An adult died 3 days after taking approximately 100 g of atrazine together with ethylene glycol and formaldehyde; 1 h after ingestion the plasma concentration was 2 mg/L (Pommery et al. 1993).
centration is a more accurate prognostic guide. Diquat is also an irritant poison that causes vomiting, diarrhoea and epigastric pain. In severe cases, liver and renal failure, convulsions and coma may ensue, but diquat ingestion does not lead to progressive pulmonary fibrosis. Blood levels A fatal outcome is usually associated with plasma paraquat concentrations greater than 0.2 mg/L at 24 h after ingestion and 0.1 mg/ L at 48 h after ingestion (Scherrman et al. 1983). In a fatal case of accidental paraquat poisoning a blood concentration of 0.64 mg/L of paraquat was reported (Ariffin, Anderson 2006). In fatal cases of diquat poisoning, plasma diquat concentrations ranging from 0.45 mg/L to 4.5 mg/L have been recorded (Vanholder et al. 1981). Phosphides Analysis The ammonium molybdate test and commercially available
detector tubes (Guale et al. 1994) may be used as qualitative and quantitative procedures for stomach contents and non-biological materials. Phosphine can also be determined in biological samples using GC and NPD detection (Chan et al. 1983; Musshoff et al. 2008). Toxicity Hydrogen phosphide is widely used as an insecticide and rodenticide (agricultural fumigant) and is usually generated by the action of water on metallic phosphides (aluminium, magnesium or zinc). Inhaled
Clarke's Analysis of Drugs and Poisons Chapter No. 16 Dated: 10/3/2011 At Time: 18:13:18
Quantification of pesticides
283
is a quantitative thallium determination in serum and/or urine. Treatment with potassium ferrohexacyanoferrate (Prussian or Berlin blue) to absorb thallium re-circulated in the bile is effective if instituted early enough; serum and urine thallium measurements are useful in monitoring the progress of the therapy. Blood levels Blood thallium concentrations in fatal cases of poisoning have ranged from 0.5 mg/L to 11 mg/L (Baselt 2004, p. 1092). The following tissue concentrations (mg/kg) of thallium were determined in three people who died after ingestion of this poison (Sadlik 1997): Woman, age 63
Man, age 58
Boy, age 17
Liver
81.0
59.2
12.1
Kidney
62.5
38.5
11.8
In a 15-year-old boy who survived the intentional ingestion of thallium sulfate, blood and urine concentrations (mg/L) examined during treatment were as follows (Lech, Sadlik 2007): Day 14
Day 24
Day 31
Blood
0.88
0.44
0.36
Urine
2.35
3.35
5.00
Uracils Figure 16.4 HPLC chromatograms of extracted human serum with UV detector (Microspher C18 column, l = 258 nm). A, blank serum; B, spiked serum sample with (I) 500 mg/L paraquat, RRT 0.75, and (II) 2500 mg/L 1,1-diethyl4,40 -bipyridyl chloride (internal standard), RRT 1.00. Reproduced with permission from Croes et al. (1993).
phosphine is readily absorbed by the lungs. Following the ingestion of metallic phosphides, phosphine is generated in the stomach and the gas acts on the gastrointestinal and central nervous systems. In severe cases abdominal pain, vomiting, convulsions and coma develop rapidly and death usually ensues within 2 h. Blood and tissue levels Postmortem blood in a man who ingested an unknown number of aluminium phosphide tablets contained 0.5 mg/L of phosphine. The tissue phosphine concentrations were 3 mg/kg in liver and 3000 mg/kg in stomach contents (Chan et al. 1983). In another case after accidental ingestion of novel rodenticide pills also containing aluminium phosphide, the following phosphine concentrations (mg/ kg) were found: Stomach contents
Nose smear
Small intestine
0.2
0.56
0.28
Urine, femoral and heart blood, liver, kidney, bile and brain samples tested negative (Musshoff et al. 2008). Thallium Analysis Several convenient colorimetric methods can be applied to determine thallium in urine, stomach contents and suspect preparations. One of these methods (Flanagan et al. 1995) is based on measuring the absorbance of a chloroform-extractable pink–red thallium–dithizone complex from an alkaline solution that contains potassium, sodium and cyanide ions to mask interference from other metal ions. It indicates the presence of thallium in urine at concentrations of 1 g/L or more. However, the method is not specific and atomic absorption spectrophotometry is a much more reliable technique (see Chapter 43). Toxicity Poisoning with thallium salts leads to delayed damage to the peripheral and central nervous systems. The cardiorespiratory system is also affected. Loss of body hair 1–2 weeks after ingestion is a characteristic sign of thallium poisoning. The most reliable diagnostic procedure
Analysis Uracils are pyrimidine herbicides that can be analysed intact
and also after derivatisation. HPLC with UV detection is a good technique for confirmation (Lawrence, Turton 1978). Quantities of 200 ng or 20–50 ng of these compounds at 254 nm or 270–280 nm, respectively, give a response equivalent to 0.01 absorbance units (Lawrence, Turton 1978). The use of HPLC and atmospheric pressure ionisation (API, both ESI and APCI) MS in positive- and negative-ion mode for qualitative analysis of uracils (e.g. bromacil) has also been published (Schreiber et al. 2000). Toxicity No serious cases of human poisoning with these compounds
have been reported. Substituted ureas Analysis These herbicides, especially phenyl- and sulfonylureas, contain nitrogen and respond to both FID and NPD. They are very thermolabile and cannot be derivatised readily. Analysis by GC is complicated, as it is impossible to avoid the formation of their numerous decomposition products because of the high injection port temperature. To reduce the formation of additional artefactual products, inert solvents (ethyl acetate or hexane) should be used to reconstitute the extracts. Protonated substituted ureas without fragmentation have been observed by LC-MS in APCI mass spectra. The appearance of spectra obtained by thermospray LC-MS is highly dependent on the analytical conditions (Niessen 1999). LC-MS(-MS) methods with positive ion and MRM modes for measurement of dichlorophenyl urea, dichlorophenylmethyl urea, diuron and linuron as markers of phenylurea herbicide exposure and of dimethoxypyrimidine, dimethylpyrimidine and methoxymethyl triazine as markers for sulfonylurea herbicide exposure in urine have been developed by Nguyen et al. (2007). Toxicity No serious cases of human poisoning as a result of ingesting this class of pesticides have been reported.
Coumarin anticoagulants Analysis Warfarin and the superwarfarin anticoagulant rodenticides
(brodifacoum, bromadiolone, coumatetralyl and difenacoum; Fig. 16.5) can be analysed either intact or after derivatisation, by either GC or GC-MS methods, these being the most sensitive and selective. For biological samples (plasma and urine), extractive methylation using the phase-transfer reagent tetrahexylammonium hydrogensulfate, methyl
Clarke's Analysis of Drugs and Poisons Chapter No. 16 Dated: 10/3/2011 At Time: 18:13:19
284
Pesticides
iodide and then SPE has been developed as part of a systematic toxicological system (Maurer, Arlt 1998). Using MS with ions of m/z 291, 294, 295, 309, 313, 322, 324, 336, 343 and 354, the presence of 4-hydroxycoumarin anticoagulants and even the hydroxy metabolites of coumachlor, coumatetralyl and warfarin can be detected (Maurer, Arlt 1998). Five of the 4-hydroxycoumarin anticoagulants (brodifacoum, bromadiolone, coumatetralyl, difenacoum and warfarin) can also be resolved and determined in serum by HPLC with fluorimetric detection (Felice et al. 1991), using the following method. 1. To 2 mL of serum add 4 mL of acetonitrile, vortex for 10 s and then centrifuge for 5 min at 700g. 2. Decant the supernatant liquid to another test tube, add 8 mL of diethyl ether and vortex mix for 2 min and then centrifuge. 3. Separate the ethereal phase (approximately 7 mL); evaporate to dryness at 50–55 C under a stream of nitrogen. 4. Dissolve the residue in 150 mL of methanol and add 50 mL of mobile phase. 5. Inject an aliquot of 50 mL into the HPLC apparatus: * Detector: fluorescence (lex¼318 nm, lem¼390 nm). * Column: C18 Ultrasphere, 250 4.6 mm. * Mobile phase: gradient mixture of 0.05 mol/L ammonium acetate buffer (with addition of 2 mL of glacial acetic acid and 2 mL of triethylamine in 1 L of aqueous phase) and methanol (38 : 62 for 2 min to 18 : 82 at 5 min for 8 min to 10 : 90 at 13.5 min for 1.5 min to 38 : 62 at 17 min). Recoveries average from 68% (warfarin) to 98% (bromadiolone). Bromadiolone and warfarin can be quantified with a precision of at least 10% at serum concentrations of 20 mg/L, and brodifacoum, coumatetralyl and difenacoum at serum concentrations of 10 mg/L. A series of procedures for the screening, confirmation and quantification of superwarfarin anticoagulant rodenticides in serum or blood by LC-MS(-MS) has been published recently. Grobosch et al. (2006) developed a method for the simultaneous determination of five superwarfarins (brodifacoum, bromadiolone, difenacoum, difethialone and
Figure 16.5 HPLC chromatograms of extracted canine serum (fluorescence detector, Ultrasphere C18 column). A, blank canine serum; B, canine serum spiked with 50 mg/L of coumatetralyl (I), difenacoum (V) and brodifacoum (VI) and 100 mg/L of warfarin (II) and bromadiolone (III, IV). Reproduced with permission from Felice et al. (1991).
flocoumafen) and five other vitamin K antagonists (acenocoumarol, coumatetralyl, coumachlor, phenprocoumon and walfarin) in 0.5 mL serum samples. Jin et al. (2007) determined bromadiolone in 0.2 mL whole blood specimens. Extraction Adamowicz and Kała (2009) evaluated the following LC-ESI-MS method for the detection and quantification of six anticoagulants: 1. To 1 mL of blood add 50 mL (10 mg/mL) of nimesulide (IS), 1 mL of acetate buffer (pH 5.5) and 6 mL of chloroform–acetone (1 : 1, v/v). 2. Shake the mixture for 20 min and then centrifuge for 5 min at 6000g. 3. Transfer 5 mL of lower organic layer to a clean tube and evaporate to dryness at 50 C on a heating block under a stream of nitrogen. 4. Reconstitute the dry residue in 100 mL of mixture consisting of acetonitrile–water (1 : 1, v/v). 5. Inject an aliquot of 20 mL into the HPLC using an autosampler. n n
n n
n
n
Chromatography Column: LichroCart Purospher RP-18e, 125 3 mm, Merck. Mobile phase, gradient mixture of 0.1% (v/v) of formic acid in water–acetonitrile (90 : 10 to 0 : 100 at 10 min for 5 min to 90 : 10 at 16 min for 4 min). Detector, triple-quadrupole mass spectrometer, operated in ESI negative-ion and selected-ion recording (SIR) modes Recorded ions (m/z): warfarin, 161 and 250; nimesulide (IS), 229; coumatetralyl, 291 and 292; bromadiolone, 525 and 527; difenacoum, 443 and 444; brodifacoum, 523 and 521; difethialone, 539 and 537. Validation parameters: 9-point (0, 10, 50, 100, 200, 500, 1000, 2000 and 5000 mg/L) calibration curves were linear from limit of quantifications (LOQs) up to 5000 mg/L; LOQs (S/N>10) (mg/L) were 10 for coumatetralyl, 15 for warfarin, 50 for bromadiolone, 60 for brodifacoum and difenacoum, and 200 for difethialone. Retention times are shown in Fig. 16.6.
Other parameters were determined at blood concentrations of 100 mg/L. Recoveries averaged from 65% (warfarin) to 81% (brodifacoum). Within-day and between-day precision was less than 15% RSD. Specificity was checked using ten blank blood samples (including postmortem samples) from different sources and no interfering peaks were observed. The same extraction procedure and HPLC-DAD method was successfully applied for the identification of above-mentioned active anticoagulant components in commercial rodenticide preparations and waters, drinks and foodstuffs (soups, sausages, ground coffee) to which a rodenticide has been added for criminal purposes, and in stomach contents taken from animals (dogs, seals, spoonbills) suspected of having been poisoned and stomach washings in emergency cases of severe poisoning (Adamowicz, Kała 2005). Toxicity Accidental and intentional ingestion of 4-hydroxycoumarin rodenticides can lead to serious poisoning manifested by bleeding in multiple organ sites. Treatment consists of supplements of vitamin K (mild cases) and, for serious cases, infusions of fresh frozen plasma or purified clotting factors until the prothrombin time returns to the normal range. Blood and tissue levels Cases of serious poisoning are generally associated with serum concentrations greater than 5 mg/L, but for some derivatives (e.g. acecoumarol and brodifacoum) toxicity appears at much lower concentrations (Geldmacher-v.Mallinckrodt 1997). A limited number of cases of non-fatal intoxications by coumarin anticoagulants have been reported where serum concentrations (mg/L) were measured. These can be summarised as follows (Grobosch et al. 2006): Brodifacoum
Difenacoum
n=3
n=2
Bromadiolone n=2
630, 710, 731
600, 970
40, 440
In a case of bromidiolone, poisoning the whole-blood concentration was 51 mg/L (Jin et al. 2007). In a fatal poisoning following ingestion of a
Clarke's Analysis of Drugs and Poisons Chapter No. 16 Dated: 10/3/2011 At Time: 18:13:19
References rodenticide containing 0.005% of brodifacoum, its concentrations (mg/ L or mg/kg) were as follows: Heart blood
Femoral blood
Bile
Liver
Spleen
Lung
2240
3919
4276
50
34
31
Brodifacoum was not detected in vitreous humour or in brain tissue. It is worth adding that in liver preserved by formalin and in formalin solution after preservation concentrations of brodifacoum were 820 mg/kg and 5440 mg/L, respectively. The most reasonable explanation for such high levels of brodifacoum in the fixed liver and formalin fixative was significant release of brodifacoum from hepatic tissues (Palmer et al. 1999).
B
C
G
A D
E F
6
8
10
12
Retention time (min) Figure 16.6 LC-MS chromatogram (mass detector, operated in ESI negativeion and SIR modes; Purospher RP-18e). The extract of control blood spiked with: A,100 mg/L of warfarin; B, 500 mg/L of nimesulide (IS); C, 100 mg/L of coumatetralyl; D, 200 mg/L of bromadiolone; E, 200 mg/L of difenacoum; F, 200 mg/L brodifacoum; G, 400 mg/L of difethialone. Reproduced with permission from Adamowicz and Kała (2009).
Organic and inorganic metallic compounds
A wide range of organic and inorganic metallic compounds is found in agricultural use. Inorganic and organometallic compounds are used as acaricides (organotin), herbicides (organoarsenic), fungicides (dithiocarbamate compounds of nickel and dithiocarbamate complexes with manganese and zinc, organic and inorganic compounds of copper and mercury) and rodenticides (magnesium, aluminium and zinc phosphides, and thallium sulfate). For some compounds, exposure to the organic form results in more serious toxicity and the features of poisoning may differ significantly from those of the inorganic compound. For example, whereas inorganic tin compounds are relatively innocuous, the organic forms (ethyl, butyl or phenyl derivatives), used mainly as molluscicides, present a serious hazard because of their lipid solubility, which allows rapid uptake into the central nervous system. This can lead to partial paralysis, visual and psychic disturbances, convulsions and death from respiratory or cardiac failure. Ingestion of inorganic mercury compounds causes gastrointestinal problems (vomiting, diarrhoea) and renal failure, whereas the lipid-soluble organic mercurials (e.g. the dimethyl and diethyl derivatives) concentrate in the central nervous system and lead to ataxia, chorea and convulsions. Numerous techniques, based on atomic absorption spectrophotometric detection and colorimetric detection, can be used to analyse these compounds
285
(see Chapters 30 and 43). Since most methods require prior destruction of the organic matrices (wet or dry), LC-MS techniques have evolved, in particular for the analysis of organotin compounds (Niessen 1999).
References Abu-Qare AW, Abou-Donia MB (2000). Simultaneous determination of pyridostigmine bromide, N,N-diethyl-m-toluamide, permethrin, and their metabolites in rat plasma and urine by high-performance liquid chromatography. J Chromatogr B Biomed Sci Appl 749: 171–178. Abu-Qare AW, Abou-Donia MB (2001). Simultaneous determination of malathion, permethrin, DEET (N,N-diethyl-m-toluamide), and their metabolites in rat plasma and urine using high performance liquid chromatography. J Pharm Biomed Anal 26: 291–299. Adamowicz P, Kała M (2005). Simple HPLC method for the identification of the most commonly used rodenticides in Poland. Probl Forensic Sci 64: 373–381. Adamowicz P, Kała M (2009). LC-ESI-MS-MS determination of six anticoagulant rodenticides in blood. Probl Forensic Sci 77: 53–63. Ageda S et al. (2006). The stability of organophosphorus insecticides in fresh blood. Leg Med (Tokyo) 8: 144–149. Al Samarraie MS et al. (2009). Lethal poisoning with ethiofencarb and ethanol. J Anal Toxicol 33: 389–392. Alder L et al. (2006). Residue analysis of 500 high priority pesticides: better by GCMS or LC-MS/MS? Mass Spectrom Rev 25: 838–865. Ameno K et al. (1995). Simultaneous quantitation of diquat and its two metabolites in serum and urine bi ion-paired HPLC. J Liq Chromatogr 18: 2115–2121. Aprea CT et al. (2002). Biological monitoring of pesticide exposure: a review of analytical methods. J Chromatogr B Analyt Technol Biomed Life Sci 769: 191–219. Ariffin MM, Anderson RA (2006). LC/MS/MS analysis of quaternary ammonium drugs and herbicides in whole blood. J Chromatogr B Analyt Technol Biomed Life Sci 842: 91–97. Arys K et al. (2000). Quantitative determination of paraquat in a fatal intoxication by HPLC-DAD following chemical reduction with sodium borohydride. J Anal Toxicol 24: 116–121. Baeck SK et al. (2007). Comparison study of the extraction methods of paraquat in post-mortem human blood samples. Arch Pharm Res 30: 235–239. Bardarov V, Mitewa M (1989). High-performance liquid and gas chromatography of dialkylphosphates, dialkylthiophosphates and dialkyldithiophosphates as their pentafluorobenzyl derivatives. J Chromatogr 462: 233–241. Barr DB et al. (2002). A multi-analyte method for the quantification of contemporary pesticides in human serum and plasma using high-resolution mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 778: 99–111. Baselt RC (2004). Disposition of Toxic Drugs and Chemicals in Man, 7th edn. Foster City, CA: Biomedical Publications. Bernardelli BC, Gennari MC (1987). Death caused by ingestion of endosulfan. J Forensic Sci 32: 1109–1112. Bidstrup PL et al. (1952). Prevention of acute dinitro-ortho-cresol (D.N.O.C.) poisoning. Lancet i: 794–795. Bissacot DZ, Vassilieff I (1997). HPLC determination of flumethrin, deltamethrin, cypermethrin, and cyhalothrin residues in the milk and blood of lactating dairy cows. J Anal Toxicol 21: 397–402. Botitsi H et al. (2007). Development and validation of a multi-residue method for the determination of pesticides in processed fruits and vegetables using liquid chromatography–electrospray ionization tandem mass spectrometry. Anal Bioanal Chem 389: 1685–1695. Brock JW et al. (1996). An improved analysis for chlorinated pesticides and polychlorinated biphenyls (PCBs) in human and bovine sera using solid-phase extraction. J Anal Toxicol 20: 528–536. Brose C et al. (1992). A thin layer chromatography program for 178 pesticides [in German]. Beitr Gerichtl Med 50: 221–228. Burton BT et al. (1991). Seizure following lindane ingestion in a patient pre-treated with phenytoin. Vet Hum Toxicol 33: 391–396. Calatayud JM et al. (2006). FIA-fluorimetric determination of the pesticide 3indolyl acetic acid. J Fluoresc 16: 61–67. Catlin DH et al. (1987). Analytical chemistry at the Games of the XXIIIrd Olympiad in Los Angeles, 1984. Clin Chem 33: 319–327. Chan LT et al. (1983). Phosphine analysis in post mortem specimens following ingestion of aluminium phosphide. J Anal Toxicol 7: 165–167. Cherstniakova SA et al. (2006). Rapid determination of N,N-diethyl-m-toluamide and permethrin in human plasma by gas chromatography–mass spectrometry and pyridostigmine bromide by high-performance liquid chromatography. J Anal Toxicol 30: 21–26. Chłobowska Z et al. (1996). Application of gas chromatography for identification of pesticides. Probl Forensic Sci 33: 20–27. Colbert DL, Coxon RE (1988). Paraquat measured in serum with the Abbott TDx. Clin Chem 34: 1948–1949. Comer SW et al. (1976). Stability of parathion metabolites in urine samples collected from poisoned individuals. Bull Environ Contam Toxicol 16: 618–625. Croes K et al. (1993). Quantitation of paraquat in serum by HPLC. J Anal Toxicol 17: 310–312.
Clarke's Analysis of Drugs and Poisons Chapter No. 16 Dated: 10/3/2011 At Time: 18:13:19
286
Pesticides
Daundkar BB et al. (2006). Detection of carbaryl insecticide in biological samples by TLC with a specific chromogenic reagent. J Planar Chromatogr-Modern TLC 19: 467–468. de Almeida RM, Yonamine M (2007). Gas chromatographic-mass spectrometric method for the determination of the herbicides paraquat and diquat in plasma and urine samples. J Chromatogr B Analyt Technol Biomed Life Sci 853: 260–264. De Alwis GK et al. (2006). Measurement of human urinary organophosphate pesticide metabolites by automated solid-phase extraction derivation and gas chromatography–tandem mass spectromy. J Chromatogr B Analyt Technol Biomed Life Sci 843: 34–41. De Zeeuw RA et al., eds (1992a). Gas Chromatographic Retention Indices of Toxicologically Relevant Substances on Packed or Capillary Columns with Dimethylsilicone Stationary Phases. Report XVIII of the DFG Commission for Clinical–Toxicological Analysis. Special Issue of the TIAFT Bulletin, 3rd edn. Weinheim: VCH. De Zeeuw RA et al., eds (1992b). Thin-Layer Chromatographic Rf Values of Toxicologically Relevant Substances on Standardized Systems. Report XVII of the DFG Commission for Clinical–Toxicological Analysis, Special Issue of the TIAFT Bulletin, 2nd edn. Weinheim: VCH. Draffan GH et al. (1977). Quantitative determination of the herbicide paraquat in human plasma by gas chromatographic and mass spectrometric methods. J Chromatogr 139: 311–320. Dulaurent S et al. (2008). Hair analysis to document non-fatal pesticide intoxication cases. Forensic Sci Int 176: 72–75. Dzgoev AB et al. (1999). High-sensitivity assay for pesticide using a peroxidase as chemiluminescent label. Anal Chem 71: 5258–5261. Eistert B et al. (1968). Methoden der organischen Chemie. Stuttgart: Thieme. Erdman F et al. (1991). A screening system for 170 pesticides [in German]. Beitr Gerichtl Med 49: 121–126. Erdmann F et al. (1990). A TLC screening program for 170 commonly used pesticides using the corrected Rf value (Rf(c) value). Int J Legal Med 104: 25–31. Fatori D, Hunter WM (1980). Radioimmunoassay for serum paraquat. Clin Chim Acta 100: 81–90. Felice LJ et al. (1991). Multicomponent determination of 4-hydroxycoumarin anticoagulant rodenticides in blood serum by liquid chromatography with fluorescence detection. J Anal Toxicol 15: 126–129. Fernandez Moreno JL et al. (2008). Multiresidue method for the analysis of more than 140 pesticide residues in fruits and vegetables by gas chromatography coupled to triple quadrupole mass spectrometry. J Mass Spectrom 43: 1235–1254. Fernandez-Gutierrez A et al. (1998). Determination of endosulfan and some pyrethroids in water by micro liquid-liquid extraction and GC-MS. Fresenius J Anal Chem 5: 568–572. Flanagan RJ et al. (1995). Basic Analytical Toxicology. Geneva: WHO. Fleisher JH, Pope EJ (1954). Colorimetric method for determination of red blood cell cholinesterase activity in whole blood. AMA Arch Ind Hyg Occup Med 9: 323–334. Fodor-Csorba K, Dutka F (1986). Selectivity and sensitivity of some thin-layer chromatographic detection systems. J Chromatogr 365: 309–314. Frenich AG et al. (2007). Multiresidue analysis of pesticides in animal liver by gas chromatography using triple quadrupole tandem mass spectrometry. J Chromatogr A 1153: 194–202. Fysh RR, Whitehouse MJ (1986). Pesticides. In: Moffat AC et al., eds. Clarke’s Isolation and Identification of Drugs, 2nd edn. London: Pharmaceutical Press, 70–86. Geldmacher-v.Mallinckrodt M (1997). Anticoagulants. In: Brandenberger H, Maes RAA, eds. Analytical Toxicology for Clinical, Forensic and Pharmaceutical Chemists. Berlin: Walter de Gruyer, 609–619. Geldmacher-v.Mallinckrodt M et al. (1974). Zur Bewertung der SerunCholinesteraseaktivitaet in Leichenblut bei Verdacht auf eine E 605Vergiftung. Z Rechtsmed 75: 191–199. Grobosch T et al. (2006). Acute bromadiolone intoxication. J Anal Toxicol 30: 281–286. Guale FG et al. (1994). Laboratory diagnosis of zinc phosphide poisoning. Vet Hum Toxicol 36: 517–519. Hara S et al. (2007). Rapid and sensitive HPLC method for the simultaneous determination of paraquat and diquat in human serum. Anal Sci 23: 523–526. Hernandez F et al. (2004). An estimation of the exposure to organophosphorus pesticides through the simultaneous determination of their main metabolites in urine by liquid chromatography–tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 808: 229–239. Hoizey G et al. (2008). Thiodicarb and methomyl tissue distribution in a fatal multiple compounds poisoning. J Forensic Sci 53: 499–502. Jadhav RK et al. (1992). Distribution of malathion in body tissues and fluids. Forensic Sci Int 52: 223–229. Jarvie DR, Stewart MJ (1979). The rapid extraction of paraquat from plasma using an ion-pairing technique. Clin Chim Acta 94: 241–251. Jin MC et al. (2007). Determination of bromadiolone in whole blood by highperformance liquid chromatography coupled with electrospray ionization tandem mass spectrometry. Forensic Sci Int 171: 52–56.
Kała M, Chacia T (1994). A thin-layer chromatography screening for commonly used pesticides. Probl Forensic Sci 30: 34–41. King JW, Zhang Z (2002). Derivatization reactions of carbamate pesticides in supercritical carbon dioxide. Anal Bioanal Chem 374: 88–92. Kłys M, Bialka J (1990). Non-fatal and fatal intoxications wih carbamate pesticide carbofuran and propoxur. Arch Med Sadowej Kryminol 40: 49–58. Kłys M et al. (1989). Non-fatal and fatal human intoxications with organophosphate pesticide chlorphenvinphos. Arch Med Sadowej Kryminol 39: 199–207. Kłys M et al. (1991). Medico-legal and clinical problems in phosalone intoxications. Arch Med Sadowej Kryminol 41: 256–266. Kłys M et al. (1992). Non-fatal and fatal human poisoning with phosphororganic insecticide fenitrothion. Arch Med Sadowej Kryminol 42: 254–265. Kraemer T et al. (1997). In: Pragst F, ed. Improvement of Sample Preparation for STA. Acceleration of acid hydrolysis and derivatisation procedures by microwave irradiation. Heppenheim: Helm-Verlag, 200–204. Kuo TL et al. (2001). Spectra interference between diquat and paraquat by second derivative spectrophotometry. Forensic Sci Int 121: 134–139. Kurt TL et al. (1986). Accidental Kwell (lindane) ingestions. Vet Hum Toxicol 28: 569–571. Lacassie E et al. (2001). Sensitive and specific multiresidue methods for the determination of pesticides of various classes in clinical and forensic toxicology. Forensic Sci Int 121: 116–125. Lawrence JF, Turton D (1978). High-performance liquid chromatographic data for 166 pesticides. J Chromatogr 159: 207–226. Lech T, Sadlik JK (2007). Thallium intoxication in humans. Toxicol Lett 172S585. Lehotay SJ et al. (2005). Validation of a fast and easy method for the determination of residues from 229 pesticides in fruits and vegetables using gas and liquid chromatography and mass spectrometric detection. J AOAC Int 88: 595–614. Liu S, Pleil JD (2002). Human blood and environmental media screening method for pesticides and polychlorinated biphenyl compounds using liquid extraction and gas chromatography–mass spectrometry analysis. J Chromatogr B Analyt Technol Biomed Life Sci 769: 155–167. Lo´pez R et al. (2007). Determination of organochlorine pesticides and polychlorinated biphenyls in human serum using headspace solid-phase microextraction and gas chromatography–electron capture detection. J Chromatogr B Analyt Technol Biomed Life Sci 846: 298–305. Mali RS et al. (2006). Thin-layer chromatography for selective detection of methomyl in forensic cases. J Planar Chromatogr-Modern TLC 19: 85–86. Maurer HH (2006). Hyphenated mass spectrometric techniques-indispensable tools in clinical and forensic toxicology and in doping control. J Mass Spectrom 41: 1399–1413. Maurer HH, Arlt JW (1998). Detection of 4-hydroxycoumarin anticoagulants and their metabolites in urine as part of a systematic toxicological analysis procedure for acidic drugs and poisons by gas chromatography–mass spectrometry after extractive methylation. J Chromatogr B Biomed Sci Appl 714: 181–195. Merck Index (2006). The Merck Index, An Encyclopedia of Chemicals, Drugs and Biologicals, 14th edn. Whitehouse Station, NJ: Merck Research Laboratories, Merck & Co., Inc. Meyer E et al. (1998). Analysis of fenthion in postmortem samples by HPLC with diode-array detection and GC-MS using solid-phase extraction. J Anal Toxicol 22: 248–252. Minakata K et al. (1990). Extraction of diquat with 1-butanol from biological materials. Forensic Sci Int 44: 27–35. Miranda EJ et al. (2006). Two deaths attributed to the use of 2,4-dinitrophenol. J Anal Toxicol 30: 219–222. Moriya F, Hashimoto Y (1999). Comparative studies on tissue distributions of organophosphorus, carbamate and organochlorine pesticides in decedents intoxicated with these chemicals. J Forensic Sci 44: 1131–1135. Moriya F et al. (1999). Pitfalls when determining tissue distributions of organophosphorus chemicals: sodium fluoride accelerates chemical degradation. J Anal Toxicol 23: 210–215. Murty AS et al. (1980). Improved ammonium molybdate method for thin-layer chromatographic detecetion of organophosphate residues. J Assoc Off Anal Chem 63: 756–757. Musshoff F et al. (2008). A gas chromatographic analysis of phosphine in biological material in a case of suicide. Forensic Sci Int 177: e35–e38. Namera A et al. (2000). Direct colorimetric method for determination of organophosphates in human urine. Clin Chim Acta 291: 9–18. Nguyen JV et al. (2007). Quantification of atrazine, phenylurea, and sulfonylurea herbicide metabolites in urine by high-performance liquid chromatographytandem mass spectrometry. J Anal Toxicol 31: 181–186. Niessen WM (1999). Liquid Chromatography–Mass Spectrometry, 2nd edn. New York: Marcel Dekker. Osselton MD, Snelling RD (1986). Chromatographic identification of pesticides. J Chromatogr 368: 265–271. Osterloh J et al. (1983). Toxicologic studies in a fatal overdose of 2,4-D, MCPP, and chlorpyrifos. J Anal Toxicol 7: 125–129. Pacioni NL, Veglia AV (2007). Determination of poorly fluorescent carbamate pesticides in water, bendiocarb and promecarb, using cyclodextrin nanocavities and related media. Anal Chim Acta 583: 63–71.
Clarke's Analysis of Drugs and Poisons Chapter No. 16 Dated: 10/3/2011 At Time: 18:13:20
Further reading Palmer RB et al. (1999). Fatal brodifacoum rodenticide poisoning: autopsy and toxicologic findings. J Forensic Sci 44: 851–855. Pang GF et al. (2006). Validation study on 660 pesticide residues in animal tissues by gel permeation chromatography cleanup/gas chromatography–mass spectrometry and liquid chromatography–tandem mass spectrometry. J Chromatogr A 1125: 1–30. Pang GF et al. (2009). Analysis method study on 839 pesticide and chemical contaminant multiresidues in animal muscles by gel permeation chromatography cleanup, GC/MS, and LC/MS/MS. J AOAC Int 92: 933–940. Park J et al. (1977). Pharmacokinetic studies in severe intoxication with 2, 4-D and mecoprop. Proc Eur Soc Toxicol 18: 154–155. Park MJ et al. (2009). Postmortem blood concentrations of organophosphorus pesticides. Forensic Sci Int 184: 28–31. Patil VB, Shingare MS (1993). Thin-layer chromatography detection of organophosphorus insecticide containing a nitrophenyl group. J AOAC Int 76: 1394–1395. Pavlic M et al. (2002). Fatal intoxication with omethoate. Int J Legal Med 116: 238–241. Perez-Ruiz T et al. (2002). Chemiluminescence determination of carbofuran and promecarb by flow injection analysis using two photochemical reactions. Analyst 127: 1526–1530. Peters FT et al. (2007). Validation of new methods. Forensic Sci Int 165: 216–224. Pfleger K et al. (2007). Mass Spectral and GC data of Drugs, Poisons, Pesticides, Pollutants and Their Metabolites, 3rd edn. Weinheim: Wiley-VCH. Poklis A et al. (1980). A fatal diazinon poisoning. Forensic Sci Int 15: 135–140. Politi L (2007). LC-MS-MS analysis of 2, 4-dinitrophenol and its phase I and II metabolites in a case of fatal poisoning. J Anal Toxicol 31: 55–61. Pommery J et al. (1993). Atrazine in plasma and tissue following atrazine–aminotriazole–ethylene glycol–formaldehyde poisoning. J Toxicol Clin Toxicol 31: 323–331. Rivers JB et al. (1970). Simultaneous gas chromatographic determination of 2, 4-D and dicamba in human blood and urine. J Chromatogr 50: 334–337. R€ ussel H (1986). R€ uckstandsanalytik von Wirkstoffen in tierischen Produkten. Stuttgart: Thieme. Russo MV et al. (2002). Determination of organophosphorus pesticide residues in human tissues by capillary gas chromatography–negative chemical ionization mass spectrometry analysis. J Chromatogr B Analyt Technol Biomed Life Sci 780: 431–441. Sadlik JK (1997). Thallium poisoning. In: Proceedings of the XXXV Annual Meeting of TIAFT. Padua: Centre of Behavioural and Forensic Toxicology, 516–518. Scherrman JM et al. (1983). [Acute paraquat poisoning: prognostic and therapeutic significance of blood assay]. Toxicol Eur Res 5: 141–145. Schreiber A et al. (2000). Application of spectral libraries for high-performance liquid chromatography–atmospheric pressure ionisation mass spectrometry to the analysis of pesticide and explosive residues in environmental samples. J Chromatogr A 869: 411–425. Sevalkar MT et al. (2000). Thin-layer chromatographic method for detection and identification of carbaryl, propoxur, and carbofuran by use of 4-aminoantipyrine. J Planar Chromatogr 13: 235–237. Simonelli A et al. (2007). Analytical method validation for the evaluation of cutaneous occupational exposure to different chemical classes of pesticides. J Chromatogr B Analyt Technol Biomed Life Sci 860: 26–33.
287
Smith DL et al. (1978). Criteria for a Recommended Standard-Occupational Exposure to Dinitro-ortho-cresol. Pub No 78–131. 1978. Washington, DC: US Department of Health, Education, and Welfare (NIOSH). Su´bova I et al. (2006). Fluorescence determination of the pesticide asulam by flow injection analysis. Anal Sci 22: 21–24. Sundberg SE et al. (2006). A simple and fast extraction method for organochlorine pesticides and polychlorinated biphenyls in small volumes of avian serum. J Chromatogr B Analyt Technol Biomed Life Sci 831: 99–104. Takayasu T et al. (2008). A fatal intoxication from ingestion of 2-methyl-4-chlorophenoxyacetic acid (MCPA). J Anal Toxicol 32: 187–191. Tarbah FA et al. (2007). Distribution of dimethoate in the body after a fatal organophosphate intoxication. Forensic Sci Int 170: 129–132. Tena MT (1992). Total and individual determination of carbamate pesticides by use of an integrated flow-injection/HPLC system. Chromatographia 33: 449–453. Tetsuya I et al. (2007). Solid phase extraction of phosphorus-containing amino acidtype herbicides and their metabolites from human blood with titania for determination by capillary electrophoresis. Anal Sci 23: 755–758. Tomlin C (2006). The Pesticide Manual, 14th edn. Alton, Hants: British Crop Protection Council. Tompsett SL (1970). Paraquat poisoning. Acta Pharmacol Toxicol (Copenh) 28: 346–358. Tutudaki M, Tsatsakis AM (2005). Pesticide hair analysis: development of a GCNCI-MS method to assess chronic exposure to diazinon in rats. J Anal Toxicol 29: 805–809. Ueyama J et al. (2006). Simultaneous determination of urinary dialkylphosphate metabolites of organophosphorus pesticides using gas chromatography-mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 832: 58–66. Vanholder R et al. (1981). Diquat intoxication: report of two cases and review of the literature. Am J Med 70: 1267–1271. Walorczyk S (2007). Development of a multi-residue screening method for the determination of pesticides in cereals and dry animal feed using gas chromatography–triple quadrupole tandem mass spectrometry. J Chromatogr A 1165: 200–212. WHO (2004). The WHO Recommended Classification of Pesticides by Hazard and Guidelines to Classification WHO Library Cataloguing-in Publication Data. Geneva: WHO. Wollersen H, Musshoff F (2007). Chromatographic techniques for measuring organophosphorus pesticides. In: Bertholf RL, Winecker RE, eds. Chromatographic Methods in Clinical Chemistry and Toxicology. Chichester: Wiley, 139–169.
Further reading Alder L (2010). Parameters for the Determination of Pesticide Residues. Berlin: Federal Institute for Risk Assessment. www.bfr.bund.de/cd/5832.(accessed 10 October 2008). Gonˇi F et al. (2007). High throughput method for the determination of organochlorine pesticides and polychlorinated biophenyls in human serum. J Chromatogr B 852: 15–21. NIOSH (2003). Pocket Guide to Chemical Hazards. Washington, DC: NIOSH, www.cdc.gov/niosh/npg/ (accessed 11 March 2010). Tomlin C, ed. (2000). The Pesticide Manual, 12th edn. London: British Crop Protection Council and Royal Society of Chemistry.
Clarke's Analysis of Drugs and Poisons Chapter No. 17 Dated: 10/3/2011 At Time: 19:14:50
CHAPTER
17
Metals and Anions R Braithwaite
General introduction Metals and anions form an important, but disparate, group of agricultural, household and industrial poisons that present many difficulties in their systematic chemical analysis (Baldwin and Marshall 1999; Yeoman 1985). Severe acute poisoning with these agents is rare in most developed countries, but remains common in many developing parts of the world. Chronic poisoning, as a result of occupational or environmental exposure to heavy metals and pesticides, occurs in many countries and can be a cause of ill-health in both adults and children. The signs and symptoms of acute poisoning may differ from those associated with chronic toxicity. Some metallic (e.g. arsenic) and anionic (e.g. cyanide) substances undergo extensive metabolism after ingestion. These factors have a significant bearing on analytical investigations applied to biological materials and their interpretation. It is important in individual cases, therefore, to know whether poisoning resulted from acute, chronic or acute-on-chronic exposure. Of equal importance is the time of specimen collection in relation to the alleged time of ingestion or exposure. The wide range of metallic or anionic poisons that might be involved in any case of suspected poisoning means that great care is required in the collection of appropriate specimens and the selection of toxicological and other tests. There is no simple systematic way to investigate cases for which the history is uncertain and the identity of the poison unknown. The investigation is often led by a process of elimination of the more likely causes of poisoning (e.g. pharmaceuticals and illicit drugs), and then a careful examination of the detailed history of the patient or deceased, in particular any access to compounds associated with industrial and agricultural use, or specific household products. Considerable advances in analytical techniques for measuring metals in biological fluids have been made since the early 1980s, particularly in electrothermal atomic absorption spectrometry (ETAAS) (Halls 1984; Slavin 1988) and inductively coupled plasma–mass spectrometry (ICPMS) (Hsiung et al. 1997). These techniques are discussed in detail in Chapter 43. The use of ICP-MS has expanded rapidly in the last 5 years and it is now the technique of choice for sensitive multi-element analysis, particularly in the investigation of suspected poisoning of ‘heavy metals’ or less common inorganic poisons. When investigating chronic exposure to toxic metals, great care must be taken to control the accuracy and precision of biological analyses. This calls for ready access to a supply of reliable internal quality-control materials and regular participation in external quality-assurance programmes. These are often not available in developing countries, but much can be done at relatively low cost with cooperation between laboratories (Braithwaite and Girling 1988; Halls 1984). The development of analytical techniques for anions in biological fluids has historically been limited. The increased availability of liquid chromatography–mass spectrometry (LC-MS) techniques has greatly improved matters. However, a number of simple methods for measuring selected poisons remain valid and have a role to play where access to more sophisticated equipment is restricted (Flanagan et al. 1995).
Specimen collection and analysis In cases of suspected poisoning admitted to hospital, specimens of blood and, where possible, urine should be taken (see Chapter 1). These should be labelled carefully (full name of patient, admission number and date 288
and time of collection) and stored in a refrigerator (4 C) pending any delay in transport to the laboratory. When blood specimens are received in the laboratory and analyses are not required immediately, it is useful to separate off plasma or serum from red cells, prior to deep freezing. However, for toxins that have a significant distribution into the red cells (e.g. lead, cadmium, mercury and cyanide), it is essential to conserve samples of anticoagulated whole blood. In postmortem examinations it is important to undertake a more systematic specimen collection and great care is required in the selection of sampling sites, method of collection and use of appropriate specimen containers (see Chapter 10). Where an industrial accident has occurred there may be access to ‘scene residues’ or materials used in a chemical process. Analysis of these materials can yield invaluable clues when the precise nature of the chemical agent is unknown. However, prior to transportation to the laboratory, separate packaging from any biological specimens is advisable to avoid the risk of contamination. Stomach contents Vomit, stomach aspirate and washout fluid are now rarely available from cases of acute poisoning admitted to hospital. Stomach washout procedures (except in rare cases) have been replaced by the administration of oral activated charcoal in most developed countries. In fatal cases, the whole stomach and its contents can be removed post mortem for detailed examination. When dealing with the initial examination of stomach contents, or vomitus, it is helpful to note any unusual smell, colour or other appearance, such as the presence of fresh or altered blood (coffee grounds; see Chapter 1). Great care should be taken when dealing with cases that involve the oral ingestion of cyanide salts or phosphides, as the contents of the stomach may represent a serious hazard and risk of secondary poisoning (a fume cupboard or safety cabinet must be used in these circumstances). The total volume of the stomach contents should be recorded. These can then be homogenised in a blender to allow two or three representative smaller specimens to be taken for both qualitative and quantitative analysis. Homogenised specimens can be filtered directly using a coarse-grade filter paper or diluted with distilled water. The clear supernatant or a representative sample of the homogenised stomach contents can be used for analysis. Quantitative analysis of the stomach contents is generally of interest only in medicolegal cases in which the quantity of chemical poison present in the stomach contents may provide useful forensic evidence. Blood Venous blood (10 mL) should be collected in all cases of suspected poisoning; for metallic poisons and anions, such as cyanide, a potassium ethylenediamine tetraacetic acid (K-EDTA) container is the most appropriate. A wide range of blood-collection tubes is available commercially; to avoid the possibility of contamination, the use of products certified as suitable for trace-element analyses is strongly recommended, particularly when dealing with environmental or subclinical exposure to agents such as lead and cadmium. Blood-collection tubes that contain gel separation barriers should not be used. Alternatively, the laboratory can purchase relatively large batches of standard blood-collection tubes
Clarke's Analysis of Drugs and Poisons Chapter No. 17 Dated: 10/3/2011 At Time: 19:14:50
Metals (all with the same batch number) and carefully test a few tubes from each batch to ensure that they are free of contamination with those metals of clinical interest (e.g. lead, cadmium and aluminium). When blood specimens are received in the laboratory in an unusual container, it may be useful to request a ‘blank’ container that can be analysed for the presence of any contaminating substance. It is equally important to ensure that reliable blood-specimen containers are used for postmortem examinations. These can be supplied to the pathologist ahead of any postmortem examination as part of the standard specimen collection kit. It is recommended that, at any postmortem examination, blood should be taken from an identified anatomical site (preferably the femoral vein) for quantitative analysis, since after death significant diffusion or redistribution of poisons may occur (see Chapter 10). Other sites (e.g. the heart) may be useful for qualitative ‘screening’ purposes. Blood should never be taken from the body cavity at the end of the postmortem examination. The main problem is that blood samples taken from different sites, e.g. femoral, subclavian, aorta, at postmortem examination show different concentrations. In cases of rapid death, where there may be large quantities of unabsorbed poison present in the stomach, or aspirated into the lungs, this can undergo significant diffusion after death, leading to very high concentrations in adjacent blood vessels. Urine An early specimen of urine (20 mL in a plain Sterilin-type container with no preservatives added) should be collected, with care taken to ensure that the sample is not contaminated during the collection process. If patients are undergoing chelation therapy, it can be useful to collect sequential 24 h urine specimens into acid-washed plastic urine containers. The total volume of urine should be recorded carefully prior to taking aliquots of specimens for analysis to determine the excretion of chelated elements and the efficacy of treatment. Other specimens Hair analysis for trace elements has often been used for diagnostic purposes in cases where an individual complains of symptoms for which no cause can be found by routine medical or pathological investigations. However, experience and published studies show that the results can sometimes be misleading (Poon et al. 2004; Seidel et al. 2001; Taylor 1986). Hair analysis has more application in surveys of population exposure and in investigations of suspicious deaths, such as those that involve arsenic or mercury. The long persistence of metals and many other chemical poisons in hair and nail samples compared with their relatively short duration in blood or urine specimens can be a major advantage in forensic cases (Daniel et al. 2004). Analysis of hair and nail sections can also sometimes yield an important chronological forensic record of when doses were administered. Specimens such as tissues and bone may sometimes be collected at postmortem examination as part of the investigation of complex medicolegal cases (Benes et al. 2000; Lech and Trela 2005; Warren et al. 2002). As with blood specimens, great care is required in selecting the site of collection and to avoid contamination. In addition, interpretation of the results without access to comparable reference specimens can be very difficult.
Quality assurance Access to appropriate ‘internal’ control or other reference materials is very important for the analysis of trace metals. A wide variety of certified reference materials, including blood, serum, urine and bone, is available commercially. Where these cannot be obtained easily, an inexpensive alternative is to prepare such materials from bovine blood, or any suitable large animal as a cooperative effort between experienced laboratories (Braithwaite and Girling 1988). This entails the preparation of large batches of samples with trace metal concentrations at, for example, normal, borderline toxic levels and toxic levels, which can then be
289
Table 17.1 Common methods of analysis for trace elements in biological fluids and tissues Colorimetric Fluorimetric Electrochemical (anodic stripping voltametry and ion-selective electrodes) Flame atomic absorption spectrometry Electrothermal atomic absorption spectrometry Inductively coupled plasma–emission spectrometry Inductively coupled plasma–mass spectrometry
analysed repeatedly by the laboratories so that the pooled results can be used to assign concentration ranges. It is equally important for laboratories to subscribe to national or international external qualityassurance schemes where these are available.
Metals Methods of analysis A wide range of techniques is available to laboratories for metals analysis (Table 17.1) and these are described in Chapter 43. A few simple qualitative and quantitative tests (e.g. the Gutzeit and Reinsch tests) can be useful, particularly where laboratory resources are severely limited (Flanagan et al. 1995). Aluminium Introduction
Aluminium is the most abundant non-essential element in the earth’s crust, but its role in human health and disease became understood only recently (Krewski et al. 2007; Martin 1986). The normal intake of aluminium from food, water and beverages is probably 2–3 mg per day, but its absorption from the gut is relatively poor and depends on the speciation of the element and the presence of other substances (e.g. phosphate and citrate) in the diet (Klein 2005). Aluminium is a powerful neurotoxin, and it is well established that excessive intravenous exposure in patients undergoing dialysis results in its significant accumulation in the body, particularly in the brain, which can cause ‘dialysis dementia’, a type of encephalopathy that can be rapidly progressive and lead to death within a few months (Alfrey et al. 1976). Use of aluminium sulfate as a flocculating agent in domestic water supplies is the major source of the metal in these patients, particularly if the water used for dialysis is not purified. The large quantities of oral aluminium salts that may be given to some renal patients to reduce the intestinal absorption of phosphate may also cause toxicity. Aluminium may also be a suspected risk factor in Alzheimer’s disease (Flaten 2001; Miu and Benga 2006). More recently, concern has developed about the harmful effects of occupational exposure to aluminium (Akila et al. 1999; Iregren et al. 2001; Meyer-Baron et al. 2007; Polizzi et al. 2002; Rifat et al. 1990). Attention has also been drawn to the possible harmful effects of aluminium present in infant formulae (Freundlich et al. 1985; Sedman et al. 1985). Specimen collection
Specimens are collected as follows: n n n n
Blood/serum/plasma – 5 mL, plain or lithium–heparin tube Water – 20 mL, plastic universal container Dialysis fluid – 20 mL, sterile plastic universal container Urine – 20 mL, sterile plastic universal container.
It is important to check that all specimen containers are ‘aluminium free’. Glass containers should never be used to collect specimens for aluminium analysis. Analysis
Constant vigilance is required to avoid contamination of specimens, reagents, tubing and equipment from aluminium in dust in laboratory areas. Cleaned glass containers should be used only to prepare concentrated stock solutions of aluminium salts. Great care is required to avoid
Clarke's Analysis of Drugs and Poisons Chapter No. 17 Dated: 10/3/2011 At Time: 19:14:50
290
Metals and Anions
Table 17.2 Recommended water and dialysate aluminium concentrations Solution
Concentration
Maximum allowable concentration (MAC) for potable water
200 mg/L (7.4 mmol/L)
Guideline concentration for potable water
50 mg/L (1.9 mmol/L)
MAC for water for preparation of dialysis fluid
10 mg/L (0.4 mmol/L)
Recommended upper limit for dialysis fluid
30 mg/L (1.1 mmol/L)
contamination when separating plasma or serum from blood specimens. Storage conditions of specimens are important (Wilhelm and Ohnesorge 1990). Aluminium is most commonly measured in plasma using ETAAS with background correction. The use of matrix-matched calibration standards and control material is essential (Taylor et al. 1994). Other methods have been described based on colorimety and fluorimetry but these are more suitable for water analysis. More recently there has been the increased use of ICP-AES and ICP-MS to measure aluminium in plasma, water and dialysate or other more complex matrices (Murko et al. 2007; Zhang et al. 2000, 2002). Interpretation
The recommended (European) guidance values for water and dialysate aluminium concentrations are shown in Table 17.2. In patients on haemodialysis, in which a water purification system (such as reverse osmosis) is in operation, it is important to monitor aluminium in preand post-water samples to detect any breakdown in the purification process that would allow contamination of the dialysis solution used to treat the patient (Berend et al. 2001). Plasma aluminium concentrations should be monitored routinely in all patients in end-stage renal failure who receive dialysis therapy, to ensure that absorption of aluminium is kept to an absolute minimum. In addition, regular testing is needed of water supplied to patients who have home dialysis. European guidelines for plasma aluminium concentrations in patients who receive dialysis therapy are given in Table 17.3. The body burden of aluminium in dialysis patients can be obtained from the measurement of plasma aluminium concentration before and after the administration of a standard dose of an aluminiumchelating agent such as desferrioxamine (Chazan et al. 1989; Marumo et al. 1987). Where renal function is normal, aluminium is excreted rapidly from the body and there is little possibility of accumulation. The reference value for urine aluminium in non-exposed healthy adults is less than 15 mg/L (0.5 mmol/L). In situations of acute or chronic occupational or environmental exposure to aluminium, measurement of blood (plasma) and/or urine aluminium is an effective way to assess the degree of exposure (Ljunggren et al. 1991; Pierre et al. 1995). Where occupational exposure involves inhalation of fine particles or dusts, aluminium may be stored in the lung tissue and leach out very slowly over several months. As a result, plasma and urine aluminium concentrations can remain elevated for several weeks or months (Ljunggren et al. 1991). Table 17.3 Recommended action limits for plasma aluminium concentrations Situation
Concentration
Normal (no history of chronic renal failure)
100 mg/L (>3.7 mmol/L)
Urgent action required; high risk of toxicity in all
>200 mg/L (>7.4 mmol/L)
There is also evidence of a dose-dependent association between increased aluminium body burden and central nervous system effects in these workers (Akila et al. 1999). Suggested thresholds for these effects in aluminium welders are 108–160 mg/L (4–6 mmol/L) in urine and 7–10 mg/L (0.25–0.35 mmol/L) in plasma (Riihimaki et al. 2000). Aluminium accumulates in the bone of dialysis patients and bone biopsy samples can be analysed to estimate the body burden (Tang et al. 1996). Extensive data are available on the aluminium content of bone in dialysis patients with aluminium-related bone disease. Bone aluminium concentrations of up to 300 mg/kg have been reported in these patients, whereas concentrations in non-exposed reference populations are very low (10 mg/L (>180 mmol/L). However, if chelation therapy has been carried out using desferrioxime, it is not possible to interpret subsequent plasma iron concentrations. A range of serum iron concentrations of 2.8–25.5 mg/L (50– 457 mmol/L) has been reported in children who survived the ingestion of up to 10 g ferrous sulfate (Baselt 2002). Children who died 3–5 days after ingesting 6–15 g of ferrous sulfate had serum iron concentrations of 19– 50 mg/L (340–900 mmol/L) on the first and second days (Baselt 2002).
n n
295
Urine – 24 h collection (aliquot) only in the case of chelation therapy Water –20 mL, plain universal container.
Analysis
Published procedures to determine lead in blood, urine and other fluids, such as tap water or tissues (bone, teeth), are numerous. One of the most successful historical methods for blood lead measurement used flame AAS with the blood specimen introduced into the air– acetylene flame in a small nickel cup (Delves cup), and is still in routine operation in several parts of the world (Delves 1970). This has been superseded by ETAAS methods, which can determine blood lead concentrations accurately (12 or 20 kg BW: 20 mg, once daily
20 mg once daily
Phenytoin
18 mg/kg (IV loading), then 2.5–5 mg twice daily (oral)
1.5–2.5 mg/kg twice daily (oral)
1.5–2.5 mg/kg twice daily (oral)
75–150 mg twice daily (oral)
Cyclophosphamide (IV)
500 mg/m2 BSA once a month
500 mg/m2 BSA once a month
500 mg/m2 BSA once a month
Propofol (IV)
2.5–4 mg/kg
2.5–4 mg/kg to 8 years, then 2.5 mg/kg
1.5–2.5 mg/kg
45 mg/kg in 3 divided doses for 24 h then 10 mg/kg daily
Up to 5 years: 35 mg/kg in 3 divided doses for 24 h then 10 mg/kg daily From 5 to 10 years: 25 mg/kg in 3 divided doses for 24 h then 6 mg/kg daily After 10 years: 0.75–1.5 mg in 3 divided doses for 24 h then 62.5–250 mg daily
0.75–1.5 mg in 3 divided doses for 24 h then 62.5– 250 mg daily
Digoxin (oral)
Neonate 2.5 kg: 45 mg/kg in 3 divided doses for 24 h then 10 mg/kg daily
BSA, body surface area; BW, body weight; IV, intravenous. (a) Doses taken from BNF for Children: these are examples of starting doses for common drugs and are not intended as a dosing recommendation; see BNF for Children for complete dosing information.
with generalised symptoms such as lethargy, vomiting, mild pyrexia and neurological signs (e.g. paralysis and fixed, dilated pupils). There may be discoloration of the urine due to myoglobinuria. Children are at greater risk from envenomation because of the relatively greater dose of toxin. Fatality rates for children are higher than for adults following snake bite (McGain et al. 2004). The risk of incurring a snake bite may also be greater in children. Children may develop signs of envenomation sooner than adults and require a higher dose of antivenom.
consistent with poisoning, increases the positive predictive value to 100%, 86% and 92%, respectively (Hwang et al. 2003). Although routine urine drug screens are not useful, urine drug screens may be useful in detecting toxins in symptomatic patients without a history of ingestion or when ingestion is denied (Belson et al. 1999), in particular where drugs of abuse, e.g. cocaine or methadone, may have been ingested by small children. Examination
Clinical aspects Assessment History
A thorough history should be recorded for each episode of poisoning. The history should include time of ingestion, probable dose, the poisons the child was exposed to and the circumstances at the time (including extent of supervision, where the episode happened, who was there, where the poisons were stored and whose medicines were ingested). It is important to establish whether the history is consistent with the patient’s developmental age. A child who is not rolling will not be able to move towards a poison hazard. A child who is not feeding himself or herself will not be able to take a poison independently. Preformatted charts aid in the collection of clinical data, improving both accuracy and completeness of the data (Buckley et al. 1999). An example of a preformatted chart is presented in Figure 27.1. Data can be collected for patient management but also for epidemiology/accident prevention purposes. Electronic databases are useful for recording the data and for performing audits. Patient management software, as is commonly found in emergency departments and hospital in-patient departments, can be modified to improve data capture for toxicology (e.g. disease-specific data entry screens). The clinical history of an episode of poisoning in young children is extremely unreliable (Hwang et al. 2003). Of children presenting with poisoning, around 60% have actually been exposed to the putative poison (Sugarman et al. 1997; Belson et al. 1999; Hwang et al. 2003). A history from an adult or another child is not more reliable than that from the child him-or herself. Physical evidence, such as an abnormal smell of the breath, staining of the clothes and symptoms/clinical signs
The initial step in the physical examination of the poisoned child is the measuring and recording of the vital signs: temperature, pulse rate, respiratory rate, blood pressure and oximetry. Temperature is an important component of the paediatric general examination as it may indicate alternative diagnoses, such as infection. There are some poisonings that affect temperature, such as hypothermia with organophosphate poisoning, and hyperthermia with serotoninergic syndrome or the anticholinergic toxindrome. Pulse rate in children should be interpreted in relation to age-appropriate norms (Wallis et al. 2005). Abnormal pulse (or heart) rate is an important sign in toxicology, indicating cardiotoxicity. Bradycardia and tachycardia are defined at relatively lower pulse rates in children than in adults. Hence, there are different intervention points. Similarly, both diastolic and systolic blood pressure are lower in children than in adults. This results in different values for defining hypotension and hypertension, and the treatment decision points. Respiration rate is also higher in infants and children than in adults (Wallis et al. 2005). Oximetry is useful in assessing respiratory function, particularly when the poison is a sedative or decreases neuromuscular function (e.g. organophosphate poisoning). Arguably, in the context of child poisoning, blood sugar level is also a vital sign. Children are at greater risk of hypoglycaemia than adults (hypoglycaemia may be a differentiating feature in some childhood poisonings compared with adults). Hypoglycaemia, if uncorrected, may lead to tissue/organ injury and long-term morbidity. It is therefore important to identify and correct hypoglycaemia in childhood poisoning. An assessment of conscious state is important in any toxicological examination because many poisons impair consciousness and this may lead to secondary injury due to impairment of respiration and airway. The Glasgow Coma Scale (GCS) is the most widely applied measure of
Clarke's Analysis of Drugs and Poisons Chapter No. 27 Dated: 10/3/2011 At Time: 12:44:24
Clinical aspects
439
Figure 27.1 Preformatted chart for the collection of clinical data in cases of poisoning. (Continued overleaf)
state of consciousness, and can be modified for paediatric use. However, the GCS was developed for the assessment and monitoring of patients with head injury and is less useful in poisoning: A neurological examination focusing upon pupils, nystagmus, muscle tone and reflexes should be performed. Pupil size and response can be important indicators of poisoning: dilated pupils occur particularly in anticholinergic poisoning, while contracted pupils occur in opioid poisoning. Unresponsive pupils can indicate profound coma, such as with barbiturate-induced coma. Horizontal nystagmus is commonly seen in sedative poisoning, and vertical nystagmus may be seen in addition to this with some anticonvulsants, e.g. barbiturates and carbamazepine. An oscillation of horizontal gaze (differentiable from
nystagmus by a lack of directional component) may be seen in serotonin syndrome. Hypotonia can also indicate sedative poisoning or neuromuscular blockade (such as organophosphate poisoning). Deep tendon reflexes may be increased as a result of serotonin syndrome, while they are decreased with sedative poisoning or neuromuscular blockade. Routine examination of the chest should be performed by listening to the sounds of the heart and breathing. Abdominal examination should be performed to determine liver size and condition, and whether there is any abdominal tenderness or rigidity and presence of bowel sounds. Presence of any signs indicating blockage of the intestine or perforation of the viscera would preclude the use of any gastrointestinal decontamination. The skin, scalp and mouth should be examined for any signs of
Clarke's Analysis of Drugs and Poisons Chapter No. 27 Dated: 10/3/2011 At Time: 12:44:25
440
Paediatric Toxicology
Figure 27.1 Continued.
injury. The general state of care of the child should be recorded, e.g. lack of hygiene, general state of hair, nails and clothing. Use of laboratory analyses Considerations for blood sampling
Children have smaller blood volumes than adults and there is greater difficulty in collecting serum, plasma and urine samples. The amount of blood that can be sampled from a child without the need for transfusion depends upon the initial haematocrit (Lister et al. 2008). Recommendations for paediatric clinical trials are for no more
than 3% of blood volume to be sampled on any one study day in a child with a normal red cell mass (haematocrit) (Kauffman 2000). In an infant or toddler this would represent 2.4 mL/kg body weight. As a guide, children can tolerate sampling of 0.25 mL/kg per day without a fall in haematocrit (Lister et al. 2008). In critically ill neonates, it is standard practice to transfuse after 10–15 mL/kg has been sampled. In critically ill patients, there may be other influences decreasing the haematocrit, such as blood loss, haemolysis, and expanded intravascular and/or extracellular volume. The potential clinical effects of the poisoning upon blood volume, haematocrit and oxygen transfer will also need to be considered.
Clarke's Analysis of Drugs and Poisons Chapter No. 27 Dated: 10/3/2011 At Time: 12:44:26
Treatment of poisoning
441
Consequently, smaller volumes are available for analysis. More sensitive assays are therefore required for the analysis of samples from children. Capillary electrophoresis (CE), and liquid chromatography– mass spectrometry (LC-MS) are newer methods that enable highly sensitive assays to be performed on small sample volumes. Topical anaesthetics (lidocaine, prilocaine, tetracaine or amethocaine) are effective in reducing the pain of venepuncture in children aged from the neonatal period through to adolescence. Distraction through the use of play therapy and/or music therapy is an effective means for reducing the stress of venepuncture (Caprilli et al. 2007). However, it should be considered whether sampling is necessary and sampling should be avoided when the results will not contribute to patient management. Samples obtained by heel-prick sampling yield results comparable with those obtained by venepuncture (Webb et al. 2007). Heel-prick samples are capillary blood as opposed to venous blood. Therapeutic drug monitoring of drugs such as ciclosporin and tacrolimus can be performed using capillary blood specimens because of a high correlation between capillary and venous concentrations. A pitfall in capillary blood sampling arises if the skin is contaminated with the analyte, e.g. if investigating an exposure where the child has had the putative agent on his or her hands and then a finger-prick specimen is obtained. In the majority of cases of paediatric poisons exposure, blood sampling will not be necessary. However, where blood concentrations are decision criteria, e.g. paracetamol poisoning, or where intentional poisoning is suspected, then blood sampling will be necessary. Blood concentrations give information about the magnitude of exposure (i.e. they are quantitative), unlike urine concentrations which give less information about the magnitude of exposure (i.e. they are qualitative). Blood concentrations can be used to determine whether to intervene, (e.g. paracetamol, lead, theophylline) or whether to monitor treatment (e.g. phenobarbital).
plasma concentration of the drug and the characteristics of the saliva (production rate, pH, salivary binding proteins and salivary enzymes) (Aps, Martens 2005). Hence, salivary assays may be more useful in documenting exposure than in determining management criteria or the extent of exposure (see Chapter 18).
Urine drug screens
Treatment of poisoning in children requires dose adjustment for size and development. This is complicated by the lack of paediatric dosing information for many antidotes. Dosing information can be obtained from the BNF for Children, as well as from electronic resources such as Toxbase, TOXINZ and Poisindex. These electronic resources require a subscription in order to obtain access, but the poisons information services will invariably have access. Decontamination, although once standard practice, is increasingly being matched to the risk posed by the exposure. Charcoal is used for carbon-based poisons, but is not effective for metals, strong acids or alkalis. Whole-bowel lavage can be used for slow-release preparations and for metals (e.g. lithium) (Buckley et al. 1995). However, there is a significant risk of aspiration with paediatric decontamination. Gastric lavage can result in aspiration in around 4% of children (Tibballs et al. 1985). Charcoal aspiration, particularly when nasogastric or orogastric tubes are used, is a hazard and can lead to respiratory failure and death (Golej et al. 2001). Whole-bowel lavage, with polyethylene glycol electrolyte lavage solutions, requires large volumes of fluid, beyond the capacity of a child to ingest orally. Hence, nasogastric or orogastric tubes are required. Care is also required for the correct placement of these tubes in order to avoid aspiration. It is accordingly important to consider the risk–benefit ratio of decontamination, and to avoid decontamination when no significant toxicity is predicted. When considering decontamination, the hazard posed by the exposure should be assessed by determining the following:
Urine drug screens may be used to confirm exposure to poisons but are of limited utility in the immediate management of acute poisoning. Urine can be collected by the clean catch method in young children, and by using collection bags in infants. Bacterial contamination of the sample is common, and if a sterile sample is required from an infant then a catheter or suprapubic aspirate sample may be collected. Many of the agents involved in paediatric poisons exposure may not be detectable by routine urine drug screens (Hwang et al. 2003), the results of the test may not be available until after the clinical presentation has resolved, and the vast majority of paediatric poisons exposures do not pose a significant hazard. Hence, performing urine drug screens routinely is not justifiable. However, where there is a diagnostic dilemma, or where deliberate poisoning by a third party is suspected, urine drug screens are warranted. When intentional poisoning is suspected it will be necessary to inform the clinicians responsible for the care of the patient, and the police or the coroner (depending on the circumstances). In most jurisdictions there is a legal requirement to report assaults on children to either the police or child protection authorities. Intentional poisoning would constitute an assault under most legal systems. It is the responsibility of the health professional who suspects that the assault has occurred to report the incident to the police/child protection authorities. Hence, reports should be made directly to the police/child protection authorities rather than to other health professionals. The police will be responsible for documenting the chain of evidence. However, the procedures will vary between legal jurisdictions and it will be necessary for the medical scientist to be familiar with procedures in each state or country in which they work. Saliva
The rate of production of saliva is influenced by hydration status, food, drugs and diurnal variations. In general, drugs are transferred into saliva by passive diffusion and the saliva : plasma ratio of drugs is influenced by the physicochemical properties of the drug (such as pKa, lipid solubility, molecular weight, spatial configuration and charge), the unbound
Hair
Hair analysis can be used to document exposure and give an indication of the timing of exposure in the medium term. When the timing of sampling is late, it may not be possible to detect drugs in blood or urine samples, but it may be possible to detect their presence in hair (Kintz et al. 2007). This approach has been used to detect drugs such as benzodiazepines, zopiclone, barbiturates, methadone, glibenclamide, trimeprazine and diphenhydramine (Kintz et al. 2006, 2007; Villain et al. 2006). More commonly, hair analysis has been used to demonstrate exposure to arsenic or heavy metals (such as mercury, lead, cadmium, chromium, and manganese). Samples of fingernail clippings and teeth (‘milk’ or deciduous teeth are shed naturally in children) can also be analysed to demonstrate heavy-metal exposure (see Chapter 19). Neonatal hair can be analysed to determine intrauterine exposure to ethanol and nicotine as well as illicit drugs such as cocaine, methamfetamine, opioids, cannabinoids and benzodiazepines (Koren et al. 2008). Meconium
In utero exposure to opioids, cocaine, benzodiazepines and ethanol can be determined from the analysis of meconium passed by the newborn (Lopez et al. 2009; Moller et al. 2010; Wang et al. 2010). ELISA and LCMS methods have been reported, as has good correlation between meconium and hair samples. The results have been used by child protection authorities to establish in utero exposure to substance misuse, and also for population studies.
Treatment of poisoning
n n n n
Does the agent have known toxicity? Has the child been exposed to a significant amount of the poison? Are there any confirmatory signs of exposure? Are there alternative safer treatments than decontamination, e.g. antidotes, intravenous fluids?
Some treatment protocols need to be modified for children. The administration of N-acetylcysteine requires relatively high amounts of dextrose solutions which for a child may result in water intoxication. For children, these administration protocols need to be modified to deliver smaller volumes of dextrose.
Clarke's Analysis of Drugs and Poisons Chapter No. 27 Dated: 10/3/2011 At Time: 12:44:26
442
Paediatric Toxicology
Conversely, some antidote doses are based upon the toxin load rather than the size of the patient. For example, with snake or spider bite envenomation, the dose of antivenom is based on the bite/quantity of venom rather than the size/age of the patient. In this case the dose should not be modified for children. However, there should be a lower threshold for treatment in children because the amount of toxin delivered by a snake bite is the same as that for an adult but the dose is proportionately higher because of their smaller body size (White 1995). When a medicine is used in an off-label or unlicensed manner, it is important to make a clinical assessment of its appropriateness. For medicines that are in common use (i.e. when it is the recognised standard of care) or when good-quality evidence is available, the medicine should be prescribed in the normal manner (Gazarian et al. 2006). Where such evidence is lacking, the use of the medicine could be considered experimental or innovative, and measures such as peer-review and informed consent should be conducted prior to use. Response to poisoning (pharmacodynamics) Pharmacodynamic differences between children and adults have been described, e.g. prepubertal children show enhanced response to warfarin compared with pubertal children and adults (Takahashi et al. 2000). The insulin requirements of children with established type 1 diabetes (after the remission phase) are around 0.7–1.0 units/kg per day, increasing at puberty to around 1.4–1.6 units/kg per day in boys and around 1 unit/kg per day in girls, and then decreasing a few years after puberty to adult requirements of around 0.7–0.8 units/kg per day. These insulin dosing requirements are influenced by rates of growth. Selective serotonin reuptake inhibitors (SSRIs) appear to have less efficacy in children and adolescents than in adults, but this may reflect difficulties in diagnosing depression in this age group. Differences in drug response between children and adults may also parallel different susceptibilities to poisoning, e.g. with aspirin causing Reye syndrome and SSRIs causing aggressive behaviour (see below). Aspirin
A 14-year-old boy presented with encephalopathy following the use of aspirin for symptom control during a mild influenza-like illness. Laboratory investigations demonstrated an elevated plasma ammonium concentration (>700 mmol/L), elevated AST (3355 U/L), elevated ALT (2488 U/L), elevated serum lactate (108 mmol/L) and a prolonged prothrombin time (23.6 s). He had a serum salicylate concentration of 232 mg/L. Despite intensive support he died as a consequence of cerebral oedema and tonsillar herniation. Postmortem findings included diffuse microvesicular steatosis and some macrovesicular steatosis (He et al. 2007). The presentation is consistent with a diagnosis of Reye syndrome, which has been linked with aspirin. The mechanism of the interaction between aspirin and a viral illness to produce this condition has been linked with b-oxidation of fatty acids, whereby some individuals are predisposed to aspirin-inhibiting fatty acid metabolism (Deschamps et al. 1991; Glasgow et al. 1999; He et al. 2007). Cytokines can also downregulate some enzymes involved in exogenous and endogenous substrate metabolism. Reye syndrome was described predominantly in children, and this resulted in recommendations to avoid aspirin in children under the age of 12 years. Selective serotonin reuptake Inhibitors
An 11-year-old boy was initially started on atomoxetine 25 mg/day, which was subsequently increased to 60 mg/day after 2 weeks, for the treatment of aggressive behaviour and attention deficit hyperactivity disorder (ADHD). After the increase in dose, he was noted to be increasingly agitated and to have an increase in mood swings. He was reported to have thrown an object at a teacher, and was uncontrollably agitated, crying and threatening to kill himself. These behaviours ceased when his medications were withdrawn (Paxton, Cranswick 2008). When treated with SSRIs, adolescents and young adults are susceptible to treatment-emergent suicidal ideation and suicide-related behaviour (Reith, Edmonds 2007). This risk appears to be age related, with the
vulnerable window being from adolescence through to early adulthood. Although this appears to be a class effect, the risk varies between individual chemical entities. Atomoxetine has its primary mode of action via the inhibition of presynaptic noradrenaline (norepinephrine) reuptake but it also has some effects on serotonin reuptake. Treatment-emergent suicidal ideation and suicide-related behaviour have also been reported with atomoxetine. Other examples of different susceptibilities to poisoning include response to hypnosedatives and ethanol. Children are more susceptible to the respiratory suppressant effects of hypnosedatives. Ethanol intoxication in children has a higher risk of hypoglycaemia than in adults.
Conclusion The approach to poisoning in childhood is determined by the nature and circumstances of the exposure, the stage of development of the child and the legal jurisdiction. Apart from the approach to sample collection, the toxicological analysis is similar to that for adults. For example, in a child who presents with suspected chloroquine ingestion (see the monograph on Chloroquine), a 1 g dose can cause death in a child compared with 3 g in an adult. Exposure to chloroquine could be confirmed by clinical signs (hypotension, QRS prolongation). Analysis of blood concentrations might be used to further confirm exposure, to make an assessment of dose, to predict outcome and to inform management (duration of observation, need for elimination enhancement). In paraquat poisoning the ingested dose might be relatively small in a child because of the unpleasant taste, and larger in an adult because of suicidal intent (see the monograph on Paraquat). Serum paraquat concentrations can be used as a guide to prognosis and as intervention criteria. Hence, the interpretation of the laboratory data can be placed within the paediatric context.
References Alcorn J, McNamara PJ (2002). Ontogeny of hepatic and renal systemic clearance pathways in infants: part I. Clin Pharmacokinet 41: 959–998. Aldridge A et al. (1979). Caffeine metabolism in the newborn. Clin Pharmacol Ther 25: 447–453. Allegaert K et al. (2005). Tramadol disposition in the very young: an attempt to assess in vivo cytochrome P-450 2D6 activity. Br J Anaesth 95: 231–239. American Academy of Pediatrics Committee on Drugs (2001). Transfer of drugs and other chemicals into human milk. Pediatrics 108: 776–789. Aps JK, Martens LC (2005). Review: The physiology of saliva and transfer of drugs into saliva. Forensic Sci Int 150: 119–131. Austin B (1996). Children and the Holocaust. Available at: www.mtsu.edu/~baustin/ children.html (accessed 25 July 2006). Balakrishnan K et al. (2007). Establishing a baseline for the monitoring of medicines availability for children in the UK. Br J Clin Pharmacol 63: 85–91. Bateman DN et al. (2008). Codeine and breastfeeding. Lancet 372: 625. Bates DW et al. (1997). The costs of adverse drug events in hospitalized patients. Adverse Drug Events Prevention Study Group. JAMA 277: 307–311. Belson MG et al. (1999). The utility of toxicologic analysis in children with suspected ingestions. Pediatr Emerg Care 15: 383–387. Binchy JM et al. (1994). Accidental ingestion of methadone by children in Merseyside. BMJ 308: 1335–1336. Bj€ orkman S (2005). Prediction of drug disposition in infants and children by means of physiologically based pharmacokinetic (PBPK) modelling: theophylline and midazolam as model drugs. Br J Clin Pharmacol 59: 691–704. Bj€ orkman S (2006). Prediction of cytochrome p450-mediated hepatic drug clearance in neonates, infants and children: how accurate are available scaling methods? Clin Pharmacokinet 45: 1–11. Black J, Zenel JA (2003). Child abuse by intentional iron poisoning presenting as shock and persistent acidosis. Pediatrics 111: 197–199. BMA (2009). End of Life – Withdrawing and withholding artificial nutrition and hydration. Available at: www.bma.org.uk/ethics/end_life_issues/ Withholdingwithdrawing.jsp (accessed 6 October 2009). Bonate PL, Howard D (2000). Prospective allometric scaling: does the emperor have clothes? J Clin Pharmacol 40: 665–670. Briggs GG et al. (2008). Drugs in Pregnancy and Lactation: A reference guide to fetal and neonatal risk, 8th edn. Philadelphia: Wolters Kluwer Health/Lippincott Williams & Wilkins. Buckley NA et al. (1995). Controlled release drugs in overdose. Clinical considerations. Drug Saf 12: 73–84. Buckley NA et al. (1999). Preformatted admission charts for poisoning admissions facilitate clinical assessment and research. Ann Emerg Med 34: 476–482.
Clarke's Analysis of Drugs and Poisons Chapter No. 27 Dated: 10/3/2011 At Time: 12:44:26
References Butte NF et al. (2000). Body composition during the first 2 years of life: an updated reference. Pediatr Res 47: 578–585. Caprilli S et al. (2007). Interactive music as a treatment for pain and stress in children during venipuncture: a randomized prospective study. J Dev Behav Pediatr 28: 399–403. Carpenter RG et al. (2005). Repeat sudden unexpected and unexplained infant deaths: natural or unnatural? Lancet 365: 29–35. Carter G et al. (2005). Repeated self-poisoning: increasing severity of self-harm as a predictor of subsequent suicide. Br J Psychiatry 186: 253–257. CDC (Centers for Disease Control and Prevention) (2007). Infant deaths associated with cough and cold medications – two states, 2005. MMWR Morb Mortal Wkly Rep 56: 1– 4. Chen G et al. (2003). Human gastrointestinal sulfotransferases: identification and distribution. Toxicol Appl Pharmacol 187: 186–197. Choonara I et al. (1990). Morphine sulphation in children. Br J Clin Pharmacol 30: 897–900. Classen DC et al. (1997). Adverse drug events in hospitalized patients. Excess length of stay, extra costs, and attributable mortality. JAMA 277: 301–306. Coulthard MG, Haycock GB (2003). Distinguishing between salt poisoning and hypernatraemic dehydration in children. BMJ 326: 157–160. Deschamps D et al. (1991). Inhibition by salicylic acid of the activation and thus oxidation of long chain fatty acids. Possible role in the development of Reye’s syndrome. J Pharmacol Exp Ther 259: 894–904. Dine MS, McGovern ME (1982). Intentional poisoning of children – an overlooked category of child abuse: report of seven cases and review of the literature. Pediatrics 70: 32–35. Edginton AN et al. (2006). A mechanistic approach for the scaling of clearance in children. Clin Pharmacokinet 45: 683–704. Fakhoury M et al. (2005). Localization and mRNA expression of CYP3A and Pglycoprotein in human duodenum as a function of age. Drug Metab Dispos 33: 1603–1607. Fanta S et al. (2007). Developmental pharmacokinetics of ciclosporin – a population pharmacokinetic study in paediatric renal transplant candidates. Br J Clin Pharmacol 64: 772–784. Fanta S et al. (2008). Pharmacogenetics of cyclosporine in children suggests an agedependent influence of ABCB1 polymorphisms. Pharmacogenet Genomics 18: 77–90. Gazarian M et al. (2006). Off-label use of medicines: consensus recommendations for evaluating appropriateness. Med J Aust 185: 544–548. Ginsberg G et al. (2004). Physiologically based pharmacokinetic (PBPK) modeling of caffeine and theophylline in neonates and adults: implications for assessing children’s risks from environmental agents. J Toxicol Environ Health A 67: 297–329. Glasgow JF et al. (1999). The mechanism of inhibition of beta-oxidation by aspirin metabolites in skin fibroblasts from Reye’s syndrome patients and controls. Biochim Biophys Acta 1454: 115–125. Golej J et al. (2001). Severe respiratory failure following charcoal application in a toddler. Resuscitation 49: 315–318. Gornall J (2006). Was message of sudden infant death study misleading? BMJ 333: 1165–1168. Habtemariam B et al. (2009). Population pharmacokinetics of valsartan in pediatrics. Drug Metab Pharmacokinet 24: 145–152. Haque IU, Zaritsky AL (2007). Analysis of the evidence for the lower limit of systolic and mean arterial pressure in children. Pediatr Crit Care Med 8: 138–144. Hawrami SA, Ibrahim N (2004). Experiencing chemical warfare: two physicians tell their story of Halabja in Northern Iraq. Can J Rural Med 9: 178–181. Haycock GB (1998). Development of glomerular filtration and tubular sodium reabsorption in the human fetus and newborn. Br J Urol 81(Suppl 2): 33–38. He M et al. (2007). A new genetic disorder in mitochondrial fatty acid betaoxidation: ACAD9 deficiency. Am J Hum Genet 81: 87–103. Holdsworth MT et al. (2003). Incidence and impact of adverse drug events in pediatric inpatients. Arch Pediatr Adolesc Med 157: 60–65. Holford NH (1996). A size standard for pharmacokinetics. Clin Pharmacokinet 30: 329–332. Hu TM, Hayton WL (2001). Allometric scaling of xenobiotic clearance: uncertainty versus universality. AAPS Pharm Sci 3: E29. Hwang CF et al. (2003). The utility of the history and clinical signs of poisoning in childhood: a prospective study. Ther Drug Monit 25: 728–734. Ieiri I et al. (2004). The MDR1 (ABCB1) gene polymorphism and its clinical implications. Clin Pharmacokinet 43: 553–576. ICH (2000). Clinical Investigation of Medicinal Products in the Paediatric Population E11. Geneva: International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use. www.ich. org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Efficacy/E11/Step4/ E11_Guideline.pdf (accessed 13 January 2011). Johnson TN et al. (2006). Prediction of the clearance of eleven drugs and associated variability in neonates, infants and children. Clin Pharmacokinet 45: 931–956. Kauffman RE (2000). Clinical trials in children: problems and pitfalls. Paediatr Drugs 2: 411–418. Kaushal R et al. (2001). Medication errors and adverse drug events in pediatric inpatients. JAMA 285: 2114–2120.
443
Kintz P et al. (2007). Hair analysis for diphenhydramine after surreptitious administration to a child. Forensic Sci Int 173: 171–174. Kintz P et al. (2006). Determination of trimeprazine-facilitated sedation in children by hair analysis. J Anal Toxicol 30: 400–402. Koren G et al. (2006). Pharmacogenetics of morphine poisoning in a breastfed neonate of a codeine-prescribed mother. Lancet 368: 704. Koren G et al. (2008). Novel methods for the detection of drug and alcohol exposure during pregnancy: implications for maternal and child health. Clin Pharmacol Ther 83: 631–634. Kunac DL et al. (2009). Incidence, preventability, and impact of Adverse Drug Events (ADEs) and potential ADEs in hospitalized children in New Zealand: a prospective observational cohort study. Paediatr Drugs 11: 153–160. Lacroix D et al. (2003). Expression of CYP3A in the human liver – evidence that the shift between CYP3A7 and CYP3A4 occurs immediately after birth. Eur J Biochem 247: 624–634. Launay-Vacher V et al. (2006). Renal tubular drug transporters. Nephron Physiol 103: 97–106. Li L et al. (2000). Fatal methadone poisoning in children: Maryland 1992–1996. Subst Use Misuse 35: 1141–1148. Linday LA (1994). Developmental changes in renal tubular function. J Adolesc Health 15: 648–653. Linday LA et al. (1981). Maturation and renal digoxin clearance. Clin Pharmacol Ther 30: 735–738. Lister P et al. (2008). Effects of blood sample volume on hematocrit in critically ill children and neonates. Paediatr Anaesth 18: 420–425. Lockhart JD, Simmons HE (1973). Hexachlorophene decisions at the FDA. Pediatrics 51: 430–434. Lopez P et al. (2009). Cocaine and opiates use in pregnancy: detection of drugs in neonatal meconium and urine. J Anal Toxicol 33: 351–355. Mahadevan SB et al. (2006). Paracetamol induced hepatotoxicity. Arch Dis Child 91: 598–603. McClure RJ et al. (1996). Epidemiology of Munchausen syndrome by proxy, nonaccidental poisoning, and non-accidental suffocation. Arch Dis Child 75: 57–61. McGain F et al. (2004). Snakebite mortality at Port Moresby General Hospital, Papua New Guinea, 1992–2001. Med J Aust 181: 687–691. Meadow R (1989). ABC of child abuse. Munchausen syndrome by proxy. BMJ 299: 248–250. Meadow R (1993). Non-accidental salt poisoning. Arch Dis Child 68: 448–452. Miles FK et al. (1999). Accidental paracetamol overdose and fulminant hepatic failure in children. Med J Aust 171: 472–475. Milroy CM, Forrest AR (2000). Methadone deaths: a toxicological analysis. J Clin Pathol 53: 277–281. Moller M et al. (2010). Opioid detection in maternal and neonatal hair and meconium: characterization of an at-risk population and implications to fetal toxicology. Ther Drug Monit 32: 318–323. Morimoto T et al. (2004). Adverse drug events and medication errors: detection and classification methods. Qual Saf Health Care 13: 306–314. Murry DJ et al. (1995). Liver volume as a determinant of drug clearance in children and adolescents. Drug Metab Dispos 23: 1110–1116. Mutlib AE et al. (2006). Kinetics of acetaminophen glucuronidation by UDPglucuronosyltransferases 1A1, 1A6, 1A9 and 2B15. Potential implications in acetaminophen-induced hepatotoxicity. Chem Res Toxicol 19: 701–709. National High Blood Pressure Education Program Working Group on Hypertension Control in Children and Adolescents (1996). Update on the 1987 Task Force Report on High Blood Pressure in Children and Adolescents: a working group report from the National High Blood Pressure Education Program. National High Blood Pressure Education Program Working Group on Hypertension Control in Children and Adolescents. Pediatrics 98(4 Pt 1): 649–658. Olhager E et al. (2003). Studies on human body composition during the first 4 months of life using magnetic resonance imaging and isotope dilution. Pediatr Res 54: 906–912. Pariente-Khayat A et al. (1997). Isoniazid acetylation metabolic ratio during maturation in children. Clin Pharmacol Ther 62: 377–383. Paxton GA, Cranswick NE (2008). Acute suicidality after commencing atomoxetine. J Paediatr Child Health 44: 596–598. Pitetti RD et al. (2008). Accidental and nonaccidental poisonings as a cause of apparent life-threatening events in infants. Pediatrics 122: e359–e362. Pragst F et al. (2006). Poisonings with diphenhydramine – a survey of 68 clinical and 55 death cases. Forensic Sci Int 161: 107–189. Reith D et al. (2009). Simultaneous modelling of the Michaelis–Menten kinetics of paracetamol sulphation and glucuronidation. Clin Exp Pharmacol Physiol 36: 35–42. Reith DM, Edmonds L (2007). Assessing the role of drugs in suicidal ideation and suicidality. CNS Drugs 21: 463–472. Reith DM et al. (1996). Relative toxicity of beta blockers in overdose. J Toxicol Clin Toxicol 34: 273–278. Reith DM et al. (2001). Childhood poisoning in Queensland: an analysis of presentation and admission rates. J Paediatr Child Health 37: 446–450. Reith DM et al. (2003a). Serious lead poisoning in childhood: still a problem after a century. J Paediatr Child Health 39: 623–626.
Clarke's Analysis of Drugs and Poisons Chapter No. 27 Dated: 10/3/2011 At Time: 12:44:27
444
Paediatric Toxicology
Reith DM et al. (2003b). Repetition risk for adolescent self-poisoning: a multiple event survival analysis. Aust N Z J Psychiatry 37: 212–218. Reith DM et al. (2003c). Adolescent self-poisoning: a cohort study of subsequent suicide and premature deaths. Crisis 24: 79–84. Rey E et al. (2001). Isoniazid pharmacokinetics in children according to acetylator phenotype. Fundam Clin Pharmacol 15: 355–359. Risk Watch (2003). Let the Clinician Beware! A death caused by a ’safe’ drug. Melbourne, Victoria: The Office of the Chief Clinical Advisor Department of Human Services. www.health.vic.gov.au/clinrisk/downloads/riskwatchedition6.pdf (accessed 28 July 2010). Rivenes SM et al. (1997). Intentional caffeine poisoning in an infant. Pediatrics 99: 736–738. Senst BL et al. (2001). Practical approach to determining costs and frequency of adverse drug events in a health care network. Am J Health Syst Pharm 58: 1126–1132. Shuman RM et al. (1974). Neurotoxicity of hexachlorophene in the human: I. A clinicopathologic study of 248 children. Pediatrics 54: 689–695. Sorbo S et al. (1984). The pharmacokinetics of thiopental in pediatric surgical patients. Anesthesiology 61: 666–670. Southall DP et al. (1997). Covert video recordings of life-threatening child abuse: lessons for child protection. Pediatrics 100: 890–891. Stevens JC et al. (2003). Developmental expression of the major human hepatic CYP3A enzymes. J Pharmacol Exp Ther 307: 573–582. Sugarman JM et al. (1997). Utility of toxicology screening in a pediatric emergency department. Pediatr Emerg Care 13: 194–197. Takahashi H et al. (2000). Developmental changes in pharmacokinetics and pharmacodynamics of warfarin enantiomers in Japanese children. Clin Pharmacol Ther 68: 541–555. Tateishi T et al. (1997). A comparison of hepatic cytochrome P450 protein expression between infancy and postinfancy. Life Sci 61: 2567–2574. Tennefors C, Forsum E (2004). Assessment of body fatness in young children using the skinfold technique and BMI vs body water dilution. Eur J Clin Nutr 58: 541–547. Tibballs J et al. (1985). Drug overdose in children. Aust Paediatr J 21: 7–11.
Tiras S et al. (2006). Nonketotic hyperglycemic coma in toddlers after unintentional methadone ingestion. Ann Emerg Med 48: 448–451. Treluyer JM et al. (1997). Developmental expression of CYP2C and CYP2C-dependent activities in the human liver: in-vivo/in-vitro correlation and inducibility. Pharmacogenetics 7: 441–452. van der Marel CD et al. (2003). Paracetamol and metabolite pharmacokinetics in infants. Eur J Clin Pharmacol 59: 243–251. Vanamo T et al. (2001). Intra-familial child homicide in Finland 1970–1994: incidence, causes of death and demographic characteristics. Forensic Sci Int 117: 199–204. Villain M et al. (2006). Hair to document exposure to glibenclamide. J Chromatogr B Analyt Technol Biomed Life Sci 842: 111–115. Wahl EF et al. (2003). Estimation of glomerular filtration rate and bladder capacity: the effect of maturation, ageing, gender and size. BJU Int 91: 255–262. Wallis LA et al. (2005). Age related reference ranges for respiration rate and heart rate from 4 to 16 years. Arch Dis Child 90: 1117–1121. Wang P et al. (2010). In utero drugs of abuse exposure testing for newborn twins. J Clin Pathol 63: 259–261. Webb NJ et al. (2007). Correlation between finger-prick and venous ciclosporin levels: association with gingival overgrowth and hypertrichosis. Pediatr Nephrol 22: 2111–2118. Wells JC et al. (2005). Prediction of total body water in infants and children. Arch Dis Child 90: 965–971. White J (1995). CSL Antivenom Handbook. Melbourne, Victoria: CSL Limited. Wong IC et al. (2004). Incidence and nature of dosing errors in paediatric medications: a systematic review. Drug Saf 27: 661–670. Woolf AD, Lovejoy FH (1993). Epidemiology of drug overdose in children. Drug Saf 9: 291–308.
Further reading Costello I et al. (2007). Paediatric Drug Handling. London: Pharmaceutical Press. Paediatric Formulary Committee (2009). BNF for Children. London: Pharmaceutical Press.
Clarke's Analysis of Drugs and Poisons Chapter No. 28 Dated: 10/3/2011 At Time: 12:52:40
CHAPTER
28
Sampling, Storage and Stability S Kerrigan
Specimen selection, sampling, storage and stability Appropriate selection, sampling and proper storage of biological evidence are important, yet sometimes overlooked, steps in forensic toxicology. These factors, in combination with drug stability, can profoundly impact the interpretation of results and the outcome of forensic casework. Criteria surrounding each of these are presented and discussed in the material that follows. Further reference to tissue sampling will also be found in other chapters in this book and will be cross-referenced within the text where appropriate.
Specimen selection Selection of the appropriate specimen is a critical component of any toxicological investigation. Circumstances surrounding the case, the availability of specimens, the nature of the investigation and even legal or statutory issues may dictate which specimens are selected, and for what purpose. Timing is an important factor in specimen collection, particularly in antemortem cases where some drugs have short detection times and therefore limited detection windows; examples include detection of an elevated concentration of D9-tetrahydrocannabinol (THC) in blood from an impaired driver, or of g-hydroxybutyric acid (GHB) following an alleged drug-facilitated sexual assault. Postmortem specimens pose additional challenges owing to autolytic and putrefactive changes. Timing is also important in death investigations because it becomes increasingly difficult to obtain good-quality specimens as the time between death and sampling (postmortem interval) increases. Factors such as embalming of the body, decomposition or burial can further complicate interpretation if tissues have been preserved, if specimens are putrefied, or if exhumation is necessary. In order to be able to select the appropriate specimen(s) the toxicologist should have access to the case history, autopsy records/pathologist’s report and all other relevant documents. Some of the important specimen selection considerations are listed in Table 28.1.
temperatures. In order to minimise sample loss, glass containers are preferred if volatile analytes such as solvents or anaesthetic gases are suspected. Plastic containers are more susceptible to interferences by plasticisers such as phthalates that might interfere with the analysis. The use of an inert plastic such as Nalgene decreases the likelihood of chemical interference, but it is good laboratory practice to evaluate all new specimen containers prior to routine use in the laboratory. If plastic containers are chosen, their integrity at low temperatures should be evaluated. Polystyrene is more susceptible to cracking at frozen temperatures than polypropylene vessels. Rubber septa or liners in screwcap containers should be avoided and replaced with inert liners (e.g. polytetrafluoroethylene (PTFE), or Teflon) to reduce leakage and minimise drug adsorption. Antemortem blood samples are generally collected into evacuated glass tubes such as Vacutainer or Venoject for forensic toxicology purposes. Collection of blood into similar glass collection vessels is also good practice in postmortem blood sampling. These tubes allow the sample to be collected into a vessel that already contains necessary additives to stabilise and preserve the matrix. Proper mixing is necessary when sodium fluoride or other additives are used to ensure that dissolution is complete. Blood Blood is one of the most important specimens of toxicological interest as it provides unique advantages over other matrices in terms of the wide variety of analytical methodologies available, the vast amount of published reference data for both antemortem and postmortem drug concentrations, and the interpretive value of the matrix from a pharmacological standpoint. However, antemortem and postmortem blood samples are notably different, and the site of the postmortem blood draw (central or peripheral) can be of critical importance. Determination of parent drug and metabolite concentrations (and their ratios) may also yield useful information pertaining to acute or chronic use. A summary of the common advantages and disadvantages of various specimens is given in Table 28.3.
Collection and sampling Specimen containers
Antemortem blood
It is important that the specimen container is appropriate for the intended use and does not compromise the analytical findings. Container size should be appropriate for the volume or weight of the specimen so that headspace is minimised. Typical specimen collection quantities are given in Table 28.2. Excessive headspace in the container can increase the chance of oxidative loss, volatilisation of analyte (e.g. ethanol and other low-boiling-point compounds) or salting out, which may occur if preservatives are present. Some analytes have a tendency to adhere to plastic or glass surfaces depending on their physicochemical properties. Silanisation of glassware can reduce adsorptive losses for drugs that are present at trace levels (10 mg/L or less). Although glass containers are preferred by many, disposable plastic containers are used routinely for a wide variety of postmortem tissues and antemortem samples, particularly urine. If glass containers are used, it is important to make use of appropriate racks for storage and transportation. One of the major disadvantages of glass is the possibility of breakage, particularly during storage at low
Antemortem blood is collected by venepuncture, typically from the antecubital region of the arm, using a syringe or evacuated container (e.g. Vacutainer, Venoject). Prior to collection, an antiseptic wipe is often used to clean the collection site. Non-alcohol-containing antiseptic wipes such as Betadine (povidone–iodine) are preferred to avoid any contamination that could interfere with alcohol analysis (see Chapter 4). Although evacuated blood tubes are typically glass, plastic tubes have also been evaluated (Karinen et al. 2010). Postmortem blood Postmortem blood collected at autopsy is quite different from antemortem blood collected by venepuncture from both qualitative and quantitative standpoints. Postmortem blood may be more viscous, may contain numerous small clots or sedimented cells, has a lower pH (as low as 5.5 owing to protein degradation), may contain 60–90% water, and is subject to varying degrees of haemolysis. The site of blood 445
Clarke's Analysis of Drugs and Poisons Chapter No. 28 Dated: 10/3/2011 At Time: 12:52:40
446
Sampling, Storage and Stability
Table 28.1 Specimen selection considerations n n n n n n n n n n n n n
Ease of use Ease of specimen collection Presence of interferences Matrix effects Parent drug and/or metabolites Detection time Stability of the drug(s) in the specimen Putrefaction Potential for automated analysis Sample volume Indication of short-term or long-term drug use Reference data Interpretive value
collection should be clearly identified on postmortem specimens and blood from different sources should never be combined. Central blood Cardiac blood samples are ideally collected following opening of the pericardial sac, removal of the pericardium, and removal of the blood from the left or right chamber after the heart has been dried. Collection of central blood by insertion of a needle through the chest wall (‘blind stick’) is practised but is discouraged. Although central blood collected in this manner may be identified to the laboratory as ‘heart blood’, it may be contaminated with pericardial fluid, fluid from the pleural cavity, or blood that has drained from the pulmonary vein or artery or the inferior vena cava (Jones 2007). Blood collected in this manner is considered non-homogeneous. Central blood may contain elevated drug concentrations as a result of postmortem redistribution or contamination (diffusion) from other body compartments (Prouty, Anderson 1990; Yarema, Becker 2005), especially following blunt force trauma. Passive drug release from reservoirs such as the gastrointestinal tract, liver, lungs and myocardium may occur immediately after death; later, cell autolysis and the putrefactive process participate in redistribution (Pelissier-Alicot et al. 2003). Drug properties such as volume of distribution, lipophilicity, protein binding and pKa play a role in the
site- and time-dependent mechanisms responsible for postmortem redistribution. Drugs with high volumes of distribution and basic character appear particularly susceptible to postmortem redistribution and their cardiac blood concentrations should be interpreted accordingly. Postmortem redistribution can account for central/peripheral blood drug concentrations that differ by 10-fold or more. Redistribution is time and concentration dependent and is very difficult to predict. Cardiac blood is typically more plentiful than peripheral blood. Although cardiac blood can be a very useful specimen for screening purposes, the relationship between cardiac blood drug concentrations and antemortem blood drug concentrations is complex. Many toxicologists therefore advise against the use of cardiac blood for quantitative and interpretative work. Peripheral blood Femoral blood is the best specimen for use in postmortem testing (Chapter 10) and should be sampled wherever possible. Blood collection from a ligated vein that has been ‘tied off’ is least likely to be contaminated by other sources of blood or a result of release of drug from tissues and organs. Typically, however, a ‘femoral stick‘ involves the collection of femoral blood from an unligated femoral vein in the groin area. Only a small volume of blood should be collected to avoid ‘milking’ the vein and drawing blood from other sources. Typically 10–20 mL of femoral blood can be collected. Over-sampling of blood from the femoral vein will draw blood from the inferior vena cava, and hence the liver, and from the larger iliac vein. Although sampling from a ligated vein is generally preferred, a comparison of drug concentrations in clamped and unclamped femoral vessels showed good correlation for eight drugs including selective serotonin reuptake inhibitors, benzodiazepines, antihistamines and one opioid (Hargrove, McCutcheon 2008). If femoral blood is not available, subclavian or iliac blood may be an alternative. Blood clots Following a fall or blunt trauma to the head, a victim may survive with circulation intact for several hours. Owing to the decreased circulation in the damaged region of the brain, drug or alcohol concentrations in blood clots (e.g. subdural, subarachnoid and/or epidural) may be influenced by incomplete metabolism. It has been suggested that intracranial
Table 28.2 Typical specimen collection quantities Postmortem
Antemortem
Specimen
Quantity
Specimen
Quantity
Blood, heart
25 mL
Blood
10–20 mL
Blood, peripheral
10–20 mL
Urine
25–100 mL
Urine
All
Amniotic fluid
5–30 mL
Bile
All
Breast milk
10–20 mL
Vitreous humour
All
Meconium
All
Cerebrospinal fluid
All
Hair
Pen-size lock
Gastric contents
All
Saliva
1–5 mL
Liver (remote proximity from liver)
All
Sweat
Microlitres (insensible sweat); 1–5 mL (sensible sweat)
Kidney
50 g
Spleen
50 g
Brain
50 g
Lung
50 g
Hair
50 g Pen-size lock (150–200 hairs or 50 mg)
Sources: Dinis-Oliveira et al. (2010); Hepler, Isenschmid (2007); Kerrigan (2002); Kidwell et al. (1998); Skopp (2004); SOFT/AAFS (2006).
Clarke's Analysis of Drugs and Poisons Chapter No. 28 Dated: 10/3/2011 At Time: 12:52:44
Collection and sampling
447
Table 28.3 Advantages and disadvantages of antemortem and postmortem biological specimens Specimen
Advantages
Disadvantages
Amniotic fluid
n n n n
n n n n
Invasive collection Risk of complications Limited reference data Collection by medical personnel
Bile
n Ease of detection of certain drugs (accumulation) n Particularly useful for conjugated drugs
n n n n
Complex matrix Interferences due to bile salts and fats Requires sample preparation/pretreatment Limited reference data
Blood (AM)
n n n n n
Widely accepted matrix Determines recent drug use (hours–days) Related to pharmacological effect Not readily adulterated Extensive reference data
n Invasive collection n Collection by medical personnel n Shorter detection time
Blood (PM)
n n n n
See above (AM) Reference data widely available Central/peripheral blood drug ratios known for some drugs Cardiac blood typically in plentiful supply but requires caution with interpretation
n n n n n
Susceptible to postmortem redistribution (central) Susceptible to postmortem artefacts and interferences Susceptible to contamination (e.g. trauma) Quality of specimen highly dependent on collection protocol Limited volume of peripheral blood
Brain
n Particularly useful for lipophilic drugs, volatiles
n n n n n
Non-homogeneous matrix Drug concentrations vary by region Complex matrix Requires sample preparation/pretreatment Limited reference data
Breast milk
n Determination of neonatal drug exposure n Not readily adulterated n Many drugs present
n n n n n
Privacy, invasive collection Limited reference data Interferences due to high lipid content Drug content varies with milk composition Variable matrix
Cerebrospinal fluid
n Determines recent drug use (hours–days) n Minimal sample preparation n Relatively few interferences
n Invasive collection n Limited reference data
Gastric contents
n Identification of acute ingestion/delayed absorption n Identification of pill fragments possible n Particularly useful for orally administered drugs/poisons
n n n n
Non-homogeneous matrix Complex matrix Requires sample preparation/pretreatment Requires total specimen collection for interpretation
Hair
n n n n
n n n n n
New technology Recent drug use not detected Environmental contamination Potential for ethnic bias Limited reference data
Kidney
n Particularly useful for non-drug analytes, e.g. metals
n Complex matrix n Requires sample preparation/pretreatment
Liver
n Ease of detection of certain drugs (accumulation) n Interpretive value for some drugs n Reference data available
n Complex matrix n Requires sample preparation/pretreatment
Lung
n Particularly important for volatile analyses
n Complex matrix n Requires sample preparation/pretreatment
Determination of prenatal drug exposure Not readily adulterated Minimal sample preparation Relatively few interferences
and centrally acting drugs
History of drug use (months) Readily available, easy collection Low potential for donor manipulation Useful for drug and non-drug analytes, e.g. metals
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 28 Dated: 10/3/2011 At Time: 12:52:45
448
Sampling, Storage and Stability
Table 28.3 continued Specimen
Advantages
Disadvantages
Meconium
n Long-term window of drug exposure n Non-invasive sample collection
n n n n n
Non-homogeneous matrix Complex matrix (waxy) Interferences Requires sample preparation/pretreatment Limited reference data
Nails
n Easy collection n History of drug use (months) n Particularly useful for metals
n n n n n
Limited data New technology Not yet widely accepted Recent drug use not detected Environmental contamination
Saliva
n n n n n n
n n n n n
New technology Short drug detection time Small sample volume (1–5 mL) Potential for oral contamination Collection method influences specimen pH and drug content
Spleen
n Particularly useful for certain analytes if no blood
n Complex matrix n Requires sample preparation/pretreatment n Limited data for most analytes
Sweat
n n n n n n
History of drug use (weeks) Cumulative measure of drug use Parent drug present Non-invasive collection Less frequent drug testing required Not readily adulterated
n n n n n n n n
Newer technology Potential for environmental contamination High intersubject variability Requires special collection device Skin irritation and discomfort Small sample volume No pharmacological interpretation possible Non-homogeneous matrix (sweat/sebum)
Urine
n n n n n
Widely accepted matrix Easy collection Plentiful supply Amenable to automated analysis Longer detection window than blood (days–weeks)
n n n n
Potential for donor manipulation Minimal parent drug Not useful for quantitative analysis Not related to impairment or pharmacological effect
Vitreous humour
n n n n n n n
Determines recent drug use (hours–days) Related to pharmacological effect Resistant to putrefaction Interpretive value for ethanol-related investigations Minimal sample preparation Relatively few interferences Useful for postmortem chemistry
n Limited data compared with blood n Small sample volume
Readily available, easy collection Parent drug present Related to free drug concentration in plasma Minimal sample preparation Many drugs determined Indicates recent drug use
is available
blood clots may serve as ‘time capsules’ prior to death, because they may reflect drug concentrations several hours prior to death, when an injury may have taken place. Vitreous humour Direct aspiration of vitreous humour using a hypodermic syringe may yield 2–3 mL of fluid per eye. The needle should be placed in the central globe and aspirated with gentle suction. Preservation with sodium fluoride is generally recommended. The eye is located within the protective environment of the orbit and, being essentially outside the body, is remote from other tissues. Vitreous fluid is therefore a particularly useful specimen owing to its anatomical isolation, affording it notable
resistance in terms of microbial invasion and degradation, as well as being remote from the central organs and subsequently less susceptible to postmortem redistribution phenomena. Vitreous humour is particularly useful for cases involving digoxin or hydrophilic analytes including paracetamol (acetaminophen) and salicylates. The equilibrium that exists between blood and vitreous fluid is slower than with other extracellular fluids, which can result in a slight delay in uptake. Furthermore, only free drugs are able to leave the blood and enter the vitreous humour. Since eye fluid is sterile and less susceptible to microbial contamination and hence postmortem alcohol production, it is routinely used for ethanol determination owing to its interpretive value from the standpoint of postmortem alcohol production and the determination of the pre- or post-absorptive phase of ethanol use
Clarke's Analysis of Drugs and Poisons Chapter No. 28 Dated: 10/3/2011 At Time: 12:52:45
Collection and sampling (Honey et al. 2005). Vitreous humour is particularly useful for postmortem analysis of glucose, urea nitrogen, uric acid, creatinine, sodium and chloride. These are important analytes for the evaluation of diabetes, degree of hydration, electrolyte imbalance, postmortem interval and the state of renal function prior to death (Coe 1977, 1993). Sodium, calcium and chloride concentrations in vitreous humour during the early postmortem interval can be used to estimate antemortem serum concentrations. It is therefore important that sodium fluoride is not added to specimens requiring vitreous chemistries. For that reason, vitreous humour is frequently collected into two separate containers: one preserved (for drug and alcohol testing) and one unpreserved (for clinical purposes). Cerebrospinal fluid Cerebrospinal fluid (CSF) can be collected either by lumbar puncture at the base of the spine using a hypodermic syringe or by withdrawal of cisternal fluid by puncturing the base of the neck. Although there are limited published reference data for quantitative drug concentrations in CSF, this clear fluid comprising mostly water is amenable to most routine methods of toxicological analysis. CSF may be of particular importance in alcohol-related cases where no vitreous humour is available, particularly if postmortem alcohol production is suspected. Like vitreous humour, CSF is anatomically isolated and less prone to contamination and bacterial invasion. Although it is more plentiful than vitreous humour, the lack of plentiful reference data limits its usefulness. However, CSF may be particularly useful in surgical death investigations. Bile Bile is generally aspirated from the gallbladder using a hypodermic syringe. It may be necessary to tie off the gallbladder prior to collection if contamination appears to be an issue. Bile should be collected prior to the liver specimen to avoid contamination. Many drugs of forensic interest accumulate in the bile, particularly those that are heavily conjugated, such as opiates, benzodiazepines and cannabinoids. Bile may also be used in cases where chronic heavy-metal poisoning is implicated. However, owing to the presence of bile salts and fats, drug extraction from this matrix can be complicated and extensive extraction and cleanup procedures are often required.
449
stomach content collected and retained by medical staff may provide valuable information concerning drugs or poisons consumed. Urine In antemortem settings, a mid-stream urine sample is usually collected into a plastic container containing sodium fluoride as preservative (Chapter 9). In some settings it may be necessary to take precautions against specimen adulteration. In postmortem settings, urine is collected by insertion using a hypodermic syringe directly into the bladder under visualisation. Puncture of the abdominal wall should be avoided to reduce the possibility of contamination. Urine is a valuable specimen for both antemortem and postmortem drug testing because it is a relatively uncomplicated matrix. However, the multiplicity of factors influencing urine drug concentrations (e.g. urine volume, clearance, metabolism, pH and time of last void) generally means that, in isolation, these results have limited quantitative value. Exceptions to this rule may include ethanol determination in a second void. Care must be exercised when considering the interpretation of urine GHB concentrations as GHB is present as an endogenous compound formed as a by-product of metabolism and may also be produced as a postmortem artefact as a consequence of the breakdown of succinic acid semialdehyde. Tissues When tissues are sampled they should be collected quickly and placed immediately into airtight containers. This is particularly important if volatiles or inhalants are suspected. Liver, kidney, brain, lung and spleen are the most frequently collected postmortem tissues. Liver
Liver is a particularly important organ because of the very large number of drugs that undergo hepatic metabolism and the fairly extensive published reference data that exist. To reduce the possibility of drug diffusion from the small bowel, tissue from deep within the right lobe is preferred (Drummer 2004). The concentrations of drugs and metabolites in liver are often elevated, hence this specimen has limited interpretive value. However, liver is particularly useful for highly protein-bound drugs and the comparison of liver/blood drug ratios may allow the differentiation of acute overdose from chronic drug use for some drugs. Kidney
Gastric contents Gastric content is a potentially valuable specimen for analysis in postmortem and clinical cases. Unabsorbed drug or tablet fragments in the gastric contents may provide valuable information concerning ingested compounds and provide an excellent material for preliminary screening (Chapter 9) owing to the potentially large amounts of drug that may be present. The absence of a drug in the gastric contents does not necessarily preclude oral administration. Odours emanating from the gastric content can provide valuable clues about what may have been consumed, e.g. pesticides and cyanides. The entire contents of the stomach should be collected and weighed. Gastric contents are non-homogeneous and should be homogenised prior to sampling. Quantitative drug determinations should be interpreted within the context of the entire contents (total quantity, rather than concentration) and it is important to take into consideration the differing absorption rates of drugs based on their physicochemical properties as well as their formulations and coatings. The presence of a drug in gastric contents, particularly at low concentration, does not necessarily indicate oral administration. Drugs may be absorbed into the stomach via gastric juices that are in equilibrium with blood or as a result of intranasal drug use. Basic drugs are more susceptible to this because they have a tendency to become trapped in the gastric compartment owing to the low pH. If heavy metals are suspected, gastric contents should be collected, together with intestinal contents. In cases of suspected poisoning where the patient may have survived for a few days in hospital prior to death and where drugs may have been metabolised and eliminated from the body prior to death, any
Most drugs pass through the kidney as a result of urinary elimination. Kidney is an important specimen in cases of suspected heavy-metal poisoning owing to accumulation in this tissue. The presence of heavy metals or ethylene glycol during toxicological tests may be accompanied by structural changes to the kidney that can be documented using histological tests. Spleen
Spleen is an important specimen for cyanide or carbon monoxide analyses, particularly in fire-related deaths where blood may be compromised or unavailable. Lung and brain are valuable specimens in cases involving volatiles or inhalants. Brain
Brain tissue is lipid rich and has a tendency to concentrate some drugs, particularly lipophilic analytes, narcotics and halogenated hydrocarbons (Skopp 2004). If quantitative drug brain concentrations are used, it is important to know the location of the specimen because the brain is a non-homogeneous matrix. Drug concentrations within the brain may vary several-fold from one region to another owing to its complex structure and differing composition. Brain is not widely used in routine toxicological analysis. Muscle
Muscle is not routinely encountered, despite the fact that it frequently contains relatively high drug concentrations, particularly for substances with high volumes of distribution. Perfusion rates between sites and drug concentrations are not consistent, and drug concentrations must be interpreted accordingly. Muscle is encountered more frequently for
Clarke's Analysis of Drugs and Poisons Chapter No. 28 Dated: 10/3/2011 At Time: 12:52:45
450
Sampling, Storage and Stability
ethanol determination in the absence of blood, or during the investigation of a suspected injection site. Hair
Hair has been used in a variety of antemortem toxicology settings to provide a history of drug exposure and has therefore found applications in workplace drug testing, in monitoring of persons on probation or on parole for drug use, in insurance testing to verify the truthfulness of statements made by applicants relating to whether they use drugs or are smokers, in child endangerment, in drug-facilitated sexual assault and in other types of criminal casework (Nakahara 1999; Kintz et al. 2006; Curtis, Greenberg 2008). Further discussion relating to the scenarios in which hair testing is employed is provided in Chapters 8, 19 and 27. One of the major advantages is the long drug detection window compared with many other specimens. Hair may allow drug exposure over several weeks or months to be determined, depending on the length of the hair. Segmenting the hair by length may allow an approximate timeline for exposure to be determined based on head hair growth rates of approximately 1 cm per month (Clauwaert et al. 2000). Hair should be cut as close as possible to the scalp from the posterior vertex region of the head, since this region shows least variation in growth rate. Typically a lock of hair equivalent to the thickness of a pen or pencil is collected. The colour, length, sampling site and any obvious cosmetic treatment of the hair should be recorded. The root (proximal) and tip (distal) sections of the hair should be clearly identified. Although head hair is the preferred specimen, hair from other sites (e.g. pubis, axillae) may be used, but interpretation of analytical findings may be more complex. The lock of hair is typically tied, wrapped in aluminium foil and stored under dry conditions in the dark at room temperature. Hair is also a useful specimen in postmortem investigations where arsenic or heavy metals are suspected. Although postmortem hair analysis is not yet widespread, there is growing interest because it may provide valuable interpretive information pertaining to the chronological sequence of toxin exposure (Cirimele et al. 2002). Hair has also proved to be useful in cases where exhumation is necessary (Tsatsakis et al. 2001). If hair is collected post mortem, it should be sampled at the very beginning of the examination to reduce the risk of contamination. Hair can provide complementary toxicological information. Issues with drug testing in hair include external contamination, ethnicity and pigmentation, chemical treatment and the use of appropriate cut-off concentrations. Contamination of the hair with drugs from other sources (external deposition, environmental contamination, sweat or sebum) is generally minimised by pretreatment of the sample using a variety of aqueous and organic rinses or wash steps prior to analysis. Other keratinised specimens such as nails can also be used to determine long-term exposure to drugs or poisons, in particular heavy metals such as thallium, arsenic or lead. However, drugs are deposited into nails at a much slower rate. External decontamination procedures should be performed prior to analysis (see Chapter 19). Injection sites Excision of skin and tissue (muscle) may be necessary in postmortem investigation of a suspected injection site. Typically a cube of muscle and skin is removed for this purpose. However, it is important to compare the drug concentrations in the suspected injection site with those in a control specimen from the same individual where there is no evidence of injection. Injection sites are not always reliable indicators of drug administration since the presence or absence of drugs in injection site tissue is dependent on the type and depth of the injection. If the injection is made directly into a blood vessel, little drug is likely to remain in the surrounding tissue. Entomological specimens The potential use of insects for detecting drugs and other toxins in decomposing tissues has been demonstrated and reviewed (Introna et al. 2001). If insects or larvae are collected from human remains they should be frozen as soon as possible. Larvae rapidly eliminate drugs when removed from the food source. Drugs, metals and pesticides have been identified in
entomological specimens including larvae and pupae. Following wash steps to remove external contamination, entomological specimens are homogenised and analysed in a manner similar to that for tissues. Saliva Saliva or oral fluid can be collected non-invasively by expectoration, by aspiration, by vacuum or by saturation of an absorbent swab (Kidwell et al. 1998). Detection times are comparable to those in blood. As much as 1.5 L of saliva per day is produced by the submandibular, parotid and sublingual glands inside the mouth. Secretions from a specific gland may be collected using a special device or by cannulation, but this is uncommon. Although specific gland secretions are advantageous from a standpoint of saliva : plasma ratio and reduced oral contamination, mixed saliva is typically collected for routine drug-testing purposes. Oral fluid can be collected non-invasively, conveniently and without invasion of privacy. Chewing an inert substance, such as Teflon tape or a rubber band, may increase salivation for the purpose of specimen collection. It should be verified that no adsorption takes place between the drug and the chewed substance. Acidic sweets or citric acid has also been used to stimulate glandular secretions. Care must be taken that residual food, drink or interfering substances inside the mouth do not interfere with the analysis. This is particularly important for drugs that are ingested orally or smoked. Owing to the ease and non-invasive nature of specimen collection, saliva is of particular interest in workplace drug testing, for insurance testing and, more recently, for roadside impairment testing. Saliva contains serous fluid derived from plasma. This ultrafiltrate of interstitial fluid contains the unbound fraction of drug at concentrations that are typically proportional to those measured in plasma. However, the predictable relationship that theoretically exists between saliva and plasma drug concentrations is influenced by many factors such as saliva flow rate, which can complicate pharmacological interpretation (Crouch 2005). A more detailed discussion on saliva and oral fluid testing is provided in Chapter 18. Sweat Moisture loss via the skin and elimination of insensible (non-visible) sweat take place during normal breathing at a rate of 0.3–0.7 L/day. Sensible sweat refers to perspiration that is actively excreted during stress, exercise or extreme temperature, at rates of 2–4 L/h. About half the total volume of sweat is eliminated from the trunk of the body. The remaining fluid is lost from the legs or upper extremities and head in approximately equal amounts (Kidwell et al. 1998). Sweat is usually collected using an adhesive absorbent patch that is placed on the surface of clean skin or by wiping the skin with a swab or gauze. Careful preparation of the skin is necessary prior to placement of a sweat patch to minimise external drug contamination or bacterial degradation of the drug once it has been retained. Use of a semi-permeable membrane to cover the absorbent pad prevents non-volatile components in the environment from penetrating the pad externally, but allows oxygen, water and carbon dioxide to diffuse through. Salts, solids and drugs that pass through the skin are trapped in the absorbent pad, where they are temporarily stored in situ, until the patch is removed. Owing to the relatively small volume (mL) of insensible sweat secreted from a small absorbent area (typically 3 5 cm), patches are typically worn for several days on the outer portion of the upper arm or back. In practice most skin wipes or sweat patches contain a mixture of sweat and sebum, the oily secretion from the sebaceous glands. As with saliva, increased flow rates can influence the quantity of drug eliminated into sweat. This specimen is particularly useful for compliance testing or monitoring long-term exposure (weeks), which might be desirable in probation or parole settings. Amniotic fluid Amniotic fluid has been used to investigate prenatal drug exposure. Its collection (amniocentesis) typically takes place between weeks 16 and 20
Clarke's Analysis of Drugs and Poisons Chapter No. 28 Dated: 10/3/2011 At Time: 12:52:45
Sample handling of pregnancy. A needle is inserted through the abdomen into the uterus where there is the least chance of touching the placenta or the foetus. The collection of amniotic fluid is typically performed in conjunction with ultrasound visualisation in order to reduce the risk of damaging the developing foetus. Although complications are rare, miscarriage occurs in a very small percentage of women. Typically 5–30 mL of amniotic fluid is removed during the procedure. Breast milk During pregnancy, oestrogen and progesterone, secreted in the ovary and placenta, cause milk-producing glands in the fatty tissue of the breasts to develop and become active. The pituitary hormone prolactin stimulates the production of fluid (600–1000 mL/day) by the milksecreting cells. Contraction of the myoepithelial cells surrounding the alveoli allows the milk to be expressed. For specimen collection purposes, a breast pump can be used. The matrix is somewhat nonhomogeneous. Colostrum, a creamy white to yellow pre-milk fluid, may be expressed from the nipples during the last trimester of pregnancy and shortly after delivery. Many drugs are excreted into breast milk and the scientific and medical literature contains numerous citations of the presence of drugs in this matrix (Chapter 27). Drugs that are extensively protein bound may not readily pass into the milk, but emulsified fats contained in the milk may concentrate highly lipid-soluble drugs. The high lipid content and natural emulsifying agents present in breast milk mean that some sample pretreatment is often required. Meconium Meconium formation begins between weeks 12 and 16 of gestation. As the first faecal matter passed by the neonate, it is typically collected within 1–5 days of birth. Analysis of drugs in meconium may provide a relatively long-term history of drug exposure during pregnancy, in particular the last 20 weeks of gestation. It provides more complete and long-term information on drug exposure than neonatal urine or cord blood. The specimen is complex and non-homogeneous. All available samples should be collected and homogenised prior to analysis. Meconium and other important matrices involved in maternal–foetal medicine have been reviewed (Gray, Huestis 2007; Lozano et al. 2007).
Sample handling Sample handling is an important consideration during the pre-analytical phase. Unlike a clinical setting, where the time between sample collection and testing is often very short, significant delays are common in a forensic setting. The pre-analytical phase may be considerable, spanning the time of death and/or discovery of a victim, autopsy and collection of specimens, sample storage, transport to the laboratory and subsequent storage prior to analytical testing. In antemortem toxicology settings, the time delay between an alleged offence and specimen collection may be short (e.g. minutes to hours in the case of most impaired driving cases) or long (e.g. hours to days in the case of some sexual assault cases). Following collection, antemortem specimens may be subject to similar delays due to shipping or transport of specimens, requests for testing made by the submitting agency and storage of samples prior to actual testing. Although the toxicologist must consider the time delay between the event (i.e. death, or committing or being the victim of an offence) and collection of a specimen for interpretation purposes, these delays are beyond the control of the laboratory. Measures can be taken, however, to preserve and maintain the integrity of specimens after collection. Sample quality plays an important role in the validity or usefulness of subsequent analytical determinations. Inappropriate sample preservation or storage may have a deleterious effect on qualitative and quantitative determinations. Preservation and storage Specimens should be stored at appropriate temperatures, with adequate preservative and in an environment accessible only to authorised
451
personnel to ensure security and integrity. Short-term storage at refrigerated temperature (4 C) is recommended for most samples, or frozen ( 20 C or lower) during long-term storage (more than 2 weeks). Exceptions to this include hair, nails or dried blood swatches on filter paper, which can be stored at ambient temperatures. Whereas clinical specimens are typically unpreserved, the use of a chemical preservative is often warranted in forensic specimens. Preservation of blood samples with sodium fluoride (2% w/v) is routine in most laboratories. Commercial evacuated blood collection tubes (e.g. grey-top tubes) contain sodium fluoride as the preservative and potassium oxalate as the anticoagulant. These are the preferred evacuated blood tubes for antemortem forensic toxicology casework. Inhibition of microorganisms and enzymes with sodium fluoride is important for commonly encountered analytes such as ethanol, cocaine and others. Fluoride acts as an enzyme inhibitor and helps prevent glycolysis. Commercial blood tubes may contain a wide variety of additives (citrate, heparin, EDTA, thrombin, acid citrate dextrose mixtures, clot activator, etc.). Although these tubes are designed for a variety of clinical uses, they are not the preferred specimen containers for drug-testing purposes. Laboratories frequently encounter these blood tubes when they are submitted from a hospital setting and special care must be taken when interpreting their results (LeBeau et al. 2000; Toennes, Kauert 2001). If an anticoagulant is to be used, potassium oxalate is preferred rather than alternatives such as EDTA, heparin or citrate. Antioxidants such as ascorbic acid (0.25% w/v) or sodium metabisulfite (1% w/v) are sometimes used to prevent oxidative losses, but these agents have the potential to act as reducing agents towards some drugs, in particular N-oxide metabolites, which may be transformed into the parent drug. In a similar fashion, adjustment of specimen pH is not generally favoured routinely, because, just as some drugs are alkali labile (e.g. cocaine, 6-acetylmorphine), others are acid labile. Sodium azide (0.1% w/v) is sometimes used as a preservative and antimicrobial agent in urine samples. Sodium azide should not be used if samples are to be analysed by enzyme-linked immunosorbent assay because it can interfere with horseradish peroxidase-mediated colorimetric detection. Although the addition of preservative should be routine for most antemortem and postmortem blood samples, an aliquot of unpreserved postmortem blood is sometimes collected. For example, fluoride preservatives should not be used if organophosphorus chemicals are suspected since this accelerates chemical degradation (Skopp, Potsch 2004). Some drugs are known to be photolabile (e.g. ergot alkaloids such as lysergic acid diethylamide and the phenothiazines). Specimens known to contain photolabile drugs should be stored in amber vials or foilcovered containers, or otherwise protected from direct sources of light. Storage of tightly sealed appropriate containers at low temperature further inhibits sample loss. Short-term storage at refrigerated (4 C) and frozen ( 20 C) temperatures is commonplace in most laboratories and repeated freeze–thaw cycles should be avoided. Labelling and specimen transfer All samples should be properly marked for identification with the case number, donor name, date and time of collection, signature or initials of the collector and specimen description. Tamper-proof containers and/ or tape bearing the collector’s initials and date of collection should be used. Specimens should be forwarded to the laboratory in appropriate leak-proof and tamper-proof packaging/shipping materials with all appropriate documentation (chain-of-custody forms, requisitions for testing, special requests, case information, medications list, police report, donor information/identifier such as date of birth or social security number, agency case number, pathologist/police officer name and contact information). Improperly packaged or identified materials should be returned to the submitting agency. Documentation accompanying the specimen(s) should list all of the specimens that were collected or available for testing. Once received by the laboratory, the specimens should be inspected and appropriately documented in terms of condition and quantity during the accessioning process.
Clarke's Analysis of Drugs and Poisons Chapter No. 28 Dated: 10/3/2011 At Time: 12:52:46
452
Sampling, Storage and Stability
Contamination There are a variety of contamination sources for both antemortem and postmortem specimens. In addition to the potential contamination issues that may result from the use of containers and external factors, a number of important exogenous and endogenous sources of contamination should be considered. Exogenous contaminants Specimens collected into plastic containers are sometimes susceptible to phthalate interferences. Numerous plasticiser interferences such as dibutylphthalate may co-extract and interfere with analytical detection by gas-chromatographic or mass-spectrometric techniques, yielding characteristic phthalate ions. All plastic containers should be evaluated prior to widespread implementation. It should be noted that contamination from phthalates may occur during the analytical process through use of disposable pipette tips, solvent containers, solid-phase extraction cartridges, tubing and numerous other sources. However, environmental exposure to these substances from household items, food, beverages and other sources can produce detectable quantities of phthalate esters or their metabolites in biological specimens including blood, serum, urine and breast milk (Silva et al. 2005; H€ ogberg et al. 2008). Embalming fluids, which typically contain a variety of alcohols and aldehydes, are a potential source of contamination in postmortem casework. These fluids not only dilute any remaining fluid in the body, but also alter drug distribution in remaining tissues. Another potential source of contamination comes from reusable syringes and containers for postmortem specimen collection. Some cleaning fluids that are used for syringes may contain alcohols that can compromise the analysis of volatiles. This highlights the importance of analysing specimens from multiple sites and using disposable syringes wherever possible. The principal concern with antemortem contamination arises from the intentional manipulation of the sample to mask the presence of drugs.This typically involves the substitution, dilution or adulteration of the biological specimen with a foreign substance. Donor manipulation occurs most frequently with urine samples in workplace drugtesting situations (see Chapter 3). As a result, specimen validity testing is required in some drug-testing programmes such as for federal employees under US Department of Health and Human Services (DHHS) guidelines. Initially, adulteration of urine for drug-testing purposes involved the use of crude household items such as soap, bleach, vinegar, ammonia or cleaning fluids. Although these substances met with some success, a wide variety of commercial adulteration reagents and kits is now widely available (Dasgupta 2007). A summary of in-vitro adulteration agents is provided in Table 28.4. Some of the most popular commercial products contain glutaraldehyde (fixative), pyridinium chlorochromate (PCC) or chromium(VI)-containing species (oxidant), nitrite (oxidant) or peroxide/peroxidase. In general, in-vitro adulterants can interfere with presumptive immunoassay tests, with the intention of producing false-negative results. However, some agents have the potential to interfere with confirmatory tests such as gas chromatography/ mass spectrometry (GC-MS) as well. Although this is less likely, studies have shown that some reagents may produce lower than expected or negative results for some analytes. Adulteration detection products are available commercially. On-site or dipstick tests are available for nitrite, glutaraldehyde, pH, specific gravity, creatinine, bleach, PCC and oxidants. Specimen dilution or in-vivo adulteration by ingestion of a substance to mask the presence of drugs is also encountered. This is commonly achieved by the ingestion of large quantities of fluid prior to the test or by administration of a diuretic. Examples of in-vivo adulteration agents are given in Table 28.5. Urine specimen substitution or dilution can be detected if specimen validity tests are performed. A specimen may be considered invalid if the pH is between 3 and 4.5 or between 9 and 11. It may be adulterated if the pH is less than 3 or greater than 11. The normal temperature range is 32–38 C. A specimen is considered dilute if the
Table 28.4 In-vitro adulteration agents n n n n n n n n n n n n n n n n n n n n n n n n n n n
Ascorbic acid Alcohols Amber-13 (hydrochloric acid) Ammonia Bleach Clear Choice (glutaraldehyde) Detergent or soap (surfactant) Drano Ethylene glycol Gasoline Glutaraldehyde Hydrogen peroxide Klear (potassium nitrite) Lemon juice Liquid soap Lime-A-Way Mary Jane Super Clean 13 (surfactant) Salt Stealth (peroxide/peroxidase) THC-Free (hydrochloric acid) UrinAid (glutaraldehyde) Urine Luck (chromium VI, oxidant) Vanish Vinegar Visine Water Whizzies (sodium nitrite)
Source: Kerrigan, Goldberger (2005).
Table 28.5 In-vivo adulteration agents Diuretics Prescription n Thiazides and thiazide-like drugs (e.g. hydrochlorothiazide, metolazone) n Carbonic anhydrase inhibitors (e.g. acetazolamide) n Loop diuretics (e.g. bumetanide, furosemide, torsemide) n Osmotic diuretics (e.g. mannitol) Over the counter (OTC) Aqua-Ban Diurex Fem-1 Midol Pamprin Premsyn PMS
n n n n n n
Other n Alcoholic beverages n Xanthines (e.g. caffeine, theophylline, 8-bromotheophylline) n Herbals and aquaretics (e.g. golden seal root, juniper) Source: Kerrigan, Goldberger (2005).
creatinine concentration is less than 200 mg/L and the specific gravity is less than 1.003. Other sources of contaminants or unexpected analytes include pyrolytic breakdown products due to thermal degradation of drugs. These may be present due to pyrolysis during administration of the drug (e.g. anhydroecgonine methyl ester following crack cocaine use) or occasionally in situ during analysis if conditions are not properly controlled or evaluated. Other sources of contamination may arise from pharmaceutical impurities or adulterants and cutting agents that are incorporated into illicit drugs prior to sale.
Clarke's Analysis of Drugs and Poisons Chapter No. 28 Dated: 10/3/2011 At Time: 12:52:46
Stability Medical artefacts Clinical therapy can sometimes produce medical artefacts that complicate toxicological findings. Medical artefacts are most common in postmortem cases where infusion pumps may continue to run after death, introducing high concentrations of drug in local body compartments. Access to hospital records and case information, and collection of peripheral blood, vitreous fluid and liver are particularly important in these types of cases. Other sources of medical artefacts may include organ harvest drugs such as the calcium-channel blocker verapamil, or papaverine, which is used to inhibit vasoconstriction during transplant surgery. If living patients are administered fluids (e.g. saline) during clinical care, blood is only contaminated (diluted) with the infusion solution if it is collected downstream from the intravenous line. Blood circulation and equilibrium with tissues is rapid, so the administration of fluids does not usually influence drug or alcohol concentrations in blood if normal precautions are taken. If downstream collection is suspected, careful review of the medical records and/or measurement of the haematocrit to determine specimen dilution may be necessary. Endogenous contaminants, artefacts and interferences By their very nature, all biological specimens are subject to endogenous interferences, regardless of whether or not they are derived from living or deceased persons. More complex biological specimens such as blood, tissue or meconium will require more extensive sample preparation to remove these interferences than less complex matrices such as vitreous humour, or cerebrospinal or oral fluid. In general, however, antemortem specimens are somewhat less susceptible to endogenous artefacts or contaminants. Ethanol, GHB, carbon monoxide, cyanide and other short-chain alcohols can be metabolically produced post mortem (Skopp 2004). The formation of toxicologically significant concentrations of cyanide in postmortem tissue (Lokan et al. 1987) has been attributed to the conversion of thiocyanate to cyanide and the breakdown of protein (Curry et al. 1967). Although in some circumstances ethanol can be produced in situ in unpreserved antemortem fluids, the same is true to a far greater extent in postmortem specimens, particularly blood. Likewise, GHB is present in antemortem fluids at very low concentrations in the absence of a serious genetic disorder such as GHBuria (Knerr et al. 2007). Differentiation of exogenous and endogenous GHB is complicated by specimen type, storage conditions, preservative and other factors. Many laboratories use a cut-off concentration to help differentiate the two, for example 10 mg/L in urine (Kerrigan 2002; LeBeau et al. 2007). Concentrations of GHB may increase in urine during storage, upon collection and storage of unpreserved blood, or in citrate-buffered antemortem blood (LeBeau et al. 2000). Although preserved antemortem blood GHB concentrations are typically lower than those in urine, numerous studies have shown forensically significant concentrations of GHB in postmortem blood. Postmortem urine and vitreous fluid appear to be less susceptible to this increase. Major changes that occur after death produce autolytic changes and putrefaction by microorganisms. Invasion of microorganisms, particularly from the gastrointestinal tract into tissues and body fluids, occurs within hours at ambient temperature. Lipids, carbohydrates and proteins are hydrolysed by microbial enzymes, the pH of blood steadily increases, and the putrefactive amines, tyramine, tryptamines, phenethylamines and other endogenous substances are liberated. Trauma is a non-preventable source of contamination in postmortem forensic toxicology. Rupture of organs or compartments within the body can compromise quantitative drug analyses owing to the mixing of fluids (e.g. of gastric contents with blood) or from the microbial action that occurs as a result. Postmortem alcohol production can also result in detectable quantities of ethanol as an artefact. Glycolysis and the presence of yeasts and microorganisms can convert a variety of postmortem substrates to ethanol. Although concentrations are typically low (18) can be extracted as non-ionised molecules at a pH range around their isoelectric zone, where the ionisation of the basic group has ended and the ionisation of the acidic group has not yet begun. In the special case that pKa þ pKb ¼ 18, the isoelectric zone shrinks to a particular point (isoelectric point) at which the molecule is not ionised. Example: n
Sulfacetamide: pKa ¼ 5.4 and pKb ¼ 12.2 (pKa þ pKb ¼ 17.6). To a great extent the molecule is not ionised at pH ¼ 3.6. (This value lies about two pH units above pKa ¼ 1.8, corresponding to pKb ¼ 12.2, and about two pH units below pKa ¼ 5.4.)
H
O
O
N S
O
For acidic substances, the maximum extraction yield is observed two pH units or more below their pKa values. Thus to extract mefenamic acid (pKa 4.2) from an aqueous solution, it should be acidified with sulfuric acid to give a pH value of 13. Ammonia solution will give an aqueous solution pH of only about 10, which is not sufficiently basic for a successful extraction. H
H
N
N
H
Sulfacetamide
Amphoteric substances with one weaker and one stronger acidic and basic function (pKa þ pKb ¼ between 18 and 10) can be extracted as non-ionised molecules at a pH range around their isoelectric point with a lower extraction yield because the molecule is also partly a zwitterion (i.e. the molecule has both its acidic and its basic functions ionised). Example: n
N
Mefenamic acid n
459
Morphine: pKa ¼ 9.9 and pKb ¼ 6.0 (pKa þ pKb ¼ 15.9). The molecule is partly non-ionised at pH ¼ 8.9 (this value lies about one pH unit above pKa ¼ 8.0, corresponding to pKb ¼ 6.0, and about one pH unit below pKa ¼ 9.9); at this pH value there also exist zwitterions that will not be extracted and which therefore reduce the overall extraction yield. Thus morphine is best extracted from aqueous media by using a freshly prepared, saturated sodium hydrogencarbonate solution, or similar compound, to give a pH value between 8 and 9. The high salt concentration of the saturated aqueous solution reduces the drug’s aqueous solubility and therefore improves extraction efficiency. H O
H O H N
H H O
H
H
H Morphine Amfetamine
The distribution behaviour of amphoteric organic electrolytes (e.g. with one acidic and one basic function) also depends on their state of ionisation, but extraction yields are more difficult to predict.
For amphoteric substances with one strong acidic and one strong basic function (pKa þ pKb ¼ 10 or < 10), zwitterions are to be expected, which results in a limited extraction yield.
Clarke's Analysis of Drugs and Poisons Chapter No. 29 Dated: 10/3/2011 At Time: 12:56:50
460
Extraction
Ion-pair extraction Strong electrolytes (e.g. compounds with a quaternary ammonium function, such as paraquat) and very hydrophilic compounds (e.g. sulfonamides) tend to stay in the aqueous phase and will not be sufficiently extracted by organic solvents. Liquid–liquid extraction can be accomplished only by ‘ion-pair’ formation with appropriate counterions. These ion pairs are much more soluble in organic solvents than the drugs themselves and so assist an efficient extraction. Anionic dyes such as bromothymol blue, bromocresol green and bromophenol blue are widely used for this purpose. Moreover, solvents such as chloroform and dichloromethane can extract basic substances as hydrohalogenides, perchlorates, nitrates, phosphates, sulfates or thiocyanates. Conversely, the extraction of acidic drugs can be improved by using quaternary ammonium compounds such as cetyltrimethylammonium bromide as ionpairing compounds. Adsorption Extraction of non-volatile drugs and poisons from biological specimens can also be accomplished by adsorption and the fundamental principles discussed above form the basis for adsorption methods as well. When adsorption is used, intermolecular interactions (van der Waal’s forces, aromatic interaction) or electrostatic interactions will enable the isolation of target compounds from the biological matrix. Hydrophobic interactions between neutral or non-polar substances are based on dispersion forces and are short-ranged, weak interactions between molecules or parts of molecules (bond strength 4–20 kJ/mol). They result from random fluctuations in local electron density distribution within molecules. Hydrophilic interactions between polar functional groups or two dipoles are based either on hydrogen bonding (e.g. hydroxyls, carboxylic acids, amines), or on dipole–dipole interactions. Like dispersion forces, they too are short ranged and relatively weak forces (bond strength 10– 40 kJ/mol). Aromatic interactions are caused by attractive forces between diffuse electron clouds in p systems; their bond strength is similar to dispersion forces. Electrostatic interactions between oppositely charged molecules have the highest bonding energies (200–1000 kJ/mol) and ionic bonding can be used for extraction via ion exchange. Extraction efficiency, recovery and internal standards On the basis of the fundamental principles described above, and on the data that can be found in the individual monographs of this book, extraction methods can be developed and optimised with respect to extraction efficiency. With a high extraction efficiency for the target compound, it should then be possible to create a complete analytical procedure with sufficient overall recovery (a complete procedure includes sample pre-treatment, extraction, fractionation, purification, evaporation, chromatographic separation, detection, identification and quantitative determination). The overall recovery can be calculated as the percentage of the analyte response after sample work-up compared with that of a solution containing the analyte at a concentration corresponding to 100% recovery. A low overall recovery can be tolerated only as long as the data for limit of quantification (LOQ) and limit of detection (LOD), precision and bias are acceptable (this is why recovery is not an essential part of method validation). However, it is good practice to determine recovery at high and low concentrations and ensure that it is greater than 50% to confer robustness to the analytical procedure. For the quantitative determination of the analyte, and in order to monitor the whole procedure when biological specimens are extracted, an internal standard has to be added at the earliest possible stage. The internal standard must mimic the physicochemical properties of the analyte as closely as possible and must follow the target compound through the entire process of extraction and subsequent analytical steps in order to compensate for any loss of the target compound.
The selection of the internal standard can be a difficult task and each individual step of the whole analytical procedure should be carefully considered when making this decision. Even minor differences in the physicochemical properties between the target compound and the internal standard can result in errors, because of their different behaviour during extraction, fractionation, purification and concentration towards the applied chromatographic system, the reagents for derivatisation or their response to the detection system. When mass spectrometry is used, stable-isotope-labelled analogues of the target compounds are the best choice as internal standards. For other analytical techniques or, if stable-isotope-labelled analogues are not available, alkyl analogues (because of their similar structure) can be used as an alternative; but it should be kept in mind that some alkyl analogues – such as morphine and codeine (methylmorphine) – can show significant differences in their physicochemical properties (the additional methyl group in codeine inactivates the phenolic function that is active in morphine). If alkyl analogues are also not available, the internal standard should be chosen on the basis of similarity in structure and functional groups (preferably from the same substance class, e.g. benzodiazepines). When glucuronides are analysed, the internal standard should also be a glucuronide. Only if there are no alternatives should active drugs be used as internal standards; in this case the presence of the active drug used as the internal standard would have to be tested for separately in the specimen. The following examples illustrate some specific considerations that should be kept in mind when choosing the internal standard: when GC is used, attention should be paid to volatility, thermal stability and reactivity with the possible derivatisation reagent; for LC, the solubility in the injection solvent (e.g. mobile phase of the LC) and the response to the detection system (e.g. UV spectrum), as well as the applicability of the ionisation technique for LC-MS should be considered. Sometimes simple problems at the beginning of the procedure can cause errors, such as the solvent used to add the internal standard to the sample (e.g. problems because of protein precipitation, or different extraction of the previously dissolved internal standard and the target compound from the sample matrix). Moreover, the added concentration of the internal standard should be similar to the expected concentration of the analyte. In conclusion, stable-isotope-labelled analogues of the target compounds mimic the analyte very closely and therefore they should be used whenever available. But it should be kept in mind that the high separation power of modern chromatographic systems can separate stableisotope-labelled analogues from the targeted analytes (stable-isotopelabelled analogues elute slightly earlier), leading to the possible influence of, for example, ion suppression on only one of the compounds. Moreover, stable-isotope-labelled analogues and targeted analytes can produce identical ion fragments, which can lead to problems during quantification. To produce accurate quantitative results, the analytical procedure should be validated (see Chapter 20). If a certified reference material to validate the analytical procedure is not available (e.g. for postmortem specimens), then the ‘method of standard addition’ can be used, where calibration and quantification are performed directly in the sample matrix, compensating for matrix effects. In this procedure different concentrations of the target compound are spiked to aliquots of the homogeneous sample prior to work-up and the detector responses are plotted as a graph (‘standard addition plot’). The initial concentration of target compound can then be calculated via extrapolation.
Practical aspects of extraction The isolation of the compounds of interest from the biological matrix is essential for their successful detection, identification and quantification. The strategy applied and the effort invested in the development of an extraction procedure depend not only on the physicochemical properties and the expected concentration of the target compounds but also on the nature of the specimen and the available equipment in the analytical laboratory. Sometimes the physicochemical properties of the target compounds allow for their direct detection after digestion of the sample matrix (e.g. metals), or for an easy separation from the less-volatile
Clarke's Analysis of Drugs and Poisons Chapter No. 29 Dated: 10/3/2011 At Time: 12:56:50
Practical aspects of extraction matrix components (e.g. gases and volatile compounds via headspace analysis). However, there is an important group of less-volatile drugs and poisons that demand more complex extraction procedures (such as liquid–liquid or solid-phase extraction) to ensure their isolation from the biological matrix. A literature review of the Journal of Analytical Toxicology over the past 20 years shows that the number of publications using liquid–liquid extraction has been quite constant and has covered a wide range of analytes, whereas, for solid-phase extraction, the number of publications has continuously increased and, in recent years, the focus has switched from drugs of abuse to applications with a wider range of target analytes. It can be concluded that, although the importance of liquid–liquid extraction in analytical toxicology has already been established, solid-phase extraction is gaining increasing importance in this field. These two important techniques for the extraction of less-volatile drugs and poisons will be discussed in more detail in this chapter. The final decision about which of these two techniques should be chosen for the particular challenge at hand is mainly based on the practical experience of the analyst and a careful consideration of the objective and the intended use of the extraction method, as well as the availability of analytical techniques. Preconditions of extraction Frequent problems in analytical toxicology include the lack of control over the sampling process and, sometimes, a limited availability of specimens. Such problems can result in insufficient or even inadequate samples. This challenge is partially resolved by close cooperation with the person responsible for sampling (e.g. the physician or pathologist) as well as appropriate quality control and educational measures. In most cases, the appropriate preparation of the specimen is a fundamental precondition for successful sample extraction. Protein-free samples (such as urine) or liquid samples with low protein content (such as serum or plasma) that are frequently used in clinical toxicology are comparable to a purely aqueous phase and therefore direct extraction can be tried. If metabolites should also be detected, hydrolytic cleavage of the conjugate bond (deconjugation) with strong acids, bases or enzymes prior to extraction can increase the recovery of these metabolites from biological fluids. This approach is particularly useful for urine and essential for drugs (e.g. laxatives) that are excreted almost exclusively as conjugated metabolites. For the differentiation of the amount of conjugated and unconjugated metabolite present in the sample, either LC-MS is required or two analyses have to be performed. In the first step the unconjugated metabolite is extracted and quantified and then the sample is re-analysed after hydrolysis, resulting in ‘total’ metabolite concentration (conjugated plus unconjugated). To obtain reliable quantitative results, appropriate standards (also conjugated) must be carried throughout the procedure to monitor the efficiency of the hydrolysis step. The use of an enzyme to cleave chemical bonds is the more specific of the different approaches mentioned above. It incurs additional cost and is more time intensive, but cleaner extracts can be achieved, which reduces the ‘down-time’ of analytical instruments. For the different preparations of purified glucuronidase and sulfatase that are available, it is crucial to pay attention to their pH and temperature optima to obtain reproducible results. A typical procedure for the enzymatic hydrolysis of glucuronides is as follows. Mix 1 mL of blood or urine with an internal standard and 1.5 mL of appropriate buffer and then add 100 mL of b-glucuronidase obtained from Helix pomatia. Mix the solution and incubate it at 37 C overnight (approximately 16 h). After incubation, the pH of the solution is adjusted appropriately for solvent or solid-phase extraction of the compounds of interest (see also Chapter 10). Acid or basic hydrolyses (Dubost, Pascal 1955), although faster and less expensive, tend to produce more artefacts due to the vigorous hydrolysis conditions and are therefore more demanding in terms of necessary clean-up procedures. Typically, strong mineral acids or alkalis are used, often with boiling or treatment in a microwave or pressure
461
cooker. To protect the analytical instruments in subsequent analyses, the extracts must be neutralised and organic solvents have to be dried prior to injection, otherwise chromatographic performance deteriorates quickly. Moreover, care should be taken to ensure the stability of the analytes under these harsh conditions of hydrolysis. If several compounds can be hydrolysed to an identical, single compound, the accurate identification of the original substance present can be precluded. For example, both the acid and the enzymatic hydrolysis of benzodiazepines result in the cleavage of conjugates, but acid hydrolysis also converts different drugs to the same benzophenone compound (e.g. diazepam, temazepam, ketazolam, medazepam and camazepam are all converted into 2-methylamino-5-chlorobenzophenone). Although the resulting compound has good chromatographic characteristics, the approach is unsuitable for those applications (such as forensic analysis) that require absolute identification of the drug ingested (see also Chapter 40). Biological materials that are not homogeneous, protein rich or degraded (such as tissue samples and postmortem samples) need homogenisation (e.g. with a blender to disrupt the cellular structure) and sometimes further sample preparation, such as deproteinisation, before extraction from the aqueous phase is possible. Homogenisation can be performed directly in a buffer solution with a physiological pH of 7.4, to avoid protein precipitation. A high dilution ratio of blood (up to 1 : 10) and tissue (up to 1 : 50) results in samples that tend not to clog tightly packed extraction cartridges and are therefore suitable for direct solid-phase extraction. Automation of the extraction process will enable the analyst to handle the large sample volumes and at the same time secures a uniform and efficient extraction by providing a homogeneous flow of the sample through the extraction cartridge. For liquid–liquid extraction of complex sample matrices, protein precipitation is generally needed before extraction. Deproteinisation can be performed with solvents such as ethanol or acetone, or with dimethylformamide, which is particularly well tolerated by most GC stationary phases. Moreover acetonitrile is frequently used for procedures where high performance liquid chromatography (HPLC) systems are applied. It is usual to use two volumes of organic phase to one volume of blood. The following procedure of a combined protein precipitation and subsequent extraction with acetonitrile under alkaline conditions works for target analyses of neutral and basic compounds in specimens of low collagen content (such as blood or brain tissue): to 0.5 mL of blood or 0.5 g of homogeneous brain tissue, add an internal standard and a freshly prepared mixture of 1 mL acetonitrile and 0.1 mL of a saturated aqueous solution of disodium hydrogenorthophosphate. After shaking, centrifugation and evaporation of the supernatant the reconstituted residue can be extracted. For solid-phase extraction, the dilution of the supernatant (to achieve a concentration of acetonitrile below 20%) is in most cases sufficient and the resulting solution can be applied directly to the sorbent. Protein precipitation may also be accomplished with acids (e.g. hydrochloric acid, perchloric acid, trichloroacetic acid and tungstic acid) and salts (e.g. sodium tungstate, ammonium sulfate, cupric sulfate and uranyl nitrate) (Curry 1988). However, all of these procedures are very time-consuming and labour intensive. Protein precipitation also risks loss of the analyte from adsorption and occlusion. When perchloric, trichloroacetic or tungstic acids are applied there is a particularly high chance that the drugs being analysed may also be co-precipitated. For the extraction of trace amounts of drugs and poisons from complex matrices, dilution and homogenisation with water or buffer solutions are therefore preferred and can prevent these problems. To prepare tissue specimens for extraction, enzymatic digestion of these samples is sometimes useful and can be achieved using pepsin, trypsin, enterokinase, lipase and b-glucuronidase. A suitable procedure is as follows: macerate 10 g of liver or other tissue with 40 mL of 1 mol/L tris(hydroxymethyl)methylamine; add 10 mg of subtilisin Carlsberg and incubate in a water bath at 50–60 C for 1 h, with agitation. Filter the digest through a small plug of glass wool to remove undissolved connective tissue. Aliquots of this digest may be substituted for the specified biological fluid in most routine screening procedures
Clarke's Analysis of Drugs and Poisons Chapter No. 29 Dated: 10/3/2011 At Time: 12:56:50
462
Extraction
(Osselton 1978, 1979; Osselton et al. 1978). The filtered digest has a pH of 8.0–9.5 (see also Chapter 10). The drawback of this procedure is that the resulting extracts contain undesirable by-products and artefacts, created during the process of digestion, and therefore a screening procedure will become more difficult. The presence of proteases may interfere with antibodies in immunoassay screening procedures. Even after extensive sample preparation, a large number of matrix components are still present in the sample and these will affect the solubility and the sorption of analytes through their association with molecules as well as by their ability to change the ionic strength of a solution. Soluble matrix components are also distributed between heterogeneous phases and can influence the completeness of equilibrium adjustment as well as the overall extraction efficiency. These considerations have to be kept in mind for the subsequent extraction procedure. Liquid–liquid extraction Direct liquid–liquid extraction (LLE) is still predominant in many laboratories when protein-free samples (such as urine) or liquid samples with low protein content (such as serum or plasma) need to be extracted, because this technique is fast, inexpensive and efficient. LLE is based on well-defined thermodynamic relationships and has a wide dynamic range. The extraction yield is strictly determined by: n n n n
The distribution equilibrium (solubility) The equilibrium of electrolytic dissociation (pH dependent) The ratio of phase volumes (organic/aqueous) The number of extraction stages.
In practical applications, the constitution of the sample matrix has an important influence on results and appropriate sample pre-treatment (e.g. protein precipitation) is a fundamental prerequisite for successful LLE. An excess of solvent and/or repetitive extraction will simply increase the amount of co-extracted impurities. Therefore, a phase ratio (organic/aqueous) of 1 to 2 is recommended. For compounds with low extractability, it is better to switch to another solvent instead of increasing the volume of the organic phase and/or to engage in multiple extractions. In order to separate target compounds from interferences as well as from each other, their varied distribution constants and different acidic or basic properties can be used for fractionation or back extraction (see below). Practical experience shows that, in order to achieve sufficient separation, the pKa values of the compounds to be separated must differ by more than four units. Fractionation in more than two or three groups is therefore ineffective, because under such circumstances substances are dragged into several fractions. It should be kept in mind that neutral compounds are to be expected in the first fraction and that co-extraction of endogenous compounds is to be expected in each extract. Therefore, in every extraction scheme, sacrifices must be made when it comes to the question of the cleanliness of extracts. For the adjustment of an appropriate pH in the aqueous solution, acids or bases can be used, but in this case the stability of the analyte and the possibility of protein precipitation should be kept in mind. Thus the use of an appropriate buffer solution for the reliable adjustment of the pH is recommended. The choice of solvent is mainly based on the solubility of the drugs and poisons that have to be extracted and follows the rule of thumb that ‘like dissolves like’. Moreover, the solubility of known interferences from the sample matrix (such as lipids: fatty acids, cholesterol, etc.) that have a negative effect on the identification and/or the quantification of the analyte should also be considered in the choice of the solvent. A major criterion for the solubility of a particular substance is the solvent’s polarity, which results from an unequal sharing of electrons within the molecule and depends on the electronegativity of its atoms and the asymmetry of the molecule. Polarity aside, solvents can also be chosen by their ability or inability to form hydrogen bonds (hydrogen donor or acceptor), their boiling point (solvent removal with the risk of evaporation losses), their pH stability (e.g. ester cleavage), as well as their water miscibility. A low solubility of water in the solvent facilitates
drying of the extract and co-extraction of water-soluble substances (e.g. salts) can be minimised. In the case of larger amounts of water dissolved in the solvent, it is possible to put the extract in a freezer to freeze the water in order to remove it. Small amounts of water can be removed by addition of anhydrous sodium sulfate, but the possible loss of trace amounts of the analyte should be kept in mind. When choosing a solvent, the potential toxicity should also be considered (e.g. chloroform) as well as flammability and explosive potential (e.g. peroxides in diethyl ether). For practical reasons the density (top or bottom layer with water), and the solvent’s UV absorbency (when spectrophotometric detection is used), as well as the grade of purity are important considerations. Before a solvent is used in a new extraction procedure, an aliquot of the solvent should be evaporated and the residue should be analysed to detect any impurities. Moreover, solvents possess different emulsifying potentials (e.g. that of chloroform is greater than that of diethyl ether), and the correct choice of solvent for LLE also includes considerations to avoid emulsion formation. The addition of neutral salts and the use of slow rotation or vortex speeds as well as using larger phase volume ratios can prevent the formation of emulsions. If emulsions are unavoidable, the phases may be separated by centrifugation, by putting the extract in a freezer and by the addition of a small amount of methanol. If emulsions are routinely obtained, the extraction method should be changed. A promising approach to avoiding emulsions, which also works well for solvents with a high density (such as dichloromethane) that are difficult to isolate from the bottom layer with water, is immobilisation of the aqueous phase (e.g. on diatomaceous earth) prior to extraction. This technique of a supported LLE can increase the extraction yield for the analyte. Some solvents can possess potential reactivity with certain analytes (e.g. some pesticides react with ethanol or acetone) and also ion-pair extraction can occur in certain solvents. This can be exploited to extract analytes (e.g. paraquat), but sometimes ion-pair extraction is an unwanted sideeffect (e.g. in the case of extraction of hydrochlorides with chloroform or dichloromethane). Finally, the analytical method applied may also influence the choice of the solvent. For example, in GC, chlorinated solvents would not be chosen if a halogen specific detector were going to be used. However, if the extract is to be evaporated to dryness and then reconstituted in another solvent before being injected into the chromatographic system (e.g. the eluent of an HPLC system), the choice of the original solvent can be wider. The boiling point and expansion volume of the solvent may also be issues (see also Table 40.6 in Chapter 40). Adding a high concentration of neutral salts (e.g. sodium chloride) to the aqueous phase can support the extraction process (the ‘salting-out effect’), depressing the mutual solubility of phases and simultaneously reducing emulsification and foaming. The evaporation of solvents (for enrichment) demands particular care in order to avoid the loss of volatile analytes (e.g. amfetamines: b.p. 200 C). To prevent the loss of volatile analytes, a small volume of acidified methanol can be added to the extract prior to evaporation. The quality and the volume of the glassware should be chosen with care, to avoid loss of the analyte by adsorption and to enable the reconstitution of the extract in a small amount of solvent (to optimise the concentration step) for further analyses. The appropriate solvent should extract as much of the target compound as possible while at the same time co-extracting only a minimum of interferences. To identify which solvents are frequently used in analytical toxicology, a literature review of the Journal of Analytical Toxicology over 5 years (2004 to 2008) was performed (Table 29.1). The most frequently used solvents were ethyl acetate, followed by 1chlorobutane, hexane and dichloromethane. Isopropyl alcohol was mainly used as a modifier. Chloroform and diethyl ether were frequently used in the past as versatile solvents and, although the severe health risks of chloroform and the fire and explosion hazards of diethyl ether are well known today, they are still in use and have not been totally substituted by dichloromethane and methyl t-butyl ether, respectively. Finally, acetonitrile was also used on a regular basis. Other solvents (such as toluene, methanol, pentane, butyl acetate, acetone and ethanol) were used only occasionally.
Clarke's Analysis of Drugs and Poisons Chapter No. 29 Dated: 10/3/2011 At Time: 12:56:50
Practical aspects of extraction
Table 29.1 Number of times a solvent was used in a paper published in the Journal of Analytical Toxicology between 2004 and 2008 inclusive Solvent
Number of citations
Ethyl acetate
38
1-Chlorobutane
28
Hexane
27
Dichloromethane
25
Isopropyl alcohol
22
Chloroform
18
Diethyl ether
15
Acetonitrile
11
Methyl t-butyl ether
7
Toluene
6
Isoamyl alcohol
6
Methanol
5
Pentane
3
n-Heptane
3
Octane
2
Dichlorothane
2
n-Butyl acetate
2
Diisopropylether
1
Isobutyl alcohol
1
Propyl acetate
1
Acetone
1
Cyclohexane
1
Tricholoroethanol
1
Ethanol
1
TOTAL
227
When a new extraction procedure is developed, and there are no data about an appropriate solvent available, the following solvents should be tested after adjusting the aqueous solution to an appropriate pH value (in order of increasing polarity): hexane, 1-chlorobutane, methyl t-butyl ether, dichloromethane, 1-butanol, ethyl acetate and acetonitrile. The basic properties of these solvents can be found in Table 29.2. If the results are not sufficiently good using a pure solvent, then modifiers can be added, such as in chloroform–2-propanol (9 : 1; e.g. for morphine), or mixtures can be used. Although so-called all-purpose extraction solvents such as ethanol–hexane–acetone (1 : 2 : 2) or dichloromethane–2-propanol–ethyl acetate (1 : 1 : 3) are used, it should be kept in mind that, in general, the more substances with different physicochemical properties are extracted, the larger the amount of interferences from the matrix that will be co-extracted. These mixtures are therefore
primarily used in the screening for a ‘general unknown’. For target analysis, a toxicologist would choose a more refined procedure with a more selective solvent. If the target compound is known, data about appropriate solvent and pH value for extraction can be found in the monographs in this publication or through a literature review. A frequently used solvent that co-extracts a relatively low amount of interferences from biological samples when compared with other solvents is 1-chlorobutane. For this solvent, extraction yields of 331 toxicologically relevant compounds are available from aqueous media at pH 9.0 (Table 29.3 and in the relevant monographs). At this pH, 228 of the 331 compounds were extracted with an extraction yield equal to or higher than 80% (represented in Table 29.3 by a yield in the organic phase with a value equal to or higher than 0.8). For the remaining compounds, sufficient extraction yields could be reached by changing the pH to acidic (e.g. for phenobarbital) and/or through the addition of a modifier (e.g. for morphine). If a target compound has to be extracted, the database gives a quick overview of the extractability of the compound with 1-chlorobutane at a defined pH value. If the extraction yield from aqueous solution is good, it can be expected that the extraction is also possible from serum. Example protocols for the extraction of various analytes can be found in various chapters of this book (e.g. Hospital Toxicology (Chapter 1), Postmortem Toxicology (Chapter 10) and High Performance Liquid Chromatography (Chapter 41)). The extraction procedure will become more complex with an increase in the complexity of the sample matrix because additional steps for purification (back extraction) have to be added. As an example of a complete extraction scheme for the extraction of bases, neutrals and acids, and to illustrate the theoretical background discussed above, the following is reproduced from Chapter 10. The pH of the specimen influences the extent to which acid and basic drugs are extracted. Addition of a weakly basic buffer, such as sodium borate (pH 9), favours the extraction of weakly basic drugs, as well as most neutral substances. Similarly, the addition of an acidic buffer, such as sodium dihydrogenphosphate, favours the extraction of acidic as well as neutral drugs. The majority of drugs of forensic interest are ‘basic’ in character, but are often present at relatively low concentrations in blood. It is, therefore, desirable to have an extraction scheme that incorporates a back-extraction step to eliminate or minimise the extraction of endogenous molecules. An example extraction scheme is shown in Fig. 29.1. The saturated sodium borate solution will force the basic drugs into the lipid-soluble un-ionised form, allowing extraction into the chlorobutane. The chlorobutane is transferred to a fresh tube and the drugs are back extracted into sulfuric acid. Neutral or acidic substances will remain in the upper chlorobutane layer, which may be pipetted or aspirated to waste. The remaining acid layer is then made basic by addition of sodium hydroxide, and the now un-ionised basic drugs are re-extracted with chlorobutane. The upper solvent layer may then be removed and concentrated under nitrogen, prior to analysis by a suitable chromatographic method. This extraction scheme will give extracts that are relatively free of interfering substances. However, it should be noted that morphine and other amphoteric drugs cannot be detected by this method since the phenolic functional group will
Table 29.2 Properties of solvents Solubility (g/100 mL water at 20 C)
Polarity index (water = 10.2)
Hydrogen acceptor
Hydrogen donor
69
0.01
0.1
No
No
78
0.07
1.0
No
No
0.74
55
0.5
2.5
Yes
No
Dichloromethane
1.34
40
2
3.1
No
No
1-Butanol
0.81
118
7.9
3.9
Yes
Yes
Ethyl acetate
0.90
77
8.6
4.4
No
Yes
Acetonitrile
0.78
82
Miscible
5.8
No
Yes
Solvent
Density (g/mL)
Hexane
0.66
1-Chlorobutane
0.89
Methyl t-butyl ether
Boiling point ( C)
463
Clarke's Analysis of Drugs and Poisons Chapter No. 29 Dated: 10/3/2011 At Time: 12:56:59
464
Extraction
Table 29.3 Extraction yields of 331 compounds of clinical or forensic interest using 1-chlorobutane from water at pH 9 (multiple observations in different laboratories were averaged)
Table 29.3 continued Compound
Extraction yield
Extraction yield
Amfetamine
0.5
0
Apomorphine
0.8
0.8
Aprindine
0.95
0.7
Articaine
1
0.9
Atenolol
0
0.8
Atropine
0.6
0.5
Azathioprine
0
2-Methylamino-1-(3,4-methylenedioxyphenyl)butane (MBDB) 1
Azinphosethyl
1
2,3-Methylenedioxyamfetamine
0.6
Azinphosmethyl
1
2,3-Methylenedioxymethamfetamine
0.9
Benperidol
1
0.6
Benserazide
0
0.3
Benzatropine
1
0.2
Benzoylecgonine
0
0.5
Betaxolol
1
0.8
Biperiden
1
0.8
Bisacodyl
0.7
2,5-Dimethoxy-4-methyl-phenethylamine (2C-D)
0.5
Bisoprolol
0.9
2,5-Dimethoxy-phenethylamine (2C-A)
0.3
Bromazepam
0.9
3-(2,3-Methylenedioxyphenyl)pentane-2-amine
0.95
Bromocriptine
1
0.6
Bromophosethyl
1
0.7
Bromophosmethyl
1
0.9
Bromperidol
1
0.5
Brotizolam
1
0.3
Budipine
1
1
Bupivacaine
1
4-Methoxyamfetamine (PMA)
0.5
Bupranolol
1
4-Methyl-2-pyrrolidinopropiophenone (MPPP)
1
Buprenorphine
1
4-Methylthioamfetamine (4-MTA)
0.6
Buspirone
0.95
1
Caffeine
0.3
1
Carazolol
0.9
1
Carbamazepine
0.95
1
Carbamazepine epoxide
0.6
0.7
Carbidopa
0
N,N-Diethyltryptamine (DET)
0.95
Carteolol
0.06
Acebutolol
0.05
Carvedilol
1
Acetaminophen (paracetamol)
0
Cathine
0.07
0
Celiprolol
0.10
0.5
Chlordiazepoxide
0.95
1
Chlormezanone
0.9
1
Chloroquine
0.95
0.95
Chlorovinphos
1
1
Chlorpromazine
1
Amantadine
0.5
Chlorprothixene
1
Amfebutamone
1
Citalopram
1
Amfepramone
1
Clobazam
1
1
Clobutinol
1
0.95
Clomethiazole
1
0.6
Clomipramine
1
1
Clonazepam
1
0.10
Clopamide
0.06
1
Clopenthixol
1
Compound 10-Hydroxycarbazepine (metabolite of oxcarbazepine) 2-(2,3-Methylenedioxyphenyl)butane-1-amine 2-(2,3-Methylenedioxyphenyl)propane-1-amine 2-(3,4-Methylenedioxyphenyl)2-methylpropane-1-amine 2-(3,4-Methylenedioxyphenyl)butane-1-amine 2-(3,4-Methylenedioxyphenyl)propane-1-amine
2,3-Methylenedioxo-N-methylphenethylamine 2,3-Methylenedioxo-N-phenethylamine 2,4,5-Trimethoxyamfetamine (TMA-2) 2,4,6-Trimethoxyamfetamine (TMA-6) 2,5-Dimethoxy-4-brom-phenethylamine (2C-B) 2,5-Dimethoxy-4-metamfetamine (DOM)
3,4-Methylenedioxyamfetamine (MDA) 3,4-Methylenedioxymetamfetamine (MDMA) 3,4-Methylenedioxy-N-ethylamfetamine (MDE) 3,4-Methylenedioxy-N-methylphenethylamine 3,4-Methylenedioxy-N-phenethylamine 4-Methoxy-2-pyrrolidinopropiophenone
N-(1-Phenylcyclohexyl)-3-ethoxypropylamine (PCEPA) N-(1-Phenylcyclohexyl)-3-methoxypropylamine (PCMPA) N-(1-Phenylcyclohexyl)-1-propylamine N-(1-Phenylcyclohexyl)-2-methoxyethylamine (PCMEA) N-Methyl-4-methoxyamfetamine (PMMA)
Adenosine Ajmaline Alfentanil Alimemazine Alprazolam Alprenolol
Amfetaminil Amiodarone Amisulpride Amitriptyline Amitriptyline oxide Amlodipine
Clarke's Analysis of Drugs and Poisons Chapter No. 29 Dated: 10/3/2011 At Time: 12:57:3
Practical aspects of extraction
465
Table 29.3 continued
Table 29.3 continued Compound
Extraction yield
Compound
Extraction yield
Clotiazepam
1
Gabapentin
0
Clozapine
1
Gallopamil
1
Cocaine
1
Gamma-hydroxybutyric acid
0
Codeine
0.8
Glibenclamide
0.2
Colchicine
0.13
Glutethimide
1
Cotinine
0.10
Haloperidol
1
Cyamemazine
1
Heptenophos
0.8
Deanol
0.4
Hydrochlorothiazide
0
Demelverine
1
Hydromorphone
0.10
Desipramine
1
Hydroxyzine
1
Detajmium
0.9
Ibuprofen
0
Dialifos
1
Imipramine
1
Diazepam
0.95
Indometacin
0
Dibenzepin
1
Ipratropium
0
Dichlorvos
1
Isofenphos
0.95
Diclophenac
0.2
Kavaine
0.7
Dihydrocodeine
0.7
Ketamine
1
Dihydroergocryptine
1
Lamotrigine
0.17
Diltiazem
1
Levetiracetam
0
Dimethoate
0.7
Levodopa
0
Dimetindene
0.98
Levomepromazine
1
Diphenhydramine
1
Lidocaine
1
Disopyramide
0.8
Lisinopril
0
Dosulepin (dothiepin)
1
Lofepramine
0.9
Doxazosin
0.95
Oprazolam
1
Doxepin
1
Lorazepam
0.85
Doxylamine
1
Lormetazepam
1
Droperidol
0.9
Loxapine
1
Enalapril
0
Lysergide (LSD)
0.95
Entacapone
0
Maprotiline
1
Ephedrine
0.2
Medazepam
1
Esmolol
0.8
Mefenorex
0.95
Ethosuximide
0.2
Melperone
1
Etomidate
1
Mepindolol
0.4
Felodipine
0.95
Mepivacaine
1
Fenethylline
0.9
Meprobamate
0.10
Fenofos
1
Meptazinol
0.9
Fentanyl
1
Mesuximide
1
Fenthion
1
Metamizole
0.4
Flecainide
0.95
Methadone
0.95
Fluconazole
0.10
Methamfetamine
0.7
Flumazenil
0.8
Methaqualone
1
Flunitrazepam
1
Methohexital
0.95
0.8
Methylphenidate
0.8
Flupenthixol
1
Metixene
1
Fluphenazine
1
Metoclopramide
0.9
Flupirtine
1
Metoprolol
0.8
Flurazepam
0.95
Mevinphos
0.9
Fluspirilene
0.9
Mexiletine
0.9
Fluvoxamine
0.8
Mianserin
1
Furosemide
0
Midazolam
0.9
Fluoxetine
table continued
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 29 Dated: 10/3/2011 At Time: 12:57:4
466
Extraction
Table 29.3 continued
Table 29.3 continued Compound
Extraction yield
Compound
Extraction yield
Mirtazapine
0.9
Piritramide
0.9
Moclobemide
0.9
Piroxicam
0
Modafinil
0.4
Prajmalium
0.9
Morphine
0
Pramipexole
0
Nadolol
0
Prazepam
1
Nalbuphine
0.8
Prilocaine
0.95
Nalorphine
0.3
Primidone
0
Naloxone
0.9
Procainamide
0.10
Nefazodone
1
Procaine
0.9
Nefopam
0.95
Procyclidine
1
Nicotine
0.9
Promazine
1
Nifedipine
1
Promethazine
1
Nicotinamide
0
Propafenone
0.95
Nimodipine
1
Propofol
0.95
Nisoldipine
1
Propoxyphene
1
Nitrazepam
1
Propranolol
1
Nordiazepam
0.95
Propyphenazone
1
Nortriptyline
1
Prothipendyl
0.95
Noscapine
1
Pseudoephedrine
0.2
Olanzapine
1
Quetiapine
1
Opipramol
1
Quinidine
0.95
Orciprenaline
0
Quinine
1
Oxazepam
0.85
Ranitidine
0
Oxcarbazepine
0.9
Reboxetine
1
Oxitriptan
0
Remifentanil
1
Oxprenolol
0.9
Risperidone
1
Oxycodone
0.95
Ropivacaine
1
Paraoxon
0.9
Salicylate
0
Parathion ethyl
1
Scopolamine (hyoscine)
0.7
Parathion methyl
1
Sertindole
1
Paroxetine
1
Sertraline
1
Pemoline
0
Sildenafil
1
Penbutolol
1
Sotalol
0
Pentazocine
0.8
Strychnine
0.9
Pentobarbital
0.2
Sulfentanil
1
Pentoxyverine
1
Sulfotep
1
Perazine
1
Sulpiride
0
Perphenazine
1
Sultiame
0
Pethidine
1
Talinolol
0.2
Phenazone
0.4
Temazepam
1
Phencyclidine
0.9
Terbufos
1
Phenobarbital
0.10
Tertatolol
1
Phenolphthalein
0.7
Tetrazepam
0.95
Phenprocoumon
0.2
Theobromine
0
Phenytoin
0.5
Theophylline
0
Pholedrine
0
Thiopental
0.9
Phosphamidon
0.8
Thioridazine
1
Phoxime
1
Tiagabine
0.5
Pimozide
1
Tiapride
0.4
Pindolol
0.4
Ticlopidine
1
Pipamperone
1
Tilidine
1
Pirimiphos
1
Timolol
0.6
Clarke's Analysis of Drugs and Poisons Chapter No. 29 Dated: 10/3/2011 At Time: 12:57:4
Practical aspects of extraction
467
Table 29.3 continued Compound
Extraction yield
Tocainide
0.3
Tolperisone
1
Topiramate
0.2
Tramadol
1
Tranylcypromine
1
Trazodone
1
Triazolam
1
Trichlorophos
1
Trifluperidol
1
Triflupromazine
1
Trihexyphenidyl
1
Trimipramine
1
Tryptophan
0
Valproic acid
0.07
Venlafaxine
0.95
Verapamil
1
Vigabatrin
0
Viloxazine
0.85
Zaleplon
1
Ziprasidone
1
Zolpidem
1
Zopiclone
0.9
Zotepine
0.95
Zuclopenthixol
1
Extraction yield = extraction yield in the organic phase (e.g. 0.3 representing an extraction yield in 1-chlorobutane of 80%). The database was compiled by the Committee on Extraction of the Society of Toxicological and Forensic Chemistry under the supervision of Dr U. Demme, and is reproduced here with the kind permission of the Society of Toxicological and Forensic Chemistry.
ionise at high pH, therefore precluding extraction into the solvent. For amphoteric drugs, the basic phase should be less than pH 9.0, and preferably pH 8.0–8.5. Although a similar extraction scheme to that used for basic drugs (but with the additions of acid and base reversed) could be used for strongly acidic drugs, such a method does not efficiently extract weakly acidic drugs, such as the barbiturates, and neutral drugs, such as meprobamate. Conversely, simple addition of an acidic buffer to whole blood and extraction with a solvent results in the co-extraction of large amounts of endogenous lipid substances. Such extracts may be ‘cleaned up’ by partitioning between immiscible solvents of different polarities, such as acetonitrile and hexane, as shown in Fig. 29.2. The more polar drugs tend to partition into the acetonitrile, whereas the endogenous lipids (fatty acids, sterols) tend to partition into the hexane. Solid-phase extraction Although method development for solid-phase extraction (SPE) is not as straightforward as for LLE, this technique offers appealing advantages. Because of SPE’s high extraction efficiency, even very small sample sizes are sufficient, thereby reducing solvent consumption. Additionally, there is no emulsion formation. Past problems with inconsistent quality of the extraction cartridges have been overcome by manufacturers’ implementation of extensive quality control measures. The increasing interest in SPE by toxicological laboratories lies mainly in its compatibility with automation. The need for automated extraction procedures is directly related to the
Figure 29.1 Extraction scheme for strong bases.
expectation that toxicological laboratories become economically selfsufficient as well as an increased demand for quality assurance and reproducibility and, therefore, comparable results between different laboratories. In SPE the analytes are isolated from the aqueous sample by adsorption onto a solid sorbent, followed by washing and elution steps. In each of these steps a mechanism of total retention or total release of the target compounds is desired and, as with LLE, this technique is based on the fundamental principles of extraction discussed above. Depending upon the choice of sorbent, the extraction of toxicologically relevant compounds is achieved via hydrophobic, hydrophilic, aromatic or electrostatic interactions; often a combination of several mechanisms is involved. In the field of analytical toxicology non-modified silica, surface-modified silica and polymer resins are used, and a more detailed description of these sorbents can be found in Chapter 41 (packing materials). To date, both selective procedures (for target analysis) and non-selective SPE procedures (for the screening for a ‘general unknown’) have been developed. Because a wide variety of different analytes can be extracted using a combination of hydrophobic and electrostatic interactions, these socalled mixed-mode sorbents (or functionalised sorbents, or hybrid extraction sorbents) are widely used in analytical toxicology. A practical
Clarke's Analysis of Drugs and Poisons Chapter No. 29 Dated: 10/3/2011 At Time: 12:57:4
468
Extraction
Dilution (phosphate buffer pH 7.4), homogenisation and centrifugation of the sample
Pre-treatment of the sorbent with ethyl acetate–2-propanol (3 : 1) followed by phosphate buffer pH 7.4
Application of the sample onto the ‘mixed-mode’ sorbent (max. 1 mL/min)
Washing with water and pH change by applying an acidic solution
Elution of acidic and neutral analytes with ethyl acetate–2-propanol (3 : 1) (max. 1 mL/min)
Figure 29.2 Extraction scheme for acids and neutrals.
example of such a procedure for the extraction and fractionation of neutral and basic metabolites of cyclizine in the urine of racehounds can be found in Chapter 7. To illustrate the fundamental principles and possible pitfalls of SPE, the following section includes a step-by-step discussion of the development of a non-selective ‘general unknown’ screening procedure for a wide variety of analytes. An overview of the procedure can be found in Fig. 29.3. The first step is the appropriate pre-treatment of viscous specimens so that the samples do not cause flow problems when passing through the tightly packed cartridges and choosing the sorbent. Tissue homogenates, especially, tend to clog the polyethylene frit (pore diameter 20 mm) holding the sorbent in place. Practical experience has shown that diluting the specimens with ten times the amount of phosphate buffer before homogenisation provides samples that can be extracted with either negative or positive pressure. Some tissues (e.g. putrefied liver) have to be diluted even more (up to 50 times). A physiological pH of 7.4 prevents protein precipitation in biological specimens and therefore avoids an unpredictable loss of analytes. After homogenisation (e.g. with a blender), and before the sample is applied to the extraction cartridge, remaining particles and cellular structures must be removed by centrifugation (cooling can prevent the loss of volatile analytes). For the handling of the sometimes large sample volumes, automation of the whole extraction process is highly recommended. The controlled and therefore homogeneous flow of the sample through the extraction cartridge in such an extraction device secures a uniform and efficient extraction, leading to more reproducible results. For this ‘general unknown’ screening procedure, a mixed-mode sorbent based on a combination of a polar modified polystyrene resin and a cationic exchanger is used. In this way analytes can be extracted by non-selective hydrophobic and aromatic interactions and, in a second step, acidic and neutral compounds can be separated from basic compounds that are retained by electrostatic interactions through their amine functionality. Polystyrene resins do not possess residual silanol groups like silica-based sorbents do (due to varying degree of
Elution of basic analytes with ethyl acetate–2-propanol–triethylamine (75 : 25 : 3) (max. 1 mL/min) Figure 29.3 Overview of a 'mixed-mode' solid-phase extraction procedure.
endcapping) Polystyrene resins have the additional advantage of being stable across the entire pH range, in contrast to the limited stability (pH 2–8) of silica-based sorbents. Moreover, polystyrene resins offer a higher capacity than silica-based sorbents and sufficient retention of a wide variety of different analytes can be achieved at a pH of 7.4. At this pH value an equilibrium of electrolytic dissociation will be reached for acidic and basic analytes in the aqueous phase. By adsorption of the non-ionised analyte to the sorbent, equilibrium must be continuously re-established and a nearly complete extraction will be reached if sufficient contact time with the sorbent is allowed and if the capacity of the sorbent is high enough. Additionally, the porous structure of polystyrene resins can be used to exclude large molecules (e.g. proteins and lipids bound in micelles) from the extraction process. The amount of sorbent that has to be used is determined by the amount of interferences that are expected in the sample matrix analysed. The second step is the pre-treatment of the sorbent. It should be washed with the strongest solvent applied in the procedure and then it must be conditioned – in the procedure described here, with a phosphate buffer of pH 7.4 – in order to make the sorbent compatible with the sample. Silica-based bonded sorbents should not dry out between the applications of different solutions. This is not an issue when working with polar modified polystyrene resins because of their additional hydrophilic properties. The third step is the application of the sample. Competitive influences of matrix components displacing the analytes from the limited surface area of the sorbent have to be considered and ‘overloading’ the extraction cartridge must be avoided. Because the mass transfer of analyte to sorbent is determined by kinetic functions and, because the intermolecular interactions (van der Waals’ forces, aromatic interactions, etc.) are short-ranged and relatively weak forces, sufficient contact
Clarke's Analysis of Drugs and Poisons Chapter No. 29 Dated: 10/3/2011 At Time: 12:57:5
References
469
Table 29.4 Possible pitfalls in the process of developing a mixed-mode solid-phase extraction procedure Steps of the mixed-mode procedure
Possible pitfalls
Specimen pre-treatment
Protein precipitation (loss of analytes), flow problems
Choice of sorbent
Capacity, undesired secondary interactions, stability
Washing and conditioning sorbent
Improper conditioning (drying of silica-based sorbents, capacity problems)
Sample application
Insufficient contact time, 'break-through', clogging
Washing and changing pH to acidic
Protein precipitation on the sorbent, insufficient acidic capacity
Elution of acidic and neutral compounds
Insufficient contact time, inappropriate solvent (solvent strength, water miscibility)
Elution of basic compounds
Insufficient contact time, inappropriate solvent (solvent strength, water miscibility), insufficient basic capacity, sorbent stability (silica-based sorbents)
time is a key factor for ensuring reproducible results. To avoid ‘breakthrough’ due to ineffective retention of analytes, the adsorption flow rate should be kept below 1 mL/min. In the fourth step, protein and other interferences are washed away with water (to avoid protein precipitation on the sorbent when subsequently applying organic solvents) and the pH is changed by applying an acidic solution so that analytes with amino functions are protonated and establish their electrostatic interactions with the charged sorbent. In the fifth step, because ionic interactions are relatively strong forces, acidic and neutral analytes as well as hydrophobic and polar interferences can be eluted with strong organic solvents (with different polarity and/or solvent strength; the first solvent should be miscible with water to be able to reach the ‘inner surface’ of the sorbent, which is loaded with water comparably to a sponge; flow rates should be kept below 1 mL/min). In the procedure described here, ethyl acetate–2propanol (3 : 1) is used. In the sixth and final step, all bonding mechanisms for the remaining basic analytes (ionic and hydrophobic interactions) have to be disrupted simultaneously and this can be done using an appropriate basic organic solvent (in the procedure described, ethyl acetate–2-propanol–triethylamine (75 : 25 : 3) is used). Elution will be incomplete if the basic solution is not strong enough to completely disrupt the electrostatic interaction. Again, flow rates should be kept below 1 mL/min, because ion-exchange binding kinetics are even slower than with hydrophobic and aromatic interactions. Volatile organic bases (such as triethylamine) have the advantage that they can be removed easily from the extract by evaporation. After evaporation of the eluent from step 5, the extract can then be analysed for neutral and acidic compounds (in the case of a very complex matrix and/or a very low concentration of the analyte, a further back-extraction step might be necessary). Simultaneously, after evaporation, the extract from step 6 can be analysed for basic compounds. The aqueous phase of the sample should not be discarded in this ‘general unknown’ screening procedure, because strong electrolytes (e.g. quaternary ammonium compounds) will not be sufficiently extracted onto the sorbent. Extraction of these compounds can be accomplished by ionpair formation (see Ion-pair extraction above). The possible pitfalls in using a mixed-mode SPE procedure are given in Table 29.4. Microextraction Miniaturisation of the extraction process simplifies the entire analytical procedure by having the extraction carried out in one vessel, eliminating the evaporation step and possible loss of volatile materials, reducing solvent use, and considerably saving in extraction time. It usually involves vigorous mixing of a small volume of solvent with a large volume of biological material (e.g. urine), centrifuging to separate the solvent layer and direct injection into a chromatograph. For example, amfetamines and related compounds may be extracted from 5 mL of urine into 100 mL of chloroform and this is injected into a GC apparatus (Ramsey, Campbell 1971). Some microextraction methods such as hollow fibre-based liquidphase extraction (Esrafili et al. 2007) and fibre-in-tube solid-phase
microextraction (Yazdi et al. 2008) have been successfully used for the analysis of antidepressant drugs in biological fluids. Microextraction by packed sorbent (MEPS) with on-line connection to GC or LC has been applied for the analysis of local anaesthetics in plasma samples (Abdel-Rehim 2004) and the analysis of amfetamines in hair (Miyaguchi et al. 2009).
Conclusions The ongoing development of more powerful and sensitive analytical instruments can only be fully utilised after the successful isolation of the target compounds from the biological specimens. Without sufficient sample extraction, either these sensitive instruments cannot be used at all or the results are unreliable owing to interferences. Although many optimised procedures for the extraction of specific target compounds (in most cases from urine or blood) can be found in the literature, nonselective, reliable and robust procedures for the simultaneous extraction of a wide range of analytes with different physicochemical properties (e.g. for the ‘general unknown’ screening) are rare, especially when more complex matrices have to be extracted. Unfortunately, the greatly desired ‘universal standard extraction procedure’ for all possible constellations of target compounds and specimens does not (yet) exist. The selection or development of a proper extraction procedure and the procedure’s adaptation to specific cases – based on analytical data of targeted compounds, available specimens, advantages and disadvantages of the various extraction techniques, and the time and resources at hand – are key tasks of the analytical toxicologist. Special knowledge and experience are needed to meet these challenges. Analytical results usually have serious medical or legal consequences: toxicologists therefore carry a high level of responsibility. To ensure the integrity of analytical results, stringent quality control measures have been implemented in modern toxicological laboratories. But such measures should not lead to overly rigid structures that limit the toxicologist to a few strictly defined extraction methods and target analytes, as this would narrow a laboratory’s flexibility to an unacceptable level. The analytical toxicologist must stay open to new extraction technologies, improvements in sample preparation and developing trends. Among these improvements is automation, which enables the unattended, reproducible extraction of samples; advances in this application of technology are expected soon.
References Abdel-Rehim M (2004). New trend in sample preparation: on-line microextraction in packed syringe for liquid and gas chromatography applications. I. Determination of local anaesthetics in human plasma samples using gas chromatography-mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 801: 317–321. Curry AS (1988). Poison Detection in Human Organs, 4th edn. Springfield, IL: Charles Thomas. Dubost P, Pascal S (1955). Determination of chlorpromazine in biological fluids; additional note. Ann Pharm Fr 13: 56–57. Esrafili A et al. (2007). Hollow fiber-based liquid phase microextraction combined with high-performance liquid chromatography for extraction and determination of some antidepressant drugs in biological fluids. Anal Chim Acta 604: 127–133.
Clarke's Analysis of Drugs and Poisons Chapter No. 29 Dated: 10/3/2011 At Time: 12:57:5
470
Extraction
Miyaguchi H et al. (2009). Rapid identification and quantification of methamphetamine and amphetamine in hair by gas chromatography/mass spectrometry coupled with micropulverized extraction, aqueous acetylation and microextraction by packed sorbent. J Chromatogr A 1216: 4063–4070. Osselton MD (1978). The release of basic drugs by the enzymic digestion of tissues in cases of poisoning. J Forensic Sci Soc 17: 189–194. Osselton MD (1979). The use of proteolytic enzymes to release high levels of drugs from biological materials submitted for toxicological analysis. Vet Hum Toxicol 21(Suppl): 177–179. Osselton MD et al. (1978). Enzymic digestion of liver tissue to release barbiturates, salicylic acid and other acidic compounds in cases of human poisoning. Analyst 103: 1160–1164. Ramsey J, Campbell DB (1971). An ultra rapid method for the extraction of drugs from biological fluids. J Chromatogr 63: 303–308. Yazdi AS et al. (2008). Separation and determination of amitriptyline and nortriptyline by dispersive liquid–liquid microextraction combined with gas chromatography flame ionization detection. Talanta 75: 1293–1299.
Further reading Franke JP, De Zeeuw RA (1998). Solid-phase extraction procedures in systematic toxicological analysis. J Chromatogr B Biomed Sci Appl 713: 51–59. Leo A et al. (1971). Partition coefficients and their uses. Chem Rev 71: 525–616. Lide RL (1996). Properties of Organic Solvents. Boca Raton, FL: CRC Press. M€ uller RK (1991). Extraction from aqueous phase. In: M€ uller RK, ed. Toxicological Analysis. Berlin: Verlag Gesundheit, 66–83. Pawliszyn J (2003). Sample preparation: quo vadis? Anal Chem 75: 2543–2558. Siek TJ (1978). Effective use of organic solvents to remove drugs from biologic specimens. Clin Toxicol 13: 205–230. Stimpfl T, Vycudilik W (2004). Automatic screening in postmortem toxicology. Forensic Sci Int 142: 115–125. Telepchak M et al. (2004). Forensic and clinical applications of solid phase extraction. In: Karch SB, ed. Forensic Science and Medicine. Totowa, NJ: Humana Press. Wille SM, Lambert WE (2007). Recent developments in extraction procedures relevant to analytical toxicology. Anal Bioanal Chem 388: 1381–1391.
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:40
CHAPTER
30
Colour Tests B Widdop
Introduction The bulk of the material in this chapter derives from that contributed by the late HM Stevens to the second edition of this publication, which was modified and expanded by Wayne Jeffrey in the third edition. The main addition in this latest version is an appendix dealing with tests for metals and anions that are not covered by the general scheme. Colour tests (sometimes referred to as chemical spot tests) provide toxicologists and drug analysts with one of the first tools for the presumptive identification of drugs and poisons. These colour tests are most usefully applied to pharmaceuticals and scene residues, and to a lesser extent to biological fluids such as stomach contents, urine. They are used to place the unknown into a specific class of compounds or to eliminate categories or classes of compounds. These colour tests remain popular for many reasons. They are simple to perform, use minimal reagents, are inexpensive and give results that can be viewed by the naked eye. They appeal particularly in parts of the developing world where laboratory facilities tend to be very limited. In many instances they can also be used as thin-layer chromatography (TLC) location reagents applied by spraying or dipping (see Chapters 1, 11, and 39). This chapter describes the colour tests cited in the monographs. For some substances, the colour reaction with a particular chemical reagent may be quite specific, but it is much more common for the colour to be produced by a class of compounds. Moreover, compounds that do not fall into the class may also give colours. For some of the tests, the colour reactions can be correlated with certain aspects of the chemical structure of a compound or group of compounds. However, anomalous responses often occur that cannot be explained on that basis. Some of these are noted in the colour tests described below, but it should be borne in mind that many others may be found. It follows that colour tests are only an indication of the presence of a compound or class of compounds and that all tests must be confirmed by more specific methods. This is especially important in forensic cases! The colour tests included here range from those that rely on reactions with certain functional groups (e.g. Folin–Ciocaltaeu for phenols), those that are almost specific for a given group (e.g. FPN reagent for phenothiazines) through to those that give diagnostic colours with a wide range of compounds (e.g. Mandelin’s test and the Marquis test).
Interpretation of colour tests Colour descriptions Colours exhibited by these tests cannot be described with any accuracy. They may vary in intensity or tincture with the concentration of compounds in the test samples and the presence of extraneous material. In addition, their assessment is always a subjective one, even in people with normal colour vision. Some of the complexes formed are unstable such that the colour changes or fades with time. Effects of ionic form Salts may give colours different from those of the corresponding acid or base. In general, free acids or bases that have been isolated from the test material by an extraction process give better colours than their salts. The colour of a salt may be modified by the nature of the other ions present. For example, all hydrochloride salts give a red colour in Mandelin’s test
and a blue colour with Koppanyi–Zwikker reagent (prior to adding pyrrolidine). Basic salts of weak acids may produce different colours because of a change in pH. Where a compound has been extracted from biological material, these factors should not create any difficulty, since it will be present in the form of the base. However, when applying the tests to pharmaceutical preparations, where the compounds are usually present as salts, this can cause problems. To overcome this, the material can be extracted in much the same way as for biological samples to derive the free base. Bromide and iodide salts can be converted into the nitrate before testing, which gives the same colour as the base, by the following method: n
n
To 0.5 mL of a 1% (w/v) solution of the salt in dilute acetic acid, add one drop of an 8% (w/v) solution of silver nitrate followed by one drop of a 2% (w/v) solution of sodium chloride to remove excess silver. Centrifuge to separate the precipitated silver halide and use the supernatant liquid, either as a solution or evaporated to dryness where necessary, for the colour tests.
The colours that are recorded in the tables and monographs are usually those obtained by testing either the free acid or the free base. Use of the colour tests lists The system adopted uses ten basic colours: the spectral colours (red, orange, yellow, green, blue and violet), together with pink, brown, grey and black. Where there is a variation in hue, this is indicated by combining two colours (e.g. red–brown). The second-named colour is considered to be the dominant one and is the main colour used in the lists. For example, red–brown is listed under brown, whereas brown–red is listed under red. When interpreting results, it is often necessary to search the lists given under two main colours (e.g. for red–brown, the lists under both red and brown should be consulted). This takes account of the subjective nature of colour assessment. An arrow between two colours (e.g. red!brown) indicates that the colour changes during the course of the test. In the monographs, the notation brown/red is used where there are two parts to a test that produce two colours. Occasionally, the colour displayed by a test solution in reflected light may be different from that in transmitted light, in which case the solution is described as dichroic. A combined colour may be obtained when more than one drug is present or the drug itself is coloured, which limits the value of the tests for biological samples.
Practical points Performing the colour tests The tests are carried out either in clear glass test-tubes or on white glazed porcelain tiles (spotting tiles), which give a uniform background against which the colours can be assessed. For drugs, the tests are designed to work on about 1 mg, either as the solid form or as a dried extract of this amount (see below), unless stated otherwise. Solutions should be made in water unless otherwise stated. Where an instruction, time, temperature, etc., appears in brackets after the drug name, such as (add water), (15 s) or (slowly at 100 C), this indicates a change in the test procedure for that particular drug. 471
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:41
472
Colour Tests
The following recommendations are most important: n
n
A sample known not to contain the compound of interest should be tested at the same time as the test sample. This enables a comparison of the colours produced by the sample and by the reagent blank. Ideally, the blank sample should have the same matrix as the test sample (e.g. for urine tests use analyte-free urine), since this takes account of the effects of extraneous materials. Otherwise, water is usually adequate. Before making a final decision on the result of a test, the reaction of the unknown should be compared with that of a reference substance tested under exactly the same conditions.
Table 30.2 is formulated to give a quick lead to those tests that can be applied to detect some of the most important drug groups and other poisons.
Table 30.2 Indication of which tests can be applied to detect some of the most important drug groups and other poisons Substance/functional group
Useful tests
Alcohols
Potassium dichromate
Alkaloids and nitrogenous bases
Dragendorff's reagent
Amides (aliphatic)
Nessler's reagent
It is essential to validate all tests and test reagents for sensitivity and specificity; O’Neal et al. (2000) have outlined a suitable method for a chemical spot test.
Aldehydes (aliphatic)
Schiff's reagent
Amfetamines
See Appendix 30.1
Application of colour tests to sample extracts
Antidepressants
Marquis test
Several solvent extraction schemes have been devised to fractionate compounds on the basis of their acidic, neutral or basic characteristics (see Chapters 1 and 10). The tests listed in Table 30.1 can be applied to the evaporated extracts.
Barbiturates
Dille–Koppanyi reagent
Validation of a colour test
Table 30.1 Tests that can be applied to the evaporated extracts Fraction
Test
Strong acid
Aromaticity Ferric chloride
Sodium nitroprusside–acetone
Koppanyi–Zwikker reagent Mercurous nitrate Vanillin reagent Zwikker reagent Benzodiazepines
Formaldehyde–sulfuric acid
Cannabis
Duquenois reagent
Carbamates (non-aromatic)
Furfuraldehyde
Cocaine
Cobalt thiocyanate
Folin–Ciocaltaeu reagent
p-Dimethylaminobenzaldehyde
Liebermann's reagent
Mandelin's test
Millon's reagent Nessler's reagent Weak acid
Scott's test Chlorinated phenols
Nitric acid (fuming) Nitric–sulfuric acid
Coniferyl alcohol
Chlorinated hydrocarbon insecticides
Diazotisation
Cyanide
Ferrous sulfate (B)
Aromaticity
Ferric chloride
Neutral
Sodium picrate
Folin–Ciocaltaeu reagent
Cyanide groups
Sodium picrate
Koppanyi–Zwikker reagent
Dithiocarbamates
Sodium nitroprusside
Liebermann's reagent
Ergot alkaloids
p-Dimethylaminobenzaldehyde
Mercurous nitrate
Halogenated hydrocarbons
Fujiwara test
Millon's reagent
Imides
Koppanyi–Zwikker test
Nessler's reagent
Ketones
Sodium nitroprusside
Aromaticity
Methadone
Cobalt thiocyanate
Furfuraldehyde
Mandelin's test
Koppanyi–Zwikker reagent
Marquis test
Liebermann's reagent
Tetrabromophenolphthalein ethyl ester
Mercurous nitrate Basic
Nessler's reagent
Mono-substituted pyridine ring
Cyanogen bromide
Amalic acid test
Nitrates and nitrites
Ferrous sulfate
p-Dimethylaminobenzaldehyde
Opiates
See Appendix 30.2
Ferric chloride
Oxidising agents
Diphenylamine
Formaldehyde–sulfuric acid
Paraquat/diquat
Sodium dithionate
Forrest reagent
Phencyclidine
Cobalt thiocyanate
FPN reagent
p-Dimethylaminobenzaldehyde
Liebermann's reagent
Tetrabromophenolphthalein ethyl ester
Mandelin's reagent Marquis reagent Nessler's reagent Sulfuric acid
Phenols
p-Dimethylaminobenzaldehyde Ferric chloride Folin–Ciocaltaeu reagent Millon's reagent
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:42
Colour test methods
Table 30.3 Colours with amalic acid
Table 30.2 continued Substance/functional group
Useful tests
Colour
Compound
Phenothiazines
Ferric chloride
Red (!violet)
Bufylline, caffeine
Formaldehyde–sulfuric acid
Pink (!violet)
Pentoxifylline
Forrest reagent
Orange (!violet)
Acepifylline piperazine, bamifylline, xanthinol nicotinate
FPN reagent Phenylpyrazolines Primary aromatic amines Primary and secondary amines
473
Nitrous acid
Pink–orange (!violet)
Fenetylline, pentifylline
Coniferyl alcohol
Yellow (!pink)
Etamiphylline
Diazotisation
Yellow (!violet)
Diprophylline, proxyphylline, theobromine, theophylline
Dragendorff's reagent Simon's test
Propoxyphene
Cobalt thiocyanate
Indications
Froehde's reagent
A red, pink, orange or yellow residue, which changes to pink, red or violet after the addition of ammonium hydroxide, indicates the presence of a xanthine (Table 30.3).
Liebermann's test Tetrabromophenolphthalein ethyl ester Quaternary ammonium compounds
Tetrabromophenolphthalein ethyl ester
Ammoniacal silver nitrate
Quaternary amines
Dragendorff's reagent
Quinines
Cobalt thiocyanate
To 20 mL of 0.1 mol/L silver nitrate add sufficient strong ammonia solution to dissolve the initial precipitate.
Reagent
Thalleioquin
Method
Quinones
Methanolic potassium hydroxide
Reducing agents
Benedict's reagent Ferric chloride
Dissolve the sample in a minimum amount of water, with the addition of ethanol if necessary, add an equal volume of the reagent and note any colour that develops. Heat the mixture in a water-bath at 100 C for 30 s.
Trinder's reagent
Indications
Salicylates Steroids
Antimony pentachloride Naphthol sulfuric acid Sulfuric acid
Sulfonamides
Copper sulfate Koppanyi–Zwikker reagent Mercurous nitrate Nitrous acid
Sulfur containing
Palladium chloride Sodium nitroprusside
Tertiary amines
Red, yellow, brown or black colours (especially at room temperature) indicate potent reducing power, which occurs when adjacent carbon atoms in a ring each bear a hydroxyl group (Table 30.4). There is no response when the hydroxyl groups are meta to each other, but there is some restoration of reducing power when they are para to each other. Some colour production is also obtained with ethynyl bonds, but not with ethylenic bonds. Ethchlorvynol and ethinylestradiol both give a white precipitate that turns yellow on heating. Carbidopa gives a silver mirror on heating.
Dragendorff's reagent Tetrabromophenolphthalein ethyl ester
Table 30.4 Colours with ammoniacal silver nitrate Colour at room temperature
Compound
Colour at 100 C
Red
Isoetharine
Brown–orange
Hexoprenaline (!brown!black)
Additional information
Isoprenaline (!red–brown)
Brown
Colour reactions given by narcotics and amfetamines with four of the so-called ‘alkaloid colour reagents’ (Marquis, Mecke’s, Froedhe’s and Mandelin’s) are listed in Appendix 30.1 and Appendix 30.2. Three colour tests have been developed for gamma-hydroxybutyric acid (GHB) and its precursor gamma-butyrolactone (GBL; see Appendix 30.3).
Rimiterol
Brown
Yellow Grey–yellow
Ethinamate
Brown
Levodopa (!brown)
Black
Hydroquinone
Brown
Brown Red–brown
Colour test methods Orange–brown
Caution: the following lists of colour tests and drugs tested are not exhaustive; the omission of a compound from a list does not indicate that no response is given, but that it may not have been tested.
Grey
Amalic acid test (test for xanthines)
Black
Red–grey
Adrenaline Methyldopa
Black
Dopamine
Black
Methyldopate
Orange–brown
Protokylol
Brown
Ascorbic acid
Method
Benserazide
–
Add to the sample a few drops of 10 mol/L hydrochloric acid followed by a few crystals of potassium chlorate, and evaporate the mixture to dryness. Observe the colour of the residue then add 2 or 3 drops of 2 mol/L ammonium hydroxide and again observe the colour.
Dobutamine
–
Dodecyl gallate
–
Noradrenaline
–
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:42
474
Colour Tests
Antimony pentachloride
Table 30.6 Colours with aromaticity test, method 2, obtained on addition of alkali to acid solution
Reagent
Colour of acid solution
Colour after addition of alkali
Compound
Colourless
Red
Clobazam (heat for 3 min with acid)
Orange
Butanilicaine, tolazoline
Method
Yellow
Place a drop of an ethanolic solution of the sample on a filter paper, add a drop of the reagent and dry in a current of warm air. Alternatively, the test may be carried out by adding a drop of the reagent to the sample on a white tile.
Amprolium, atropine methobromide, hyoscine butylbromide, hyoscine methobromide, ketoprofen, pipazethate, tetrahydrozoline, tetramisole, trimetaphan
Violet
Atropine methonitrate (transient), hyoscine methonitrate (transient)
Brown
Isopropamide
Red
Aminacrine, benzonatate, tetracaine (amethocaine), trimethoprim
Dry some antimony trichloride over phosphorus pentoxide, melt the dried material (m.p. 73 C), and pass dry chlorine gas into the melt until a yellow fuming liquid is obtained. Add this liquid to about 10 times its volume of chloroform, filter the solution into a dark glass-stoppered bottle and store in a desiccator.
Indications
Various colours are obtained with the cardiac glycosides, their aglycones and certain oestrogens and corticosteroids (Table 30.5). No colour is obtained with beclometasone, cortisone, fluocinolone, fludroxycortide, prednisolone, prednisone, progesterone, testosterone or triamcinolone.
Yellow
Orange–red
Amicarbalide, carbocromen, dyclonine, glibenclamide, levallorphan, metocurine, padimate, propanidid, quinuronium, salinazid
Brown
Dibromopropamidine, dichlorophen tubocurarine
Brown–violet
Dequalinium
Aromaticity Method 1
Place a portion of the sample in each of two ignition tubes, and to one tube add some solid sodium hydroxide. Heat both tubes carefully, allow the water vapour to escape, insert into the vapours in each tube an open capillary tube that contains Marquis reagent, and observe the colour of the reagent.
Indications
Indications
Red or orange colours indicate that the sample is aromatic in nature. The colours probably result from the liberation of traces of aromatic hydrocarbons, phenols, etc. Colours obtained after heating with sodium hydroxide generally indicate the presence of aromatic acids. Colours obtained after heating without sodium hydroxide generally indicate the presence of phenols, phenolic acids and aldehydes that contain more than one hydroxyl group. A negative result does not necessarily imply that the substance is nonaromatic. Method 2
Add 2 or 3 drops of concentrated nitric acid to the sample, heat in a water-bath at 100 C for 1 min, cool the mixture, dilute 3–4 times with water and make the solution alkaline by the addition of a 40% (w/v) solution of sodium hydroxide.
Table 30.5 Colours with antimony pentachloride Colour
Compound
Red
Dienestrol, diethylstilbestrol
Orange
Cholesterol (!brown), desoxycortone, dydrogesterone, fludrocortisone, hydrocortisone, hydroxyprogesterone, strophanthin-K (!red)
Yellow
Red
Dextromethorphan, haloperidol
Orange
Alfadolone
(!brown)
Androsterone, digitoxigenin, digoxigenin
(!brown!black–violet)
Digitoxin, digoxin, lanatoside C, ouabain (very weak)
Green–yellow
Fluocortolone
Green
Betamethasone (!brown), dexamethasone, mestranol, pancuronium
Brown
Carbenoxolone, dimethisterone, estradiol, estriol, estrone, ethinylestradiol (!black), fluoxymesterone, norethandrolone, norethisterone, oxymetholone, rotenone
Orange–brown
Enoxolone (!violet)
Green–brown
Noretynodrel
A change from colourless or yellow in acid solution to darker colours (e.g. orange or red–orange) after the addition of sodium hydroxide indicates the presence of a benzene ring in the molecule, probably though the production of a nitrophenol or other nitro compound. Certain compounds (e.g. diazepam, methaqualone) give a negative result. Orange colours are given by certain non-aromatic corticosteroids (e.g. cortisone), by substances that contain sulfur and by compounds that already contain an aromatic nitro group (e.g. nifursol). Colour changes are given in Table 30.6. Certain substances give distinct colours with cold nitric acid, but the colours fade on heating; these are listed in Table 30.7. Benedict's reagent Reagent
Dissolve 1.73 g of copper sulfate in 10 mL of water. Dissolve 17.3 g of trisodium citrate and 10 g of anhydrous sodium carbonate in 80 mL of water with the aid of heat; pour this solution into the copper sulfate solution and dilute the mixture to 100 mL. Method
Add 0.5 mL of the reagent to the sample and heat in a water-bath at 100 C for 3 min. Indications
The formation of red cuprous oxide occurs with strong reducing agents, such as ascorbic acid, dithionites, certain phenolic compounds that contain two hydroxyl groups para to each other, and compounds that contain at least four hydroxyl groups on a non-aromatic ring (e.g. glucose, tetracyclines). Table 30.7 Colours with aromaticity test, method 2, obtained on addition of cold nitric acid to sample, which fade on heating Colour
Compound
Red
Aminacrine (15 s), clozapine, dropropizine, medazepam, trimethoprim
Brown
Metocurine
Pink–brown
Diethylthiambutene (changing to green)
Black
Tubocurarine
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:43
Colour test methods A weak response (orange–brown or brown colours) is given by streptomycin, hydroxylamine and substituted hydrazines (e.g. phenelzine). No colour is obtained with beclometasone, cardiac glycosides and estriol (two hydroxyl groups) or clindamycin (three hydroxyl groups). Carbon disulfide Method
Mix the sample with 1 mL of water and 0.1 mL of a 1% (w/v) solution of sodium tetraborate, add 0.2 mL of a 10% v/v solution of carbon disulfide in ethanol and heat in a water-bath at 100 C for 3 min; cool the solution and add 3 drops of 0.1 mol/L silver nitrate. Indications
A brown colour indicates the presence of a dithiocarbamate, which suggests that the original substance was an aliphatic or heterocyclic primary or secondary amine. The original sample should be tested to ensure that it does not give a brown colour with silver nitrate alone. Chromotropic acid Reagent
1. Dissolve 20 mg of chromotropic acid in 10 mL of concentrated sulfuric acid. 2. Dissolve 1 g of sodium nitrite in 10 mL of concentrated sulfuric acid.
Cobalt thiocyanate (see Scott's test) Reagent
1. A 2% (w/v) solution of cobalt thiocyanate in water 2. Phosphoric acid 3. 1 g H2PtCl66H2O in 20 mL of H3PO4. Mix 9 parts of solution 1 and 3 parts of solution 2, add 1 part of solution 3 and mix well. Add 9 parts distilled water and mix. When the solution turns pink it is ready for use. Method
Add a few drops of the reagent to the sample to be tested. Indications
See Table 30.10. Limit of detection (LOD): cocaine-HCl 60 mg, methadone-HCl 15 mg. Table 30.10 Colours with cobalt thiocyanate Colour
Compound
Blue (flaky precipitate)
Cocaine
Brilliant greenish blue
Benzfetamine-HCl, brompheniramine maleate, chlordiazepoxide-HCl, chlorpromazine-HCl, doxepin-HCl, hydrocodone tartrate, methadone-HCl, methylphenidate-HCl
Strong greenish blue
Diacetylmorphine-HCl, ephedrine-HCl, phencyclidine-HCl, procaine-HCl, propoxyphene-HCl, pseudoephedrine-HCl
Strong blue
Quinine-HCl
Method 1
Add a small amount of sample, either solid or in solution, to 1 mL chromotropic acid reagent. Note any colour that may be produced, and then add the solution dropwise to 0.5 mL of water, with cooling. Substances that give a colour with cold sulfuric acid must be excluded. Indications
475
Coniferyl alcohol (primary aromatic amines) Reagent
See Table 30.8.
Warm 0.1 g of coniferyl alcohol until it melts (m.p. 74 C), dissolve in 3 mL of ethanol and dilute to 10 mL with ethanol.
Table 30.8 Colours with chromotropic acid
Method
Colour
Compound
Place 1 drop of a solution of the sample on a filter paper, add 1 drop of the reagent and expose the paper to hydrochloric acid fumes.
Red (before dilution)
Formaldehyde, paraformaldehyde (reacts slowly)
Violet (after dilution)
Hydrochlorothiazide, hydroflumethiazide
Method 2 (for chlorophenoxy herbicides)
Add 1 mL of 1 mol/L hydrochloric acid to 10 mL of sample and extract with 20 mL of toluene for 5 min. Centrifuge for 5 min and remove the toluene layer. Repeat with a further 20 mL of toluene and evaporate the combined extracts to dryness. Dissolve the residue in 0.2 mL of concentrated sulfuric acid and divide between two wells of a spotting tile. Add 0.1 mL of sodium nitrite solution to one well and 0.1 mL of chromotropic acid reagent to the other. Heat the tile at 80 C and observe any colour development.
Indications
An orange colour indicates the presence of an aromatic primary amine in which the amino group is attached directly to a benzene ring. An anomalous reaction is obtained with diphenylamine (bright orange). Copper sulfate Method 1
Dissolve the sample in a minimum volume of 0.1 mol/L sodium hydroxide and add a 1% (w/v) solution of copper sulfate, drop by drop, until the colour change is complete. Indications
Green, blue or brown colours indicate the presence of a sulfonamide (Table 30.11).
Indications
See Table 30.9.
Table 30.11 Colours with copper sulfate, method 1 Colour
Compound
Green
Phthalylsulfathiazole, succinylsulfathiazole (!violet), sulfachlorpyridazine, sulfadimethoxine, sulfadimidine (!brown), sulfadoxine, sulfaethidole, sulfamerazine (!brown), sulfamethizole, sulfamethoxazole, sulfametopyrazine, sulfapyridine (!brown–green), sulfaquinoxaline, sulfasomidine
Blue
Phthalylsulfacetamide, sulfacetamide, sulfaguanidine, sulfanilamide, sulfaphenazole, sulfaurea, sulfinpyrazone, sulthiame
Table 30.9 Colours with sodium nitrite and chromotropic acid Compound
Sodium nitrite
Chromotropic acid
2,4-Dichlorophenoxyacetic acid
Brown
Purple
2,4-Dichlorophenoxypropionic acid
Dark brown
Light purple
4-Chloro-2-methylphenoxyacetic acid
Light brown
Light purple
2-(4-Chloromethylphenoxy)propionic acid
Light brown
Purple
2,4,5-Trichlorophenoxyacetic acid
No reaction
Purple
2-(2,4,5-Trichlorophenoxy)propionic acid
No reaction
Light pink/ purple
Brown Orange–brown
Sulfafurazole Sulfasalazine
Green–brown
Sulfamethoxypyridazine, sulfamoxole
Violet–brown
Sulfadiazine, sulfamethoxydiazine, sulfathiazole
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:43
476
Colour Tests
Method 2
Method
Add 1 or 2 drops of a 1% (w/v) solution of copper sulfate to the sample on a white tile.
Add 2 drops of solution 1 to the drug, followed by 1 drop of solution 2.
Indications
A light purple (blue–violet) colour indicates the presence of a barbiturate. Other reacting compounds are hydantoins, sulfonamides, pyrimidine, piperidine, methyprylon. The LOD is 25 mg or lower.
A blue colour indicates the presence of an alkali salt of a fatty acid, such as sodium cromoglicate (1–2 min) or valproate. The colours are not produced by a change of pH (some of the alkali salts will change the pH), as negative results are obtained with sodium bicarbonate. Cyanogen bromide
Indications
p-Dimethylaminobenzaldehyde (Wasicky reagent or Van Urk reagent; a general test for ergot alkaloids) Reagent
Reagent
1. Decolorise bromine solution by the addition of solid potassium cyanide and then add more bromine solution until the solution is pale yellow. 2. Prepare a saturated solution of aniline in water. Solutions 1 and 2 are stable for 1 week. Mix equal volumes of the two solutions immediately prior to the test. Method
Add 1 drop of the mixed reagent to the sample on a white tile. Indications
Red, orange or yellow colours indicate the presence of a mono-substituted pyridine ring. Increasing chain length of the substituent group weakens the response; a delayed response is obtained when the pyridine ring is substituted by nitrogen adjacent to the ring nitrogen; a weak response is obtained where there is a C¼O substituent adjacent to the ring nitrogen. There is no response to the test if the pyridine ring is bound to another ring, if it is substituted in more than one position or if the nitrogen in the ring is substituted. Anomalous results are obtained with azatadine (pink), bisacodyl (no response) and tropicamide (violet– pink) (Table 30.12).
Dissolve 2.0 g of p-dimethylaminobenzaldehyde (p-DMAB) in 50 mL of 95% ethanol and 50 mL of concentrated hydrochloric acid. The reagent should be freshly prepared. Method
Add the reagent to the sample in a test-tube, warming if necessary. Observe any colour produced, then carefully dilute with water or spray dried spots on filter paper and heat. Indications
Colours are given by a number of substances, which include ergot alkaloids, dimethyltryptamine, psilocin, psilocybine (gives a violet colour), cannabinols and certain indoles in which the indole ring is not bonded to another conjugated ring (red changing to violet on dilution), and certain phenols and phenolic amines (red or orange, usually changing to violet on dilution). Some other types of compound also respond. See Table 30.13. The LOD for lysergide (LSD) is 6 mg. Diphenylamine test Reagent
Diazotisation
Mix 0.5 g of diphenylamine in 20 mL of water and dilute to 100 mL with concentrated sulfuric acid.
Method
Method
Dissolve the sample in 2 mol/L hydrochloric acid, and to 1 drop on a white tile add 1 drop of a 1% solution of sodium nitrite, and 1 drop of a 4% solution of naphth-2-ol in 2 mol/L sodium hydroxide.
Indications
Indications
A bright red or orange–red colour indicates the presence of a primary aromatic amine. Diphenylamine does not give a reaction; aminonitrothiazole (solid) gives a violet colour. Dille–Koppanyi reagent modified (a general test for barbiturate-like compounds) Reagent
1. Dissolve 0.1 g of cobalt(II) acetate dihydrate in 100 mL of methanol. Add 0.2 mL of glacial acetic acid and mix. 2. Add 5 mL of isopropylamine to 95 mL of methanol.
Apply the reagent to the sample on a white tile or in a test-tube. A blue colour indicates the presence of an oxidising agent such as bromate, chlorate, chromate, dichromate, iodate, lead(IV), manganese (III, IV, VII), nitrate, nitrite, permanganate or vanadate. This test has been modified for use on blood samples to detect ethchlorvynol (Caughlin 1991). Blood (0.5 mL) is mixed with 1.0 mL of acetone and vortex mixed. The sample is centrifuged and 50 mL of the supernatant is added to 50 mL of diphenylamine reagent and 25 mL of chloroform. The mixture is vortex mixed and allowed to stand. A pink colour that develops in the chloroform layer indicates ethchlorvynol. Dragendorff reagent (a general reagent for nitrogenous bases) Reagent
Table 30.12 Colours with cyanogen bromide
(a)
Dissolve 1 g of bismuth subnitrate in 3 mL of 10 mol/L hydrochloric acid with the aid of heat. Dilute to 20 mL with water and dissolve 1 g of
Colour
Compound
Red
Zimeldine (30 s)
Table 30.13 Colours with p-dimethylaminobenzaldehyde
Pink
Azatadine
Colour
Compound
Orange–pink
Carbinoxamine, dimetindene, doxylamine, iproniazid, phenyramidol, triprolidine (1–2 min)
Red (changing to violet on dilution)
Cannabinols, phenazone (100 C, 5 min), pindolol, psilocin, psilocybine, tryptamine
Violet–pink
Tropicamide
Red (no violet on dilution)
Benserazide, cocaine (100 C, 3 min), feprazone, harmine, phencyclidine (100 C, 3 min)
Orange (changing to violet on dilution)
Dobutamine, dopamine, diamorphine, morphine, orciprenaline, phenol, terbutaline, tyramine
Violet
Ergot alkaloids (dihydroergotamine, ergometrine, ergotamine, ergotoxine, lysergide, methysergide), dimethyltryptamine, psilocin, psilocybine
Yellow
Primary aromatic amines, e.g. aminosalicylic acid, anileridine, aniline, procaine, benzocaine
Orange
Red–orange Yellow (a)
Azaperone, brompheniramine, chlorphenamine, isoniazid, metyrapone, nicametate, nicotinamide, nicotine, nicotinic acid, nifenazone, nikethamide, pheniramine, xanthinol nicotinate Benzyl nicotinate Halopyramine, mepyramine, tripelenamine
Anomalous results are obtained with azatadine (pink), bisacodyl (no response) and tropicamide (violet–pink).
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:44
Colour test methods potassium iodide in the mixture. If black bismuth triiodide separates, add 2 mol/L hydrochloric acid and more potassium iodide to dissolve it.
Table 30.15 Colours with ferric chloride Colour
Compound
Method
Red
Acetates, phenazone, propionates
Dissolve the sample in 3 drops of 2 mol/L hydrochloric acid, add 2–3 mL of the reagent and dilute to 10 mL with water.
Brown–red Orange
Indications
An orange, red–orange or brown–orange precipitate suggests the presence of an alkaloidal base (precipitated as the alkaloidal bismuth iodide). Primary, secondary, tertiary and quaternary amines give positive results. This reagent is commonly used as a spray or locating agent to detect alkaloids on TLC plates.
Green–yellow Green
Blue–green
1. Add 2.5 mL of acetaldehyde and 2.0 g of vanillin to 100 mL of 95% ethanol 2. Concentrated hydrochloric acid 3. Chloroform. Method
Place the solid sample, or an evaporated petroleum ether (or other organic solvent) extract of the sample, in a test-tube and add 3 drops of solution 1. Shake for 1 min and add 3 drops of solution 2. Agitate gently and observe the colour produced. Add 9 drops of solution 3, vortex mix gently and note whether the colour is extracted from the mixture. Indications
Ferric chloride (general reagent for phenols, e.g. salicylates) Reagent
Dissolve 5 g of anhydrous ferric chloride, or 8.25 g of ferric chloride hexahydrate, in 100 mL of distilled water.
Chlorpromazine, hexoprenaline, propyphenazone, valproate Paracetamol Adrenaline, betanaphthol, dobutamine, dopamine, etamivan, ethylnoradrenaline, hexylresorcinol, hydroquinone, hydroxyquinoline, isoetarine, isoprenaline, levodopa, methyldopa, methyldopate, noradrenaline, paraphenylenediamine, phenothiazine, protokylol, rimiterol, thioridazine Chlorquinaldol
Blue
Apomorphine, dodecyl gallate, gallic acid, morphine, parachlorophenol, pethidine, phenol, tannic acid
Violet
Aminosalicylic acid, diflunisal, dipyrone, hexachlorophene (transient), labetalol, salicylaldehyde, salicylamide, salicylic acid, salicyluric acid
Blue–violet Brown
Aminophenazone, salicylamide (after hydrolysis), salicylic acid Aloin, carbidopa
Yellow–brown
Salinazid
Green–brown
Benserazide
Black Violet–black
A colour change from grey to green through blue to violet–blue suggests the presence of cannabis, but differentiation from roasted coffee and patchouli oil is required. The colour change is best seen with fresh drug material. The violet colour is extracted into the chloroform layer only when cannabis is present (Table 30.14). The LOD is 350 mg of tetrahydrocannabinol (THC). No colour is obtained with other natural products, such as basil, bay leaf, eucalyptus oil, mace, marjoram, rosemary, sage, thyme or tobacco.
Nifenazone
Yellow
Duquenois reagent, modified Reagent
477
Ethyl gallate (!blue–black)
Ferrous sulfate A (test for nitrates and nitrites) Reagent
To 1 volume of a 10% (w/v) solution of ferrous sulfate (FeSO4,7H2O) add 5 volumes of concentrated sulfuric acid with cooling. Method
Add the sample to 0.5 mL of the reagent. Indications
A red or pink colour is given only by nitrates and nitrites (e.g. glyceryl trinitrate). Ferrous sulfate B (test for cyanide)
Method
Add ferric chloride solution to the sample or an ethanolic solution of the sample. Indications
Red, orange, green, blue, violet or brown colours indicate the presence of a phenolic compound, fatty acid or a phenylpyrazoline. High quantities of phenothiazines can also cause this test to be positive. Salicylates give a violet colour. Many phenols give no colour with ferric chloride when water is used as a solvent, but give positive tests when anhydrous solvents such as chloroform are used. Aspirin (acetylsalicylic acid) does not give a positive result unless first hydrolysed with concentrated sodium hydroxide to give salicylate. Colours are listed in Table 30.15.
Reagent
Dissolve 10 g of ferrous sulfate in 100 mL of freshly boiled and cooled water (prepare fresh). Method
Dilute 1 mL of sample with 2 mL of 10% (w/v) sodium hydroxide solution and add 2 mL of ferrous sulfate solution. Add sufficient 10% (v/v) hydrochloric acid to dissolve the ferrous hydroxide precipitate. Indications
A blue colour is given by cyanide. There are no common sources of interference. Folin–Ciocaltaeu reagent (test for phenolic compounds)
Table 30.14 Colours with modified Duquenois reagent
Reagent
Compound
Initial colour
Colour extracted by chloroform
Cannabis
Violet–blue
Violet
For the stock solution, dissolve 100 g of sodium tungstate and 25 g of sodium molybdate in 800 mL of water in a 1500 mL flask, add 50 mL of phosphoric acid and 100 mL of concentrated hydrochloric acid, and reflux for 10 h. Cool, add 150 g of lithium sulfate, 50 mL of water and 4–6 drops of bromine, and allow to stand for 2 h. Boil for 15 min to remove the excess bromine, cool, filter and dilute to 1000 mL with water. This stock solution should be stored at a temperature not exceeding 4 C and used within 4 months of its preparation; it has a yellow colour and must not be used if any trace of green colour is present.
Coffee (roasted)
Violet–brown
Nil
Nutmeg
Pale reddish purple
Nil
Patchouli oil
Violet
Nil
Tea (leaves)
Green–blue
Nil
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:44
478
Colour Tests
For use, dilute 1 volume of this stock solution with 2 volumes of water.
Table 30.17 Colours with Forrest reagent Colour
Compound
Method
Red
Acepromazine, carfenazine, chlorpromazine, diethazine, dimetotiazine, mequitazine, mescaline, mesoridazine, piperacetazine, prochlorperazine, promethazine, propiomazine, thiazinamium, thiopropazate, thioproperazine, thioridazine
Add the diluted reagent to the sample and make the mixture alkaline with 2 mol/L sodium hydroxide. Indications
A blue colour indicates the presence of a phenolic compound. The reaction is progressively inhibited with increased halogenation of the phenol nucleus. Formaldehyde–sulfuric acid Reagent
To 4 volumes of concentrated sulfuric acid add 6 volumes of formaldehyde solution (using a pipette with the tip below the surface of the acid) with stirring and adequate cooling. When the reagent is warm it remains clear for about 1 h. If turbidity develops, this may be dispelled by heating in a water-bath at 100 C for about 1 min (note that this reagent is not the same as that used in the Marquis test). Method
Mix the sample with the reagent and heat at 100 C for 1 min. Indications
Benzodiazepines generally give an orange colour with the exception of bromazepam and clozapine (a benzodiazepine-like compound), which both give yellow, and flurazepam (pink). Other indications include phenothiazines, tetracyclines and thioxanthenes. Tryptamine (brown) and zomepirac (red) also react. Those marked with an asterisk in Table 30.16 fluoresce orange under ultraviolet (UV) light (l ¼ 350 nm). No response is obtained with chlordiazepoxide, dimethoxanate or proquamezine. Some of the newer benzodiazepines have not been tested.
Violet–red
Perphenazine
Brown–red
Alimemazine
Pink
Profenamine
Orange
Fluphenazine, phenothiazine, trifluoperazine, triflupromazine
Pink–orange
Acetophenazine
Red–orange
Methdilazine
Brown–orange
Perazine, pericyazine
Green Blue–green
Clomipramine, desipramine, imipramine, ketamine, opipramol, thiethylperazine, thioridazine, trimipramine
Violet
Levomepromazine, proquamezine (!red!orange)
Brown
Metopimazine
Red–brown
Promazine
Forrest reagent Reagent
Mix together equal volumes of a 0.2% (w/v) solution of potassium dichromate, a 30% (v/v) solution of sulfuric acid, a 20% (w/w) solution of perchloric acid and a 50% (v/v) solution of nitric acid. Method
Table 30.16 Colours with formaldehyde–sulfuric acid Colour
Compound(a)
Red
Chlorprothixene*, clopenthixol*, flupentixol*, fluphenazine, metopimazine, pericyazine, promazine, thiothixene, triflupromazine, zomepirac
Brown–red
Lymecycline, oxytetracycline, tolmetin
Pink
Flurazepam, thioproperazine, trifluoperazine
Orange
Clonazepam, clorazepic acid, demeclocycline (!brown–red), demoxepam, diazepam, flunitrazepam, ketazolam, lorazepam, lormetazepam, medazepam (add water), metixene, nitrazepam, nordazepam, oxazepam, prazepam, temazepam, tetrazepam
Red–orange Yellow Green–yellow
Methacycline Bromazepam, clozapine, dimethothiazine, doxycycline Rolitetracycline (!yellow–brown), tetracycline (!yellow–brown)
Green
Thiethylperazine
Blue
Carphenazine, levomepromazine, thioridazine
Violet
Mesoridazine, perphenazine
Red–violet
Indications
Red, pink, orange, blue or violet colours are obtained with phenothiazines. A blue colour is obtained with certain dibenzazepines. The blue colour is inhibited by the presence of phenothiazines, so an excess of reagent must be added to overcome this. Colours are listed in Table 30.17. FPN reagent (general reagent for phenothiazines) Reagent
Mix together 5 mL of 5 % (w/v) ferric chloride solution, 45 mL of a 20% (w/w) solution of perchloric acid and 50 mL of a 50% (v/v) solution of nitric acid. Method
Dissolve the sample in a minimum volume of 2 mol/L hydrochloric acid (or use 1 mL of urine) and add an equal volume of the reagent. Indications
A variety of colours, from pink, red, orange, violet to blue, indicate the presence of phenothiazines (Table 30.18). Froehde reagent Reagent
Blue–violet
Acepromazine, acetophenazine, piperacetazine, promethazine, propiomazine
Brown–violet
Thiazinamium
Dissolve 1.0 g of molybdic acid or sodium molybdate in 100 mL of hot concentrated sulfuric acid.
Tryptamine
Method
Chlortetracycline, clomocycline
Add a drop of the reagent to the sample on a white tile.
Brown Orange–brown (a)
Alimemazine, chlorpromazine, diethazine, mequitazine, methdilazine, perazine, phenothiazine, prochlorperazine, profenamine, thiopropazate
Dissolve the sample in a minimum volume of 2 mol/L hydrochloric acid and add an equal volume of the reagent. To test urine, add 1 mL of reagent to 0.5 mL of urine.
Compounds giving colours that fluoresce under UV light (l = 350 nm) are indicated by an asterisk.
Indications
Colours are listed in Table 30.19.
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:45
Colour test methods Table 30.18 Colours with FPN reagent
Furfuraldehyde (general reagent for carbamates)
Colour
Compound
Reagent
Red
Chlorpromazine, dimetotiazine, mesoridazine, methdilazine, prochlorperazine, thiazinamium, thiopropazate
A 10% (v/v) solution of furfuraldehyde in ethanol.
Orange–red
Mequitazine
Violet–red
Perphenazine, proquamezine (!red!orange)
Brown–red
Alimemazine
Orange
Acetophenazine, diethazine (!yellow), fluphenazine, metopimazine, morphine, pericyazine, phenothiazine, profenamine, promethazine, thioproperazine, trifluoperazine, triflupromazine
479
Method
Dissolve the sample in ethanol, place a drop of the solution on a filter paper, add 1 drop of the reagent and expose the paper to hydrochloric acid fumes for 2–3 min. Indications
A black spot indicates the presence of non-aromatic carbamates. N-Substituted carbamates do not react. The LOD is 1 mg.
Red–orange
Carfenazine
Iodine test
Pink–orange
Propiomazine (!red!fades)
Method
Brown–orange
Acepromazine, perazine, piperacetazine
Blue
Clomipramine, imipramine, thiethylperazine, thioridazine, trimipramine
Mix the sample with an equal volume of manganese dioxide and heat the mixture carefully to dull redness over a small flame. Repeat the test by heating the sample alone.
Violet
Levomepromazine
Indications
Promazine
The appearance of violet vapour indicates the presence of iodine in the molecule. Better results are sometimes obtained when the manganese dioxide is omitted (e.g. with amiodarone).
Brown Red–brown
Table 30.19 Colours with Froehde reagent Colour Yellow Blue–yellow
Compound
Iodoplatinate test (general test for alkaloids and nitrogenous heterocyclic compounds)
Hydrocodone, pethidine
Reagent
Oxycodone-HCl
Add 2 mL of a 5% (w/v) solution of platinic chloride and 5 g of potassium iodide to 98 mL of water and shake until dissolved. This reagent is often used as a locating agent in TLC.
Orange
Diphenhydramine, flurazepam, promazine, trifluoperazine, triflupromazine
Green
Chlorphentermine, codeine, mescaline, oxycodone, phenyltoloxamine
Yellow–green
Lysergide
Method
Dissolve the sample in 2 drops of 2 mol/L hydrochloric acid, add 2–3 mL of the reagent and dilute to 10 mL with water.
Blue
Pentazocine
Indications
Red
Amfetamine, chlorpromazine-HCl
A violet, blue–violet, brown–violet or grey–violet precipitate suggests the presence of an alkaloidal base (precipitated as the alkaloid–iodoplatinate complex). The clearest colours are obtained with tertiary and quaternary amines; primary amines give indistinct colours and amines of small relative molecular mass generally do not react.
Grey–red
Propoxyphene-HCl
Purple–red
Alimemazine, diacetylmorphine, promethazine, propylhexadrine, salicylic acid, tetracycline, thioridazine
Brown Red–brown
Ephedrine, mescaline Doxepin-HCl
Koppanyi–Zwikker test
Black Brown–black
Opium
Reagent
Green–black
Methylenedioxyamfetamine (MDA)-HCl
A 1% (w/v) solution of cobalt nitrate in ethanol. Method
Fujiwara test (general reagent for halogenated hydrocarbons)
Dissolve the sample in 1 mL of ethanol, add 1 drop of the reagent followed by 10 mL of pyrrolidine and agitate the mixture.
Reagent
Indications
Freshly prepared 20% (w/v) sodium hydroxide solution.
A violet colour is given by substances that contain the following structures:
Method
Mix together 2 mL of the reagent and 1 mL of pyridine. Add the sample (1 mL of urine) and heat in a water-bath at 100 C for 2 min with shaking. Indications
A red–pink colour in the pyridine layer indicates the presence of compounds that possess at least two halogen atoms bound to one carbon atom. These include chloramphenicol, chlorbutanol, chloroform, dichlorophenazone, trichloroethane, trichloroethanol, trichloroacetic acid and trichloroethylene. Chloral hydrate and dichlorophenazone do themselves react but are excreted in urine as trichloroacetic acid. No colour is given by dicophane (DDT) or carbon tetrachloride, although massive exposure to the latter solvent may lead to a positive urine test because of the presence of chloroform as a contaminant. 2,2,2-Trichloroethanol gives a yellow colour. The LOD is 1 mg/L.
n n
Imides, in which C¼O and NH are adjacent in a ring (e.g. barbiturates, glutethimide, oxyphenisatine and saccharin). Sulfonamides and other compounds with free –SO2NH2 on a ring (e.g. clopamide, furosemide, sulfanilamide and thiazides), or with –SO2NH2 in a side-chain (e.g. chlorpropamide), or with –SO2NH2 that links a benzene ring with another ring other than a pyrazine, pyridazine, pyridine or pyrimidine ring (e.g. sulfafurazole and sulfamethoxazole). These latter structures give pink or red–violet colours (e.g. sulfadiazine and sulfadimethoxine).
No response is obtained with compounds with other substituents on the nitrogen atom. Anomalous responses are obtained with paramethadione and theophylline (violet), and with cycloserine, idoxuridine, mephenytoin, niridazole and riboflavin (no response). Note that hydrochlorides give a blue colour before the addition of pyrrolidine.
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:45
480
Colour Tests
Liebermann's reagent Reagent
Add 1 g of sodium or potassium nitrite to 10 mL of concentrated sulfuric acid with cooling and swirling to absorb the brown fumes. Method
Add 2 or 3 drops of the reagent to the sample on a white tile. Occasionally it is necessary to carry out the test in a tube and heat in a water-bath at 100 C. Many substances give colours with sulfuric acid alone and the test should be repeated using sulfuric acid instead of the reagent. Indications
the response to the reagent at the violet end of the spectrum are, in decreasing order of efficacy: ring sulfur (with or without aromatic ring); ring oxygen (with aromatic ring); extra-ring oxygen or sulfur (with aromatic ring); aromatic compounds that consist entirely of C, H, N. Thus, there is a tendency for the response to the Marquis reagent to move gradually towards longer wavelength (i.e. through green to orange and red) as the ratio of C, H, N to the other groups in the molecule rises (Table 30.22). The LOD values are: 1 mg for codeine sulfate, mescaline sulfate, methadone-HCl; 5 mg for lysergide tartrate, metamfetamine-HCl and morphine; 10 mg for amphetamine-HCl and diamorphine-HCl.
This test was originally developed to give intense colours with phenols:
McNally's test
n
Reagents
n
n
Orange colours are given by substances that contain a monosubstituted benzene ring not joined to C¼O, N–C(¼O)– or to a ring that contains a C¼N–O– group. Orange or brown colours are given by some substances that contain two monosubstituted benzene rings (or some disubstituted compounds in which fluorine is the second substituent) that are joined either to one carbon atom or to adjacent carbon atoms. A wide range of colours is given by compounds that contain –OH, O–alkyl or –O–CH2O– groups attached to a benzene ring or to a ring in a polycyclic structure that contains a benzene ring. The benzene ring must not bear –NO2, or be halogenated, or contain an –O– substituent ortho to the oxy groups. Compounds that contain ring sulfur give a similar range of colours.
Colours are listed in Table 30.20. Note that a yellow colour is given by a variety of other compounds.
1. A 0.5% solution of copper sulfate in 10% acetic acid. 2. A freshly prepared 2% (w/v) solution of sodium nitrite. Method
Dissolve the sample (1 mg) in a few drops of acetone, and add 1–2 mL of water. Add 3 drops of solution 1 and an equal volume of solution 2. Shake and heat in a water-bath at 100 C for 3 min. Indications
A red colour indicates the presence of free salicylic acid. Aminosalicylic acid gives a brown precipitate, and diflunisal gives a violet colour. Certain acids produced during the putrefaction of tissues also give red colours in this test: p-hydroxyphenylacetic acid, p-hydroxyphenylpropionic acid and p-hydroxyphenyl-lactic acid. Mecke's reagent (useful test for opium alkaloids)
Mandelin's test (useful test for amfetamines and antidepressants)
Reagent
Reagent
Method
Dissolve 1.0 g of ammonium vanadate in 1.5 mL of water and dilute to 100 mL with concentrated sulfuric acid.
Add a drop of the reagent to the sample on a white tile.
Method
An immediate blue or green colour is indicative of opiates (see Table 30.23).
Add a drop of the reagent to the sample on a white tile.
Dissolve 1.0 g of selenious acid in 100 mL of concentrated sulfuric acid.
Indications
Indications
When interpreting the result of this test, account should be taken of the colour given by sulfuric acid and by Liebermann’s test. Hydrochlorides give a red colour with this reagent. When the colours differ from those given with sulfuric acid or Liebermann’s test, this indicates an aromatic ring together with a saturated 5-, 6- or 7-membered ring that contains only one nitrogen atom. The heterocyclic ring must not contain a second nitrogen atom or an oxygen atom. It must not be substituted or bound by –CONH– to the aromatic ring. The aromatic ring must not have –CF3 as a substituent. Colours are also produced if sulfur is in a ring, provided that the ring does not contain more than one nitrogen atom (Table 30.21). LOD values are: codeine sulfate 5.0 mg, amphetamine-HCl 10.0 mg, diamorphine-HCl 20 mg, metamfetamine 150 mg, morphine 5 mg and strychnine 0.05 mg.
Melzer's reagent (general reagent for hallucinogenic mushrooms)
Marquis test
Mercurous nitrate (general reagent for barbiturate-like compounds)
The Marquis test is a useful broad-spectrum test used mostly for opium alkaloids and amfetamines. Reagent
Carefully mix 100 mL of concentrated sulfuric acid with 1 mL of 40% (v/v) formaldehyde solution (stable for several weeks if protected from light). Method
Add a drop of the reagent to the sample on a white tile. Indications
Various colours that represent the whole of the visible spectrum are given by a large number of compounds. Structures that tend to maintain
Reagent
Dissolve 1.5 g of iodine in 100 mL of an aqueous solution that contains 5 g of potassium iodide and 100 g of chloral hydrate. Method
Place a few drops of the reagent on the mushroom spores or mushroom tissue to be tested. Indications
A blue, bluish-grey or black–grey colour indicates amyloid mushrooms. A slight yellow or no change indicates that the mushrooms are nonamyloid. Psilocybes are always non-amyloid.
Reagent
To a saturated solution of mercurous nitrate, add solid sodium bicarbonate until effervescence ceases and the precipitate formed becomes yellow. The precipitate then changes to a biscuit colour. This reagent should be freshly prepared and should be shaken immediately before use, and should not be kept for more than 1 h. Method
Dissolve the sample in the minimum amount of ethanol, add 1 drop of the opaque reagent, shake and examine at intervals during 2 min. A blank solution that contains only ethanol and reagent should be treated similarly at the same time.
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:45
Colour test methods
481
Table 30.20 Colours with Liebermann's reagent Colour
Compound
Red
Acepromazine, ajmaline, alprenolol, aminacrine (100 C), antazoline, brucine, chlorprothixene, clopenthixol, flupentixol, mestranol, oxytetracycline, prajmalium, thiazinamium, tiotixene, tolmetin (100 C), trifluoperazine, xylazine
Violet–red
Indapamide
Brown–red
Methylchlorophenoxyacetic acid
Pink Brown–pink Orange
Trichlorophenoxyacetic acid (!brown) Prazosin (100 C !red–orange) Aletamine, alverine, ampicillin, atropine methobromide, atropine methonitrate, baclofen, benactyzine (!brown), bethanidine (!brown), broxyquinoline, butanilicaine, chloroquine (100 C), clidinium (!brown), cyclandelate, cyclizine, dazomet, decoquinate (slow), diethylthiambutene (100 C), dimefline, diuron, doxapram, dyclonine (100 C), fenclofenac (100 C, !brown), fenitrothion, fenpipramide, glibenclamide (100 C, 15 s), hyoscine butylbromide, hyoscine methonitrate, linuron, loxapine (50–60 C), metindizate (!brown), methylphenidate, metolazone (!green–brown), monolinuron, nomifensine, phenazone (100 C), phenelzine, propham, salinazid, sulfinpyrazone, tolazoline, trimetaphan, tripelennamine (!brown), triprolidine, xipamide, zomepirac (100 C)
Red–orange
Acetanilide, amfetamines, aniline, atropine, bamipine, beclamide, benethamine, caramiphen, carbetapentane, chlorcyclizine, cinchophen, cycrimine, diphenylpyraline, doxylamine, dropropizine, ephedrines, famprofazone, fencamfamin, glutethimide, hyoscine, hyoscyamine, isoaminile, isocarboxazid, levamisole, meclozine, mephentermine, methoin, methyl benzoquate, methylphenobarbital, metixene, metomidate, morazone, nialamide, pentapiperide, pethidine, phenacemide, phenbutrazate, phendimetrazine, phenglutarimide, pheniramine, phenmetrazine, phenobarbital, phensuximide, phenylmethylbarbituric acid, phenytoin, prolintane, tofenacin, tranylcypromine, triamterene, triphenyltetrazolium, warfarin
Brown–orange
Ambutonium, bumetanide, diphenhydramine, fenuron, feprazone (100 C, !brown), ibuprofen, labetalol, mepivacaine, methadone, nefopam (!brown), tetrahydrozoline
Yellow Brown–yellow Green Blue–green
Amicarbalide (100 C), clonidine (100 C, !orange), dequalinium (100 C, !orange), diethylpropion, diloxanide, ethoxzolamide, fenfluramine (100 C), flavoxate, gliclazide, metoclopramide, nifenazone (100 C), piroxicam, propachlor, tropicamide Amiodarone Bialamicol, chlorotrianisene, colchicine, dextromoramide (100 C), diamthazole, hydrastine, mequitazine, naphthols, phenol, phenothiazine, thiocarlide Hydrochlorothiazide, hydroflumethiazide, pindolol
Brown–green
Cyclopenthiazide
Grey–green
Azapropazone
Black–green Blue
Green–blue Violet Red–violet Black–violet Brown
Red–brown
Naproxen Amidopyrine (100 C), bendroflumethiazide, benzonatate (100 C), chromonar (100 C, 3 min), clomipramine, diphenylamine, dipyrone (100 C), imipramine, mefenamic acid, mefruside, oxypertine, padimate (100 C), procarbazine (100 C, 15 s), propyphenazone (100 C; red with water), tetracaine (100 C), yohimbine Amiphenazole (100 C) Methocarbamol, mianserin, paracetamol, penthienate methobromide, phenacetin, propiomazine, resorcinol, timolol (100 C), trazodone (100 C; transient) Chloroxuron Methoxychlor Acepromazine, acetophenazine, adiphenine, azacyclonol, barban, benzilonium, benzyl nicotinate, biperiden, clemastine, clofenotane, clomifene, cyclothiazide, dextropropoxyphene, dichlorprop, diperodon, diphemanil, difenidol, emepronium, etenzamide, fenpiprane, flurbiprofen, haloperidol, mepenzolate, methylpiperidyl benzilate, mexiletine, nadolol, penfluridol, phenaglycodol, phenylbutazone (100 C), phosalone, pimozide, pipazethate (100 C, !red), pipoxolan, pyrrobutamine, rotenone, sotalol (100 C), sulindac, veratrine, zimeldine Benzthiazide, bisacodyl, carfenazine, chlorpromazine, diclofenac, dosulepin, profenamine, etisazole, fenbufen, fenoprofen, methapyrilene, perphenazine, polythiazide
Pink–brown
Metoprolol
Orange–brown
Benazolin, diphenadione, maprotiline, methiocarb, piperidolate
Green–brown
Methdilazine, norbormide, promazine, thiopropazate
Violet–brown
Bamethan, clofibrate, dichlorophen
Black–brown
Mecoprop
Grey
Isopropamide
Black
Acetomenaphthone, aloin, aminophenols, amodiaquine, apomorphine, atenolol, benorilate, benzquinamide, buprenorphine, butorphanol, carbaryl (!green), carbidopa, cepha€ eline, chloroxylenol, chlorphenesin, clomocycline, clorgyline, codeine, cotarnine, cresol, cyclazocine, dextromethorphan, diamorphine, dibromopropamidine, diprenorphine, doxepin, emetine, ethamivan, ethinylestradiol, estradiol, estriol, estrone, etilefrine, furosemide, glycopyrronium, guaiphenesin, hexobendine, hydroxyephedrine, hydroxystilbamidine, ibogaine, indometacin, levallorphan, mebeverine, mescaline, methylchlorophenoxyacetic acid, methylenedioxyamfetamine, morantel, morphine, naloxone, 1-naphthylacetic acid, narceine, nicergoline, normetanephrine, noscapine, noxiptiline, octafonium, oxprenolol, oxyphenisatine, papaverine, pholcodine, pizotifen, practolol, profadol, propanidid, protokylol, pyrantel, rimiterol, ritodrine, rotenone, salbutamol, terbutaline, tetrabenazine, tetracycline, thymol, trimethobenzamide, trimetozine, tubocurarine, verapamil, viloxazine
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:46
482
Colour Tests
Table 30.21 Colours with Mandelin's test Colour
Compound
Red
Ajmaline, amfetamine, azacyclonol, chlorprothixene, diperodon (!green), dofamium (!brown), flupentixol, gelsemine (!green), indapamide, mequitazine, methotrexate, nialamide, pericyazine, prajmalium, prolintane, sodium cromoglicate, tiotixene, xylometazoline
Brown–red Orange
Diacetylmorphine-HCl, doxepin-HCl, nadolol, propoxyphene-HCl Brompheniramine, dropropizine (slow), ethylnoradrenaline, hydrastinine (!green), lachesine (!green), levamisole (!grey–green), methanthelinium, metixene, methyldopa, methyldopate, methylpiperidyl benzilate (!brown!green), noradrenaline, orphenadrine, pipenzolate (!green), poldine metilsulfate (!green!violet), procaine-HCl, propantheline, proquamezine (!violet), solanidine (!violet!blue), solanine (!violet!blue), strychnine (blue!purple!violet!red!red–orange), sulindac, thenalidine (!brown)
Red–orange
Cotarnine (!brown), doxepin
Green–orange
5-Methyltryptamine
Brown–orange Yellow
Mexiletine Azaperone, benzatropine, broxaldine, chelidonine (!green), conessine, deptropine, desipramine (!blue), dihydralazine, diphenhydramine, difenidol, diphenylpyraline, dropropizine (!orange), halquinol, homidium, lidoflazine, methacycline (!orange– violet), paraphenylenediamine, penicillamine, protokylol (!brown), tofenacin, tylosin (!yellow–brown), veratrine (!orange!violet– brown), viprynium
Orange–yellow
Cocaine-HCl, hexoprenaline, methaqualone, methylphenidate-HCl
Green–yellow
Methoxamine, oxycodone-HCl
Yellow–brown
Mescaline-HCl
Green
Acepromazine (!red), adiphenine (!blue), amfetamine, benorilate, bephenium hydroxynaphthoate, bibenzonium, buclosamide (blue rim), bunamidine, chlorpromazine (!violet), clefamide (!brown), codeine (green!blue), colchicine, cyclazocine, cyclomethycaine (!brown), debrisoquine, diaveridine, dibenzepin, diethazine (!violet with excess reagent), diethylthiambutene (!green–blue), dimethindene, dimethoxanate (!brown), dimoxyline, dipipanone (!blue), dosulepin, doxorubicin, doxycycline (!yellow), fenpiprane, guanoxan, harman, hydroxyephedrine, isoxsuprine, metanephrine, methadone (!blue), methdilazine (!violet), methocarbamol, methoxyamfetamine, methylenedioxyamfetamine (!blue), a-methyltryptamine (!orange), metopimazine, monocrotaline, niclosamide, nitroxoline, norharman (!yellow), normetanephrine, obidoxime (!blue), oleandomycin, oxymetazoline, paracetamol, pecazine (!violet), pentazocine, perazine (!violet), phenazone, phenazopyridine, phenformin, phenindamine, phenoxybenzamine (!violet), phenyltoloxamine, pindolol, piperacetazine (!red!violet), pipoxolan (!brown), prenylamine, profenamine (!violet), proflavine, promazine (!violet), promethazine (!violet), propranolol, reserpine, ritodrine, salicylic acid, thenium, thenyldiamine, thiocarlide (!yellow), tranylcypromine (!violet), trifluomeprazine (!red–violet), trihexyphenidyl
Yellow–green
Benzfetamine-HCl, metamfetamine-HCl, normethadone, opipramol
Blue–green
Amfetamine-HCl, benzoctamine, berberine (!brown), edrophonium, hydroxystilbamidine, ketobemidone, methoxyphenamine, phentolamine, profadol (!green), viloxazine
Brown–green
Benzydamine, chlorphenesin
Grey–green
Alverine, azapropazone, diamphenethide, diethyltryptamine (!yellow), dihydrocodeine, guaifenesin, hordenine, levomethadyl acetate, normorphine, oxyphencyclimine, papaverine, terbutaline, trihexyphenidyl
Blue
Green–blue Violet
Bamethan (!green), clomipramine, deserpidine (!green), desferrioxamine (!violet), doxapram, droperidol (!green), harmine (!green), imipramine (add water), maprotiline, mebhydrolin, metaraminol, phenaglycodol, phenyramidol, pyridoxine (!grey–green), salbutamol (blue rim!brown rim), strychnine (blue!purple!magenta!red!red–orange!orange), thioridazine (!violet), trimipramine (add water), triphenyltetrazolium (slow), xipamide, xylazine, yohimbine (!green) Chlophedianol, labetalol Alimemazine, amidefrine, benperidol, bezitramide (!orange), bisacodyl, captodiame, cefaloridine, chloropyrilene (!orange), clomifene (!orange–brown), clomocycline (!brown), denatonium, dipyridamole, guanoclor (!orange!brown–yellow), guanoxan, hexobendine, hydromorphone (!orange), mepacrine (!yellow), mepyramine, metisazone (!yellow), mianserin, morantel, naloxone (!brown), oxyclozanide (!orange), oxyphenisatine, oxytetracycline (!red!orange), penthienate, perphenazine, phenylbutazone, pizotifen (!green), prilocaine, primaquine (!orange), propiomazine, pyrantel, pyrrobutamine, rolitetracycline (!red!orange), tetracycline (!red!orange), thiethylperazine, thiopropazate, triacetyloleandomycin (slow), tridihexethyl, trimetazidine
Red–violet
Antazoline, carfenazine, dimethothiazine, histapyrrodine, thonzylamine
Blue–violet
Alcuronium, hexocyclium, levomepromazine
Brown–violet
Alprenolol, bitoscanate, butaperazine, naphazoline
Grey–violet
Methadone-HCl, methoserpidine, oxprenolol, tricyclamol
Black–violet
Methylenedioxyamfetamine-HCl, methapyrilene
Brown
Amitriptyline (!green), azapetine, bamipine, carbetapentane (slow), clidinium (!green), cyclopentolate, diphemanil, dipyrone, doxepin, embutramide, fluanisone, fluphenazine, isoetarine, isometheptene, isoprenaline, metindizate, methyl benzoquate, methysergide, metoclopramide, norpipanone (!blue), nortriptyline (!green), opium, phenelzine, phenylephrine, pimozide, piperidolate, prochlorperazine (!violet), propoxycaine, rescinnamine, salinazid, stanozolol, tetrabenazine, thioproperazine (!green!violet), tolnaftate, tolpropamine, tramazoline, tubocurarine
Red–brown
Benzthiazide, clioxanide, cycrimine, decoquinate, diclofenac, ethomoxane, hydrastine (!red), trifluoperazine, triflupromazine
Pink–brown
Metoprolol
Orange–brown
Rifampicin, spiramycin, thebaine
Yellow–brown
Clemastine, clofazimine, physostigmine, rifamycin SV, trimethoprim, tripelennamine
Green–brown
Etenzamide, harmaline, lysergic acid, mesoridazine, narceine, pentazocine, phenyltoloxamine, syrosingopine
Violet–brown
Chlortetracycline (!yellow), cyproheptadine, demeclocycline, dihydroergotamine, ergotamine, lymecycline (!yellow), methylergometrine, nicergoline (!brown), octaphonium, oxethazaine, protriptyline, trimethobenzamide
Grey–brown
Dextropropoxyphene, mephenesin carbamate
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:47
Colour test methods
483
Table 30.21 continued Colour
Compound
Grey
Dihydromorphine, diprenorphine, etilefrine (!green!brown), ibogaine (!violet), indometacin, lobeline, lysergide, morphine, oxypertine, propranolol, trazodone (!violet)
Blue–grey Black Grey–black
Alphaprodine, diamorphine, morphine Procyclidine Flurazepam
Table 30.22 Colours with the Marquis test Colour
Compound
Red
Alprenolol, benzylmorphine (!violet), buphenine, dimethothiazine, etenzamide, etilefrine, fenclofenac (slow), fenpiprane, fluphenazine, flurbiprofen, hexoprenaline, labetalol (!brown–red), maprotiline, mephenesin carbamate, mequitazine (slow), mesoridazine (!violet), methoxyphenamine, metopimazine, mexiletine, nadolol, pentazocine (!green), pericyazine, phenazopyridine, phenoperidine, phenylephrine, piperacetazine, prenylamine, thebaine (!orange), thiethylperazine (!green), thioproperazine, tiotixene, tolpropamine, tranylcypromine (!brown), vinblastine
Orange–red
Alverine, amfetamine-HCl, bethanidine, diphemanil, flupentixol, metamfetamine-HCl
Violet–red
Thioridazine (!blue–green)
Black–red
Doxepin-HCl
Brown–red
Alphaprodine, doxepin, trihexyphenidyl
Pink
Alimemazine, fenoprofen, fluopromazine, metoprolol, promazine, promethazine, trifluoperazine
Orange
Adrenaline (!violet), aletamine, amfetamine (!red!brown), anileridine (slow), benactyzine (!green!blue), benzethonium, benzilonium (!green!blue), benzfetamine, bunamidine (!red), carbetapentane (slow), carfenazine (!red–violet), chlorphentermine, clidinium (!blue), cyclandelate (slow), cycrimine (!red), dehydroemetine, dimethyltryptamine, dipyridamole, ethacridine (!red), ethoheptazine, ethylnoradrenaline (!brown), famprofazone, fenbufen (!brown), fencamfamin, fenethylline, fentanyl, harmine, indapamide (!violet), indometacin, isothipendyl, ketobemidone, lachesine (!green!blue), lymecycline, mepenzolate (transient), mephentermine (!brown), mescaline, metamfetamine, metanephrine (!violet–brown), methacycline, methanthelinium, methindizate (!green), methylphenidate, methylpiperidyl benzilate (!green!blue), 5-methyltryptamine (!brown), a-methyltryptamine (!brown), N-methyltryptamine, nefopam (!brown), nomifensine (slow), normetanephrine (!violet–brown), oxeladin, oxytetracycline, pentapiperide, pethidine, phenethylamine, phenformin (!brown), phentermine, piminodine, pipenzolate (!green!blue), piperidolate, pizotifen (!red), poldine methylsulfate (!green!blue), primaquine, profadol (!red–brown), prolintane (!brown), propantheline, prothipendyl, psilocybine, rolitetracycline, spiramycin, tetracycline, trimethoprim, trimethoxyamfetamine, tryptamine, veratrine, xylometazoline
Red–orange
Chlorprothixene
Pink–orange
Diuron
Yellow–orange
Orphenadrine, pipradrol
Brown–orange Yellow
Amitriptyline Acriflavine (!red), amiloride, azacyclonol, benzquinamide, benzatropine, bromazine, broxaldine, broxyquinoline, caramiphen, chlordiazepoxide, chlorphenoxamine (!green), chlortetracycline (!green), chlortalidone, cinchophen, clefamide, clemastine (green rim), colchicine, conessine (!orange), cyclizine, demeclocycline (!green), deptropine, diethyltryptamine (!brown), 2,5-dimethoxy-4methylamfetamine, diphenhydramine, diphenidol, diphenylpyraline, doxycycline, ethoxzolamide, ethylmorphine (!violet!black), furaltadone, halquinol, hydrocodone (!brown!violet), hydromorphone (!red!violet), hydroxyephedrine, isoetarine (!orange), lidoflazine, lorazepam, mepacrine, methyldopa (!violet), methyldopate (!violet), norcodeine (!violet), orciprenaline, oxycodone (!brown!violet), oxyphenbutazone, phanquone, phenbutrazate (slow), phentolamine, phenyramidol, pindolol (!brown), pramoxine (!green), proflavine (!orange), salbutamol, salinazid, sodium cromoglicate, solanine (!violet), terbutaline, tetrabenazine, thebacon (!violet), tofenacin, triamterene, trimetazidine (fades), vancomycin, viprynium embonate, zomepirac (100 C, !orange)
Orange–yellow
Methylphenidate-HCl, stanozolol
Pink–yellow
Methadone-HCl
Green
Berberine, carbaryl, chelidonine, harman, norharman, oleandomycin, propranolol, protriptyline, pseudomorphine, sulindac (slow)
Yellow–green
Acepromazine (!red), verapamil (!grey)
Blue–green
Tolnaftate
Brown–green
Harmaline
Grey–green Blue Grey–blue Violet
Cyproheptadine, deserpidine, naphazoline, oxypertine, phenindamine, protokylol, rescinnamine, reserpine (!brown) Clofibrate, embutramide, nicergoline (!grey) Mebhydrolin, 1-naphthylacetic acid Alimemazine, apomorphine (!black), azatadine, benorilate, bisacodyl, buprenorphine, butriptyline, captodiame, chloropyrilene, chlorpromazine, clofazimine, codeine, diamorphine, diethylthiambutene, dihydrocodeine, dimethindene (!blue), dimethoxanate, doxorubicin, doxylamine, etoxazene, guaifenesin, guanoxan, hexocyclium metilsulfate, mepyramine, 6-monoacetylmorphine, morphine, nalorphine, normorphine, oxprenolol, oxycodone-HCl, oxyphenisatine, pecazine, penthienate, pentazocine, perazine, perphenazine, phenoxybenzamine, phenyltoloxamine, pholcodine, pimozide, pipoxolan (!grey), prochlorperazine, procyclidine, profenamine, promazine, promethazine, proquamezine, solanidine, thenium, thiopropazate, tricyclamol, viloxazine
Red–violet
Acetophenazine, benzoctamine, bephenium hydroxynaphthoate, cefaloridine, chlophedianol (!brown), dihydromorphine, ethomoxane, isoxsuprine, lobeline, methdilazine, propiomazine, tramazoline, trifluomeprazine, trifluoperazine, triflupromazine, trimeperidine
Blue–violet
Methocarbamol, levomepromazine, morantel, neopine, noscapine (fades), pyrantel table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:47
484
Colour Tests
Table 30.22 continued Colour
Compound
Brown–violet
Butaperazine, dopamine, methylenedioxyamfetamine, tridihexethyl
Grey–violet
Diprenorphine, oxymorphone, pyrrobutamine, thenalidine, trihexyphenidyl
Black–violet
Dextropropoxyphene (!green), methapyrilene, thenyldiamine
Brown
Bibenzonium, carbidopa, cyclazocine (!green), diclofenac (slow), dimoxyline, dosulepin, doxepin, ergometrine, ergotamine, erythromycin, hordenine (!green), ibuprofen (100 C, !orange), isoprenaline (!violet), lysergamide, methadone, naloxone (!violet), naproxen, narceine (!green), noradrenaline, pethidine-HCl, phentermine, phenazocine, rimiterol (!black), serotonin (slow), syrosingopine, tyramine (!green)
Red–brown
Biperiden, debrisoquine, methyl benzoquate, oxetacaine, phenprobamate, trimetozine, tripelennamine
Orange–brown
Benethamine (!brown), clomocycline, nortriptyline
Yellow–brown
Moxisylyte, ritodrine, triacetyloleandomycin, tylosin
Green–brown
Alcuronium, bufotenine, psilocin
Violet–brown
Clomifene, diethazine, levomethadyl acetate (!grey–green), methoxamine (!green)
Grey–brown Grey Blue–grey Black
Dihydroergotamine, methylergometrine, octafonium Butorphanol, diaveridine (!violet–brown), ibogaine (!orange), lysergide, methoserpidine, methysergide, pholedrine (!green) Acetorphine (!yellow–brown), etorphine (!yellow–brown) Methylenedioxyamfetamine
Blue–black
Methylenedioxyamfetamine
Green–black
Lysergide
Table 30.23 Colours with Mecke's reagent Colour
Compound
Green
Diacetylmorphine, mescaline-HCl, morphine, oxycodone-HCl
Blue–green
Codeine, diacetylmorphine-HCl, hydrocodone tartrate, methylenedioxyamfetamine-HCl
Brown–green
Methadone
Orange
Alimemazine, diphenhydramine, fluopromazine, pethidine, phenyltoloxamine, promazine, promethazine, propoxyphene, trifluoperazine, triflupromazine
Indications
A change from colourless or from a pale colour to red, orange, yellow, green or blue is given by quinones, diones that possess an aromatic ring, phenols with adjacent hydroxy groups and compounds that contain nitro groups on a ring (Table 30.24). Many of these compounds are coloured already and give pale or colourless solutions in methanol. Millon's reagent (general reagent for phenols) Reagent
Dissolve 3 mL of mercury in 27 mL of fuming nitric acid and add an equal volume of water with stirring.
Yellow
Amfetamine, procaine
Method
Red
Doxepin-HCl
Add 0.5 mL of reagent to the sample and warm the mixture.
Black–red
Chlorpromazine-HCl
Purple–red
Tetracycline
Violet (dark blue)
Methylenedioxyamfetamine, thioridazine
Brown
Ephedrine
Red–brown
Propoxyphene-HCl
Black Green–black
Table 30.24 Colours with methanolic potassium hydroxide Colour
Compound
Red
Benserazide, isoetarine (!orange!yellow), metronidazole, nitrofurazone, phenindione
Orange–red Lysergide, opium
Indications
A dark grey or black colour indicates a ring imide group or sulfonamides with an additional ring. The speed and intensity of the reaction varies between different compounds. The following ring imides react in decreasing order of intensity: barbiturates, bemegride, phenytoin > benperidol, cycloserine, pimozide > glutethimide, oxyphenisatine > saccharin, sulfinpyrazone. In the case of sulfonamides, succinylsulfathiazole, sulfamoxole, sulfanilamide, sulfasomidine and sulfathiazole react with greater intensity than all others. Chlorpropamide and tolbutamide give a moderate response. If used as a spray, the LOD is 1–5 mg for barbiturates.
Pink
A 20% (w/v) solution of potassium hydroxide in methanol. Method
Add a few drops of the reagent to a solution of the sample in methanol and heat if necessary to boiling point to develop the colour.
Levodopa (!red–brown)
Orange–pink
Rimiterol
Brown–pink
Dobutamine
Orange
Acinitrazole, barban, carbidopa, dinitro-orthocresol (100 C), dinobuton, dinoseb, dodecyl gallate, hexoprenaline (!brown), isoprenaline (!yellow), nifedipine, nifuratel, nifursol, obidoxime, protokylol (!yellow)
Pink–orange
Adrenaline(!brown)
Yellow–orange
Nitrofurantoin
Yellow
Methanolic potassium hydroxide Reagent
Fenitrothion, quintozene, tecnazene, trifluralin
Green–yellow
Acebutolol, diphenadione, methyldopa (!orange), metolazone, niclosamide, niridazole, nitroxoline, nitroxynil, phanquone (!brown–violet), sodium cromoglicate Phytomenadione (!violet!brown)
Green
Apomorphine (!red), dinitolmide
Blue
Dopamine (!orange!brown), methyldopate (!orange), noradrenaline (!orange)
Violet
Dimetridazole (when boiled)
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:48
Colour test methods Indications
A red or orange–red colour indicates the presence of a phenolic substance. Primary aryl amines also react. Some basic compounds that contain a phenolic group do not react to this test; a combination of this test with the Folin–Ciocaltaeu reagent is therefore advised for phenolic compounds. Phenols that contain more than one hydroxyl group do not give the typical red colour. This reagent does not react with phenols substituted with Cl, Br or I. Naphthol–sulfuric acid This test should be carried out in conjunction with the sulfuric acid test. Reagent
Mix 1 g of naphth-2-ol with 40 mL of concentrated sulfuric acid and heat in a water-bath at 100 C, with occasional stirring, until the naphth2-ol is dissolved. Method
Table 30.25 Colours of steroids with naphthol–sulfuric acid Colour with hot reagent
Steroid
Colour after dilution
Red
Mestranol
Red
Orange–red
Desoxycortone
Blue–black
Dydrogesterone
–
Hydroxyprogesterone
Blue, violet (dichroic)
Noretynodrel
Brown–red
Brown–red
Ethinylestradiol
Pink
Orange
Norethisterone
Orange–brown
Orange, green (dichroic)
Norethandrolone
Red–orange
Yellow
Diethylstilbestrol
Orange
Testosterone
Green, brown (dichroic)
Green–yellow
Fluoxymesterone
Yellow
Green
Beclometasone
Brown–yellow
Fluocinolone
Yellow
Mix the sample with 1 mL of the reagent, heat in a water-bath at 100 C for 2 min and note any colour produced. Cool, add 1 mL of water and note the colour again.
Yellow–green
Dexamethasone
Yellow
Indications
Green, yellow (dichroic)
Estriol
Orange
Estrone
Orange
Triamcinolone
Yellow
A range of colours is obtained with steroidal structures (Table 30.25). A positive response to this test combined with a positive response to the sulfuric acid test is indicative of the presence of a steroid. Compounds other than steroids that give colours with this test include chloral hydrate and chloramphenicol (brown–yellow), starch and tartaric acid (green). Nessler's reagent
Green, brown (dichroic)
Fludroxycortide
Yellow
Blue–green, yellow (dichroic)
Estradiol
Orange
Violet
Fludrocortisone
Brown
Brown
Oxymetholone
Pink–orange
Prednisolone
Brown
Prednisone
Orange
Reagent
1. Dissolve 50 g of mercuric chloride and 35 g of potassium iodide in 200 mL of water and cool. 2. Dissolve 50 g of sodium hydroxide in 250 mL of water and cool. Add the cold solution 2 to the cold solution 1 and make up to 500 mL. Allow the mixture to stand and decant the clear supernatant (stable for many months) for use. Store in dark brown bottles away from the light.
Red–brown
Orange–brown
Method
Add the reagent (3 drops) to the sample (3 drops), agitate and heat the mixture to 100 C in a water-bath, examining it every minute for 10 min. A blank solution should be treated similarly at the same time. Indications
A brown–orange colour is produced quickly by aliphatic amides and thioamides. The presence of an aromatic ring slows the reaction. The nearer the amide group is to the ring, the more the reaction is inhibited. Substituents in the ring may cause a weak reaction. An immediate black colour is produced by substances that contain ortho- or para-hydroxy groups and by substances that contain an –NH–NH– or –NH–NH2 group in an aliphatic side-chain. Some compounds must be heated to 100 C to produce blackening. Colours are given in Table 30.26.
485
Yellow–brown
Green–brown
Progesterone
Yellow
Dimethisterone
Brown–green
Enoxolone
Orange
Fluocortolone
Red–brown
Alfadolone
Orange
Androsterone
Orange
Cortisone
Orange
Dienestrol
Yellow
Carbenoxolone
Orange
Cholesterol
Violet
Hydrocortisone
Yellow–brown
Betamethasone
Orange–brown
obtained (amantadine, rimantadine). Gentamicin gives a violet colour after heating for 4 min. Nitric acid, fuming
Ninhydrin Reagent
Dissolve 0.5 g of ninhydrin in 40 mL of acetone. Method
Dissolve the sample in methanol, place a drop of the solution on a filter paper, add 1 drop of the reagent and dry in a current of hot air. Indications
A violet colour that appears rapidly indicates the presence of an aliphatic primary amine or an amino acid group. The presence of an aromatic ring inhibits the response, and the inhibition increases the nearer the amino group is to the ring, as for amfetamine (pink–orange), procainamide and proxymetacaine (both yellow). If the amino group is associated with a saturated ring, a positive but weak pink–violet colour is
Method
Mix the sample with 3 drops of fuming nitric acid, heat at 50 C for 30 s and observe any colour produced. Cool the mixture, add 2 drops of it to 2 mL of concentrated sulfuric acid and observe the colour. To the remainder of the cooled mixture, add 2 mL of water followed by 2 mol/L sodium hydroxide, dropwise, until pH 8 is reached (use an indicator paper). Indications
Chlorinated phenols give a series of colours in the three parts of this test (Table 30.27). Nitric acid–sulfuric acid (Erdmann's reagent) Mix 1 mL of concentrated nitric acid with 30 mL of concentrated sulfuric acid.
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:49
486
Colour Tests
Table 30.26 Colours with Nessler's reagent
Table 30.28 Colours with nitrous acid
Colour
Compound
Colour
Compound
Orange
Acebutolol (slow), carbidopa (!black), methotrexate
Orange
Sulfafurazole
Acetylcarbromal (slow), bromvaletone, carbromal, chloramphenicol, dinitolmide, etenzamide (weak), ethionamide, fluoroacetamide, nicotinamide, phenacemide (slow), pheneturide (slow), protionamide, pyrazinamide, salicylamide (weak), urea
Yellow
Sulfadoxine, sulfachlorpyridazine, sulfadimidine, sulfaethidole, sulfamethizole, sulfamethoxazole, sulfamethoxydiazine, sulfamethoxypyridazine, sulfametopyrazine, sulfamoxole, sulfaphenazole, sulfapyridine, sulfasomidine, sulfathiazole, sulfinpyrazone
Green
Phenazone
Blue
Dipyrone (transient)
Violet
Amidopyrine (transient)
Brown–orange
Yellow
Dihydrostreptomycin (!brown), penicillamine
Brown
Demeclocycline, mebutamate (slow), nadolol, paracetamol (slow)
Yellow–brown
Atenolol (slow)
Black (immediate)
Adrenaline, apomorphine, ascorbic acid, benserazide, dihydralazine, dobutamine, dodecyl gallate, dopamine, ethylnoradrenaline, hexoprenaline, hydralazine, iproniazid, isocarboxazid, isoetarine, isoniazid, isoprenaline, levodopa, mebanazine, methyldopa, methyldopate, nialamide, noradrenaline, phenelzine, procarbazine, protokylol, rimiterol
Black (at 100 C)
Cimetidine, gentamicin, labetalol, meprobamate (grey–black), methallibure, salinazid, thiacetazone
Table 30.27 Colours with fuming nitric acid Colour Part 1
Part 2
Part 3
Compound
Orange–red
Orange
Orange–brown
Hexachlorophene
Red
Red
Brown–violet
Pentachlorophenol
Method
Mix the sample with 1 mL of the reagent and heat at 100 C in a waterbath for 2 min. A blank solution should be treated similarly at the same time. Indications
Red, orange, yellow, brown or black colours are given by aliphatic compounds that have a sulfur atom in the chain, and by aromatic compounds that have a sulfur atom in the side-chain. However, no colour is given when an S-alkyl chain is present, unless the chain is terminated by an halogenated group. No response is obtained if the sulfur is in a group that links two rings. Reducing agents such as ascorbic acid, chloral hydrate, chloroform and glucose, and compounds that contain a chain with a hydrazine link (–NH–NH–, –NH–NH2), give a translucent dark-grey or black colour, but do not give the gradual yellow to orange to brown colour seen with sulfur-containing compounds. Compounds that contain adjacent hydroxyl groups on an aromatic ring give orange colours that turn brown (Table 30.29).
Method
Dissolve the sample in 1 mL of ethanol, add a pellet of potassium hydroxide and evaporate to dryness (100 C in a water-bath). To the residue add 0.5 mL of water and 1 mL of carbon tetrachloride, shake and allow to separate. Decant the lower carbon tetrachloride layer and shake it with 1 mL of the reagent. Indications
A red colour in the acid layer suggests the presence of clofenotane or its metabolite, dichlorodiphenyldichloroethylene (DDE). The red colour changes to orange and then to green. Weak pink colours are given by aldrin, dieldrin and endrin. A red colour is also given by dichlorodiphenyldichloroethane (DDD, mitotane), but the colour does not change. Note that the substance should be tested to ensure that it does not give a colour with sulfuric acid alone.
Phosphorus test Method
To the sample add 0.5 mL of concentrated nitric acid and 0.2 mL of concentrated sulfuric acid and heat at 100 C in a water-bath for 30 min. Cool, add 1 mL of a 10% (w/v) solution of ammonium molybdate and replace in the water-bath at 100 C for 5 min. A blank solution should be
Table 30.29 Colours with palladium chloride Colour
Compound
Red
Gloxazone
Orange
Adrenaline (!brown), benserazide (!brown), bitoscanate, captopril, carbidopa (!brown), carbimazole, disulfiram, dobutamine (!brown), ecothiopate, isoetarine (!brown), levodopa (!brown), methallibure, thiamazole, polythiazide, rimiterol, thiacetazone, thiopental
Nitrous acid Method
Dissolve the sample in a minimum volume of water, and add an amount of solid sodium nitrite equal in volume to the sample followed by a few drops of 2 mol/L hydrochloric acid.
Brown–orange Yellow
Indications
Orange or yellow colours are given by certain sulfonamides, and green, blue or violet colours by certain phenylpyrazolines (Table 30.28). No response is obtained with succinylsulfathiazole, sulfacetamide, sulfadiazine, sulfadimethoxine, sulfaguanidine, sulfamerazine, sulfaquinoxaline, sulthiame or propyphenazone.
Orange–yellow Brown
Orange–brown
Palladium chloride Reagent
Dissolve, with the aid of heat, 0.1 g of palladium chloride in 5 mL of 2 mol/L hydrochloric acid and dilute the solution to 100 mL with water. Mix together equal volumes of this solution and 2 mol/L sodium hydroxide. The mixed reagent is stable for several weeks.
Black–brown
Demeton-S Clindamycin, dazomet, dimercaprol, dimethoate, methisazone (!orange!brown), penicillamine Thialbarbital Ambazone, azinphos-methyl, dihydrostreptomycin (slow), ethionamide, malathion, noxythiolin, parathion, phosalone, protionamide, spironolactone, thiram Chlorthiamid, diazinon, disulfoton, fenitrothion, formothion, phorate, vamidothion Di-allate, dichlofluanid, tri-allate
Grey
Chlorfenvinphos
Black
Ascorbic acid, captan, chloroform, cloral hydrate, mebanazine, nifuratel, phenelzine, procarbazine, sulfasalazine, sulfaurea, trichlorfon
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:49
Colour test methods Table 30.30 Colours with potassium dichromate
487
once. Add 1 drop of solution 2 and shake (the blue colour disappears and a clear pink solution develops). Add several drops of solution 3.
Colour
Compound
Red
Carbidopa
Yellow (!brown)
Phenol (2 min)
Green (!brown)
Adrenaline, dopamine, hexoprenaline, isoetharine, isoprenaline, levodopa, methyldopa, methyldopate, noradrenaline, rimiterol
Simon's test (modified sodium nitroprusside test)
Aniline (2 min)
Reagent
Benserazide, o-cresol (30 s), m-cresol (2 min), orciprenaline (slow), protokylol (!red–brown on warming), terbutaline (slow)
1. Dissolve 1 g of sodium nitroprusside in 100 mL of water and add 2 mL of acetaldehyde to the solution with thorough mixing. 2. Freshly prepared 2% sodium carbonate in distilled water.
Dobutamine
Method
Blue–green Brown
Green–brown
Indications
The chloroform layer develops an intense blue colour if cocaine is present. Methadone also reacts. The LOD is 60 mg cocaine-HCl and 15 mg methadone-HCl.
Add 1 drop of solution 1 to the sample, followed by 2 drops of solution 2. treated at the same time. For some compounds, the reaction may occur after shorter heating times than those stated above. Indications
A bright yellow solution or precipitate indicates the presence of phosphorus and suggests an organophosphorus pesticide, especially if the sample is a water-immiscible liquid. Cyclophosphamide and triclofos also react. Potassium dichromate
Indications
A dark-blue colour indicates a secondary amine (e.g. metamfetamine, ephedrine, 3,4-methylenedioxymetamfetamine (MDMA)) or an unsubstituted heterocyclic amine as its free base. A deep blue colour indicates the presence of metamfetamine. Primary amines (e.g. amfetamine, methylenedioxyamfetamine (MDA)) yield a slow pink to cherry-red colour. Sodium dithionite Reagent
Method 1
A 5% (w/v) solution of sodium dithionite in a 10% (w/v) solution of sodium hydroxide.
Dissolve the sample by shaking in 0.5 mL of 2 mol/L hydrochloric acid and add a few crystals of potassium dichromate.
Method
Indications
An immediate brown colour, or a green colour that changes to brown, indicates the presence of an aminophenol or of a phenol that has two or more hydroxyl groups in adjacent positions on the ring (Table 30.30). Monophenols, halogenated phenols and phenols with hydroxyl groups meta to each other react more slowly or not at all. Method 2
If the sample is a liquid, add 1–2 drops to 1 mL of water followed by 1 mL of a saturated solution of potassium dichromate in 50% v/v sulfuric acid. Indications
A green colour is given by acetaldehyde, ethanol, methanol, propan-1-ol and propan-2-ol. Schiff's reagent Reagent
Dissolve 0.2 g of basic magenta (fuchsin, CI 42510) in 120 mL of hot water, cool, add 20 mL of a 10% (w/v) solution of sodium hydrogensulfite and 2 mL of concentrated hydrochloric acid, and dilute to 200 mL. Store at 4 C and protect from light.
Apply the reagent to the sample, either on a white tile or as a solution in a test-tube. A blank solution should be treated similarly at the same time. Indications
Colours are produced by bis(pyridyl) compounds (Table 30.31). Dark colours are likely to be given by certain metallic solutions because of reduction. Sodium hypobromite test (for carbamazepine) Reagent
Dissolve 0.5 mL of bromine in 5 mL of a 40% w/v solution of sodium hydroxide with shaking and cooling. (this should be freshly prepared). Method
Add 1 mL of 2 mol/L hydrochloric acid to 5 mL of sample and 5 mL of chloroform. Vortex mix for 1 min and centrifuge for 5 min. Remove the upper layer, add 1 mL of the chloroform extract to 0.2 mL of sodium hypobromite reagent and mix for 30 s. Indications
Carbamazepine forms a blue to violet colour in the chloroform layer. The test has a sensitivity of 250 mg/L.
Method
Add the sample to 1 mL of the reagent.
Sodium nitroprusside
Indications
Reagent
A violet colour indicates the presence of an aliphatic aldehyde. The longer the carbon chain, especially if it is branched, the weaker the response to the test.
A 1% (w/v) solution of sodium nitroprusside.
Scott's test (see also Cobalt thiocyanate) Reagent
1. Cobalt thiocyanate dissolved in water (2% w/v) and then diluted 1 : 1 with glycerine 2. Concentrated hydrochloric acid 3. Chloroform. Method
Add a small amount of the sample to be tested to a test-tube, add 5 drops of solution 1 and shake. If cocaine is present a blue colour develops at
Method 1
Add the sample to 2 mL of the reagent followed by a drop of 2 mol/L sodium hydroxide. Indications
Orange colours are given by ketones and red colours by acetaldehyde. Table 30.31 Colours with sodium dithionate Colour
Compound
Green
Diquat
Blue
Paraquat
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:49
488
Colour Tests
Method 2
Indications
Mix the sample with a minimum volume of 2 mol/L sodium hydroxide, evaporate to dryness, dissolve the residue in 2 drops of water and add 0.5 mL of the reagent.
A red colour that appears in the lower acid layer indicates the presence of dieldrin (colour develops quickly) or aldrin (colour develops slowly). A pink–orange colour is obtained with endrin.
Indications
A violet colour is given by substances that contain labile sulfur in the molecule and by unsubstituted dithiocarbamates.
Tetrabromophenolphthalein ethyl ester
Method 3
Dissolve 50 mg tetrabromophenolphthalein ethyl ester (TBPE) in 100 mL chloroform, shake the solution for 2 min with 1 mL of 10% (v/v) hydrochloric acid and discard the aqueous phase. Dry the organic layer with anhydrous sodium sulfate. Separate the drying agent by filtration. Store the reagent in an amber bottle at 4 C.
Carry out Method 2 above, but after evaporation to dryness heat the residue until it is yellow or orange in colour before proceeding. Indications
A violet colour is given by certain substances that contain labile sulfur and do not react to method 2 (e.g. clomethiazole, lincomycin and monosulfiram). Sodium nitroprusside–acetone Reagents
1. Dissolve 2 g of sodium nitroprusside in 5 mL of water and add 45 mL of methanol 2. 2% (w/v) sodium carbonate 3. Acetone 4. 10% acetaldehyde. Method 1
Add a drop of solution 1 followed by a drop of solution 2 to 3–4 mg of sample dissolved in solution 3 on a spot plate. A purple colour is indicative of amfetamine. The LOD is 30 mg. Method 2
Reagent
Method
Place 0.5 mL of sample to be tested in a conical test tube, add 100 mL phosphate buffer (10 mmol/L, pH 8.0) and vortex mix. Add 50 mL of the TBPE reagent and vortex mix. After 2–3 min note the colour of the chloroform layer. If the sample to be tested is a solid, dissolve 1–2 mg of the material in 0.5 mL of buffer and proceed. Indications
A deep blue colour indicates quaternary ammonium compounds. An orange, brown, red or purple colour indicates the presence of basic drugs. This test is most sensitive to tertiary amines (e.g. tricyclics, propoxyphene, phenothiazines, diphenhydramine, phencyclidine, methadone, pethidine). Its LOD is 1 mg/L. Thalleioquin test
Add a drop of solution 1 followed by a drop of solution 4 to 1–2 mg of sample dissolved in solution 3. An immediate blue colour is indicative of metamfetamine. The LOD is 5 mg.
Method
Sodium picrate (Steyn test)
Indications
Reagent
A green colour indicates the presence of a quinine-type structure (e.g. hydroquinidine, hydroquinine, quinidine, quinine). Cinchonidine and cinchonine do not respond.
Prepare a solution of 5 g sodium bicarbonate and 0.5 g picric acid in 100 mL of water.
Dissolve the sample in a minimum volume of 2 mol/L hydrochloric acid, add 2 drops of bromine solution, place 1 drop of the mixture on a piece of filter paper and expose the paper to ammonia fumes.
Method
Mix the sample with a few drops of chloroform and concentrated sulfuric acid to hasten the reaction while holding a piece of filter paper, impregnated with the reagent, in the vapours that issue from the tube, and heating the contents to 30 C. Indications
The yellow colour of the filter paper changes from orange to brown– orange and then to orange–red or red in the presence of cyanide. Positive results are given by compounds that contain cyanide groups (e.g. cimetidine, diphenoxylate and isoaminile). Sulfuric acid Method
Apply concentrated sulfuric acid directly to the sample on a white tile or in a test-tube. Indications
A range of colours is obtained with compounds of various types. Steroids give orange or yellow colours, many of which fluoresce under UV light (l ¼ 350 nm) either immediately or after dilution (Table 30.32). Thioxanthenes give red or orange colours that fluoresce under UV light (l ¼ 350 nm) (Table 30.33). Sulfuric acid–fuming sulfuric acid
Trinder's reagent (see Ferric chloride) Reagent
The solution is prepared as follows: 40 g of mercuric chloride and 40 g of ferric nitrate are dissolved in 850 mL of distilled water; 10 mL of concentrated HCl is added and the solution is diluted to 1 L. This solution is stable for 1 year. Method
A few drops of the reagent are added to a few drops of urine. A purple colour indicates the presence of a salicylate. This test was devised for the quantitative assay of salicylates in serum, with the mercuric chloride serving as a protein precipitant. The ferric chloride test has been modified for use on blood samples (Asselin, Caughlin 1990). Blood (0.5 mL) is mixed with 1.0 mL of acetone and vortex mixed. The sample is centrifuged, and 50 mL of the supernatant is added to 50 mL of ferric chloride. A purple colour at the interface indicates salicylates. Vanillin reagent Reagent
Dissolve 1 g of vanillin in 20 mL of concentrated sulfuric acid, warming if necessary. Method
Mix together 7 mL of concentrated sulfuric acid and 3 mL of fuming sulfuric acid.
Add 2 drops of the reagent to the sample, heat in a water-bath at 100 C for 30 s and note any colour that is produced. Dilute the cooled mixture by adding a few drops of water and note any change of colour.
Method
Indications
Dissolve the sample in a minimum volume of toluene and add 1 or 2 drops of the reagent.
Many compounds of different chemical structure react with this reagent. However, for barbiturates, the reaction appears to be a steric
Reagent
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:50
Colour test methods
489
Table 30.32 Reactions of steroids with sulfuric acid Initial colour
Compound
Fluorescence at 350 nm
Fluorescence after dilution
Dienestrol
Nil
Nil
Dimethisterone
Nil
Yellow
Mestranol
Yellow
Orange (pink in daylight)
Dexamethasone
Nil
Nil
Prednisolone
Nil
Green (red in daylight)
Beclometasone (slow)
Nil
Nil
Cholesterol
Nil
White
Dydrogesterone
Green–yellow
Green–yellow
Fludrocortisone
Green
Green (dichroic in daylight)
Norethandrolone
Green–yellow
–
Norethisterone
Orange
Orange (violet in daylight)
Norethynodrel
Orange
Orange
Oxymetholone
Nil
Nil
Spironolactone (!yellow–green)
Yellow–green
Green
Diethylstilbestrol
Nil
Nil
Triamcinolone
Nil
Nil!green (slow)
Red–orange
Ethinylestradiol
Orange
Orange (red in daylight)
Pink–orange
Betamethasone
Nil
Nil
Green, orange (dichroic)
Hydrocortisone
Green
Green
Alfadolone
Nil
Nil
Androsterone
Nil
White
Carbenoxolone
Nil
Yellow
Cortisone
Green
Green
Desoxycortone
Green–yellow
Yellow (violet in daylight)
Red Orange–red
Pink Orange–pink Orange
Yellow
Enoxolone
Nil
Green–yellow
Fluocinolone
Green
Quenched
Fluoxymesterone
Green
Quenched
Fludroxycortide
Green
Quenched
Hydroxyprogesterone
Green
Quenched
Estradiol
Green
Green (orange in daylight)
Prednisone
Green
Green
Progesterone
Green
Quenched
Orange–yellow
Fluocortolone
(Weak)
(Weak)
Green–yellow
Estrone
Green
Green–yellow (orange in daylight)
Estriol
Yellow–green
Quenched (orange in daylight)
Testosterone
Green
Nil
No colour
phenomenon that depends on the structure of the side-chain at the 5-position. Dark colours, which are either dispelled or changed to violet, blue or green by dilution, are produced when either side-chain is greater than two carbon atoms in length or contains a cycloalkene ring. Branching can be proximal to the pyrimidine ring, but not distal. No colour is obtained if both side-chains are less than three carbon atoms in length or if either is branched distally or contains an aryl nucleus. Long, straight, saturated chains also appear to hinder reaction. Hydroxybarbiturates give positive responses (Table 30.34), but bemegride, glutethimide, phenytoin and primidone do not respond. No response is obtained with amobarbital, aprobarbital, barbital, butobarbital, enallylpropymal, hexethal, ibomal, idobutal, metharbital, methylphenobarbital, nealbarbital, phenobarbital or phenylmethylbarbituric acid. With cold reagent, an orange colour is produced by pentobarbital, secobarbital and thiopental, and a brown colour by cyclopentobarbital.
Zwicker reagent (alkaline cobalt test) This is a general test for barbiturate-like compounds. Reagent
1. Dissolve 0.5 g of copper(II) sulfate pentahydrate in 100 mL of distilled water. 2. Add 0.5 mL of pyridine to 95 mL of chloroform. Method
Add a few drops of solution 1 to the sample to be tested, followed by a few drops of solution 2 and then heat. Indications
The presence of a violet–blue colour indicates barbiturates (Table 30.35). The LOD is 1000 mg for phenobarbital.
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:51
490
Colour Tests
Table 30.33 Colours of thioxanthenes with sulfuric acid (a)
Colour
Compound
Purple
Oxytetracycline, tetracycline
Red
Caramiphen (when warmed), dantron, fenitrothion, methenamine (when warmed), mequitazine (slowly at 100 C), methacycline, methylprednisolone (after 1 min), metopimazine, nuarimol, ouabin, phenothiazines, prednisolone (after 1 min), pipoxolan
Table 30.34 Reactions of barbiturates with vanillin reagent Colour after heating
Compound
Colour after dilution
Red
30 -Hydroxybutobarbital
Violet (transient)
Violet–red Brown–red
Heptabarbital
Colourless
30 -Hydroxyamylobarbital
Colourless
Cyclobarbital
Green
Orange–red
Oxprenolol*, quinomethionate
Cyclopentobarbital
Green
Violet–red
Morantel, oxytetracycline
Pentobarbital
Violet
Pink
Doxylamine, indapamide (slow)
Secobarbital
Violet
Orange
Alprenolol, amitriptyline, benactyzine, benzilonium, benzquinamide, benzyl nicotinate, chlorprothixine*, clidinium, clopenthixol*, cyclothiazide (!red–brown), diethylthiambutene, diphemanil, diphenhydramine, diphenidol, doxepin, flupentixol*, indometacin, mazindol, mebanazine, mecoprop, methapyrilene, methindizate, methixene*, methyclothiazide, methylpiperidyl benzilate, naproxen, nefopam, nifedipine, nortriptyline, orphenadrine, penthienate methobromide, polythiazide, pyrantel (!violet), rotenone, tiotixene*, tofenacin
Thiopental
Violet
Butalbital (weak)
Colourless
Yellow
Orange–yellow Green
Acebutolol, amiloride, amiodarone, benzthiazide, broxaldine, broxyquinoline, cinchophen, clefamide, clemastine (green rim), cyclopenthiazide, diphenadione, doxycycline, enoxolone, fenbufen, furosemide, halquinol, hydroquinidine*, hydroquinine*, lorazepam, methyl benzoquate, 5-methyltryptamine, a-methyltryptamine, N-methyltryptamine, metolazone, minocycline, piperacetazine (!red), procyclidine, quinidine*, quinine*, rimiterol, salbutamol, salinazid, sodium cromoglicate, trichlormethiazide, veratrine (!violet), zomepirac
Orange Brown–orange
Brown Violet–brown
Secbutobarbital (weak)
Violet
Allobarbital
Violet (transient)
Brallobarbital
Brown–orange
Talbutal
Violet
Thialbarbital
Violet (transient)
Hexobarbital
Violet
Methohexital
Colourless
Vinbarbital
Colourless
30 -Hydroxypentobarbitone
Colourless
Table 30.35 Colours with Zwicker reagent Colour
Compound
Blue
Diphenylhydantoin
Ethyl biscoumacetate, hexoprenaline, pizotifen (!violet)
Green
Diacetylmorphine, pseudoephedrine
Bromodiphenhydramine, cyclizine, diphenhydramine, diphenylpyraline, phenothiazine, protriptyline
Yellow
Tetracycline
Blue Brown–blue Violet
Red–violet Brown Red–brown
Chlortetracycline, demeclocycline, thioridazine Bendroflumethiazide, chlorotrianisene, chromonar*, clofazimine, cyproheptadine, dosulepin, mesoridazine (!blue), methylenedioxyamfetamine, nicergoline, perazine, phenindione, rolitetracycline, tetracycline Trifluomeprazine Chelidonine, sulindac Lymecycline
Orange–brown
Biperiden, ouabain (slow)
Yellow–brown
Tylosin (slow)
Grey–brown
Octaphonium
Black Blue–black
Clomocycline
(a) Compounds giving colours that fluoresce under UV light (l = 350 nm) are indicated by an asterisk.
Basic tests for drug substances and products The WHO has published texts Basic Tests for Drugs (WHO 1998), which includes pharmaceutical substances, medicinal plant materials and dosage forms, and Basic Tests for Pharmaceutical Substances (WHO 1986). The basic tests described, which are designed to verify the identity of drug substances and medicinal products and to detect gross contamination, use a limited number of readily available reagents and equipment. Overall, the combined texts offer compound-specific tests for approximately 500–600 products that are based on a combination of organoleptic checks and simple physicochemical tests, such as colour reactions and melting-point determinations.
Semi-quantitative TLC methods have been developed as basic tests using a limited number of solvent systems and detection systems. References to these tests are given in (WHO 1998). It should be remembered that basic tests are not, in any circumstances, intended to replace pharmacopoeial requirements, but should be used as a rapid, inexpensive means to verify identity and strength of drugs and medicinal products, and possibly to detect poor-quality counterfeit and other substandard products. In the event that suspect products are detected, these should be tested for compliance against pharmacopoeial requirements. Colour reagents and thin-layer chromatography Many common colour-test reagents are used routinely as spray locating reagents in TLC (e.g. acidified iodoplatinate solution, Dragendorff’s reagent, Marquis reagent, Van Urk’s reagent). It is worth noting that the preparation of the spray equivalent of a colour reagent may differ slightly from that of the colour reagent preparation itself (see Chapters 1 and 11 and Index of Reagents). It is also true that the reaction and resultant colour that marks the presence of a certain substance obtained from spraying a reagent on TLC plates may differ from that obtained from a direct colour test and, in some cases, will not yield any results. This is because of pH effects – that is, whether the TLC plate has been dipped in 0.1 mol/L sodium hydroxide and the substance tested is acidic in character (i.e. the free acid of a salt post extraction). However, this aspect of colour reagents and their use in TLC as sprays can often give clues as to the drug or substance’s chemistry. A list of substances and their colour reactions to various TLC spray reagents is given in the Index of Analytical Data.
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:52
Appendix 30.2. Colour reactions of narcotics
Appendix 30.1. Colour reactions of amfetamine-like compounds Compound
Marquis
Mecke
Froehde
Mandelin O!R!G/Br
D-Amfetamine
R/O!R/Br
Y
Css
Benzfetamine
R!Br
G!Y
Y!G
Diethyltryptamine
Y!Br
B!G/Bk
2,5-Dimethoxyamfetamine
lrY!R/Br
Br/G
ltG
ltG
Dimethoxymethylamfetamine
Y/G
Y!G!dkR/Br
Y/G
Y!G!Br
2,3-Dimethylamfetamine
M R/V
V dkV
V Gy/V
V Gy/G
2,4-Dimethylamfetamine
V O/R
G G/Br
Y Br/Y
Y Br/Y
2,5-Dimethylamfetamine
Y/G
Br/G
Y/G
Y/G
2,6-Dimethylamfetamine
Css
Br/G
Css
Br/Y
3,4-Dimethylamfetamine
Y/Br
G!Br/G
Lt G
NR
3,5-Dimethylamfetamine
Y
O/Br!Br/G
Css
Gy/V
Dimethyltryptamine
Y!G!Br
Y!G/Bk
ltY
G!Br
Css
ltBr
Ephedrine
Css
Css
Mephentermine
O!RBr
G
Mescaline
O/R
O!G/Br
ltY
Y!G!Br
Metamfetamine
R/O!R/Br
NR
NR
G!B/G
3-Methoxy-4,5-methylenedioxyamphetamine
O!M
dkB
B!dkB
O!O/R
3,4-Methylenedioxyamfetamine
B/BK!dkV
G!dkB
G!dkV
R/V!dkV
3,4-Methylenedioxymethylamfetamine
B!V!Bk
G!dkB
Y/G!dkB
B!V!Bk
Paramethoxyamfetamine
Effervescence
ltG
G!R/Br
Phendiamine
V!Bk
ltG
ltV
G
Phendimetrazine
NR
NR
NR
NR
Pheniramine
NR
NR
NR
NR
Phenmetrazine
NR
ltY
NR
NR
Phentermine
ltO
ltO
Br
G
Phenylephrine
O
O/Br
B
G
Phenylpropano lamine
ltY
ltY
Br
NR
Pseudoephedrine
Css
Css
Css
ltBr
3,4,5-Trimethoxyamfetamine
R/O!O
G/Br!Br
Y
NR
B, blue; Bk, Black; Br, brown; Css, colourless; dk, dark; G, green; Gy, grey; lt, light; M, magenta; NR, no reaction; O, orange; R, red; V, violet; Y, yellow.
Appendix 30.2. Colour reactions of narcotics Compound
Marquis
Mecke
Froehde
Mandelin
Anileridine
O
NR
NR
NR
Codeine
V
dkG
ltG
ltG
Dihydromorphinone
V/Br
Br
V
Css
Ethylmorphine
O!V
dkG
Y!ltG!B
G
Fentanyl
O!Br
G
Diamorphine
V
ltG
M
NR
Hydrocodone
V
G!B
ltY
NR
dkB!Gy
Hydromorphine
Y!R!V
Y!O!G
Levorphanol
Gy!Bk
ltGy!Bk
B
G
Meperidine (pethidine)
O(slow)
ltY(slow)
NR
NR
Methadone
ltV
G!Br
B
Morphine
M!V
G!B
V!Gy
ltGy
Nalorphine
O!R!V
ltG!dkG
M
G
Oxycodone
O/Y!ltV
Y!dkG
Y!YBr
Y!G
Oxymorphone
M!V
Y!Br
B!V
Bk
Papaverine
M
dkG
G
G/Br
Pentazocine
M
ltGy!V
dkB
G!Br
Thebaine
V/Br
Br
Br
V/Br
B, blue; Bk, black; Br, brown; Css, colourless; dk, dark; G, green; Gy, grey; lt, light; M, magenta; NR, no reaction; O, orange; R, red; V, violet; Y, yellow.
491
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:52
492
Colour Tests
Appendix 30.3. Chemical test for gamma-hydroxybutyric acid (GHB) and gamma-butyrolactone (GBL)
Appendix 30.4. Tests for metals and anions not covered by the general scheme
Reagents
Metals
Chlorophenol red
Reinsch test (for antimony, arsenic, bismuth and mercury)
0.04 g chlorophenol red in 100 mL water, adjust to pH 7 with 0.01 mol/L sodium hydroxide.
Method
Modified Schweppes
1. 2 g of dextrose in 20 mL of water 2. 2.4 g of aniline hydrochloride in 20 mL of ethanol. Mix solutions 1 and 2 together and dilute to 80 mL with methanol. Bromocresol purple
0.04 g bromocresol purple in 100 mL of water. Adjust to pH 7 with 0.01 mol/L sodium hydroxide. Bromothymol blue
Use a 5 5 mm piece of copper foil or mesh. Clean the copper in 50% (v/v) nitric acid until it develops a shiny surface, rinse with water and proceed as follows: 1. Place 20 mL of sample and 10 mL of concentrated hydrochloric acid in a 100 mL conical flask and add the copper foil or mesh. 2. Heat on a boiling water-bath for 1 h and add as necessary dilute hydrochloric acid to maintain the volume of the solution. 3. Cool, remove the copper, wash gently with water and examine the surface. Indications
Bromocresol green
n n n n
0.03 g bromocresol green in 100 mL of methanol–water 4 : 1. Adjust to pH 7 with sodium hydroxide 0.01 mol/L.
Note that other elements, (e.g. selenium and tellurium) also give black deposits and sulfur compounds may give a speckled discoloration.
Methyl orange
Confirmatory test
0.04 g bromothymol blue in 100 mL of water. Adjust to pH 7 with 0.01 mol/L sodium hydroxide.
0.01 g methyl orange in 100 mL of methanol. Adjust to pH 7 with sodium hydroxide 0.01 mol/L.
A purple–black stain indicates antimony. A dull black stain indicates arsenic. A shiny black stain indicates bismuth. A silvery deposit suggests mercury.
This is applied to the stained copper foil or mesh derived as described above. Reagents
Colour test 1 Reagent
Mix chlorophenol red and modified Schweppes reagent in a 3 : 1 ratio. Method
Place 0.5 mL of a liquid sample in a test tube. Adjust to pH 5–8 with 0.01 mol/L sodium hydroxide. Add 2 drops of the test reagent and swirl. An immediate colour change (orange–red to dark red) indicates GHB. GBL gives a yellow colour. Colour test 2 Reagent
Mix bromocresol purple and bromothymol blue in a 1 : 1 ratio and mix the combined reagent with modified Schweppes reagent in a 7 : 1 ratio. Method
Same procedure as for colour test 1. GHB gives a purple colour and GBL gives a yellow colour. Colour test 3 Reagent
Mix bromocresol green and methyl orange in a 1 : 1 ratio and mix the combined reagent with modified Schweppes reagent in a 3 : 1 ratio. Method
Adjust the pH of the test solution to neutral if necessary and add 2 drops of test reagent. A dark-green colour indicates GHB; GBL gives a yellow– orange colour. Colour test 4 Reagent
1% cobalt nitrate solution. Method
Place 0.5 mL of a liquid sample in a test tube. Add a few drops of the test reagent. A pink-to-violet colour indicates GHB.
1. 2. 3. 4.
Aqueous potassium cyanide solution (100 g/L) Freshly prepared aqueous sodium sulfite solution (50 g/L) 3 mol/L nitric acid Quinine/potassium iodide reagent. Dissolve 1 g of quinine in 100 mL of water containing 0.5 mL of concentrated nitric acid. When the quinine has dissolved add 2 g of potassium iodide.
Method
1. Leave the copper in potassium cyanide solution for 10 min. 2. Wash any undissolved stain with water and add 1 mL of sodium sulfite solution followed by 1 mL of 3 mol/L nitric acid. 3. Shake the mixture frequently for 5 min and add 1 mL of water and 1 mL of potassium iodide reagent. Indications Stains caused by the presence of arsenic dissolve in potas-
sium cyanide solution whereas stains due to antimony and bismuth remain. The slow formation of an orange–brown suspension is seen with the quinine/potassium iodide reagent if a stain due to bismuth is present. The LOD for arsenic is about 5 mg/L and for antimony and bismuth is 2 mg/L. A more specific test for arsenic, the ‘Gutzeit test’, involves the conversion of arsenic to arsine and subsequent reaction of the gas with reagents such as silver diethyldithiocarbamate to give a coloured product. A modified version of the Gutzeit apparatus is available from Fischer Scientific which allows quantitative measurement of arsenic in stomach contents, food, water and other materials. Confirmatory test for mercury
This is applied to the silver stained foil or mesh from the Reinsch test. Reagent Cuprous iodide suspension. Dissolve 5 g of copper sulfate and 3 g of ferrous sulfate in 10 mL of water with continuous stirring and add 7 g of potassium iodide dissolved in 50 mL of water. Allow the cuprous iodide precipitate to form, filter, and wash with water. Transfer the precipitate as a suspension in water to a brown glass bottle. Method Add 0.1 mL of copper(I) iodide suspension to a filter paper, place the foil on the suspension, cover and leave for 1–12 h. Indications A salmon-pink colour due to the formation of cuprous mercuric iodide suggests the presence of mercury and positive results may appear within 1 h, but with low concentrations colour development may take up to 12 h. The LOD for mercury is about 5 mg/L.
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:53
Appendix 30.4 Barium
Thallium
Reagents
Test 1
1. Concentrated hydrochloric acid 2. Platinum wire.
Reagents
Method
1. Dip the end of the platinum wire into the hydrochloric acid and then into the test material. 2. Insert the wire into the hot area of a micro-burner flame and observe any changes in the flame colours. Indications
Barium salts impart a green flame; copper and thallium salts also give a green flame in this test.
1. Cyanide reagent: dissolve 1.6 g of sodium hydroxide, 1.2 g of potassium sodium tartrate and 1.36 g of potassium cyanide in 10 mL of water. 2. Prepare a fresh solution of dithizone (250 mg/L) in chloroform. Method Add 1 mL of cyanide reagent to 5 mL of urine in a stoppered glass test-tube and vortex mix for 20 s. Then add 2 mL of dithizone solution, vortex mix for 1 min and centrifuge (5 min). Indications The presence of thallium is indicated by a pink–red colour in the chloroform layer. The test will detect thallium at 0.1 mg/L. A number of other metal ions give colours with this test.
Confirmatory test
Test 2
Reagents
Reagents
1. 2. 3. 4.
1 mol/L sulfuric acid 100 g/L aqueous lead acetate solution 50 mL/L aqueous acetic acid Solid ammonium acetate.
Method
1. To a mixture of 2 mL of lead acetate solution and 2 mL of dilute sulfuric acid add enough ammonium acetate to dissolve the lead acetate precipitate. 2. Add 0.1 mL of dilute acetic acid to 1 mL of sample followed by 1 mL of the lead sulfo-acetate solution (from step 1) and vortex mix for 5 s. 3. Centrifuge for 2 min and observe the tube against a dark background. Indications Barium salts yield either a white turbidity or white precip-
itate. Calcium and strontium salts interfere. The LOD for barium is approximately 100 mg/L. Copper Reagents
1. 10 g/L solution of dithiooxamide in methanol 2. Concentrated ammonium hydroxide. Method
Place 0.1 mL of sample onto a filter paper to give a spot of around 1 cm in diameter. Expose the paper to ammonia fumes and add 0.1 mL of the dithiooxamide reagent to the spot. Indications
An olive green stain is seen with copper salts. A green stain due to chromium is usually visible before addition of the dithiooximide reagent. Yellow–brown or yellow–red colours are given by several other metals. The LOD for copper is approximately 1 mg/L.
493
1. 2. 3. 4. 5.
Bromine water (saturated) 6 mol/L hydrochloric acid 20% (w/v) aqueous sulfosalicylic acid solution 0.1% (w/v) aqueous methyl violet solution Toluene.
Method To 1 mL of urine carefully add 2 drops of hydrochloric acid and 5 drops of bromine water. Leave to stand for 5 min, add 5 drops of sulfosalicylic acid solution and 0.5 mL of toluene, and shake gently. Indications A transient blue–green colour in the toluene layer suggests the presence of thallium. The LOD for thallium is 1 mg/L.
Anions Borate Reagents
1. 10 g/L solution of turmeric in methanol 2. 1 mol/L hydrochloric acid 3. 4 mol/L ammonium hydroxide. Method
1. Prepare turmeric test papers by soaking strips of filter paper (1 5 cm) in the turmeric solution and drying at room temperature. 2. Acidify a portion of the sample with the dilute hydrochloric acid and apply to a strip of turmeric paper. 3. When the paper is dry moisten it with dilute ammonium hydroxide. Indications
A positive sample will impart a red–brown colour to the turmeric paper which strengthens as the paper dries. When the paper is moistened with dilute ammonium hydroxide a green–black colour is produced. Oxidising agents that bleach turmeric (e.g. bromates, chlorates, iodates and nitrites) interfere with the test. The LOD for borate is 50 mg/L. Confirmatory test
Iron
Reagent 0.5 g/L solution of carminic acid in concentrated sulfuric acid.
Reagents
Method Add 0.5 mL of filtered stomach contents or scene residue to a
1. 2 mol/L hydrochloric acid 2. 10 g/L aqueous potassium ferricyanide solution 3. 10 g/L aqueous potassium ferrocyanide solution. Method
1. Add 0.1 mL of 2 mol/L hydrochloric acid and 0.05 mL of potassium ferricyanide solution to 0.1 mL of sample. 2. Add 0.1 mL of 2 mol/L hydrochloric acid and 0.05 mL of potassium ferrocyanide solution to a further 0.1 mL of sample. 3. Agitate both mixtures for 5 s, leave for 5 min at room temperature and centrifuge for 5 min. Indications
Ferrous salts give a deep blue precipitate with potassium ferricyanide and ferric salts give a deep blue precipitate with potassium ferrocyanide. The LOD for both ferrous and ferric salts is about 10 mg/L.
10 mL glass tube and slowly pour 0.5 mL of carminic acid solution down the inside of the tube so that a layer is formed underneath the sample. Indications Borate is indicated by the formation of a blue–violet ring at the junction of the two layers. Note that strong oxidising agents (e.g. bromates, chlorates, iodates and nitrites) also give positive results. Bromides Reagents
1. 2 mol/L nitric acid 2. 10 g/L solution of silver nitrate 3. Concentrated ammonium hydroxide. Method
1. To 1 mL of clear tests sample add 0.5 mL of 2 mol/L nitric acid and mix for 5 s.
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:53
494
Colour Tests
2. Add 0.1 mL of silver nitrate solution. 3. Centrifuge any precipitate and decant off the supernatant. 4. Add 0.1 mL of concentrated ammonium hydroxide.
3. Calcium chloride (solid) 4. Powdered silica. Method
1. Saturated solution of fluorescein in aqueous acetic acid (600 mL/L) 2. Concentrated sulfuric acid 3. Solid potassium permanganate.
1. To 5 mL of sample in a porcelain crucible add 100 mg of calcium chloride and gently evaporate to dryness over a burner. 2. Destroy the organic material by strong heating to leave a white ash. 3. Mix the residue with 200 mg of powdered silica. 4. Apply a drop of sodium chloride solution to a microscope slide, add 1 mL of concentrated sulfuric acid to the contents of the crucible and immediately position the slide such that the sodium chloride drop is suspended over the crucible. 5. Rest a small beaker of ice on the slide and heat the crucible gently for 5 min over a burner. 6. Remove the slide after 5 min and examine the sodium chloride solution under a microscope.
Method
Indications In the presence of fluorine-containing compounds, silicon
Indications
Bromide gives an off-white precipitate that is slightly soluble in ammonium hydroxide. A white precipitate that dissolves in ammonium hydroxide indicates chloride and a yellowish precipitate insoluble in ammonium hydroxide suggests iodide. The LOD for bromide is 50 mg/L. Confirmatory test Reagents
1. Soak a strip of filter paper in fluorescein solution. 2. Transfer 2 mL of test solution to a 10 mL glass tube and add about 2 mg of potassium permanganate. 3. Add 0.2 mL of concentrated sulfuric acid and hold the fluorescein paper at the mouth of the tube. Indications Any bromide is oxidised to free bromine, which then reacts
with the fluorescein dye (yellow) to give tetrabromofluorescein (eosin) which has a pink–red colour. The LOD for bromide is 50 mg/L. Chlorate Chlorates and other oxidising agents can be detected by the diphenylamine test described previously. The following tests can also be applied. Test 1 Reagents
1. 6 mol/L sulfuric acid 2. 1% w/v aqueous solution of indigo carmine 3. Solid sodium sulfite. Method To 1 mL of sample add 4 mL of 6 mol/L sulfuric acid followed by 1 mL of indigo carmine reagent. Indications A deep blue colour indicates the presence of chlorates. The colour fades after adding several crystals of sodium sulfite. The same reactions are given by bromates and hypochlorites.
tetrachloride is produced, which dissolves in the sodium chloride solution to form sodium silicon tetrafluoride. This forms small hexagonal crystals as the water evaporates from the slide, which sometimes have a pink hue. The crystals are seen at the edge of the drop and appear before the larger cubic crystals of sodium chloride. Test 2 Reagents
1. Concentrated sulfuric acid 2. Calcium hydroxide (solid) 3. Paraffin wax. Method
1. Repeat steps (1) and (2) described for test 1. 2. Smear a film of paraffin wax on one side of a glass microscope slide and expose part of the surface by making an identifiable sign on the paraffin film. 3. Add 5 mL of concentrated sulfuric acid to the crucible and cover it with the slide, with the paraffin film on the inside. 4. Heat the crucible gently for 20 min and then remove the slide. 5. Remove the paraffin film with toluene and examine the slide. Indications Hydrogen fluoride is generated from fluorine-containing compounds and etches the glass to give a mark corresponding to that made in the paraffin film. Both tests have a LOD of approximately 100 mg/L of fluoride.
Test 2 Reagents
1. Manganous sulfate reagent: a 1 : 1 mixture of saturated aqueous manganous sulfate and orthophosphoric acid 2. A 10 g/L solution of diphenylcarbazide in methanol. Method
1. Add 0.2 mL of manganous sulfate reagent to 0.1 mL of test sample and warm the mixture gently over a burner. 2. Cool and add 0.1 mL of diphenylcarbazide solution. Indications Chlorate yields a purple colour that intensifies after cooling
and adding diphenylcarbazide. A similar reaction is given by persulfates and periodates. Note: after ingestion of chlorate the blood turns brown owing to the formation of methaemoglobin by oxidation of the ferrous ion of haemoglobin. Add 0.2 mL of a 10% (w/v) of aqueous potassium cyanide solution to 1 mL of blood and an immediate red colour confirms the presence of methaemoglobin. Exposure to a wide range of other substances (e.g. aniline, nitrites, nitrates, aniline, dapsone, benzocaine, urea herbicides) causes methaemoglobinaemia.
Hypochlorite Test 1 Reagents
1. Glacial acetic acid 2. 50 g/L aqueous lead acetate solution. Method
1. To 1 mL of test solution add acetic acid dropwise to reach a pH of approximately 6 (test with universal indicator paper). 2. Add 0.5 mL of lead acetate solution and boil for 3 min. Indications Hypochlorite forms a brown precipitate. An immediate
brown to black precipitate is given by sulfides with lead acetate. Test 2 Reagents
1. 100 g/L aqueous potassium iodide solution 2. Glacial acetic acid 3. Solid starch. Method
Test 1
1. To 0.1 mL of test solution add 0.1 mL of acetic acid followed by 0.1 mL of potassium iodide solution. 2. Mix and add about 20 mg of starch.
Reagents
Indications Hypochlorite gives a blue colour. (Note: hypochlorite also
1. A 50 g/L aqueous solution of sodium chloride 2. Concentrated sulfuric acid
gives a positive reaction in the diphenylamine test for oxidising agents described above.)
Fluoride
Clarke's Analysis of Drugs and Poisons Chapter No. 30 Dated: 10/3/2011 At Time: 13:2:54
Appendix 30.4 Iodides Reagents
1. 10 g/L aqueous silver nitrate solution 2. 2 mol/L nitric acid 3. Concentrated ammonium hydroxide. Method
1. To 1 mL of clear test solution add 0.1 mL of nitric acid and 0.1 mL of silver nitrate solution. 2. Centrifuge down any precipitate, remove the supernatant and add to it 0.1 mL of ammonium hydroxide. Indications
A curdy yellow precipitate that is insoluble in ammonium hydroxide is given by iodides. Chlorides give a white precipitate that dissolves in ammonium hydroxide, and an off-white precipitate that is sparingly soluble in ammonium hydroxide suggests bromides. Confirmatory test
495
2. To 1 mL of test sample in a test-tube add 3 mL of sulfuric acid and insert the lead acetate paper into the neck of the tube. 3. Heat the tube in a boiling water-bath for 5–10 min. Indications
Hydrogen sulfide fumes that turn lead acetate paper black are produced by sulfides. The LOD for sulfide is 50 mg/L. Thiocyanates Reagent
A 50 g/L aqueous solution of ferric chloride. Method
Mix 0.5 mL of ferric chloride solution with 0.5 mL of sample. Indications
A deep red colour is given by thiocyanates. The LOD for thiocyanate is 50 mg/L.
Reagents
1. 2 mol/L hydrochloric acid 2. 100 g/L freshly prepared sodium nitrite solution 3. Solid starch. Method Mix thoroughly about 20 mg of starch with 0.1 mL of test solution, 0.1 mL of hydrochloric acid and 0.1 mL of sodium nitrite solution in a test-tube. Indications A blue colour confirms the presence of iodide.
Oxalates
References Asselin WMA, Caughlin JD (1990). A rapid and simple colour test for detection of salicylate in whole hemolyzed blood. J Anal Toxicol 14: 254–255. Caughlin JD (1991). A rapid colour test for detection of ethchlorvynol in whole hemolyzed blood. Can Soc Forensic Sci J 24: 111–114. O’Neal CL et al. (2000). Validation of twelve chemical spot tests for the detection of drugs of abuse. Forensic Sci Int 109: 189–201. WHO (1986). Basic Tests for Pharmaceutical Substances. Geneva: World Health Organization. WHO (1998). Basic Tests for Drugs: Pharmaceutical substances, medicinal plant materials and dosage forms. Geneva: World Health Organization.
Reagents
1. 100 g/L aqueous calcium chloride solution 2. 30% (v/v) solution of acetic acid 3. 2 mol/L hydrochloric acid. Method
1. Add 1 mL of calcium chloride solution to 2 mL of test solution and mix. 2. If a precipitate forms, add 1 mL of acetic acid. 3. If the precipitate does not dissolve, separate it by centrifugation and add 1 mL of dilute hydrochloric acid. Indications
A white precipitate that is insoluble in acetic acid but dissolves in hydrochloric acid indicates the presence of oxalates. Confirmatory test Reagents
1. Thiobarbituric acid 2. Concentrated ammonium hydroxide. Method
1. Add 50 mL of test solution to 100 mL of ammonium hydroxide in a micro test-tube and mix thoroughly. 2. Gently evaporate the mixture to dryness over a micro-burner. 3. Add about 200 mg of thiobarbituric acid and reheat gently to about 150 C. Indications Oxalates give a bright red product that is soluble in methanol. The LOD for oxalate is 250 mg/L. Sulfides Reagents
1. 10% (v/v) aqueous solution of sulfuric acid 2. 10% (w/v) solution of lead acetate in boiled and purified water 3. 2 mol/L acetic acid. Method
1. Immerse a strip of white filter paper in a mixture of 10 volumes of lead acetate solution and 1 volume of acetic acid and allow to dry.
Further reading Bamford F (1951). Poisons, Their Isolation and Identification, 3rd edn. London: Churchill. Bentley KW (1954). The Chemistry of Morphine Alkaloids. Oxford: Clarendon Press. Clarke EGC (1962). The isolation and identification of alkaloids. In: Lundquist F, eds. Methods of Forensic Science, Vol. 1. London: Wiley, 1–241. Enders PW (1985). A simple color test on quaternary ammonium compounds. In: Brandenberger H, Brandenberger R, eds. Reports on Forensic Toxicology. Mannedorf: Branson Research, 195–198. Fiegl F (1966). Spot Tests in Organic Analysis, 7th edn. Amsterdam: Elsevier. Fiegl F, Anger V (1972). Spot Tests in Inorganic Analysis, 6th edn. New York: Elsevier. Flanagan RF et al. (1995). Basic Analytical Toxicology. Geneva: World Health Organization. Gonzales TA et al. (1954). Colour reactions for the identification of non-volatile organic poisons. In: Legal Medicine, Pathology and Toxicology, 2nd edn. New York: Appleton–Century–Crofts, 1191–1255. Johns SH et al. (1979). Spot tests: a colour chart reference for forensic chemists. J Forensic Sci 24: 631–649. Johnson CA, Thornton-Jones AD, eds. (1966). Drug Identification. London: Pharmaceutical Press. Kaye S (1980). Handbook of Emergency Toxicology, 4th edn. Springfield, IL: Charles C Thomas. Musshoff F et al. (2000). Hallucinogenic mushrooms on the German market – simple instructions for examination and identification. Forensic Sci Int 113: 389–395. Saker EG, Solomons ET (1979). A rapid inexpensive presumptive test for phencyclidine and certain other cross-reacting substances. J Anal Toxicol 3: 220–221. Sangalli BC (1989). A new look at qualitative toxicology. Spot tests in the emergency department. Vet Hum Toxicol 31: 445–448. Stair E, Whaley M (1990). Rapid screening and spot tests for the presence of common poisons. Vet Hum Toxicol 32: 564–566. Sunshine I, ed. (1975). Methodology for Analytical Toxicology. Cleveland: CRC Press. US Department of Justice (1978). NILECJ Standard for Chemical Spot Tests for Preliminary Identification of Drugs of Abuse. Washington DC: US Department of Justice. US Department of Justice (1981). NILECJ Standard for Chemical Spot Tests for Preliminary Identification of Drugs of Abuse. Washington DC: US Department of Justice.
Clarke's Analysis of Drugs and Poisons Chapter No. 31 Dated: 11/3/2011 At Time: 8:29:59
CHAPTER
31
Immunoassays RS Niedbala and JM Gonzalez
Introduction The first immunoassay principles were published decades ago (Yalow, Berson 1959). Since that time immunoassays have become a routine part of day-to-day life for scientists in various disciplines as well as for general consumers. For example, every over-the-counter pregnancy and fertility test is based upon immunoassay results. The impact has been even greater in broader fields where analysts use immunoassays for the routine screening of samples for a host of target analytes. Fields such as veterinary medicine, environmental testing, anti-terrorism and human clinical diagnostics all use various types of immunoassays (Huckle, Wild 2005). For instance, all the applications that exist, immunoassays have limitations that are generally created by the antibodies used. Antibodies may have poor specificity that results in cross-reactivity with structurally similar compounds, creating false results. Antibodies may also be affected by the matrix used. For example, hair testing for drugs of abuse is challenged by the treatments that consumers use on their hair and scalp. It is also difficult to generate antibodies reliably against target compounds. It requires a ‘make it and test it’ scenario that can take years. Nevertheless, immunoassays have become an integral part of routine clinical testing. When developed carefully, immunoassays can detect minute amounts of target analyte in complex samples. They can be robust and capable of working in harsh conditions along the roadside or in remote areas. Immunoassays will remain an important tool for anyone who needs to detect analytes reliably in a cost-effective and rapid manner. The focus of this chapter will be to explain the principles of immunoassays and practical considerations in applying them in the field of toxicology. Immunoassays can generally be constructed for target molecules with molecular weights as low as 250, as well as for large, complex proteins and cellular components. Each immunoassay is formatted to the usage situation and its analytical requirements. Some are designed to be highly sensitive in detecting and quantifying extremely small amounts of target analyte. Others are designed only for qualitative analysis to detect the presence or absence of analytes. Thus, researchers and developers have refined immunoassays to become reliable, robust, accurate tests that can be manufactured on a large scale. Parallel to its development, and in response to it, societies have embraced the use of immunoassay technologies. For example, many countries have enacted legislation defining cut-offs as well as specific targets for immunoassays aimed at the detection of drugs of abuse in various situations where individuals or the general public may be at risk (US Department of Health and Human Services 2006; US Department of Transportation 2000). These legislated assay parameters regulate the implementation of millions of tests that use urine, saliva, hair or blood as matrices. The results from an immunoassay test followed by confirmation using alternative technologies can be used as forensic evidence in a court of law. In several countries individuals are tested for drug use prior to starting a new job, for suspicion of use during employment (Chapter 3) or following treatment or incarceration. Other countries are seeking to allow police to perform roadside testing of suspected drivers who may be under the influence of drugs of abuse (Parliament of Victoria, Australia 2003) (Chapter 5). Efforts are under way to develop and validate robust on-site immunoassays that can meet specific requirements of law and technical performance. Although they are not 496
yet available for all desired target drugs, much progress has been made (Maes et al. 1999; Moeller et al. 1999; Samyn et al. 1999b; Verstraete, Puddu 2000; Walsh et al. 2004). Immunoassays may take a variety of formats, although they all contain common elements regardless of design (Table 31.1) The first element is an antibody that has been targeted against a specific analyte. A target analyte can be a large molecule, an infectious agent or a small molecule that can elicit an immune response. The quality of the antibody will ultimately determine the potential performance of any immunoassay. The antibody is relied upon to be specific for the desired target analyte, insensitive to the test matrix being used, and stable over long periods of storage when packaged into an immunoassay test kit. The second element in an immunoassay is the reporter that will generate a signal that can be used to determine the immunoassay results. A variety of systems based on radioactive, enzymatic and chemiluminescent labels as well as colloidal particles are available for this task (Table 31.2). Each has advantages and disadvantages in immunoassay design and use. These strengths and weaknesses will be discussed later in this chapter. Once a quality antibody and a reporter have been identified, the platform for performing the immunoassay may be selected. There are many commercial platforms that are proprietary to corporations. However, basic researchers can utilise microtitre plates or rapid test formats such as lateral flow (Peace 2000; Niedbala et al. 2000; Perrigo, Joynt 1995; The Walsh Group 2002; Verheijen 2002; Volkov et al. 2009). Finally, the buffers and packaging for any immunoassay must meet the requirements for stability and storage of the test. Buffers also serve another key purpose, which is to prepare the sample matrix to be tested for compatibility with the antibody and test format being used. Various issues confront an immunoassay test developer when trying to use samples as diverse as urine or saliva. Each matrix has unique characteristics that can interfere with an immunoassay causing falsenegative or false-positive results. Immunoassay developers have the challenge first of developing and then of thoroughly validating any test before it is used by collaborative researchers or the general public. When specifically evaluating the performance of a toxicology assay it is worth examining what makes a good immunoassay test. First of all the purpose of the test should be considered. For instance, is the test to be used for qualitative screening or for monitoring concentrations of a particular drug in biological fluids? The difference in analytical requirements between, for example, a test for therapeutic drug monitoring (TDM) purposes and a qualitative screening test can be substantial. A TDM test is performed to optimise the level of drug therapy and therefore the immunoassay test result must be very accurate. In some cases, changing the dose of the drug incorrectly can result in serious or fatal outcomes (Baselt 2005; Hardman et al. 2001; Porter 2006; Uhlenhuth et al. 1990) (Chapter 2) Conversely, a screening test must simply detect the presence or absence of a target compound or a class of compounds reliably above or below a designated cut-off concentration. In such tests the cut-off concentration is specified; however, the actual performance of the test will have some variation around the cut-off. It is worth discussing two aspects of immunoassay design and performance. The terms sensitivity and specificity are often used and require some explanation. These terms relate to analytical sensitivity/specificity or clinical sensitivity/specificity. The term ‘analytical sensitivity’ is used to describe the assay’s ability
Clarke's Analysis of Drugs and Poisons Chapter No. 31 Dated: 11/3/2011 At Time: 8:30:0
Basic principles and issues of immunoassays
Table 31.1 Elements of an immunoassay Element
Purpose
Representation
Antibody
Targets analyte
497
principles described below should allow researchers to achieve optimal performance for any immunoassay they may develop or evaluate.
Basic principles and issues of immunoassays Antibody development and production
Reporter
Amplifies result
Hapten derivative
Drug derivative to link to reporter
Analyte
Target for assay
Buffers
Conditions, pH Sample
Solid phase
Location to immobilise antibody
Matrix
Describes type of sample being analysed
to detect the lowest level of target drug while ‘analytical specificity’ describes how precise it is at targeting a compound. ‘Clinical sensitivity’ describes how reliably a test identifies positives among a tested population, while ‘clinical specificity’ is a measure of whether or not identified positives are truly positive. For example, in the case of a screening test for drugs of abuse, a test that was clinically 100% sensitive and specific would have correctly identified all presumptive positives and all of them would have been confirmed positive for the target drug by an alternative and completely specific technique. The reality is that no screening immunoassay test is 100% accurate in identifying positives or negatives. In the case of drugs of abuse assays, positives are confirmed by other techniques such as gas chromatography–mass spectrometry (GC-MS). This is often expensive, but necessary, since antibody-based immunoassays are often subject to interferences or lack target analyte specificity. A well-designed screening immunoassay will therefore have a high degree of clinical specificity, meaning that positive immunoassay results are likely to be confirmed quickly and cheaply by GC-MS, thereby minimising costs by identifying samples that do not contain the analyte of interest. A final consideration before exploring the specifics of immunoassay is the metabolism and disposition of any target analyte drug and the matrix in which it is to be detected or quantified. Toxicological assays are increasingly performed on a variety of matrices including hair, saliva, blood or urine. The reason for choice of matrix may be ease of collection (saliva), a long window of detection (hair), compliance with regulations (urine) or forensics (stool or blood). Prior to developing an assay, it is critical to know which metabolites are the most prevalent and reliable as target analytes in a given type of sample. References are available to help any immunoassay developer outline their strategy for development (Baselt 2005). As immunoassay use has expanded, so have the tools and the understanding of the principles for developing them. Application of the Table 31.2 Various reporters used in immunoassays Reporter
Sensitivity achieved in immunoassay (molecules detected)
Enzymes
107
Fluorescence
106
Chemiluminescence
105
Radioimmunoassay
105
The development of an immunoassay for opiates (diamorphine, morphine, etc.) serves as a good example with which to describe in detail the principles and issues of immunoassay development. Diamorphine, morphine and related opiates are used throughout the world and their benefits and deleterious effects on health are well known (Baselt 2005; Hermes 1993). However, developing an immunoassay to identify specific opiates can be tedious and requires an appropriate strategy. The first step when developing such an assay is to consider the target molecule and the human matrix that will be tested. Diamorphine and morphine have been thoroughly studied and their pharmacokinetics are well understood (Moore et al. 1984). Figure 31.1 shows the metabolism of diamorphine and the major metabolites formed. It can be seen that morphine is a primary metabolite in urine, saliva and blood. It also has a relatively long half-life and is known to be stable in biological matrices. Therefore, morphine becomes the target analyte against which antibodies will be needed. Antibodies are proteins in mammals with a primary purpose to fight infection. They are generated by beta-lymphocytes following exposure to an immunogen. Immunogens are materials such as foreign proteins or cells that trigger the immune response. There are several antibody subtypes including IgG, IgA, IgM, IgE and IgD. Each functions within various compartments of the human body and may act to attack new infections or as sentinels if an infection reappears. For our purpose, antibodies are deliberately generated by introducing a designed immunogen (or antigen) into a host cell or animal. Beta cells within the host respond to the antigen by producing antibodies with affinity for the target antigen. Antibodies may be generated using a number of methods. The two major techniques produce monoclonal or polyclonal antibodies (Howard, Kaser 2007; Levine 2003). Polyclonal antibodies are often produced in rabbits, sheep, chickens and even llamas (Frenken et al. 2000). Each species has particular characteristics. Sheep, for example, may be bled to yield substantial quantities of blood for large-scale needs. Chickens are easy to use since antibodies may be isolated from eggs. No matter what the choice of species, the disadvantage of polyclonal antibodies is that there may be a mixture of antibody subtypes isolated with varying performance. In contrast to polyclonal antibodies, monoclonals are produced following fusion of a polyclonal cell with bacterial cell lines (Kohler, Milstein 1975). The fused line will produce a highly specific antibody that is immortalised in the cell line. Thus, supply is never an issue as long as the cell line is carefully maintained. Newer methods that are alternatives to traditional polyclonal and monoclonal antibody production include phage display as well as targeted engineering of protein domains with antibody-like attributes (Binz et al. 2005; Chiswell, McCafferty 1992; Ryan 2003). These newer techniques have primarily been used against large proteins and have not yet been developed sufficiently for use against small drug molecules. Following the example of morphine, an immunogen must first be developed that will be used to trigger production of antibodies. Morphine is a small molecule and, by itself, is not immunogenic. Usually a compound must have a molecular weight greater than 2000 before it will trigger an immune response. To solve the problem of immunogenicity, a morphine derivative is first linked to a carrier protein by a process known as haptenisation. The aim of this haptenisation process is to conjugate multiple, chemically modified, or derivatised, drug molecules (the hapten) to the carrier protein. Figure 31.2 shows the structure of morphine. Examining the chemical structure, it can be seen that morphine has amino or hydroxyl groups that can be used as sites for conjugation. The specificity of the antibodies generated can be determined by the position on the molecule used for conjugation. The site of conjugation is usually hidden from the immune system, so that changes made to the molecule at this position will have no effect on antibody binding. In the case of morphine, if an
Clarke's Analysis of Drugs and Poisons Chapter No. 31 Dated: 11/3/2011 At Time: 8:30:0
498
Immunoassays O CH3C N
HO
CH3
N
O
CH3
O
CH3C
CH3C
O
O
heroin
6-acetyl morphine Glucuronide
HO
HO NH
O
HO normorphine
N
CH3
O
HO O morphine
Figure 31.1 Structure of diamorphine (heroin) and its major metabolites.
immunogen is produced via conjugation with the hydroxyl group at position 3, this ceases to be a determinant against which antibodies are generated. As a result, the antibodies are likely to have cross-reactivity towards the major urinary metabolite of both diamorphine and morphine, morphine-3-glucuronide, and they will also recognise codeine (3-O-methylmorphine). Urine screening immunoassays to detect abuse of diamorphine or morphine usually employ antibodies raised by this means. Conversely, conjugation via the hydroxyl group at position 6 will yield antibodies that have a greater specificity towards morphine relative to morphine-3-glucuronide and that also display good cross-reactivity towards 6-monoacetylmorphine and morphine-6-glucuronide. These antibodies are well suited to formulating saliva assays for detecting diamorphine abuse, since this sample contains large amounts of 6monoacetylmorphine as well as of morphine itself. Various methods are available to covalently couple through these groups or to add an extension linker prior to linking it to a carrier protein (Van Regenmortel et al. 1988). Carriers include large proteins such as bovine serum albumin (BSA), bovine gamma globulin or keyhole lymphocyte cyanin. A plethora of other possible carrier proteins exists, but in general the selected carrier proteins are dissimilar in structure to the reporter to be used in the assay and large enough to trigger the immune response. Once antibodies are generated they are usually purified using simple precipitation techniques or by isolation using protein-A. Protein-A is a lectin that specifically binds to antibodies and allows rapid isolation of the purified proteins (Hober et al. 2007). Immunoassay design The antibody is a key element in the construction of any immunoassay and requires detailed consideration. Once an antibody is available it must be evaluated in the format in which it will be used. Assay formats are generally divided into two categories. The first is called a heterogeneous immunoassay, in which the differentiating characteristic is the requirement of a step
to separate bound from free material. The alternative is called a homogeneous immunoassay and is distinguished from heterogeneous assays by the fact that it does not require a separation step. Figure 31.3 shows the basic elements of a heterogeneous immunoassay. Heterogeneous immunoassay/examples The simplest form of heterogeneous immunoassay involves a competitive assay using a solid phase. Most commonly, a microtitre plate coated with immobilised antibodies against the target drug can be used. In this assay format, sample, buffer and a hapten–drug–reporter conjugate are added to a microtitre well. The mixture is incubated for some time during which the conjugate and free drug compete to bind to the solid phase. The solid phase is then washed to remove unbound material. Substrate is added, after which the signal is measured in each reaction well. The signal generated is inversely proportional to the concentration of free drug in the sample. Enzyme-linked immunosorbent assay (ELISA) is the most common format used for the detection of large molecules or proteins, but it has also been adapted to detect small-molecule drugs. These types of heterogeneous assay are sometimes called ‘sandwich’ assays because the target analyte is captured between two antibodies. Figure 31.4 shows a diagram of a typical heterogeneous sandwich assay. In the first step, an aliquot of sample or calibrator is mixed with buffer and incubated with the capture antibody that has been conjugated or adsorbed onto a solid surface such as a microtitre plate. After Analyte
Reporter–hapten conjugate Antibody
Solid phase Figure 31.2 Structure of morphine.
Figure 31.3 Basics of an immunoassay.
Clarke's Analysis of Drugs and Poisons Chapter No. 31 Dated: 11/3/2011 At Time: 8:30:0
Basic principles and issues of immunoassays
Step 1
Large molecular weight analyte
Step 1
Capture antibody on solid phase
Antibody
Analyte
Hapten on solid phase
Step 2
Incubate + Wash Step 2
499
Reporter Anti-antibody
Incubate + Wash
Enzyme Secondary antibody
Incubate + Wash
Step 3 Add substrate
Colour is produced
Step 3
Incubate + Wash
Add substrate
Color is produced Step 4
Measure signal generated
Figure 31.4 The basic scheme for an ELISA assay. Note the wash between steps. This is the distinctive feature of a heterogeneous assay.
incubation, the surface is washed to remove whatever material did not specifically bind to the antibodies on the solid surface. In the second step, another solution is added that contains a secondary antibody, often labelled with a reporter. Following this incubation, the surface is again washed to remove unbound material. Next, a substrate solution or developer solution is added to generate a signal. Once sufficient signal has been generated, the reaction is stopped and the signal is measured. The amount of signal generated is proportional to the amount of target material that was captured on the surface. Heterogeneous assays are often extremely sensitive and may be titred by adjusting the concentration of reagents and sample. The disadvantages of heterogeneous assays include the long incubation periods and multiple wash steps. In the field of toxicology a number of heterogeneous assay techniques have become available and are in routine use. These techniques include ELISA, radioimmunoassay (RIA), chemiluminescent immunoassay (CIA), fluorescent immunoassay (FIA) and finally lateral flow assay (LFA). Figure 31.5 shows a scheme for a typical heterogeneous assay targeting a small-molecule drug. In the first step, sample and buffer are mixed with anti-drug antibodies. If target drug is absent, the antibody will bind to the drug linked to the solid phase. The solid phase can be a microtitre plate, bead or membrane made of nitrocellulose, polymers or glass. Once this initial incubation is complete, the surface is washed and a
Step 4 Measure signal generated Figure 31.5 The scheme shows a typical competitive solid-phase immunoassay commonly used to detect drugs of abuse. It is a heterogeneous format using washing steps to remove materials that did not bind immunologically. Signal is inversely proportional to the concentration of free drug in the sample.
second reagent is added containing reporter-labelled antibodies against the first antibody targeting the drug of interest. A sandwich is formed between the drug conjugate on the solid phase and the secondary antibody–reporter conjugate. After incubation, unbound secondary antibody is washed away and substrate or signal is measured in the reaction mix. The signal detected is inversely proportional to the concentration of drug in the sample. There are a number of variations on the two assay schemes described above. In most cases the differences are in either the reporter used or the solid surface (see Table 31.2). Various reporters used include radioactive (RIA), fluorescent (FIA) and chemiluminescent (CIA) labels. The key to each heterogeneous assay is the wash step that removes excess reagent and lowers the background signal from the sample or other interferents. In this way heterogeneous assays achieve maximum analytical sensitivity. Lateral flow is a type of heterogeneous assay that has become prevalent in emergency rooms and laboratories and in law enforcement to test on-site for drugs of abuse (Ulti-Med 2002). These tests are capable of multiplexed detection of a panel of drugs of abuse from a single aliquot of urine, blood or saliva (Inoue, Seta 1992; The Walsh Group 2002). The basic scheme for a multiplexed lateral flow assay is shown in Fig. 31.6. A lateral flow assay is constructed from various materials that are assembled to form a test strip. A sample pad is the first component and is where a small aliquot, usually 50–100 mL, of sample is placed onto the strip. The sample pad often contains buffer salts to condition the sample to the correct pH and sometimes to remove cells or debris. These pads are often glass fibre or cellulose (Verheijen 2002; Volkov et al. 2009). The next material beyond the sample pad is the conjugate pad. It will usually hold the reporter–antibody conjugate targeting each drug of interest. The reporter most commonly used is colloidal gold. Gold particles are small, usually 10–100 nm in size, but are easily seen with the naked eye as they accumulate in a detection zone on a strip. Purified antibodies can easily be adsorbed onto the surface (Verheijen 2002; Volkov et al. 2009). Once a sample is added to a test strip it flows through the sample pad, the conjugate pad and then onto a nitrocellulose strip. Nitrocellulose as a raw material acts to bind capture
Clarke's Analysis of Drugs and Poisons Chapter No. 31 Dated: 11/3/2011 At Time: 8:30:0
500
Immunoassays Sample applied to absorbent pad Assay reagents
Sample wicked up into absorbent pad
Lateral flow membrane Control lines Test lines (target capture) Figure 31.6 The figure shows the basic design of a lateral flow assay. It contains a series of material in linear order designed to contain reagents that flow along with the sample past capture zones. The immunological result is read in each capture zone usually by visual interpretation.
reagents and to allow liquids to flow by capillary action along a test strip. The nitrocellulose strip can vary in length according to the desired goals of the test. For example, a longer strip will allow multiple analyte test lines (see Fig. 31.6). However, it will take longer for fluids to move along the strip, thus increasing the time of the assay. The nitrocellulose is also where the immunological reaction takes place. Capture zones at the control line and test lines are created by spraying a solution of the capture protein derivative onto the nitrocellulose. The proteins then adsorb onto the nitrocellulose and become immobilised (see Fig. 31.6). The usual capture zone material is target hapten derivatised and conjugated to a carrier protein such as BSA. As the buffered sample and reporter antibody reach the reaction zone there is competition between drug in the sample and hapten conjugate on the strip to bind the reporter antibody. If there is a large concentration of drug, there will be little reporter conjugate available to bind to the reaction zone and the line will have no colour. If the sample is negative for drug, the line at the capture zone will be intense and easily seen with the naked eye. Use of lateral flow assays has grown exponentially in the last decade. Once the basic chemistry has been developed, the system must be packaged into a housing (Fig. 31.7). The housing may be integrated into a urine cup, dip stick or a saliva collector (Draeger Safety UK Ltd 2008; Orasure Technologies 2008). The challenge for lateral flow assays used in toxicology is that they must reliably detect a panel of drug targets using dynamic flow, and binding of reagents all moving through a series of striped materials is governed solely by capillary forces. From a purely analytical perspective, lateral flow assays are often imprecise and insensitive. It is not uncommon for lateral flow assays to have relative standard deviations (RSDs) of 15–30% when testing replicates of sample or calibrator. Also, numerous field evaluations have highlighted performance shortcomings (Peace 2000; The Walsh Group 2002). Even so, lateral flow is an accepted method used by those performing on-site testing of drugs of abuse. It is expected that, as commercial use continues to grow, new instrumentinterpreted methods will improve the performance of lateral flow assays (Alverix Inc. 2010; Faulstich et al. 2007). Visual QC window (control lines)
Sample port
Test window (test lines) Side view Figure 31.7 Lateral flow assay in a housing.
Homogeneous immunoassay/examples Homogeneous immunoassays, by definition, do not require a step to separate bound from free target analytes and reagents. In most cases, homogeneous assays are used on automated platforms that require little operator interaction. This is something of a disadvantage since such equipment is not easily portable. Thus, homogeneous assays are used routinely on large laboratory-based instrument platforms capable of analysing hundreds or thousands of samples per day. The availability of these assays at a very low cost has revolutionised immunoassay-based drugs of abuse testing. Homogeneous drug assays are often competitive assays where free drug in a calibrator or sample competes with a reporter conjugate to bind to a specific antibody on the target. After a brief incubation, the change in signal is measured spectrophotometrically, fluorescently or through chemiluminescence. The following are descriptions for some homogeneous assay methods available for drugs of abuse testing. The EMIT (enzyme-multiplied immunoassay technique) has been a staple in drug testing since the 1970s when the method was first developed (Kabakoff, Greenwood 1981; Rubenstein et al. 1972; Ullman 1994; Ullman, Maggio 1980). An EMIT assay is a competitive format wherein hapten-labelled glucose-6-phosphate dehydrogenase (G6PDH) competes with free drug in a sample or calibrator to bind to a specific antibody raised against the hapten target. If the hapten-G6PDH binds to the antibody, there is a conformational change in the enzyme that leads to a decrease in activity (Fig. 31.8). This change in activity is proportional to the concentration of free drug present in the sample. EMIT assays have been developed for a large number of drugs of abuse and TDM analytes. Over time a wide variety of instrumentation has also been made available to laboratory workers, allowing EMIT to quickly become a method of choice. However, it is worth noting that, as with all immunoassays, EMIT assay performance is dependent upon the antibody used. In addition, EMIT assays are subject to interference from sample adulteration and non-specific cross-reactivity (Colbert 1994; Rollins et al. 1990; Rossi et al. 2006). Somewhat comparable in operating principles, CEDIA (cloned enzyme donor immunoassay) relies on the modulated activity of mutant beta-galactosidase. (Henderson et al. 1986) developed a genetically engineered form of beta-galactosidase which exists as two components. One fragment is called the enzyme acceptor (EA) and the other is called the enzyme donor (ED). The mutant enzyme becomes enzymatically active when both the ED and the EA are present. A homogeneous assay can be developed by conjugating a hapten to the ED. The hapten-ED will compete with free drug in the presence of antibody against the hapten (Fig. 31.9). When high concentrations of target hapten-drug are present, the hapten-ED is available to bind to the EA and an increase in betagalactosidase will be detected. Thus, like EMIT, the amount of enzymatic activity is proportional to the concentration of free drug in the sample. Additionally, CEDIA assays can be run on the same automated spectrophotometric equipment that also runs EMIT assays. Other types of homogeneous assays exist and are based upon reporters that are non-colorimetric. In this category, fluorescence
Clarke's Analysis of Drugs and Poisons Chapter No. 31 Dated: 11/3/2011 At Time: 8:30:0
Basic principles and issues of immunoassays
501
In the presence of free drug Substrate
Antibody Product
Hapten
Free drug
G6DPH
In the absence of free drug Antibody Substrate Hapten
G6DPH Figure 31.8 EMIT assay scheme; signal is directly proportional to the concentration of analyte.
polarisation, microparticle agglutination and new techniques such as LOCI (luminescent oxygen channelling immunoassays) (Ullman 2005) have all been demonstrated and used routinely in commercial laboratories. Fluorescence polarisation is a relatively simple technique that utilises the competitive binding to antibodies between a hapten–fluoroscein In the presence of free drug
Hapten ED
conjugate and free drug (hapten; Fig. 31.10) (Abbott Laboratories 2005; Colbert et al. 1985; Dandliker et al. 1973). The hapten–fluorescein conjugate rotates rapidly when not bound to an antibody. When it is bound to the antibody, the rotation is slowed dramatically compared with the unbound molecule. To generate the assay signal, a fluorimeter shines light at the excitation wavelength for fluorescein through a vertical polarising filter. Rapidly rotating unbound hapten–fluorescein molecules emit light in a different plane to the incident light. The
In the presence of free drug
Product
Antibody
High rotation/low polarisation Antibody
EA
F Hapten–fluorescein Free drug Substrate Free drug In the absence of free drug In the absence of free drug
Low rotation/high polarisation
Antibody
F Substrate
Hapten ED
Hapten–fluorescein
EA
Antibody Figure 31.9 CEDIA assay scheme; signal is directly proportional to the concentration of analyte.
Figure 31.10 Fluorescence polarisation immunoassay assay scheme; signal is inversely proportional to the concentration of analyte.
Clarke's Analysis of Drugs and Poisons Chapter No. 31 Dated: 11/3/2011 At Time: 8:30:0
502
Immunoassays In the presence of free drug Low agglutination/low light scattering Antibody
Micro particle
Hapten
Free drug
In the absence of free drug High agglutination/high light scattering
Micro particle
Hapten
Figure 31.11 KIMS assay scheme signal is inversely proportional to the concentration of analyte.
relatively stationary antibody-bound hapten–fluorescein molecules, however, return light in a similar plane. This is detected via the polarising filter. Drug added via the sample competes for binding to the antibody with the fluorescein-labelled hapten, thereby reducing the amount of fluorescein bound to the antibody, resulting in less emitted fluorescence being detected via the polarised filter. Fluorescence polarisation assays require specialised equipment and are the basis of the Abbott ADx system. The background fluorescence found with many biological samples means that it is necessary to take a blank reading of the sample and reagents before the addition of the fluorescent tracer to the mixture. Another popular homogeneous technique used for drugs of abuse assays is based upon microparticle agglutination. The KIMS assay (kinetic interaction of microparticles in solution) relies on the old but reliable principles of latex particle agglutination (Fig. 31.11) (De Giovanni, Fucci 2006; Feldman et al. 2004). In the KIMS technology, polystyrene latex microparticles have been coated with a drug-conjugate. These particles are dispersed in solution, after which sample and antibodies to the drug (hapten)-conjugate are added. In the presence of high drug concentrations, the antibody-binding sites are occupied by free drug and a low amount of agglutination takes place. In the presence of low target drug concentrations, the antibodies bridge between the particles and begin to agglutinate particles into large clusters. These clusters scatter light. Therefore, the change in signal is inversely proportional to the concentration of free target drug in the sample. KIMS is the basis of the Roche Abuscreen Online system, which involves monitoring the rate of agglutination by spectrophotometric means. The challenge to any homogeneous assay technique is sensitivity. These assays usually take seconds to minutes to perform and are often limited by kinetics or antibody quality. LOCI is a chemiluminescent system that overcomes many limitations of other assay reporters (Ullman 2005). The reagents used to produce a signal in the assay are encapsulated in latex spheres which prevent interference from sample matrices. One population of latex spheres contains a chemiluminescer and the second a photosensitiser capable of exciting oxygen when exposed to 680 nm light. When the two beads are in close proximity, excited oxygen diffuses out of the bead and into the chemiluminescer bead, producing detectable photons. The LOCI assay has been tested with a variety of analytes and each one has shown superior sensitivity to other competing technologies (Ullman 2005).
Immunoassay optimisation Earlier in this chapter the methods used to develop an antibody against a chosen hapten drug target were reviewed. Assuming that a viable candidate antibody has been generated and an assay format chosen, the next step is to begin developing and optimising the immunoassay. The goal of this section is to outline some of the key experiments followed by an explanation of important assay parameters that should be considered. Again the target analyte to be used as an example in this section will be morphine. The structure for morphine is shown in Fig. 31.1. Morphine is one of a large class of opiate compounds that are often abused. Therefore, the goal of this hypothetical assay is to detect a broad class of compounds in a single sample. The format for the assay to be developed is lateral flow and the sample matrix will be saliva. Studies of the metabolism of opiates have shown that both the parent drug and its major conjugated metabolites appear in oral fluids and these have previously been shown to be detectable using a laboratory-based immunoassay (Niedbala, Kardos 2005). This suggests that a lateral flow assay is plausible. To start developing the lateral flow assay, morphine is derivatised through the hydroxyl group before being conjugated to BSA (Verheijen 2002; Volkov et al. 2009). Using a Kinematic Linomat striper, the morphine–BSA conjugate can be adsorbed onto the surface of a 10 mm pore size nitrocellulose sheet by sequentially dispensing small amounts of conjugate to form a capture zone (Verheijen 2002; Volkov et al. 2009). Additionally, a reporter such as colloidal gold, fluorescent latex particles or upconverting phosphors can be labelled with purified anti-morphine as described elsewhere (Niedbala et al. 2000, 2001). Finally, phosphatebuffered saline, N-(2-hydroxyethyl)piperazine-N 0 -2-ethanesulfonic acid (Hepes), or an alternative buffer may be chosen as long as it adequately controls the pH of the final assay mixture without negatively affecting the assay. Once all the reagents are prepared and a standard curve showing a response over various concentrations of morphine has been run, experiments can be conducted to optimise the concentration of each assay component. Usually the components that are varied are the loading of reagents onto the capture zone, the level of reporter conjugate to control detectable signal, and finally the sample volume to obtain the best precision and optimal dynamic range.
Clarke's Analysis of Drugs and Poisons Chapter No. 31 Dated: 11/3/2011 At Time: 8:30:0
Basic principles and issues of immunoassays
503
80,000 Average peak area
70,000 60,000 50,000
0 ng/mL
40,000
10 ng/mL
30,000
20 ng/mL
20,000
40 ng/mL
10,000 0 A
B
C
Coupling protocol ID Figure 31.12 This graph shows the results from three experiments loading various amounts of morphine-BSA onto a lateral flow strip. The optimum level was reached when the signal at zero concentration of morphine was highest.
For example, Fig. 31.12 shows the effect of three different line striping protocols for the capture zone on the nitrocellulose strip. As the loading of morphine-BSA increased there was a maximum amount of signal that could be generated. Note, however, that the overall displacement between zero and the various concentration of free drug remained about the same. Next, the concentration of reporter conjugate may be optimised. Figure 31.13 shows an example of a lateral flow assay utilising up-converting phosphor conjugates as the reporter. Note that, as the level of reporter increased, so did the signal at 0 ng/mL free morphine. This large separation helps to ensure discrimination between negative and positive samples. Finally, the assay sample size was adjusted and the assay was tested for precision by running replicates at a number of morphine levels. As shown in Fig. 31.14, the assay developed was capable of easily discriminating between 0 and 40 ng/mL morphine, the target concentrations appropriate for an oral fluid-based test. Immunoassay performance parameters
immunoassays for toxicological investigations (Green, Isenchmid 1995; Kwong et al. 1988; Linnet, Brandt 1986). Parameter 1: Precision
Precision is a measure of the variation that occurs either when replicate samples are tested in a single run (intra-assay precision) or when replicate samples are tested and compared between runs (inter-assay precision). Whether intra- or inter-assay precision, the value is always reported as the RSD. It is important to plot the RSDs of any test and look for the deviation around each point tested. In the author’s laboratory it is usual to plot the mean plus 2 standard deviations above and below each assay point on the curve. If it overlaps the standard deviation from the calibrator above, optimisation of the assay is continued to improve performance. It is important that precision data should be examined as either the precision of the signal generated in an assay or transformed numbers on a standard curve. Most toxicology assays are qualitative and therefore use signal only. However, transformed numbers are more stringent and are an excellent way to show robust assay performance. Parameter 2: Limit of detection
Having discussed the major aspects of test reagents and development, it is appropriate now to turn to parameters used to evaluate immunoassay performance. It is important to note that there are a number of ways to determine each of the parameters considered. Those discussed here are relevant to scientists developing or evaluating commercial
Many toxicology screening assays require cut-off levels that challenge the development of any immunoassay. Analytes such as LSD, buprenorphine or tetrahydrocannabinol (THC) must be detected at extremely low concentrations in oral fluids. The assay parameter that indicates the lowest level detectable is the limit of detection (LOD). The LOD for an
Integrated peak area response
80,000 70,000 60,000 50,000 0 ng/mL 40,000
10 ng/mL
30,000
20 ng/mL
20,000
40 ng/mL
10,000 0 4 µg
8 µg
12 µg
16 µg
Amount of phosphor per test Figure 31.13 This graph shows results of varying the conjugate reporter in the lateral flow assay being designed to detect morphine. Signal increased as more conjugate was added. This produced a steeper curve and improved the performance of the assay.
Clarke's Analysis of Drugs and Poisons Chapter No. 31 Dated: 11/3/2011 At Time: 8:30:1
504
Immunoassays
Average peak area (n=3)
18,000 16,000 14,000 12,000 10,000 8,000 6,000 4,000 2,000 0 0
10
20
30
40
50
60
70
80
90
Drug concentration [ng/mL] Figure 31.14 Once optimised, the morphine lateral flow assay was tested for precision. This graph shows that the optimised assay had little variation around each calibrator concentration. There was also a large separation in signal between calibrators.
immunoassay is often determined in one of two ways. The first approach is to run replicates of the zero calibrator in an assay and then take the mean plus 2 or 3 standard deviations and plot it onto the assay standard curve. This concentration is then assigned as the LOD. The second method, which is more robust, is to run replicates of a variety of concentrations along the assay curve and, similarly to determining precision, plot 2 standard deviations above and below each point. The lowest concentration that can separate 2 standard deviations above zero and 2 standard deviations below the replicate concentration is then denoted the LOD. Parameter 3: Specificity/cross-reactivity
This parameter is extremely important for toxicology assays. As stated earlier, some assays are targeted for certain compounds, such as THC, while others will ideally detect a broad class of compounds, such as benzodiazepines. In addition, assays should also be free from interference from more common over-the-counter medications. Using the example of a morphine assay, Table 31.3 lists a number of opiates of interest along with their cross-reactivities in an immunoassay. In addition to cross-reactivity, the table also shows calculations for morphine equivalents and the concentration that may produce a positive result. Each of these values for cross-reactivity, equivalents and concentration that may produce a positive result is derived by running various concentrations of potential cross-reactants in the assay of interest and comparing the results against the cut-off calibrator, which, in this case, is morphine. Each of the parameters can be determined by comparing concentrations against the cut-off.
Parameter 4: Interferences/adulteration
Even if an immunoassay is performed correctly, it can provide an incorrect result if the sample was somehow adulterated or contained an interfering substance. The literature or product package inserts for assays that use blood or urine will frequently warn that haemolysis, lipaemia or high levels of protein in a sample can interfere with homogeneous immunoassays. More recently, orally based diagnostic tests have looked at the food or drinks that can interfere with assays, while hair testing has considered the effects of various treatments, soaps or hair colorants. In all of these cases the assay designers must be prepared to realistically evaluate such interferences. At a minimum, immunoassay developers should document and inform users of their assays about potential interferents. Adulterants are related to, but different from, interfering compounds. Interferents are defined as normal day-to-day materials used by individuals that can cause aberrant results. Adulterants are materials wilfully added to a sample in order to disrupt a toxicology immunoassay. Such materials have been used in recent years to target many of the homogeneous assays used to initially screen urine samples for drugs of abuse (Wu et al. 1999). Materials such as bleach (sodium hypochlorite), salts or detergents have been reported to be very effective and their potential for disruption should be evaluated in any newly developed assay. Parameter 5: Stability
Any immunoassay is expected to perform over a relatively long period. Commercial immunoassay kits are usually stable from 6 months to
Table 31.3 Morphine immunoassay cross-reactants Compound
Concentration (ng/mL)
Morphine equivalents (ng/mL)
Percentage cross-reactivity (%CV)
Concentration that may produce a positive result (ng/mL)
6-Acetyl morphine
40
34.8
87.0
46
Codeine
40
24.1
60.3
66
Diacetylmorphine
100
>73.7
>73.7
54
Hydrocodone
100
60.5
60.5
66
Hydromorphone
100
>55.9
55.9
72
10 000
68.8
0.7
5,714
100
80.0
80.0
50
Meperidine b–Morphine-3-glucuronide
1
36.8
>100
1
Normorphine
100
34.3
34.3
117
Oxycodone
100
43.1
43.1
93
Oxymorphone
100
36.3
36.3
110
Nalorphine
Clarke's Analysis of Drugs and Poisons Chapter No. 31 Dated: 11/3/2011 At Time: 8:30:1
References several years. Laboratory-based tests are somewhat easier to maintain and control, since cold-room storage is not a problem. Point-of-care tests that are used in remote locations without room temperature control or perhaps no facilities at all face some of the greatest challenges to stability. In these cases the stability of a test must always be related to the functional temperature range that can be tolerated. Any immunoassay developer should field test their immunoassay under various conditions of humidity, temperature and even altitude. Limitations can then be reported to potential users of the test. Such studies are difficult and often highlight the limitations of an immunoassay. However, they are essential so that the incidence of false results is minimised in the field or in the laboratory. Parameter 6: Quality control
There are various ways to approach quality control for any immunoassay. Some laboratory methods look at the precision of controls from one run to another. Other immunoassays use the standard curve generated to see whether the spread between calibration points is appropriate. These internal assay controls are all good measures that ensure that the final result is correct. Additional quality control steps should include external controls. Some vendors sell urine, blood or saliva samples that have been spiked at various concentrations. Proficiency testing programmes are a very useful means by which an individual laboratory can ensure that its assays are performing correctly and for detecting any problems with a particular method. These schemes regularly send samples to a large number of participant laboratories to be tested on a blind basis. A number of organisations exist that can supply urine, saliva, hair or sweat proficiency samples. Examples include RTI International, the College of American Pathologists or Cardiff Bioanalytical Services Ltd. Once tested, the results for each proficiency sample are returned to the co-coordinators of the scheme and each participant laboratory receives a report showing how its findings have compared with those of the others (Cone 1992). Immunoassay automation Great strides have been made over the last few decades in the automation of immunoassays. The use of homogeneous assays such as EMIT, CEDIA or KIMS for the rapid analysis of large numbers of samples for drugs of abuse has been described above. In addition, specialised instrumentation has been developed by commercial companies to utilise reporters such as fluorescence, radiolabels and chemiluminesence. In every case these immunoassay platform instruments allow for precise pipetting and incubation of samples. This degree of control is essential in assuring consistent and reliable results. It is interesting to see the overall sensitivity of each system (see Table 31.2). Future systems will be required not only to perform rapid analyses on large numbers of specimens but also to test different specimen types. This is an enormous challenge for immunoassay design and matching instrumentation. Future systems may be as much as 1000 times more sensitive than current techniques. Alternative fluids for toxicology immunoassays The latest area of drug immunoassay innovation has been concerned primarily with applications to alternative matrices. Traditionally, toxicological assays for drugs of abuse have been developed for use with urine, whereas TDM assays almost always utilise blood, serum or plasma. Urine testing remains the dominant matrix for drugs-of-abuse screening, although it is now recognised that this sample can be easily adulterated, requires private collection facilities, and is affected by overhydration (Cone et al. 2003). In recent years, more assays are being performed using fluids or matrices such as hair, sweat and oral fluid. Although considered several years ago, it is only recently that interest in using alternative fluids for routine screening of drugs of abuse has arisen (Cone 1992; Malamud, Niedbala 2007; Samyn et al. 1999a; Schramm et al. 1992; Wong 2008) There are several reasons behind this trend and they vary for different countries. For some there is a desire to allow law enforcement to screen for drugs of abuse at the roadside (Parliament of
505
Victoria, Australia 2003). This is not practical with any specimen except an oral sample (Verstraete et al. 1999c). In other situations, the drug concentration in the sample collected must have some correlation with that in blood so that some idea of impairment can be demonstrated. Again, this has led to the increased use of saliva testing (Cone et al. 2002; Thompson et al. 1987; Toennes et al. 2005) (Chapter 18). Where the goal is to have the longest window of detection to identify drug abuse, hair is the specimen of choice (Chapter 19).
Conclusions Immunoassays for the detection or measurement of drugs and their metabolites are routinely used in commercial laboratories worldwide. Automated immunoassay instrumentation capable of analysing thousands of samples per hour has now become routine. Although there are ways in which immunoassays can be flawed, the great majority perform reliably when used correctly. Future directions in immunoassay development are expected to improve upon methods to develop antibodies and format assays. However, these improvements will still utilise the basic principles outlined in this chapter.
References Abbott Laboratories (2005). Abbott AxSYM System. Cocaine metabolite product insert sheet. Abbott Park: Abbott Laboratories. Alverix Inc. (2010). San Jose, CA, USA. www.alverix.com. Baselt RC (2005). Disposition of Toxic Drugs and Chemicals in Man, 7th edn. Foster City, CA: Chemical Toxicology Institute. Binz HK et al. (2005). Engineering novel binding proteins from nonimmunoglobulin domains. Nature Biotechnol 23: 1257–1268. Chiswell DJ, McCafferty J (1992). Phage antibodies: will new ‘coliclonal’ antibodies replace monoclonal antibodies? Trends Biotechnol 10: 80–84. Colbert DL (1994). Drug abuse screening with immunoassays: unexpected crossreactivities and other pitfalls. Br J Biomed Sci 51: 136–146. Colbert DL et al. (1985). Single-reagent polarization fluoroimmunoassay for amphetamine in urine. Clin Chem 31: 1193–1195. Cone EJ (1992). Saliva Testing for Drugs of Abuse. New York: New York Academy of Sciences. Cone EJ et al. (2002). Oral fluid testing for drugs of abuse: positive prevalence rates by Intercept immunoassay screening and GC-MS-MS confirmation and suggested cutoff concentrations. J Anal Toxicol 26: 541–546. Cone EJ et al. (2003). Urine testing for cocaine abuse: metabolic and excretion patterns following different routes of administration and methods for detection of false-negative results. J Anal Toxicol 27: 386–401. Dandliker WB et al. (1973). Fluorescence polarization immunoassay. Theory and experimental method. Immunochemistry 10: 219–227. De Giovanni N, Fucci N (2006). Hypothesis on interferences in kinetic interaction of microparticles in solution (KIMS) technology. Clin Chem Lab Med 44: 894–897. Draeger Safety UK Ltd (2008). Draeger Drug Check. Blyth: Draeger Safety. Faulstich K et al. (2007). Developing rapid mobile POC systems. IVD Technology 13: 47–53. Feldman M et al. (2004). Evaluation of Roche diagnostics ONLINE DAT II, a new generation of assays for the detection of drugs of abuse. J Anal Toxicol 28: 593–598. Frenken LG et al. (2000). Isolation of antigen specific llama VHH antibody fragments and their high level secretion by Saccharomyces cerevisiae. J Biotechnol 78: 11–21. Green KB, Isenchmid DS (1995). Medical review officer interpretation of urine drug testing results. Forensic Sci Rev 7: 41–60. Hardman JG et al. (2001). Goodman, Gilman’s the Pharmacological Basis of Therapeutics, 10th edn. New York: McGraw-Hill Professional. Henderson DR et al. (1986). CEDIA, a new homogeneous immunoassay system. Clin Chem 32: 1637–1641. Hermes WJ (1993). Substance abuse. In: The Encyclopedia of Health. New York: Chelsea House. Hober S et al. (2007). Protein A chromatography for antibody purification. J Chromatogr B Analyt Technol Biomed Life Sci 848: 40–47. Howard GC, Kaser MR (2007) Making and Using Antibodies. Boca Raton, FL: CRC Press. Huckle D, Wild D (2005). Market trends. In: Wild D, ed. The Immunoassay Handbook, 3rd edn. New York: Elsevier. Inoue T, Seta S (1992). Analysis of drugs in unconventional samples. Forensic Sci Rev 4: 90–107. Kabakoff DS, Greenwood HM (1981). Homogenous enzyme immunoassay. In: Alberti KGMM, Price CP, eds. Recent Advances in Clinical Biochemistry. Edinburgh: Churchill Livingstone, 1–30.
Clarke's Analysis of Drugs and Poisons Chapter No. 31 Dated: 11/3/2011 At Time: 8:30:1
506
Immunoassays
Kohler G, Milstein C (1975). Continuous culture of fused cells secreting antibody of predefined specificity. Nature 256: 495–497. Kwong TC et al. (1988). Critical issues in urinalysis of abused substances: Report of the substance-abuse testing committee. Clin Chem 34: 605–632. Levine B (2003). Principles of Forensic Toxicology, 2nd edn. Washington, DC: AACC Press. Linnet K, Brandt E (1986). Assessing diagnostic tests once an optimal cutoff point has been selected. Clin Chem 32: 1341–1346. Maes V et al. (1999). Drugs and medicines that are suspected to have a detrimental impact on road user performance. Roadside Testing Assessment (ROSITA) D1 DG VII PL98-3032. www.rosita.org. Malamud D, Niedbala RS (2007). Oral-based Diagnostics. New York: New York Academy of Sciences. Moeller M et al. (1999). Operational user and legal requirements across EU member states for roadside drug testing equipment. Roadside Testing Assessment (ROSITA) D3, DG VII 98-SC.3032. www.rosita.org. Moore RA et al. (1984). Sensitive and specific morphine radioimmunoassay with iodine label: pharmacokinetics of morphine in man after intravenous administration. Ann Clin Biochem 21(Pt4): 318–325. Niedbala RS et al. (2001). Detection of analytes by immunoassay using up-converting phosphor technology. Anal Biochem 293: 22–30. Niedbala RS, Kardos K (2005). Oral fluid drug testing using the Intercept device. In: Wong R, Tse H, eds. Drugs of Abuse: Body Fluid Testing. Totowa, NJ: Humana Press. Niedbala RS et al. (2000). Multiphoton Up-Converting Phosphors for Use in Rapid Immunoassays, 7th edn. Proceedings of SPIE, Vol. 3913. Bellingham, WA: SPIE. Orasure Technologies (2008). OraQuick Advance. Bethlehem, PA: Orasure Technologies. Parliament of Victoria, Australia (2003). Road Safety (Drug Driving) Act. 31-102003. Peace MR et al. (2000). Performance evaluation of four on-site drug-testing devices for detection of drugs of abuse in urine. J Anal Toxicol 24: 589–594. Perrigo BJ, Joynt BP (1995). Use of ELISA for the detection of common drugs of abuse in forensic whole blood samples. Can Soc Forensic Sci J 28: 261– 269. Porter WH (2006). Clinical toxicology. In: Burtis CA et al., eds. Tietz Textbook of Clinical Chemistry and Molecular Diagnostics, 4th edn. St Louis, MO: Elsevier Saunders, 1287–1369. Rollins DE et al. (1990). Investigation of interference by nonsteroidal antiinflammatory drugs in urine tests for abused drugs. Clin Chem 36: 602–606. Rossi S et al. (2006). Characterization of interference with 6 commercial delta9tetrahydrocannabinol immunoassays by efavirenz (glucuronide) in urine. Clin Chem 52: 896–897. Rubenstein KE et al. (1972). “Homogeneous” enzyme immunoassay. A new immunochemical technique. Biochem Biophys Res Commun 47: 846–851. Ryan TA (2003). Fluorescent proteins with ties that bind. Nature Biotechnol 21: 1447–1479. Samyn N et al. (1999a). Analysis of drugs of abuse in saliva. Forensic Sci Rev 11: 1–19.
Samyn N et al. (1999b). Inventory of state-of-the-art road side drug testing equipment. Roadside Testing Assessment (ROSITA) D2, DG VII PL98-3032. www. rosita.org. Schramm W et al. (1992). Drugs of abuse in saliva: a review. J Anal Toxicol 16: 1–9. Thompson LK et al. (1987). Confirmation of cocaine in human saliva after intravenous use. J Anal Toxicol 11: 36–38. Toennes SW et al. (2005). Screening for drugs of abuse in oral fluid – correlation of analysis results with serum in forensic cases. J Anal Toxicol 29: 22–27. Uhlenhuth EH et al. (1990). International study of expert judgement on therapeutic use of benzodiazepines and other psychotherapeutic medications IV: Therapeutic dose dependence and abuse liability of benzodiazepines in the long-term treatment of anxiety disorders. J Clin Psychopharmacol 19: 23S–29S. Ullman EF (1994). Homogenous immunoassays. In: Wild D, ed. The Immunoassay Handbook. New York: Stockton Press, 212-230. Ullman EF (2005). Homogenous immunoassays. In: Wild D, ed. The Immunoassay Handbook, 3rd edn. New York: Elsevier. Ullman EF, Maggio ET (1980). Principles of homogenous enzyme-immunoassay. In: Maggio ET, ed. Enzyme Immunoassay. Boca Raton, FL: CRC Press, 105–134. Ulti-Med (2002). Assay for the qualitative detection of drug of abuse in saliva. St Paul, MN: Ulti-Med. US Department of Health and Human Services (2006). National Survey on Drug Use and Health: National findings. Office of Applied Studies NSDUH Series H-32. DHHS publication No. SMA 07-4293. US Department of Health and Human Services, Substance Abuse and Mental Health Service Administation (SAMHSA) (2008). Mandatory guidelines for fedral workplace drug testing programs. Fedral Registry 73: 71858–71907. US Department of Transportation Procedures for transportation workplace drug and alcohol programs: Final rule 49 CFR Part 40. Federal Register 65: 79462–75579. Van Regenmortel MHV et al. (1988). Laboratory Techniques in Biochemistry and Molecular Biology, Synthetic Polypeptides as Antigens. Amsterdam: Elsevier. Verheijen R (2002). Immunological strip tests. Methods and tools in biosciences and medicine. Anal Biochem 4: 134–166. Verstraete A, Puddu M (2000). General conclusions and recommendations. Roadside Testing Assessment (ROSITA) D5, DG VII 98-SC.3032. www.rosita.org. Volkov A et al. (2009). Rapid prototyping of lateral flow assays. In: Rasooly A, Herold K, eds. Methods in Molecular Biology: Biosensors and Biodection, vol 504. New York: Humana Press, 217–235. Walsh JM et al. (2004). Developing global strategies for identifying, prosecuting and treating drug-impaired drivers. Bethesda, MD: The Walsh Group, sponsored by the US Office of National Drug Control Policy, June 2004. The Walsh Group (2002). An Evaluation of Oral Fluid Point of Collection Testing Devices. Bethesda MD: The Walsh Group. Wong DT (2008). Salivary diagnostics. Am Sci 96: 37–43. Wu AH et al. (1999). Adulteration of urine by “Urine Luck”. Clin Chem 45: 1051–1057. Yalow RS, Berson SA (1959). Assay of plasma insulin in human subjects by immunological methods. Nature 184(Suppl 21): 1648–1649.
Clarke's Analysis of Drugs and Poisons Chapter No. 32 Dated: 11/3/2011 At Time: 8:39:17
CHAPTER
32
Ultraviolet, Visible and Fluorescence Spectrophotometry J Cordonnier and J Schaep
Introduction and theoretical background General introduction Analytical absorption spectroscopy in the ultraviolet (UV) and visible regions of the electromagnetic spectrum has been widely used in pharmaceutical and biomedical analysis for quantitative purposes and, with certain limitations, for the characterisation of drugs, impurities, metabolites and related substances. By contrast, luminescence methods, and fluorescence spectroscopy in particular, have been less widely exploited, despite the undoubted advantages of greater specificity and sensitivity commonly observed for fluorescent species. However, the wider availability of spectrofluorimeters able to present corrected excitation and emission spectra, coupled with the fact that reliable fluorogenic reactions permit non-fluorescent species to be examined fluorimetrically, has led to a renaissance of interest in fluorimetric methods in biomedical analysis. UV and visible spectrophotometry: theoretical background General considerations
Molecular absorption in the UV and visible regions arises from energy transitions that involve the outer orbital or valence electrons. Spectra in liquid media are usually broad, relatively featureless bands, a result of the large number of closely spaced vibrational and rotational transitions. The fundamental band shape approximates Gaussian or lognormal Gaussian curves. Given the broad, overlapping profiles commonly encountered, the shape and precise location of individual bands are of limited usefulness in qualitative analysis. However, any fine structure detected in the spectra, coupled with solvent and pH effects, can be of diagnostic value. More informative spectra can be obtained for some volatile molecules of toxicological interest, such as benzene and polynuclear aromatic hydrocarbons; when examined in the vapour phase, vibrational and rotational fine structure can readily be seen superimposed on the broad spectral profiles. This is illustrated in Fig. 32.1 for 1,2,4,5-tetrazine. However, most drugs, metabolites and related compounds are relatively non-volatile; their spectra are observed necessarily in solution, or possibly in the solid phase by reflectance, or by compression to form a KBr disc, as used in infrared spectrophotometry. UV and visible spectrophotometry find their primary application in quantitative analysis. The scope of absorption spectroscopy can be extended significantly by the use of colour reactions, often with a concomitant increase in sensitivity and/or selectivity. Such reactions are used to modify the spectrum of an absorbing molecule so that it can be detected in the visible region, well separated from other interfering components in the UV spectrum. Moreover, chemical modification can be used to transform an otherwise non-absorbing molecule into a stable derivative that possesses significant absorption. Spectral selectivity can be enhanced further by a number of chemical or instrumental techniques, which include difference, higher-derivative and dual-wavelength spectrophotometry. Such methods, and certain graphic techniques such as the Morton–Stubbs method, can contribute in different ways to reducing the general problem of spectral interference in quantitative spectroscopy. Spectral interference can arise from socalled ‘irrelevant’ non-specific absorption, and also from absorption by other materials and impurities that may be present. When interference
arises specifically from the spectral overlap of two or more well-defined components, a number of methods can be applied to measure the individual concentrations. These methods include the Vierordt multiwavelength technique, least-squares deconvolution and second- or higher-derivative spectrophotometry. Spectral selectivity, and in some cases detection sensitivity, can be enhanced significantly by the various chemical and instrumental techniques outlined above. Such methods should, of course, be validated by applying the conventional analytical criteria of accuracy (against a reference method), linearity, precision and independence from interfering substances. The scope of UV and visible spectrophotometry can be further extended when combined with a chromatographic separation step, such as high performance liquid chromatography (HPLC). The development of rapid-scanning detectors based on the linear photodiode array permits spectra to be acquired during the elution of peaks. Computer-aided manipulation of these spectra has led to new strategies for the examination of chromatographic peak homogeneity, based on classic techniques in spectroscopy. The use of microcomputers enables the development of archive-retrieval methods for spectral characterisation (Fell et al. 1984). Nomenclature
In the UV and visible spectrum, the energy of photons associated with electronic transitions lies in the range 147–630 kJ/mol. This energy (DE) can be expressed in terms of the principal parameters that define electromagnetic radiation, namely frequency m (Hz), wavelength l (nm) and wavenumber n (cm1): DE ¼ hm ¼
hc ¼ hcn l
ð32:1Þ
where h is Planck’s constant and c is the velocity of radiation in vacuo. The positions of peaks are sometimes described in terms of wavenumber, which has the advantage of being a linear function of energy, but this term is much more frequently used in infrared spectrophotometry. The practical unit most often used in UV and visible spectrophotometry is wavelength, usually expressed in nanometres (nm). The units that have previously been used for wavelength – millimicron (mm) and angstr€ om (A ) – are not recommended terms. The position of maximum absorbance of a peak is designated lmax. The wavelength span is conventionally divided into two ranges: the UV extends from 200 nm to about 400 nm; the visible range extends from about 400 nm to 800 nm. Outside these limits, the ‘far UV’ or ‘vacuum UV’ extends from 100 nm to 200 nm, and the ‘near infrared’ from 1 mm to about 3 mm. A molecular grouping specifically responsible for absorption is described as a chromophore, and is usually a conjugated system with extensive delocalisation of electron density. Any saturated group with little or no intrinsic absorption of its own, but that modifies the absorption spectrum when attached directly to a chromophore, is described as an auxochrome, examples being –OR, –NR2, –SR. Auxochromes are considered to exert their effect through partial conjugation of their polarisable lone-pair electrons with those of the adjacent chromophore. If, however, the lone pair of electrons is involved in bonding as, for example, in the case of a protonated quaternary ammonium group, the 507
Clarke's Analysis of Drugs and Poisons Chapter No. 32 Dated: 11/3/2011 At Time: 8:39:17
508
Ultraviolet, Visible and Fluorescence Spectrophotometry The logarithmic term is linearly related to concentration and path length, and is referred to as absorbance (A). The older terms extinction (E) and optical density (OD) are not recommended, although they are often found in the literature. Transmittance (T ¼ I/I0) and percentage transmittance (%T ¼ 100(I/)0)) are not linear functions of concentration and path length, and can be related readily to absorbance: A ¼ log
Io 1 ¼ log ¼ 2 logð%TÞ T I
ð32:3Þ
The molar absorptivity, e, is a fundamental property of a molecule that tells how much light is absorbed at a particular wavelength. It has two connotations in European usage, and a third according to American convention. If the concentration is expressed in g/L, e is described as the absorptivity (k, L/g per cm). When concentration is expressed in g/100 mL, k is described as the specific absorbance and given the symbol A1% 1 cm or A(1%, 1 cm), defined as ‘the absorbance of a 1% w/v solution in a cell of 1 cm path length’. It is usually written in the shortened form A11 and is widely used in analytical chemistry. It was formerly known as the ‘specific extinction coefficient’, symbol E1% 1 cm or E(1%, 1 cm). American convention recognises the constant k as ‘absorptivity’ (a, L/g/cm) defined as ‘the absorbance of a 1 g/L solution in a cell of 1 cm path length’. These terms for absorptivity can readily be interconverted: a¼ Figure 32.1 UV absorption spectra of 1,2,4,5-tetrazine.
auxochromic effect vanishes. This property can be used for molecular characterisation, as discussed below. Laws of absorption spectrophotometry
The extent of absorption of radiation by an absorbing system at a given monochromatic wavelength is described by the two classic laws of absorptiometry, which relate the intensity of radiation incident on the absorbing system (I0) to the transmitted intensity (I) (Fig. 32.2). Lambert’s (or Bouguer’s) law concerns instrumental factors, and states that, at a given concentration (c) of a homogeneous absorbing system, the transmitted intensity (I) decreases exponentially with increase in path length (b). The complementary Beer’s law deals with concentration and states that, for a layer of defined path length (b), the transmitted intensity (I) decreases exponentially with the increase in concentration (c) of a homogeneous absorbing system. Combination of these observations gives the familiar Beer–Lambert law: log
I0 ¼ cb I
ð32:2Þ
where e is the molar absorptivity or molar extinction coefficient of the system, defined as ‘the absorbance of a one molar solution in a cell of 1 cm path length’. The concentration c is here expressed in mol/L.
A11 ¼ 10 Mr
ð32:4Þ
where Mr is the relative molecular mass. Thus, a compound with an Mr of 100 and absorptivity a of 20 at wavelength l in a particular solvent at a defined pH (if aqueous) and at a specified temperature, has a corresponding specific absorbance A11 of 200 and a molar absorptivity e of 2000. Absorbance and absorptivity are often expressed in logarithmic form in cases where spectra are to be compared. The logarithmic form of the Beer–Lambert law expresses the effects of the molar extinction coefficient (e), concentration (c) and path length (b) as additive terms logA ¼ logþlogcþlogb
ð32:5Þ
The value for log e is typically in the range 1–5. Since only the molar extinction coefficient (e) is a function of l, the shape of a logarithmic absorption curve is independent of concentration and path length. Their only effect is to shift the log A spectrum along the log A axis. A disadvantage of the log A plot is that fine structure near the top of the peak is compressed. Validity of the Beer–Lambert law
The validity of the Beer–Lambert law is affected by a number of factors. If the radiation is non-monochromatic, i.e. if its spectral bandwidth is greater than about 10% of the drug absorption bandwidth at halfheight, the observed absorbance will be lower than the ‘true’ limiting value for monochromatic radiation. Thus, sharp bands are more susceptible than broad bands to absorbance error on this account. Moreover, if the absorbing species is non-homogeneous, or if it undergoes association, dissociation, photodegradation, solvation, complexation or adsorption, or if it emits fluorescence, then positive or negative deviations from the Beer–Lambert law may be observed. Stray-light effects and the type of solvent used may also lead to non-compliance with the Beer–Lambert law. Stray-light effects
Figure 32.2 Attenuation of a beam of radiation by an absorbing solution.
Stray light is radiation at wavelengths different from those desired. It may arise from light scattering or other defects within the instrument, or it may be caused by external radiation. If the stray light is not absorbed, the observed absorbance tends to a constant value as the concentration of drug is increased, yielding a negative deviation from the Beer– Lambert law.
Clarke's Analysis of Drugs and Poisons Chapter No. 32 Dated: 11/3/2011 At Time: 8:39:18
Introduction and theoretical background
509
Stray-light errors are more likely to be observed near the wavelength limits of an instrument, at which the radiation intensity of the source and the efficiency of the optical system are reduced, especially below 220 nm and at the crossover point between the UVand the visible lamps (about 320–400 nm). Errors may become serious if the solvent absorbs strongly or if a strongly absorbing sample is measured by difference spectrophotometry. Solvent effects
The solvent often exerts a profound influence on the quality and shape of the spectrum. For example, many aromatic chromophores display vibrational fine structure in non-polar solvents, whereas in more polar solvents this fine structure is absent because of solute–solvent interaction effects (see also Fig. 32.1). A classic case is phenol and related compounds, which have different spectra in cyclohexane and in neutral aqueous solution. In aqueous solutions, the pH exerts a profound effect on ionisable chromophores because of the differing extent of conjugation in the ionised and the non-ionised chromophore. The quality of spectral measurement is affected directly by the type and purity of the solvent used. Each solvent has a cut-off wavelength (which corresponds to about 10% transmittance) and this varies with solvent purity (Table 32.1). A solvent should not be used below its cutoff wavelength, even though reference-cell compensation is employed, because of the greater risk of stray-light effects. The UV spectra of some solvents are illustrated in Fig. 32.3. Some cautionary comments may be appropriate at this point. It is better to use single- or double-distilled water, and to avoid deionised water, which can be contaminated with absorptive fragments of ionexchange resin or contain bacterial metabolites; these can contribute significantly to non-specific absorption at low wavelengths. Ethanol is normally used as the 96% v/v strength, since dehydrated alcohol is usually contaminated with traces of benzene added to form the azeotropic mixture for distillation. Acetonitrile can vary noticeably in quality, depending on the supplier; the grade supplied for use in HPLC is usually to be recommended. Acetone, sometimes used to clean cells, is highly absorptive and not always easily removed, despite its volatility and aqueous solubility. Chloroform and carbon tetrachloride absorb strongly at about 250 nm and should therefore be used only for
Table 32.1 Cut-off points equivalent to 10% transmittance for spectroscopic solvents
Figure 32.3 UV absorption of solvents (HPLC grade unless otherwise stated): A, acetonitrile (far-UV grade); B, methyl t-butyl ether; C, acetonitrile; D, 1chlorobutane; E, methylene chloride; F, acetic acid (AR grade); G, ethyl acetate; H, acetone; I, hexane; J, iso-octane; K, methanol; L, tetrahydrofuran; M, chloroform; N, diethylamine (AR grade).
measurements at wavelengths above about 280 nm. Given the safety considerations of chlorinated solvents, use of these is best avoided if possible. Ether, although transparent down to 220 nm, presents particular problems because of its volatility (unstable standard solutions) and inflammability. Although absorptivity is considered to be relatively insensitive to temperature changes, organic solvents in general suffer from high temperature coefficients of expansion, so that for ultimate precision a cell provided with a thermostat may be required.
Solvent
Wavelength (nm)
Water (distilled) or dilute inorganic acid
190
Acetonitrile (HPLC, far-UV grade)
200
Acetonitrile
210
Butyl alcohol
210
Cyclohexane
210
Fluorescence spectrophotometry: theoretical background
Ethanol (96% v/v)
210
General considerations
Heptane
210
Hexane
210
Isopropyl alcohol
210
Methanol
210
Ether
220
Sodium hydroxide (0.2 mol/L)
225
Ethylene dichloride
230
Methylene chloride
235
Chloroform (stabilised with ethanol)
245
Carbon tetrachloride
265
N,N-Dimethylformamide
270
Benzene
280
Pyridine
305
Acetone
330
Molecular fluorescence is an emission process in which molecules are excited by the absorption of electromagnetic radiation. The excited species then relax to the ground state, giving up their excess energy as photons. There are several ways in which an excited molecule can give up its excess energy and relax to its ground state. Two of the most important of these mechanisms are non-radiative relaxation and fluorescent relaxation. Non-radiative relaxation can occur through collisions between excited molecules and molecules of the solvent, giving excess energy to solvent molecules. When relaxation takes place by fluorescence, bands of radiation are produced as the excited molecules relax to several energy states, which are very close in energy level and thus in wavelength (Fig. 32.4). Fluorescence occurs only from the lowest vibrational level of an excited electronic state. Note that molecular fluorescence bands are made up largely of lines that are longer in wavelength (lower in energy) than the band of absorbed radiation responsible for their excitation. This shift to
Clarke's Analysis of Drugs and Poisons Chapter No. 32 Dated: 11/3/2011 At Time: 8:39:18
510
Ultraviolet, Visible and Fluorescence Spectrophotometry
Figure 32.4 Energy-level diagram showing some of the energy changes that occur during (A) absorption, (B) non-radiative relaxation and (C) fluorescence by a molecular species.
longer wavelength is sometimes called the Stokes shift. For that reason, the absorption or excitation spectrum and the fluorescence spectrum for a compound often appear as approximate mirror images of each other. The most useful region for the fluorescence technique is 200– 800 nm. Fluorescence spectrophotometry is usually the method of choice for quantitative analytical purposes if applicable. It has assumed a major role in analysis, particularly the determination of trace contaminants in our environment, industries and bodies, because for applicable compounds fluorescence spectrometry gives high sensitivity and high specificity. The selectivity of fluorescence methods is greater than that of absorption methods, as fewer substances fluoresce than absorb radiation in the UV or visible region. Furthermore, fluorescence is more selective because both the emission and the absorption spectra can be obtained. Fluorescence is usually also more sensitive than absorption methods, as it is always easier to measure a small signal against a very small zero background than to measure a small difference between large signals. However, the phenomenon of fluorescence itself is subject to more rigorous constraints on molecular structure than is absorption.
fluoresces whereas nitrobenzene does not). The molecular grouping responsible for fluorescence is sometimes described as a fluorophore. Fluorescence is particularly favoured in rigid molecules, as molecular rigidity reduces deactivation by non-radiative processes (there being fewer internal vibrations). This is also why certain organic chelating agents are more fluorescent when complexed with a metal ion. Laws of fluorescence spectrophotometry
The power of fluorescent radiation If is proportional to the radiant power of the excitation beam absorbed by the system: 0
I f ¼ K ðI 0 IÞ
ð32:6Þ
The constant K0 depends upon the quantum efficiency of the fluorescence. Beer’s law can be used to relate If to the molar concentration c of the fluorescing molecule: I ¼ 10 bc I0
ð32:7Þ
Nomenclature
The term quantum efficiency used in fluorescence is quantified by the quantum yield (i.e. the ratio of the number of molecules that fluoresce to the total number of excited molecules). Highly fluorescent molecules can have quantum efficiencies that approach unity. Many drugs possess rather high quantum efficiencies for fluorescence, such as quinine and lysergic acid diethylamide (LSD). All absorbing molecules have the potential to fluoresce. They do so if fluorescent emission occurs at a greater rate than relaxation by nonradiative pathways. The kind of relaxation process is highly dependent on the molecular structure. Compounds that contain aromatic rings give the most intense and most useful fluorescence emission. Substitution on an aromatic ring causes shifts in the excitation wavelength spectrum and in fluorescence efficiency. Substituents such as –NH2, –OH, –OCH3 and –NHCH3 groups often enhance fluorescence, while –Cl, –Br, –I, –NO2 or –COOH are electron-withdrawing groups that can lead to reduction or absence of fluorescence (e.g. aniline
Substituting equation (32.7) into (32.6) we obtain: 0 I f ¼ K I 0 1 10 bc
ð32:8Þ
After expansion of the exponential term, and provided that ebc 150, (b) 93, (c) 63, (d) 45 and (e) 32 mm. A, original spectra; B, spectra after standard normal variate (SNV) transformation.
accuracy of 0.3 nm, while an FT instrument (data stored at 12 cm1 intervals) has an accuracy of 2 cm1. Some form of wavelength interpolation between stored data points is essential when locating the position of minima (see Wavelength repeatability). NIST and NIST-traceable wavelength standards for transmission measurements are also available; however, an inexpensive alternative is to use trichloromethane, which exhibits sharp absorption peaks at 1152.1, 1410.2, 1619.9 and 1861.2 nm (Busch et al. 2000). The British Pharmacopoeia recommends the use of methylene chloride (dichloromethane) as a standard. Figure 34.8 shows the spectrum of methylene chloride with the important absorption peaks labelled. For instruments in which transmittance or transflectance measurements are not possible, a spectrum may be measured by adding titanium dioxide to a few millilitres of methylene chloride and recording the diffuse reflectance spectrum.
wavelengths of minima or maxima in the second-derivative absorbance spectra. Materials with well-defined narrow second-derivative peaks, such as the mixed lanthanide oxides wavelength standard (suitable for wavelengths below 2200 nm), are ideal. Alternatively, compounds such as ascorbic acid, aspartame, benzoic acid, salicylic acid, sucrose and talc may be used. Some form of wavelength interpolation between stored data points is essential when locating the position of peaks. Fitting a quadratic curve to three consecutive data points that encompass a peak and calculating the exact position from the equation works well. Let the fitted equation be A ¼ a þ bl þ cl2, where A is the second-derivative absorbance and a, b and c are the fitted coefficients; then peak ¼ b/2c. Typical standard deviations for peak wavelengths are 500 data points) should be carried out. While this is possible with two or three wavelengths, the time taken for larger numbers of wavelengths makes this impractical. For complex samples, in which many components vary, whole-spectrum techniques, such as PCR and PLSR, are preferred. Care must be taken not to over-fit the calibration data by using too many parameters in the model. The final step, validation, is the most important. The model produced is used to predict the values for the test set that can then be compared against their reference values. Only if the values predicted for this independent data set are satisfactory can the model be considered acceptable. Plots of NIR predicted values versus reference values for both the calibration and test sets (e.g. Fig. 34.18) should be examined. Visual
Figure 34.18 Typical validation plot of near-infrared predicted value against reference value.
inspection easily reveals any problems with the data sets and model, such as outliers or curvature. Ideally, both plots should be straight lines with slope unity and intercept zero. A good calibration plot but poor validation plot suggests over-fitting of the model. Numerically, the fit can be assessed by calculating the standard error of calibration (SEC) and standard error of prediction (SEP): sffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi Pn ^ 2 i¼1 ðX i X i Þ SE ¼ D ^ is the NIR predicted value for the where Xi is the reference value and X ith sample of the calibration set (for SEC) or validation set (for SEP), n is the number of samples in the data set being considered, and D is the number of degrees of freedom (for the calibration set this will be n minus the number of parameters fixed in the model, e.g. number of wavelengths used for MLR plus 1, number of components used plus 1 (if data centred) for PCR or PLSR). For the validation set, D ¼ n. For a good model, SEC and SEP should be similar and not much larger than the standard error of the reference method. SEP is a measure of the accuracy (how close the NIR predicted values are to the reference values) for the method and, of course, needs to be compatible with the intended purpose to which the procedure is to be put. Moffat et al. (2000) have shown how quantitative NIR can meet the requirements of the International Conference on Harmonisation’s Guidelines on Validation of Analytical Procedures for the assay of an intact pharmaceutical product in terms of specificity, linearity, range, accuracy, precision and repeatability.
Near-infrared imaging The coupling of an NIR spectrometer and scanning microscope allows spectroscopic imaging of surfaces. Not only can small samples be identified, but information about the distribution of different chemical components and their particle size can be obtained. Commercial reflectance NIR microscopy mapping systems are based on one of three data acquisition methods. In point mapping, a single detector is used to measure a spectrum at a single point on the sample surface and then the sample position is moved and the process is repeated until the complete area has been scanned. Line mapping works in a similar way except that a linear array of detectors is used so that data are more quickly acquired. In global mapping, the whole sample area is measured simultaneously using some form of tuneable filter to control the wavelength selection. Typically spectral resolution of 4–16 cm1 and spatial resolution of >10 mm can be achieved. Because of the nature of NIR diffuse reflection, it is most probably not meaningful to work at spatial
Clarke's Analysis of Drugs and Poisons Chapter No. 34 Dated: 10/3/2011 At Time: 14:9:54
Resources
Figure 34.19 NIR image of a cross-section of a ranitidine tablet. Each pixel is 30 30 mm2. The image is 60 60 pixels (i.e. 1.8 1.8 mm2). The map shows the value (coded in different shades of grey) of the correlation coefficient of the NIR spectrum at each pixel when compared with that of a pure sample of ska.) ranitidine. (Reproduced with permission of RA Watt and A Duszyn
resolutions of 99
—
Relative sensitivity — —
1
1/2
99.98
600.00
1.00
3
1/2
0.0
639.98
1.21
C
1/2
1.11
150.86
15
N
1/2
0.37
60.80
19
F
1/2
H H
13
29 31 2
Si P
H
14
1/2 1/2 1
N
1
23
Na
3/2
35
Cl
3/2
O
5/2
17
1.59 102 1.04 103
100.0
564.46
8.30 101
4.7
119.19
7.84 10
100.0
242.88
6.63 10
0.015 99.63 100.0 75.53 0.037
92.10 43.34 158.71 58.79 81.34
565
Fragment
d(1H)
Cyclopropyl CH2
–0.2–0.8
CCH2C
0.4–2.4
CH3C
0.5–2.0
CNH
0.5–3.0
COH
0.5–5.0
CH3C=C
1.5–2.3
CCH2C=C
1.6–2.0
CH3S; CH3CO
1.7–2.7
3
CH3Ph
20–30
2
CH3N
22–31
CCH2S
23–30
9.65 104 1.01 101 9.25 102 4.70 103 2.91 102
important in biology, including many studies in vivo. Many other spin-1/2 nuclei, such as 29Si, 119Sn, 129Xe, 195Pt and 199Hg, have found much use in specialist applications. Nuclei with I > 1/2 are quadrupolar and, in general, give broad NMR lines, but in some cases useful information can be gleaned. Examples include 2H NMR in liquid crystals, 14 N NMR in heterocyclic chemistry, and 23Na NMR studies of intraand extracellular sodium ions.
C=CCH2C=C
23–30
CCH2CO
23–35
CCH(Ph)C
25–30
CCH2Ph
25–32
CCH2N
25–33
C=CCH2CO
28–37
C=CCH2Ph
30–38
CH3O
32–41
NCH2CO
32–45
CCH2Cl; CCH2O
34–43
CH3OCO
35–38
COCH2Ph
35–42
CHNCO
35–43
Parameters from an NMR spectrum
CCH2OCO
35–45
Chemical shifts
NCH2N
35–51
As mentioned earlier, not all nuclei of a given isotope resonate at exactly the same frequency. This is because, in a molecule, a given atomic nucleus is surrounded by electrons which also possess a magnetic moment, and these provide a fluctuating magnetic field that opposes the main field of the NMR magnet. As a consequence, the nuclei are shielded from the main magnetic field and require a higher field to bring them to resonance and thus they can be considered to have higher Larmor frequencies. The degree of shielding depends on the electron distribution around the nucleus and hence on the chemical environment. The different degrees of shielding are known as chemical shifts. Thus, interpretation of chemical shift values allows identification of molecular structural fragments. Chemical shifts are measured relative to that of a reference substance usually placed into the sample. For 1H and 13C shifts in organic solvents, this is tetramethylsilane (TMS). The chemical shift is then defined as d(H) ¼ (difference in the resonance frequency in hertz between the analyte and TMS) 106/(operating frequency of the spectrometer). Chemical shifts are thus quoted in parts per million (ppm) and are independent of the operating frequency of the spectrometer, which allows comparisons irrespective of magnetic field strength. For aqueous samples, an alternative reference compound is used, of which trimethylsilyl [2,2,3,3-2H4]propionic acid sodium salt (TSP) is the most common example. The chemical shifts for TMS and TSP are set arbitrarily to zero. Typical 1H and 13C NMR chemical shifts of a variety of important molecular fragments are shown in Tables 36.2 and 36.3, respectively.
NCH2Ph
36–46
C=CH
36–85
C=CCH2O
40–52
Indirect (J) spin–spin coupling
The resonance lines of individual nuclei can show further splitting because of indirect spin–spin coupling. Given the symbol J, this is measured in hertz and is independent of the observation frequency. Such spin coupling arises from a magnetic interaction between NMRactive nuclei and is transmitted via the intervening electrons, hence the term ‘indirect’. Coupling is observed only within a molecule. Thus for
COCH2O
40–55
OCH2O
45–63
OCH2Ph
47–56
NCH2O
48–59
NHCO
50–90
Aromatic H
60–90
OCHO
80–83
C=NOH
80–110
CHO
95–100
COOH
95–130
two spin-1/2 nuclei, such as protons, the resonance line for each proton is split into a doublet, the two lines corresponding to the two possible orientations of the adjacent proton relative to the magnetic field. For extended coupling chains, each component of a doublet can be split further into doublets of doublets and so on. If a given proton is adjacent to two equivalent other protons (as in a CH2 group) then, of the four possible spin orientations of the two protons, two of them are identical and a 1 : 2:1 triplet results. For such ‘first-order’ systems, the multiplicity can be deduced on the basis of Pascal’s triangle according to the number of equivalent coupled nuclei. In situations where the chemical shift difference between the protons is large compared with the J-coupling, this simple rule applies. For situations where the chemical shift in hertz between coupled partners is not large compared with the magnitude of the coupling constant (d/J C4 and lactones
g-BP
Amino acids, some primary amines and furans
b-PM
Largest range of acids, alcohols, barbiturates, diols, epoxides, esters, halocarbons, ketones, lactones, terpenes; first choice for chiral method development
2,3-di-O-methyl-6-O-t-butyldimethylsilyl are also available embedded (usually 25–35% by weight) in 20% phenyl-PSX, another intermediate polarity stationary phase. These columns are useful for separating positional isomers (phenols, xylenes, etc.), as well as enantiomers. Solute–stationary phase interactions
For liquid stationary phases, three major types of interaction determine chromatographic elution: dispersion, dipole–dipole interaction and hydrogen bonding. Table 40.2 shows the contribution of each of these interactions for the common types of liquid stationary phases. It should be remembered that hydrogen bonding interactions are considerably stronger than dipole– dipole interactions, which are themselves stronger than dispersion interactions. Thus, although the dispersion interaction between the various stationary phases is listed as strong or very strong, and the hydrogen bonding interactions as weak or moderate, if the analyte has functional groups that can undergo hydrogen bonding with the particular stationary phase employed, the hydrogen bonding interaction is likely to be stronger than interaction by dispersion. However, the reverse is true if the analyte also has a high proportion of groups in its molecular structure that can participate in dispersion interactions, as would be the case, for example, for long-chain fatty acids. Dispersion is the dominant interaction for all PSX and PEG stationary phases; it can be simplified into the concept that the more volatile the compound (the lower its boiling point), the more likely it is to be in the mobile phase and so the faster it elutes from the column. Although this holds true for groups of compounds with similar functional groups or
Table 40.2 Contribution of different types of interactions to solute separation on GC stationary phases Functional group
Type of interaction Dispersion
Dipole–dipole Hydrogen bonding
Methyl-PSX
Strong
None
None
Phenyl-PSX
Very strong
None
Weak
Cyanopropyl-PSX
Strong
Very strong
Moderate
Trifluoropropyl-PSX
Strong
Moderate
Weak
PEG
Strong
Strong
Moderate
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:4
640
Gas Chromatography
within homologous series, it cannot be applied universally. In general, a difference of 30 C in boiling point is sufficient to predict and maintain elution order, but differences of less than 10 C can be overturned by the influence of other interactions. Dipole–dipole interactions of PEG phases and the cyanopropyl- and trifluoropropyl-substituted PSXs enable these phases to separate solute molecules that have different dipole moments. Such solutes are those with positional isomers of electronegative groups, such as pesticides, halocarbons and many pharmaceuticals. Moderate hydrogen bonding is exhibited by PEGs and cyanopropylsubstituted PSXs, with less marked effects shown by phenyl- and trifluoropropyl-substituted PSXs. Functional groups that exhibit strong hydrogen bonding include alcohols, carboxylic acids and amines; aldehydes, esters and ketones generally have less effect; hydrocarbons, halocarbons and ethers produce negligible hydrogen bonding. Although the amount of separation obtained through dipole–dipole interactions or through hydrogen bonding can be difficult to predict, resolution of compounds with smaller differences in dipole moments or in hydrogen bonding strengths requires larger percentages of siloxane substitution. McReynolds constants
The retention behaviour of five carefully selected probe compounds (benzene, butanol, pentan-2-one, nitropropane and pyridine) has traditionally been used to classify stationary phases in terms of their polarity (McReynolds 1970; Rorschneider 1966). The retention indices of each of these five reference compounds are measured on the stationary phase being tested, and then compared with those obtained under the same conditions on squalene (a standard non-polar phase). The
differences in the retention indices between the two phases (DI) for the five probe compounds are added together to give a constant, known as the McReynolds constant, which is used to compare the ability of stationary phases to separate different classes of compounds. Phases that provide McReynolds values of 4 can be substituted freely for each other; those differing by 10 units generally yield similar separations. Phases with McReynolds values below 100 are considered non-polar, those above 400 indicate a highly polar phase and values between 100 and 400 an intermediate polarity. Table 40.3 shows the McReynolds constants, operating temperature range and example applications for the most popular stationary phases. DI values for individual probes indicate the deviation from boiling point order and consequently represent the contribution of forces other than dispersion to elution for that probe. The probes are chosen to represent different functional groups as follows: n n n n n
Benzene for aromatics and olefins (p-type interactions) Butan-1-ol for alcohols, nitriles, carboxylic acids and diols (electronattracting effect) Pentan-2-one for ketones, ethers, aldehydes, esters, epoxides and dimethylamino derivatives (dipole–dipole effect) Nitropropane for nitro and nitrile derivatives (electron-donating effect) Pyridine for bases (non-bonding electron attraction and hydrogenbonding effects).
Moffat et al. (1974a) devised a system to assess the effectiveness of liquid stationary phases in packed columns by calculating the discriminating power, and examined a number of phases commonly used in toxicology
Table 40.3 Polarity (McReynolds values) of some common stationary phases, and example applications P Temperature McReynolds values(b) Capillary phase(a) DI Applications range y0 z0 u0 s0 x0 (min./max.) SPB-octyl
60/300
3
14
11
12
11
51
*-1
60/320
4
58
43
56
38
199
Amines, hydrocarbons, pesticides, PCBs, phenols, sulfur compounds, flavours, fragrances
*-5
60/320
19
74
64
93
62
312
Alkaloids, drugs, FAMEs, halogenated compounds, aromatic compounds
*-1301
20/280
69
113
111
171
128
592
Aroclors, alcohols, phenols, volatile organic acids
0/300
101
146
151
219
202
728
Aroclors, amines, pesticides, drugs Aroclors, herbicides, pesticides, trimethylsilyl (TMS) sugars
*-35
Separates by boiling point, polychlorinated biphenyls (PCBs)
*-1701
10/280
67
170
153
228
171
789
*-50, *-17
30/310
125
175
183
268
220
971
*-210
45/250
178
204
208
305
280
1175
*-225
40/230
146
238
358
468
310
1520
FAMEs, alditol acetates, neutral sterols
*-23
40/250
228
369
338
492
386
1813
cis–trans-FAMEs, stereoisomers
*-wax, *-20M
35/280
305
551
360
562
484
2262
Alcohols, free acids, essential oils, ethers, glycols, solvents, primary amines
*-FFAP
50/250
340
580
397
602
627
2546
Acids, alcohols, aldehydes, acrylates, nitriles
Nukol
60/200
314
569
372
578
504
2337
Alcohols, free acids, essential oils, ethers, glycols, solvents
*-2330
10/250
382
610
506
710
591
2799
cis–trans-FAMEs, positional isomers
*-2380
10/275
402
629
520
744
623
2918
cis–trans-FAMEs, positional isomers, alditol acetates
*-2340
25/250
419
654
541
758
637
3009
cis–trans-FAMEs, positional isomers
—
20/200
496
746
590
837
835
3504
Acids, esters, phenols, terpenoids
—
100/200
537
787
643
903
889
3759
TMS or methyl sugars, acidic drugs
TCEP
10/145
594
857
759
1031
917
4158
a-Cyclodextrin in 35% phenyl-PSX
30/240
102
243
142
221
170
878
Enantiomers and isomers (see Table 40.1)
b-Cyclodextrin in 35% phenyl-PSX
30/240
119
264
154
134
187
858
Enantiomers and isomers (see Table 40.1)
(a)
Drugs, glycols, pesticides, steroids Aldehydes, ketones, organochlorines, organophosphates
Flavours, fragrances, essential oils
* is the proprietary prefix for the phase, for example: * = HP supplied by Hewlett Packard/Agilent; * = DB supplied by J&W; * = CPSil supplied by Chrompack; * = RT supplied by Resteck; * = SP supplied by Supelco; * = OV supplied by Ohio Valley. This list is not intended to be exhaustive. x = benzene; y 0 = butan-1-ol; z 0 = pentan-2-one; u 0 = 1-nitropropane; s 0 = pyridine.
(b) 0
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:4
Gas chromatography columns
641
Installing, conditioning and maintaining columns
onto the column. Portions can be cut periodically from the top of the guard column as deterioration in chromatography requires, without any appreciable loss of resolution from the analytical column. A retention gap is used to improve peak shape when poor chromatography is the result either of a large injection volume (>2 mL) or of solvent–stationary phase polarity mismatches. Greatest improvement is seen in early eluting peaks, or for solutes with similar polarity to that of the solvent.
Column installation
Maximum operating temperatures
(Moffat et al. 1974b). Contrary to popular belief, it was shown that one column could be used to elute all the drugs studied, and that for screening purposes a single column, either SE-30 or OV-17 (100% dimethyl-PSX or 5% phenyl-PSX capillary equivalents), was sufficient for the reliable identification of drugs.
A GC column is attached at one end to the injector and at the other end to the detector. Attachment is typically via a nut and ferrule, the nut attaching to a screwthread on the injector and detector. As the nut is tightened, the ferrule is compressed and helps produce a gastight fitting. Fused-silica capillaries should have their ends freshly cut after insertion through ferrules, to eliminate blockages. The injector end of the column should be fitted first, adjusting the height of the protrusion above the ferrule according to the type of inlet being used, then tightening the fittings just enough to prevent leakage when tested with a proprietary leak-testing fluid (not soap solution, which leaves a residue). The detector end of the capillary column may be immersed into a small tube of methanol to ensure adequate flow, and the capillary end re-cut and then attached to the detector and checked for leaks. The detector is activated and the column tested at room temperature with an injection of 1 or 2 mL of methane, when a needle-sharp peak should be obtained. When the carrier gas pressure has been adjusted to give a flow of approximately 1–2 mL/min of carrier gas, the column may be heated and a test mixture injected. Commercial columns are invariably supplied with a chromatogram obtained from a test mixture, and it should be possible to obtain a performance at least equal to that supplied. Various test mixtures are used, including a mixture of dimethylphenol and dimethylaniline with straight-chain paraffins. Any acidity or alkalinity of the column is apparent in loss of the peak shape of the amine or phenol. The efficiency obtained is a function of the entire chromatographic system. Poor efficiency or peak shape often results from a non-swept volume somewhere in the system. It may be necessary to add an additional gas supply to the column outlet to ensure that the detector is purged effectively, because most detectors are designed to operate with packed columns and a flow rate of about 30 mL/min, as opposed to the 1 or 2 mL/min delivered by a capillary column (see Detector systems below). Column conditioning
Modern capillary columns require only minimal conditioning before use to remove volatile impurities that remain from the manufacturing process. For thorough conditioning, the column should be installed in the injector port only, with the detector end disconnected. With the column at room temperature, a low carrier gas pressure (14–35 kPa) should be maintained for half an hour to purge oxygen from the system. The temperature may then be raised by about 1 /min until a temperature about 10 C above the desired maximum operating temperature has been reached, and the column is maintained at this for 2 h. Care must be taken not to exceed the maximum operating temperature. After conditioning, the column is connected to the detector, and a period of further conditioning undertaken only if the background signal is excessive. Some phases are particularly oxygen sensitive and can be ruined by careless conditioning. A constrictor fitted to the detector helps prevent back diffusion of oxygen if air or oxygen is supplied to the detector. Guard columns and retention gaps
A guard column and a retention gap are essentially the same thing, but they are installed to serve different purposes. These 1–10 m lengths of fused-silica tubing are attached to the front of the chromatography column via a press-snap connector or zero dead-volume union, and then installed into the injector port. The surface of the silica is deactivated to minimise solute interactions, but no stationary phase is added. The tubing diameter should be the same as that of the column, but if different it should ideally be of a wider bore. The function of a guard column is to trap deposits of non-volatile residues, preventing them from contaminating the analytical column. Solutes are not retained by the guard column (since there is no stationary phase) and pass directly
Maximum operating temperatures for stationary phases are usually quoted assuming isothermal operation with a flame ionisation detector (see Table 40.3). Other detectors may impose different limits, the mass spectrometer being much more susceptible to bleeding of the stationary phase than the thermal conductivity detector. All phases bleed at high temperatures through loss of smaller-sized (and hence lower-boiling) polymer chains, although normally this is not noticeable. Operating temperature has a profound effect on column life, particularly for capillary columns. Loss of stationary phase or breakdown of the thin film into pools exposes part of the tubing surface and results in serious loss of performance. Additionally, in columns that contain PSX phases with two different functional groups, one group (usually that which confers additional polarity) is preferentially lost. This results in a change of relative separation (or in RI – see discussion below) as well as a loss of resolution. The temperature limit of a column may be influenced by the deactivation procedure used in production, rather than by the stationary phase itself. Newer silica columns have a very low metal oxide content, thought to act as a catalyst for the degradation of both sample and stationary phase, and thus enable phases to be run at higher temperatures. Fused-silica capillary columns have a protective external coating of polyimide that is slowly degraded at elevated temperatures (maximum temperature originally 360 C, now up to 400 C), which can also limit column life. However, separations are usually achieved at much lower temperatures. Temperature programming
For complex mixtures with components of widely varying retention characteristics, it is often impractical to choose a column temperature that allows all the components to be resolved. Increasing the column temperature throughout the analysis dramatically reduces the time taken for higher-boiling compounds to elute, and simultaneously improves the sensitivity of the assay, as the peaks are remarkably sharper. If the early eluting compounds are resolved inadequately, a lower starting temperature or slower initial ramp should be used, taking care to observe the temperature requirements of the type of injector used. All instruments currently manufactured are available with a temperature program option, and a multi-ramp programmer is particularly useful for capillary chromatography. The first ramp can be used during splitless injection (see later) to bring the column rapidly up to the initial chromatography temperature, followed by a slower analytical ramp to perform the separation. One problem with temperature programming is that the backpressure increases with temperature and reduces the carrier gas flow if a mass-flow controller is not used. For polar stationary phases, the polarity increases with temperature, which causes distortion of RI data (see discussion on the use of RIs below). Column bleed also increases, which results in a rising baseline that can be mitigated somewhat by adequately conditioning the column before use. Evaluating column performance A column’s performance is assessed on the basis of its efficiency (the narrowness of a peak), the peak shape (whether it tails or fronts) and its ability to resolve compound mixtures. This section deals with separation theory, and the reader may find it useful to refer at intervals to Fig. 40.3. Retention time
Retention time (tR) is the time taken for a given solute to travel through the column, and is the time assigned to the corresponding peak on the chromatograph. It is a measure of the amount of time that the solute spends in the column, and is therefore the sum of time spent in both the stationary and the mobile phases.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:5
642
Gas Chromatography
Figure 40.3 A and B are symmetrical peaks that show the measurement of significant parameters: W, width at the base of the peak; W1/2, width at half peak height; tR(A) and tR(B), retention times of peaks A and B, respectively; DtR, the difference in retention time between A and B; XYZ, a line drawn at 10% of peak height. Peak C is symptomatic of column overload with solute. Peak D is symptomatic of degradation of a thermally unstable solute. Peak E is symptomatic of adherence to active sites in the injection port or on the column. Resolution of two compounds A and B. F and G: peaks A and B have identical retention times (5.59 and 5.77 min, respectively), but in G the peaks are narrower (W1/2 = 0.071 versus 0.126 min, respectively), and are fully resolved (Rs = 1.50 versus 0.84, respectively). H: the peak widths of A and B are the same as in G, but the retention time of peak B is later (5.83 versus 5.77 min, respectively). Again, the peaks are fully resolved and Rs is larger than in G (Rs = 1.99 versus 1.50).
Retention time of a non-retained compound or hold-up time (tM or t0) The retention time, tM, is the time taken for a non-retained solute to
travel along the column; it represents the transit time for the mobile phase (carrier gas) in the column and is a column-specific parameter, applicable only under the prevailing conditions of gas flow and oven temperature. It is the same for all solutes on the column, and no other peak can be expected to elute earlier than this time. tM is obtained by injecting a non-retained compound suitable for the detector system being used (butane or methane for flame ionisation detection (FID) or thermal conductivity detection (TCD); acetonitrile for nitrogen– phosphorus detection (NPD); methylene chloride for electron-capture detection (ECD); vinyl chloride for photoionisation detection (PID) or electrolytic conductivity detection (ELCD)). Average linear velocity
The average linear velocity (m) represents the average speed of carrier gas through the column, usually expressed in cm/s, and is considered more meaningful than measuring the flow (usually expressed in mL/min) at the column effluent, since flow is dependent on column diameter. This term directly influences solute retention times and column efficiency. Velocity is controlled by altering the column head pressure, and is calculated from equation (40.1): mðcm=sÞ ¼
L tM
ð40:1Þ
where L is the column length (cm), and tM is the retention time (in seconds) of a non-retained solute. Retention factor
The retention factor (k) is the ratio of the amount of time a solute spends in the stationary and mobile phases and is calculated from tR and tM using equation (40.2): k¼
tR tM t0 R ¼ tM tM
ð40:2Þ
where tM is the retention time of a non-retained solute, tR is the retention time of the solute and t 0 R is the adjusted retention time of the solute. Since all compounds spend an identical time in the mobile phase, k is a measure of retention by the stationary phase. A compound with a retention factor of 4 spends twice as much time in the stationary phase (but not twice as much time on the column) as a compound with a retention factor of 2. Thus, k provides relative rather than absolute information, and is to a large degree independent of the operating conditions. Separation factor
The separation factor (a) is a measure of the time interval between two peaks. If a equals 1, then the peaks have the same retention time and coelute. Separation factor is calculated using the equation: k2 ð40:3Þ a¼ k1 where k1 is the retention factor of the first peak, and k2 the retention factor for the second peak. The value of a does not indicate whether the peaks are resolved completely from one another, however. Two peaks may have only 0.01 min between them on one column but still be resolved completely, while on another column they may have 0.1 min between them but not be resolved adequately (refer to Fig. 40.3). Number of theoretical plates or column efficiency
The theoretical plate is an indirect measure of peak width at a specific retention time. Higher plate numbers indicate greater column efficiency and narrower peaks. The number of plates per metre of column (N) is calculated from either form of equation (40.4): ð40:4aÞ N ¼ 16ðt R =wb Þ2 N ¼ 5:54ðt R =wh Þ2
ð40:4bÞ
where tR is the time from injection to peak maximum for the solute, wb is the peak width at base in units of time, and wh is the peak width at half height in units of time.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:5
Gas chromatography columns Efficiency is thus a function of the column dimensions (diameter, length, film thickness or loading), the type of carrier gas and its flow, and the chemical nature of the solute and the stationary phase. For most applications in drug analysis, the chromatogram contains only two or three compounds, but an efficient column maximises the probability that a peak consists of only one compound and that it is the compound of interest. Other more complex separations (e.g. of flavours or trace residues in foods) may require two-dimensional chromatography or heartcutting (see later) to obtain sufficient separation.
Table 40.4 Relationship of capacity (ng solute)(a) of GC columns with diameter and film thickness Film thickness df (mm)
Column internal diameter (mm) 0.18–0.20
0.25
0.32
0.53
25–50
35–75
50–100
50–100
75–125
100–250
100–200
125–250
250–500
250–500
500–1000
400–600
500–800
1000–2000
1000–1500
1200–2000
2000–3000
0.10
20–35
0.25
35–75
0.50
75–150
Resolution
1.00
150–250
200–300
Resolution (R) takes into account both retention time and the peak width. For any pair of compounds, resolution can be calculated using either form of equation (40.5): t R2 tR1 ð40:5aÞ R ¼ 1:18 wh1 wh2
3.00
—
5.00
—
R¼2
t R2 tR1 wb1 wb2
where a ¼ separation factor, k ¼ retention factor, N ¼ theoretical number of plates. Phase ratio (b) The phase ratio of a column is a calculated term relating the column radius (r) in millimeters and the film thickness (df ) in micrometres: r 2df
ð40:6aÞ
If all other conditions are held constant, then changes in phase ratio can be used to predict expected shifts in retention of solutes using equation (40.6b): kb ¼
kr 2df
(a)
Capacity is defined as the maximum amount of solute that can be injected without peak broadening by >10% at half-height. Approximate values are given for capacity (ng) per component. Actual sample capacity depends on the operating conditions, and the polarity of the stationary phase and the solute (polar phases and solutes give lower values).
ð40:5bÞ
where suffixes 1 and 2 refer to the two peaks being evaluated; tR ¼ retention time; wh ¼ peak width at half height in units of time; wb ¼ peak width at base in units of time An R value of 1.5 indicates baseline resolution, with numbers above 1.5 indicating the presence of baseline between the peaks (higher values indicate less overlap between peaks). In practice a value of about 1.2 is needed to be able to distinguish between two peaks occurring independently and a value of >1.0 to be able to quantify either peak reliably in the presence of the other. Sometimes the parameter percentage resolution is used, as this concept is easier to visualise (calculated by dividing the height of the valley between the peaks by the total peak height). equations (40.5a) and (40.5b) allow the operator to calculate resolution directly from the chromatogram, but they give little indication of the factors that contribute to it, or the parameters that can be modified by the analyst. A preferred relation is equation (40.7c), which shows that resolution is a product of three parameters, selectivity, capacity and the square root of the efficiency. pffiffiffiffi a1 k N R¼ ð40:5cÞ a kþ1 4
b¼
643
ð40:6bÞ
where k ¼ retention factor. Thus, to increase solute retention the phase ratio must be decreased, which can be brought about either by decreasing column diameter or by increasing film thickness. Sometimes it will be necessary to alter either the column diameter (e.g. to reduce flow though a detector) or to alter film thickness (e.g. to increase efficiency), but as long as the phase ratio remains constant then these changes can be accomplished without compromising separation. Table 40.8 shows calculated phase ratios for the most common sizes of capillary columns and loadings. Peak shape or asymmetry
A well-designed GC system should give symmetrical peaks, as tailing or fronting adversely affects resolution. Tailing may result from non-swept
volume in the system or from component–stationary phase or component–support interactions. Tailing of polar compounds can often be remedied by the use of a more polar stationary phase. Fronting (shark’s fin peaks) is usually caused by overloading, particularly with capillary columns, and can be resolved either by making a smaller injection or by using a column with a higher stationary phase ratio. Column capacity is the maximum amount of a solute that can be chromatographed successfully without loss of peak shape. Table 40.4 shows the relationship between column capacity, film thickness and column diameter. Peak fronting caused by thermal decomposition can be reduced by either lowering the injection temperature or using a cold on-column injector system. Peak shape is usually expressed by the peak asymmetry (As). In Fig. 40.3, the peak asymmetry factor for substance B is given by equation (40.7): As ¼
YZ XY
ð40:7Þ
where a vertical line is drawn through the peak maximum and XYZ is drawn at 10% of the peak height. A symmetrical peak has As ¼ 1. Use of retention indices If gas chromatographic retention data are to be exchanged between laboratories, they must be independent of the instrument used. The concept of retention index (RI) has been shown to be more reliable than that of relative retention time (i.e. the retention time of the solute relative to that of a reference compound). The RI system uses a homologous series of compounds (i.e. a series of compounds that increase in size by an additional methylene unit) to provide the reference points on the scale. The most commonly used is the system described by Kovats (1961) using straight-chain saturated hydrocarbons (n-paraffins or nalkanes). For any column temperature and stationary phase, the elution times of members of a series of n-alkane homologues are assumed to increase by an index of 100 for each additional methylene unit. On this scale, H2 has an index of zero, methane has an index of 100, ethane 200, and so on up the scale of alkanes. The RIs of unknown substances are measured against this scale, obviating the need to correct data between laboratories because of variations in retention time. The method is illustrated in Fig. 40.4, in which phenobarbital has a retention time of 4.5 min and an RI of 1957. RI ¼ 100ðPzþn Pz Þ
log t R ðxÞ log t R ðPz Þ log t R ðPzþn Þ log t R ðPz Þ
ð40:9aÞ
where tR is the retention time, Pz is the carbon number of the smaller nalkane, Pzþn is the carbon number of the larger n-alkane and x is the unknown solute.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:5
644
Gas Chromatography
Figure 40.4 Calculation of the RI of phenobarbital from a plot of a series of homologous n-alkanes. (A) Packed column, isothermal conditions, retention time for phenobarbital is 4.5 min, RI = 1953; (B) capillary column, temperature programmed to run at 10 /min, retention time for phenobarbital is 7.01 min, RI = 1960.
Retention indices collected from many sources show remarkable agreement, even when measurements were made on different (though equivalent) phases and at different temperatures (Ardrey, Moffat 1981). Most capillary GC is performed in temperature-programmed mode and the relationship between the retention time and chain length is almost linear, provided that the ramp rate is constant throughout the run and n >7. The simplified equation (40.8b) is used to calculate RI (see Fig. 40.4 for an illustration). RI ¼ 100ðPzþn Pz Þþ
t R ðxÞ t R ðPz Þ t R ðPzþn Þ t R ðPz Þ
ð40:8bÞ
Other homologous series have been proposed and good results were found with the alkan-2-ones and n-aldehydes. Use of other series (nalkyl esters, n-alkylbenzenes, n-alkyl iodides) has been less successful. When using a specific detector to identify compounds, the accuracy is increased by splitting a small percentage of the column effluent to a flame ionisation detector to give a ‘real-time’ calculation of RI, rather than relying on either historical retention times for n-alkanes or using an alternative homologous series. Alternatively, Franke et al. (1993) have proposed the use of an RI reference mix (a selected group of compounds that structurally resemble those under investigation) rather than a homologous series. Retention indices for unknown compounds can then be normalised relative to the known RI values of the reference compounds. The main advantage of this approach is that both temperature-dependent and column-ageing effects on RI, which arise from polarity mismatch between the homologous series and the investigated solutes, are somewhat reduced and the data are therefore more reproducible over time. As would be expected, there is better agreement for non-polar (most hydrocarbon-like) compounds than for polar compounds, and also for non-polar versus polar stationary phases where there is less difference in retention time between the n-alkane index markers. Using capillary columns, reproducibility can be as good as to within one RI unit for a non-polar phase, and to within a few units for a more polar one. Good temperature, pressure and flow control, and precise measurement of the injection time and peak elution, are essential for accurate measurement of RIs. The carrier gas flow rate and the polarity of the stationary phase are temperature dependent. Thus, the partitioning of polar compounds into the stationary phase is affected by temperature to a greater extent than that for the n-alkanes. Differences between constant-flow and constant-pressure modes of operation are exaggerated when the column is ramped in several stages. It is common practice to have an initial fast rate of increase in temperature followed by one or more slower ramp rates, since the number of low-boiling compounds of
interest is usually lower than the number of higher boiling compounds. Inaccuracies in RI calculation can also arise with high solute concentrations because of problems identifying the crest of the peak. This can also result in a trace component that elutes on the back of the concentrated one, as the major component begins to take part in the separation process by acting as a ‘dynamic stationary phase’. This delays the elution of the trace component. Column deterioration with use can lead to a preferential destruction or loss of the more labile component of a mixed phase. For example, in columns that contain mixed cyanopropyl- and methyl-PSX phases, the cyanopropyl group is preferentially lost, so that the column polarity is reduced. The elution of the index markers (n-alkanes) remains unchanged, but the progressive loss of cyanopropyl substituents results in a poorer interaction with polar compounds and an apparent decrease in their RI. With single-component stationary phases, the effect is still present, though less noticeable, as there is loss of retention of both RI markers and polar compounds.
Inlet systems The inlet system provides the means of introducing the specimen into the GC. Obtaining a narrow sample band at the start of the chromatographic process is critical to achieve good resolution, since broad sample bands usually produce broad peaks, especially for analytes that elute early. The choice of injector depends on the characteristics of the specimen or residue, the quantity and characteristics of the analytes to be separated, and the temperature and nature of the stationary phase and the column. Solids may be dissolved in a suitable solvent and injected with a micro-syringe. It is best to keep the solution as concentrated as possible to reduce the size of the solvent peak. Liquids can be injected using a micro-syringe, but with sensitive detection systems the sample should be dissolved in a suitable solvent to reduce the sample size and avoid overloading the detector. Gases and vapours may be introduced by injection through the inlet port septum using a gas-tight syringe. The three common types of GC injectors are split, splitless and cold on column. In reality, splitless injection is an extreme example of split injection and both are carried out using the same hardware. Conventional glass syringes of 1–10 mL volume with stainless-steel needles can be used on all but cold-on-column injectors, and the injection is made by piercing a silicone rubber septum. Care must be taken to select septa that have low bleed characteristics at the operating temperature, and those with Teflon backs are most reliable in this respect. Unstable materials can be decomposed by the high temperature of the injection system, particularly if the system is constructed of metal. For labile substances, cold-on-column injection is preferred, but clean extracts must be used to minimise column contamination.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:6
Inlet systems Split and splitless injectors Split injectors are used for more concentrated samples, since only a fraction of the sample actually enters the column. An inlet splitter allows a high flow of carrier gas through the injector while maintaining a low flow (1–4 mL/min) through the column: the excess gas and associated sample components are vented to the atmosphere through the split line. The ratio of these two flows (the split ratio) controls the proportion of the injected sample that reaches the column. The total flow through the injector may be from 10 mL/min to 100 mL/min, which gives split ratios of 10 : 1 to 100 : 1. A good splitter should be linear, i.e. it should split high- and low-boiling point compounds equally. The function of the splitter is not primarily to reduce sample volume, but rather to ensure that the sample enters the column as a compact plug. Split injections, therefore, produce some of the most efficient chromatographic separations, and allow the use of very narrow capillary columns. A lower split ratio channels a larger fraction of the injected sample down the column and may result in column overload. High split ratios waste large amounts of carrier gas and insufficient analyte may reach the column. In splitless injection, all the carrier gas passes to the column. This is useful for very volatile compounds, for low sample concentrations or for trace analysis. The flow rate in the injector is the same as that in the column (1–4 mL/min), and the only path for the injection to take is into the column, since the split vent is closed. At a fixed time after injection (usually 15–60 s), the injector is purged by opening the split vent to introduce a much larger flow of carrier gas through the injector (typically 20–60 mL/min) and any remaining sample in the injector is discarded through the split vent. Since the rate of sample transfer onto the column is so slow (because of the low gas flow), peaks are usually somewhat broader than for split injections. Care should also be taken to ensure that the volume of the injector liner is not exceeded by the expanding solvent injected (Table 40.5), otherwise splitting of early peaks will be observed. Temperature conditions can be adjusted to narrow or focus the sample band at the top of the column. Splitless injections should therefore be made with the initial column temperature at least 10 C below the boiling point of the solvent (Table 40.5), and the initial temperature should be held at least until after the purge activation time. Solvent condenses on the front of the column and traps the solute molecules, which focuses the sample into a narrow band (known as the solvent effect). Individual solutes with a boiling point 150 C above the initial column temperature condense and focus at the top of the column in a process known as cold trapping. Either the solvent effect or cold trapping must occur before efficient chromatography can be obtained. Some newer chromatographs have the option of a pulsed splitless injection. In this mode, the column head pressure is increased immediately upon injection (typically to 174 kPa) and held there
Table 40.5 Boiling points and expansion volumes for commonly used injection solvents Solvent
Boiling point ( C)
Expansion volume (mL) per L of solvent(a)
Suggested GC oven starting temperature ( C)
Methylene chloride
40
330
15–30
Carbon disulfide
46
355
15–30
Acetone
56
290
30–45
Methanol
65
525
40–55
n-Hexane
69
165
40–60 45–65
Ethyl acetate
77
215
Acetonitrile
85
405
55–75
iso-Octane
99
130
70–90
Water
100
1180
70–90
Toluene
111
200
80–100
Values are given at 250 C and 105 kPa head pressure.
(a)
645
for 30–60 s, before returning to the normal operating pressure. This facilitates band sharpening and, while the process is not guaranteed to increase the fraction of the injection delivered onto the column, sensitivity is often improved because of improved chromatography. Glass liners for split and splitless injectors come in a variety of shapes and volumes and it is prudent to start with a straight liner and to investigate some of those that cause turbulence (e.g. the inverted cup style) later if this is unsatisfactory. A plug of deactivated glass wool in the liner helps prevent the deposition of non-volatile or particulate material on the column, but may cause some peak deterioration, and for the best results needs to be placed at a consistent position in the liner. Packing of splitless injection liners with deactivated glass wool may decrease the chromatographic performance, but this must be weighed against the potential for damage to the stationary phase from the repeated injection of non-volatile or particulate material. Large-volume injectors The analysis of trace amounts of components or contaminants in complex matrices such as foods, beverages and environmental samples is difficult. Adequate sensitivity to detect trace components is provided by specific detectors such as the NPD or ECD, but regulatory standards require positive identification of these compounds by mass spectroscopy (MS). To overcome the inferior sensitivity of MS, large-volume injectors have been developed. Examples include the Apex pre-column separating inlet (PSI), the temperature-programmed sample inlet (PTV) from Gerstel and time-coupled time-resolved chromatography (TCRC). The inlet typically consists of a length (10–50 cm) of standard (2 mm i.d.) glass chromatography column that can be deactivated or packed with traditional materials. The first two injectors are mounted directly in the GC injector port; the latter is a free-standing column coupled by a four-way valve into the GC inlet. Injection volumes range from 125 mL for the PSI, 1 mL for the PTV and up to 20 mL for the TCRC. Injection of larger volumes (up to 60 mL) is possible for some applications, but result in discrimination in favour of high-boiling components and loss of volatiles. Large-volume injectors remove the solvent from the sample prior to its introduction onto the capillary column, typically by low-temperature evaporation through the split vent. As the sample is concentrated towards the bottom of the injector, the injector is heated, the split vent is closed and the analytes are introduced onto the GC column in splitless mode. Those injectors that can be heated selectively and cooled allow the precise introduction of selected components only from the sample, and thus reduce the quantity of non-volatile components (e.g. sugars) that might overload or destroy the analytical column. The TCRC has a small mobile oven (2–8 mm width) that can be scanned along the length of the column to produce band compression. Prior to the next injection, the injector columns are usually baked to vent high-boiling compounds to waste. Sensitivity can often be improved 50- to 100-fold and time is saved in sample preparation, since extensive clean-up or extraction procedures are no longer required. Cold-on-column injection Cold-on-column injection is most suited to compounds that are thermally labile. The injection needle must be fine enough to enter the column bore, usually fused-silica or stainless steel with a fusedsilica insert. The top of the column is held at a temperature low enough for the solvent that contains the sample to condense, usually by an air- or carrier gas-cooled sleeve. The solvent temporarily swamps the stationary phase and ensures that the sample components concentrate in a narrow band. Any solvent or sample that remains in the injector is backflushed with carrier gas, often by automatic valves. The proximal end of the column is then brought rapidly to the operating temperature, when the solvent vaporises and chromatography begins. The potential for rapid column contamination or deterioration means that cold-on-column injection is usually restricted to those applications where its use is essential.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:6
646
Gas Chromatography
Volatiles interface The volatiles interface allows automated analysis of gaseous samples. The interface is a low-volume highly inert switching block, and is ideally suited to trace-level detection. A portion of the carrier gas supply is diverted through the specimen sampler and released under controlled conditions onto the column. The remainder of the carrier gas goes to a flow sensor, which prevents fluctuations in column gas flow that would otherwise occur when the switching valves are opened and closed. The interface can be run in split, splitless or cold-on-column modes as described in the sections above. Samples may be introduced from external devices, such as air samplers or purge-and-trap devices (see section below), or from headspace analysis, which permits analysis of volatile substances in a liquid sample while minimising contamination of the column. This technique is used in the assay of ethanol and other solvents in blood and for complex household preparations, such as polishes, which contain volatile substances (see Chapter 14).
pressure. If the volume of the liner is smaller than the expanded solvent volume (see Table 40.5), some of the sample is propelled out of the injector in a process known as backflush. This can appear as a broad tailing solvent front, since it now takes longer to flush the expanded solvent out of the injector and carrier gas line. Backflush can also cause injector contamination, since the analytes condense in the cooler carrier gas line, from where they may bleed continuously into the injector and cause high background or spurious peaks. Carryover or peak ghosting can occur when the next injection backflushes and carries previously condensed compounds back into the vapour phase and onto the column. Backflush can usually be solved by using a smaller injection volume, a less expansive solvent, a lower injector temperature, a liner with an upper restrictor, or a faster carrier gas flow. The use of an adjustable septum purge gas (usually 0.5–1 mL/min) also decreases the potential for backflush, as components that would normally condense on the cooler septum and travel into the carrier gas lines are swept away by the septum purge. Too high a purge flow results in loss of highly volatile components.
Thermal desorption and purge-and-trap injection The analysis of samples that have been pre-concentrated onto solid adsorbents is common in the fields of industrial air monitoring, analysis of residues in food, soil and water, petrochemical analysis and environmental monitoring. The methods of preparing samples for analysis are described in the section on specimen preparation. These samples require special interfaces with GCs to ensure good chromatography. In some instances the sample preparation device and injector are manufactured as stand-alone pieces of equipment that require very little modification of conventional injectors, while others must be dedicated pieces of equipment. Once collected, the concentrated sample must be desorbed into the chromatograph using the heated injector port. The major problem here is the possible introduction of water into the chromatograph from moisture adsorbed during collection from high-humidity samples. Release of solutes from the adsorbent should be as rapid and complete as possible to allow for rapid and sensitive analysis and for a narrow sample band to be introduced into the chromatograph. This is achieved either by cooling the column oven cryogenically to refocus the sample in the injector prior to injection or by using a dry purge system coupled to the gas chromatograph via a volatile interface (see above) designed to operate above ambient temperature. Here, the specimen is thermally desorbed from the collection tube onto a narrower (1 mm i.d.) tube of the same adsorbent material. The concentrated solute is then released into the chromatograph, ensuring rapid and complete sample introduction. Adsorbents must be thermally stable to reduce interference from background contaminants. With solid phase microextraction (SPME) the adsorbed sample is introduced into the heated injector port via a special sleeved needle (see under Specimen preparation). This technique requires the injector liner to be narrow (usually 0.75 mm as opposed to 2 or 4 mm) to increase the linear velocity of carrier gas through the liner and ensure that a narrow band of sample is introduced onto the column. Solid injection When solvent interference is serious the sample may be injected as a solid. The ‘moving needle’ injector has found application in steroid analysis and for the determination of anticonvulsant drugs. A solution of the material to be injected is placed on the tip of the glass needle with a syringe. A small flow of carrier gas sweeps the solvent out of the top of the device to waste. The dry residue is then introduced by moving the needle into the heated injection zone of the chromatograph with a magnet. This form of injection can be used only with compounds that do not volatilise with the solvent. Backflush Upon vaporisation, the injected sample undergoes considerable expansion, sometimes up to 100 to 1000 times its original volume, which creates a pulse of pressure that often exceeds the column carrier gas
Injector discrimination Injector discrimination occurs because not all the compounds in the sample vaporise at the same rate. Since the sample remains in the liner for a limited time, this usually results in some loss of higher-boiling solutes. This can be alleviated by increasing the residence time of the sample within the injector, or by using a higher injector temperature or smaller injection volume. However, there is usually a compensatory loss in lower-boiling compounds. Discriminating behaviour can usually be managed by making reproducible injections. Gas pressure and flow control For accurate and reproducible GC, either a constant carrier gas flow or a constant carrier gas pressure must be maintained. Under isothermal conditions, simple pressure control is adequate for packed or capillary columns and back pressure can be monitored by a pressure gauge between the flow controller and the injector. A decrease indicates a leaking septum and an increase suggests contamination of the injector liner or the top of the column. This also ensures that the flow controller is performing correctly. Since the back pressure rises to equal the supply pressure, flow becomes pressure controlled. Flow control is highly desirable, if not essential, during temperature programming with packed columns and can be used to advantage with capillary columns. The added convenience of a digital (electronic) flow controller may be worthwhile. Since the carrier gas becomes less viscous as the column oven temperature rises, the gas pressure must be increased as the run progresses to maintain constant velocity (or constant flow) throughout the analysis. Fig. 40.5 shows the effects of increasing the column temperature on the carrier gas flow and velocity if the head pressure is held constant during the run. As flow and velocity do not respond identically to increasing temperature (see Fig. 40.5D), late-eluting analytes are recovered more quickly using constant flow than under constant-pressure conditions. Furthermore, since column efficiency is a function of the carrier gas velocity (Fig. 40.6), resolution at the end of the chromatogram is improved under constant flow conditions. Switching between conditions of either constant flow or constant pressure can sometimes resolve otherwise co-eluting compounds. Table 40.6 shows the relationship between flow and pressure for various lengths and diameters of capillary columns. It shows the calculated head pressure (kPa) required to achieve the stated gas velocity or flow through a 25 m column operating at 150 C. Note that head pressure values above 280 kPa are not usually practicable using standard pressure regulators. Increasing the column length has a direct and proportional increase on head pressure for both velocity and flow calculations. The way in which carrier gas velocity affects column efficiency is best demonstrated by reference to the van Deemter curves in Fig. 40.6. These demonstrate that the optimum column efficiency (minimum height equivalent of a theoretical plate, HETP) occurs at intermediate
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:6
Detector systems
647
Figure 40.5 Effect of temperature on carrier gas flow and velocity. (A) and (B) are under conditions of constant carrier gas head pressure (140 kPa). (A) shows the change in column flow (mL/min) with change in temperature from 50 C to 300 C. (B) shows the change in velocity (cm/s) with change in temperature from 50 C to 300 C. (C) and (D) are under conditions of constant carrier gas flow (1 mL/min). (C) shows the change in carrier gas velocity (cm/s) with change in temperature from 50 C to 300 C. (D) shows the change in column head pressure (kPa) with change in temperature from 50 C to 300 C. All calculations are for a 25 m column of 0.25 mm i.d. operating at atmospheric pressure and 150 C.
velocity, and that column efficiency is compromised at both low and very high velocities. A small loss in efficiency for a shorter analysis time is usually tolerated. Curves are shown for the three most common carrier gases (helium, nitrogen and hydrogen), and it can be seen that the chromatography is much less tolerant to changes in nitrogen
velocity than to helium. Helium is favoured by most users, as analysis times are half that with nitrogen, with only a slight loss in efficiency. While hydrogen gives the best dynamic range and shortest analysis times, there are safety issues relating to its use. While the gas used for the carrier gas should always be of the highest purity available, a lower-quality gas can sometimes be used for the makeup or detector, since these do not contribute to column deterioration by oxidation. Regardless of quality, it is advisable always to use a scrubber (to remove oxygen and hydrocarbons) followed by a dryer (to remove water vapour) between the supply and the instrument. Metal trap bodies are recommended, as plastics are permeable to impurities in laboratory air, especially when large amounts of organic solvents are used. Most traps have an indicator to show when they are saturated, and they can be changed without interruption to the gas flow. Stainless steel or copper tubing is recommended for plumbing of all gases, as plastics are permeable to moisture and oxygen, and Teflon, nylon, polyethylene, polypropylene and PVC contain contaminants that degrade gas purity.
Detector systems
Figure 40.6 Van Deemter plots for a 25 m 0.25 mm i.d. WCOT OV-101 column. HETP = height equivalent of a theoretical plate.
The choice of chromatography detector for an application depends on factors such as cost, ease of operation, consumables supply, sensitivity, selectivity and the linear working range. Some detectors respond to almost all solutes, while others (selective detectors) respond only to solutes with specific functional groups,
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:6
648
Gas Chromatography
Table 40.6 Relationship of gas chromatography column diameter to column flow, velocity and head pressure (kPa) Column internal diameter (mm) 0.20
0.25
0.32
0.53
93.2
10.8
74.5
28.7
10.4
654.1 181.5
144.9
90.4
54.2
19.5
724.5 201.5
160.0
100.1
60.0
215.3
Hydrogen
675.5 157.3
115.9
58.2
24.8
3.66
Nitrogen
959.1 240.8
181.5
95.9
43.2
6.76
193.2
102.8
46.9
7.38
0.10
0.18
Hydrogen
329.1
Nitrogen Helium
Velocity 30 cm/s
Flow 1 mL/min
Helium
1007.4 254.6
Data for a 25 m column operating at 150 C.
atoms or structural configurations. Additional functional groups can often be added to solutes, generally after extraction (see below under Derivatisation), to achieve a response from a selective detector and gain additional sensitivity and selectivity. The use of detectors such as the ECD to identify amenable compounds, and the NPD to detect compounds that contain phosphorus and nitrogen, removes many of the extraneous peaks frequently observed when using non-selective detectors, such as the FID. However, these selective detectors have also led to the detection of substances such as plasticisers from blood-collection tubes or transfusion lines, which interfere in many toxicological analyses. Detectors that detect the presence of a solute and also give information about its structure are increasingly popular and MS, Fourier transform infrared spectroscopy and atomic emission spectrometry have been invoked to achieve this goal. Detector sensitivity is measured as signal-to-noise ratio, in which the signal corresponds to the height of the peak, and the noise to the height of the baseline variability. A signalto-noise ratio of 8 to 10 is considered sufficient to confirm the presence of a peak. Each type of detector has a linear operating range in which the response obtained is directly proportional to the amount of solute that passes through, although this can be modified slightly by the nature of the solute and the chromatographic conditions (mobile phase type and flow, detector temperature). The linear operating range is considered to be exceeded when the incremental response obtained from the detector varies by more than 5% from that expected. Most detectors (except MS) rely on gas other than the mobile phase (combustion, reagent or purge gas) for their operation. Usually, a total flow of at least 30 mL/min is necessary to sweep the solute molecules physically through the body of the detector at sufficient speed to prevent refluxing and produce narrow peaks. Thus, the addition of a ‘makeup’ gas is invariably required with capillary columns. Recommended gases and their flows for each detector are included in the manufacturer’s instruction manuals, and it is important to follow these guidelines (and those on maintenance) to achieve the stated performance. Here, only the most widely used detectors are considered in detail. Several other types of detectors are available; for a more detailed discussion, the reader is referred to the text by Scott (1996). Flame ionisation detector This is the most widely used of all detectors, since it responds to nearly all classes of compounds. The effluent from the column is mixed with hydrogen and the mixture is burnt at a small jet in a flow of air. A polarising current is applied between the jet and an electrode situated above it. When a component elutes from the column it burns in the flame to create ions that carry a current between the electrodes and provide the signal. The background current and noise are both low. Any of the usual carrier gases can be used and minor changes in gas flow are without effect. Sensitivity is moderate (0.1–10 ng), with linearity extending sometimes as high as six orders of magnitude. The response of the FID is dependent on the number of carbon atoms in the molecule, but the response is lowered if oxygen or nitrogen is also present in the
molecule. It responds to all organic compounds that contain carbon– hydrogen bonds with the exception of formic acid. Both the sensor design and electronics are simple, and manufacturing cost is therefore low. The FID is easy to clean, and when operating with capillary columns it is virtually maintenance free. With packed columns, however, there is a tendency for a build-up of stationary phase bleeding from the column, which must be removed periodically. The insensitivity of the detector to water is a useful feature that allows aqueous solutions to be used. Nitrogen–phosphorus detector or alkali flame ionisation detector The introduction of alkali metal vapours (usually supplied by an electrically heated bead of rubidium chloride or caesium chloride) into the flame or ‘plasma’ of an FID confers an enhanced response to compounds containing phosphorus and nitrogen. By adjustment of the plasma gases the detector can be made virtually specific for phosphorus compounds (e.g. a phosphorus : carbon response ratio of 50 000 : 1 and a phosphorus : nitrogen response ratio of 100 : 1). Even when optimised for nitrogen compounds, it retains its response to phosphorus (e.g. a nitrogen : carbon response ratio of 5000 : 1 and a nitrogen : phosphorus response ratio of 10 : 1). This detector is particularly useful for drug analysis, since most drugs contain nitrogen, while the solvent and the bulk of the co-extracted material from a biological sample do not. The NPD is ideal for detecting pesticides that contain phosphorus, and therefore has wide application in environmental and regulatory analysis (air, soil, water and residues in food). The extreme sensitivity to compounds that contain phosphorus can be further exploited by the preparation of derivatives that contain this element. Sensitivity is excellent (1–10 pg), with a good linear range of up to four or six orders of magnitude. A disadvantage is the need for the supply of three gases and, unlike with the FID, their control is absolutely critical to selectivity. The detecting element (bead) lasts between 1 and 3 months depending on usage. Stationary-phase bleeding from packed columns coats the bead and collector assembly but can be rinsed off using methanol or dilute (0.1 mol/L) sulfuric acid. Most of the early problems that arose from poor reproducibility in bead coating have now been resolved, and the most stable detectors nowadays have a geometry that enables the bead to be located and fixed in its optimal position with relative ease. Electron-capture detector The early form of this detector consists of a small chamber with a pair of electrodes and a radioactive source, usually 63Ni, placed close to the cathode to ionise the carrier gas. Potential applied to the electrodes produces a steady background current. Electron-capturing solutes arriving in the chamber remove some of the electrons and reduce the detector current. The response of the detector is therefore a loss of signal rather than an increase, as is given by most other detectors. Although the ECD can be polarised from a suitable low-voltage direct-current supply, it is more sensitive when a pulsed power supply is used, and in modern detectors the polarising pulses are modulated to maintain a constant current. A voltage that depends on the modulation frequency is generated as the output signal. Additional carrier gas is necessary, even with packed columns, to obtain a flow of at least 60 mL/min to purge the detector adequately and avoid peak broadening and distortion. Sensitivity can also be improved dramatically by raising the operating temperature of the detector, and decreasing the makeup gas flow. The ECD is a selective detector with a very high sensitivity to compounds that have a high affinity for electrons; for many compounds, the sensitivity of the ECD often exceeds that of MS, and sometimes even that of the NPD. Compounds that contain a halogen, nitro group or carbonyl group are detected at 0.1–10 pg, 1–100 pg and 0.1–1 ng, respectively. This makes it very useful for compounds such as the benzodiazepines or halogenated pesticides and herbicides. Alternatively, the great sensitivity of the detector may be utilised by preparing derivatives with halogenated reagents, such as trifluoroacetic, heptafluorobutyric or pentafluoropropionic (PFP) anhydrides. Linearity (at best only two or three orders of magnitude) is a limiting factor for quantitative analysis.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:7
Detector systems In older models, the addition of a small amount of quench gas, such as methane, improves stability and linearity, and is essential if argon or helium carrier gas is used. Newer models can be operated successfully with helium as both carrier and detector gas. The ECD, because of its high sensitivity, can be contaminated easily: an impure cylinder of gas can damage a detector beyond repair in a matter of only a few hours. Cleaning is difficult, although some material can be removed by heating the detector to its maximum operating temperature overnight, and the injection of water in 100 mL aliquots through an empty glass column can also help. However, if contamination is avoided, it is virtually maintenance free. The radioactive source requires special handling procedures that may be subject to federal legislative regulations. More recently, it has been shown that this detector can work with greater sensitivity and operate over an increased linear range using a helium plasma in place of the radioactive source.
649
700 amu) several times per second. The abundance of each mass at a given scan time produces the mass spectrum, which can be summed and plotted versus time to obtain a total ion chromatogram. The MS detector can be operated either in full scan mode (collecting all the ions within a given mass range) or selected-ion monitoring (SIM) mode, which collects only pre-selected masses characteristic for the compound under study. Sensitivities for the two modes of operation are quite different: 1–10 ng for full scan, increasing to 1–10 pg in SIM because of the dramatic decrease in background noise. The linear range is excellent and often spans five or six orders of magnitude. Recent advances in computer technology, coupled with improved detector design, have revolutionised the use of the MS detector from a research tool to one of routine application. This technique is described in more detail in Chapter 37. Ion-trap mass spectrometer
Fourier-transform infrared detector In the Fourier-transform infrared detector (FTIRD), the column effluent is conducted through a light pipe and swept by a scavenging gas into the path of an infrared light beam that has been processed by an interferometer. The interferometer directs the entire source light to a beam splitter, which sends the light in two directions at right angles. One beam takes a fixed path length to a stationary mirror, while the other takes a variable path length to a computerised moving mirror. The two beams are recombined, and the difference in path lengths creates constructive and deconstructive interference, or an interferogram. The recombined beam is then passed through the sample. Analyte molecules absorb light energy of specific wavelengths from the interferogram, and the sensor reports variation in energy versus time for all wavelengths simultaneously. For molecules to be infrared active they must be able to undergo a change in dipole moment with the transition to their excited state. As a result, many compounds that are symmetrical do not respond. Fourier transformation refers to the mathematical computation that converts the data from an intensity versus time plot into an intensity (percentage transmission) versus frequency spectrum. Each dip in the spectrum corresponds to light absorbed, and can be interpreted as characteristic of specific functional groups in the molecule. Computer libraries allow for easy and rigorous comparison of spectra. FTIR can be fully quantitative, but it is relatively insensitive (10 ng range). Its advantages are that it is non-destructive, and it can distinguish between isomers (MS cannot). Because of the logistical difficulties of combining FTIR with GC, this combination of techniques has started to emerge only recently. Atomic emission detector With the atomic emission detector (AED), carrier gas that elutes from the column delivers solutes into a high-temperature helium plasma, where heat energy is absorbed by the constituent elements. In returning to their ground state, they emit energy as light, the wavelength of which is characteristic for each element. Emitted light is focused by a quartz lens and spherical mirror onto a diffraction grating, and the dispersed light is focused onto a diode array that is continuously scanned (wavelength usually 170–800 nm). Typically, some 15 elements can be monitored simultaneously, and each is plotted against time. The composite chromatogram allows the percentage elemental composition of each peak to be determined. Sensitivity is very good, but the detector is complex and expensive to operate and is not widely used. Mass spectrometer A gas chromatograph is an almost ideal inlet device for quadruple MS. The detector is maintained under vacuum, and in the most common technique of electron impact (EI) the column effluent is bombarded with electrons. Compounds absorb energy, which causes them to ionise and fragment in a characteristic and reproducible fashion. The resultant ions are focused and accelerated into a mass filter that allows fragments of sequentially increasing mass to enter the detector stepwise. The mass filter scans through the designated range of masses (usually up to about
As with other forms of mass spectrometers, EI or chemical ionisation (CI) is used to produce an ion source, but this is focused into the iontrap mass spectrometer in pulses rather than continuously. The fundamental difference is that all the solute ions generated over the entire pulse period are trapped in the detector and are then sequentially ejected in increasing mass number from the trap into the electron multiplier. The addition of helium into the trap (133 mPa) contracts the ion trajectory to the centre of the trap, where it is further focused by the ring electrode, to form dense ion packets that are expelled more efficiently than diffuse clouds, and thus greatly improve resolution. The spectral patterns can be quite different from those produced by mass filter spectrometers, and are often characteristic of the conditions under which the instrument is run, which makes comparison difficult between instruments. However, because the ion collection period is longer, the sensitivity of the ion trap in full scan mode is similar to that obtained in SIM on the average MS. Furthermore, an improved mass range (sometimes up to several thousand atomic mass units) gives this type of detector many applications, particularly for quantitative trace analysis, and for higher mass components. This technique is described in more detail in Chapter 37. Dual detector systems The simultaneous use of a combination of a universal detector (FID) with a specific detector to monitor the effluent of a column can provide useful information about the properties of functional groups and substituents in a molecule. The FID response is roughly dependent on the number of carbon atoms in a molecule and is quite predictable. However, the ECD response varies widely for different compounds is dependent on the electron-deficient part of the compound and is difficult to predict. The NPD response of a compound depends to some extent on the number of phosphorus or nitrogen atoms in a molecule, but it also depends on their environment. Thus, by using the FID as a reference, and measuring the ECD or NPD response relative to it, another characteristic for identification is obtained in addition to retention behaviour. Dual detector systems can be used in several ways. The column can be split at the detector end and the effluent passed into two different detectors that operate in parallel. This approach allows the most flexibility, since the choice of detectors is wide, and the effluent can be split in proportion to the sensitivity required from each detector. For capillary columns this is accomplished easily with zero-dead-volume press-fit tee connectors, but it is a more complicated operation for packed columns. Additional makeup gas may be required to ensure a good flow through the detectors, and care should be taken to use tubing of a total area smaller than or equal to the analytical column to avoid loss of peak shape through refluxing at the detector. Alternatively, the GC oven houses two completely separate but identically matched columns, each connected to a single detector. This is not an ideal approach, as matching columns is difficult and has to be checked at frequent intervals. Another approach is to stack the detectors in series, and some manufacturers deliberately provide detectors in identical modules for this purpose. There are limitations to the choice of possible detector combinations, as the first
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:7
650
Gas Chromatography
detector must always be a non-destructive detector, such as the ECD, AED or FTIRD.
Specimen preparation Prior to chromatography, it is usually necessary to isolate the compound(s) of interest from either a biological matrix (plasma, urine, stomach contents, hair or tissue) or some other matrix, such as soil, air or water. Removal of extraneous material and concentration of the compounds of interest usually take place simultaneously. The high water solubility of some drug metabolites (e.g. glucuronide conjugates) requires chemical conversion to a less polar entity to permit isolation from waterbased samples, and a hydrolysis procedure is often used for this purpose. Isolation and concentration Protein precipitation
If the analyte is present in blood in high concentration, a simple protein precipitation step often provides a suitable extract, although the possibility of losing significant amounts of analyte with the precipitate must be considered. Mixing with a solution of mercuric chloride or barium sulfate readily precipitates plasma proteins, and centrifugation provides a supernatant for direct injection onto the chromatography column. Use of perchloric or trichloroacetic acids (10%) is not advised, unless the resultant solution is neutralised prior to injection. Dimethylformamide is a good organic precipitation reagent that is well tolerated by most GC stationary phases. Other organic precipitating agents are methanol, acetone and acetonitrile, all of which should be added in the proportion of two volumes to each volume of blood. While the extract is still water based, most columns with a high stationary-phase loading (5 mm film thickness) can tolerate the injection of 1 mL of water. If the column is not water tolerant, it is possible to evaporate small volumes of the supernatant to dryness for reconstitution in a more suitable solvent. Liquid–liquid extraction
Liquid–liquid extraction is the most frequently used method to isolate and concentrate solutes for GC. The pH of the specimen is adjusted to ensure that the compounds to be extracted are not ionised (basic for bases, acid for acidic compounds). Bearing in mind that some portion of the aqueous acid or base will dissolve in the solvent, the use of strong mineral acids or alkalis is not advised as this adversely affects column performance. Best results are obtained with acidic buffers (phosphate or acetate) and with ammonium hydroxide or basic buffers (borate), using a 5 : 1 ratio of solvent to specimen. The solvent chosen should be sufficiently polar to partition the compound of interest without co-extracting excessive amounts of polar contaminants. For more water-soluble drugs, such as beta-blockers, the addition of 2–10% of a polar solvent (e.g. isopropanol or butanol) is helpful, or solid sodium chloride can be added to ‘salt out’ the analyte. If a derivatisation step is to be carried out subsequently, the use of a solvent compatible with the derivatisation eliminates the need for an evaporation step. Use of solvents with a higher density than the sample (e.g. dichloromethane) can lead to difficulty in isolation of the organic phase. Purification of extracts by back extraction (re-extraction of the analytes from the organic solvent at the opposite pH followed by re-extraction into solvent at the original pH) may be helpful for trace analysis. The use of a small volume of solvent for the final extraction serves as a concentration step without the need for separation and evaporation of the organic phase. Solid–liquid or solid-phase extraction
Solid–liquid extraction uses a polypropylene cartridge with a small amount (200 mg to 3 g) of high-capacity (1–20 mL) silica-based packing at the base of the reservoir. On introduction of the sample matrix, the compounds of interest are withheld by the packing. Impurities are then rinsed selectively from the column, and the final elution releases the compound of interest. Evaporation followed by reconstitution in a suitable solvent provides a clean, concentrated sample ready for analysis by GC. Bonded-phase packings that have been modified by the addition of various functional groups are available. The mechanisms of interaction for the matrix, analytes and packings are similar to those in LC
(see Chapter 38). Polar stationary phases retain polar analytes (normal phase) and are eluted with organic solvents, while non-polar stationary phases retain non-polar analytes (reversed-phase) and are eluted with aqueous solvents. Ion-pair extraction uses a non-polar stationary phase and polar analyte, with a counter-ion added to the sample solution, and allows retention of the (now neutral) analyte by a reversed-phase mechanism. In ion-exchange extraction, the adsorbent surface is modified with ionisable functionalities. Analytes with ionic charges opposite to those on the packing are retained. Solvents that contain counter-ions of greater strength are used to elute the analytes of interest from the tube. Solid-phase microextraction
Solid-phase microextraction (SPME) requires no solvents or complicated apparatus and can concentrate volatile and non-volatile compounds in both liquid and gas samples. The unit consists of a fusedsilica fibre attached to a stainless-steel plunger coated with a stationary phase (mixed with solid adsorbents as required). The plunger is inserted through a septum into a vial that contains the sample, and the fibre is exposed by depressing the plunger either into the liquid or into the headspace for 20–30 min. The retracted fibre is inserted into the injection port of the GC, and is desorbed when the plunger is depressed. The unit may be reconditioned and used 50 to 100 times. For field analysis, adsorbed samples can be stored and transported in the needle sealed in a special container for subsequent analysis by GC (or LC). Pesticides recovered from water samples have been shown to be more stable when stored in this way than in water. The special small-volume injection liner fits any model of chromatograph, and produces sharper peaks because of the higher linear gas velocity, with little or no backflush. Suitable stationary phases are: n
n n n n
100 mm dimethyl-PSX film for low-molecular-weight compounds or volatiles, or a thinner film (7 mm) for higher-molecular-weight semivolatile compounds 85 mm polyacrylate film for polar compounds 65 mm film of dimethyl-PSX-divinyl benzene for volatile alcohols and amines For surfactants, 50 mm Carbowax-templated resin For trace-level volatiles, a 75 mm Carbowax-carboxen phase is suitable.
An alternative approach uses a small magnetic stir bar encapsulated in glass and coated with a layer of dimethyl-PSX. The bar is left to stir in the sample for 30–120 minutes and then removed and placed in a thermal desorption tube. From there, it is introduced onto the GC as described in the section Thermal desorption. Both approaches give similar performance for higher-boiling compounds (>350 C), but SPME is inferior for lower-boiling compounds such as naphthalene and fluorene (b.p. 218 C and 298 C, respectively). Supercritical fluid extraction
A supercritical fluid (SCF) is a substance that is maintained above its critical temperature and pressure, where it exhibits physicochemical properties intermediate between those of a liquid and those of a gas. Properties of gas-like diffusivity, gas-like viscosity and liquid-like density combined with a pressure-dependent solvating power provided the impetus to apply SCFs to analytical separation. The initial applications most often involved isolation of flavours and contaminant residues from food and soil. These have now been extended to the isolation of drugs from blood and other aqueous-based media by using adsorbents added in-line (such as molecular sieves, diatomaceous earth, silica gel, etc.) to filter proteinaceous material and adsorb water. It is possible, by adding small volumes of co-solvent to the SCF, to extract highly polar solutes with excellent efficiency. In contrast to the conventional extracting solvents, the fluid most often used in supercritical fluid extraction (SFE), supercritical CO2, is non-polluting, non-toxic and relatively inexpensive. Additionally, extractions are carried out quickly at temperatures that avoid degradation of temperature-sensitive analytes and provide clean extracts with extremely high efficiency. Several dedicated SFE analysers are available; each consists of a gas supply, pump and controller used to pressurise the gas, temperature-controlled oven, extraction vessel, internal diameter regulator and collection device.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:7
Specimen preparation
651
The CO2 supply is compressed to a selected pressure (e.g. 28 000 kPa) and its is temperature adjusted (e.g. 50 C). As the supercritical CO2 passes through the sample material, the solutes are extracted to an equilibrium solubility level, typically about 10% (w/w). The gaseous solution that leaves the extractor is passed through the pressure-reduction valve, where the pressure (and thus the dissolving power) of the CO2 is reduced. The solutes precipitate in the separator, and the CO2 is recycled through the system several times until the extraction is completed, when it is vented to waste.
pyrolysis unit has been devised that sits in the GC oven (Gorecki, Poerschmann 2001). This is a silicosteel metal capillary (0.53 mm diameter) connected through butt connectors to a fused-silica restrictor inserted in the injection port which prevents backflush, and to the analytical column. The unit can be heated up to 750 C in 13 ms, and can be cooled again to ambient temperature in 4 s. The much reduced discrimination traditionally related to transfer of higher-boiling fractions is thus overcome, and this arrangement greatly extends the application of pyrolysis as a means of sample introduction into GC.
Headspace analysis
Tissues and hair
This method of isolation is used for analytes with volatility higher than that of the common extraction solvents. A detailed description of the technique is given in Chapter 14.
Tissues and hair require treatment prior to drug extraction to break down the biological matrix and enable a good recovery of the drug. For solid tissues, good results are obtained by incubation of a portion of the tissue with a mixture of a collagenase, a protease and a lipase in a buffer of suitable pH. For small amounts of tissues (100 mg), overnight treatment at room temperature suffices, although gentle agitation or occasional mixing speeds up the process. Larger amounts of tissue benefit from mechanical homogenisation prior to incubation. For the analysis of hair, an initial washing to remove residues from cosmetic products or environmental contaminants is recommended, followed by incubation with either caustic alkali (for basic drugs) or mineral acid (for acidic drugs). After adjustment of the pH, drug recovery can proceed by the usual procedures established for the specific compounds under investigation. For additional information see Chapter 10 and Chapter 19.
Purge and trap
Purge and trap is a powerful procedure for extracting and concentrating volatile organic compounds from soil, sediment, water, food, beverages, etc. It is especially useful for poorly water-soluble compounds and those with boiling points above 200 C. The procedure involves bubbling an inert gas (nitrogen or helium) through an aqueous sample or suspension at ambient temperature, which causes volatile organic compounds to be transferred into the vapour phase. During the purge step, purge gas sweeps the vapour through a trap containing adsorbent materials that retain the volatilised compounds. Water vapour may be removed by dry purging. The trap is rapidly heated to 5–10 C below the desorption temperature. The valve is then switched to join the trap flow to the carrier gas flow, and the trap heated to its desorption temperature for a fixed time. Adsorbent tubes are usually packed with multiple beds of sorbent materials, each one more active than the preceding one, which allows compounds with a wide range of boiling points and polarities to be analysed simultaneously. During the purge, the smaller and more non-polar solutes are readily carried down the beds and, since the carrier gas passes in the opposite direction during the desorption phase, the larger and more polar compounds do not come into contact with the innermost active beds, from which their release may be difficult to effect. Thermal desorption
This technique is used extensively for air monitoring in industrial hygiene, environmental air, indoor air or source-emission monitoring. The device may be portable or fixed and of varying size. Air is pumped continuously through the device at a fixed rate, during which time components are extracted gradually and concentrated onto the adsorbent beds; the arrangement of the beds is the same as described above for the purge and trap, and prevents potentially irreversible binding of large molecules. The direction of the flow is simply reversed during desorption. Analysis requires a special interface to the GC, which is described above in the section Thermal desorption and purge-and-trap injection. The adsorbents must have high capacity to remain active during the entire sampling period, and show an acceptable pressure drop during sampling. Ideally, a minimal amount of unwanted analytes should be absorbed, as these will contribute to the background noise. Pyrolysis
Analytical pyrolysis can be a very useful tool for characterization of complex materials, including synthetic polymers (e.g. plastics) and natural organic polymers such as humic organic matter (HOM). Conventional pyrolysis with a unit connected to the exterior of the GC injection port is of only limited use for HOM, first because of the formation of much carbonaceous residue of virtually zero diagnostic value, and second because of loss of high-boiling pyrolysis products with great diagnostic value during the sample transfer from the pyrolysis unit to the GC column. Such products are long-chain alkanes, alkylbenzenes, fatty acids and dicarboxylic acids, as well as steranes and hopanes. These large compounds originate in the HOM as they are not formed during pyrolysis, and are diagnostically distinct from smaller products such as phenols that might originate either from the HOM itself or equally from the breakdown of lignin, carbohydrates and proteins, which are the starting material for HOM. To circumvent this deficiency, an in-column
Hydrolysis Recovery of conjugated drug metabolites from biological fluids can be increased by hydrolytic cleavage of the conjugate bond prior to extraction. This offers a vast improvement in sensitivity for qualitative analysis, particularly from urine, and is essential to identify drugs (e.g. laxatives) that are excreted almost exclusively as conjugated metabolites. However, reliable quantitative analysis of conjugated metabolites requires that the unconjugated metabolite must first be removed or quantified, and then the total (conjugated plus unconjugated) metabolite be measured after hydrolysis in a subsequent separate procedure. For quantitative work, appropriate standards that contain conjugated metabolites must be carried through the procedure to monitor the efficiency of the hydrolysis step. Enzymatic hydrolysis
The use of a specific enzyme to cleave chemical bonds is the more specific of the two approaches but it incurs additional cost and time. It also provides cleaner extracts, and therefore prolongs the life of the chromatography column. There are a number of commercial preparations of purified glucurase and sulfatase harvested from different species. It is important to pay attention to the pH and temperature optima of the specific enzyme preparation. Temperature-tolerant preparations allow heating up to 60 C, which permits relatively short incubation times (2 h). Chemical hydrolysis
This quicker and less expensive approach can provide suitable extracts for chromatography for some analytes, although they are generally more demanding in terms of clean-up procedures. Typically, strong mineral acids or alkalis are used, often with boiling or treatment in a microwave or pressure cooker. Extracts must be neutralised, otherwise the chromatography column deteriorates quickly. Care should be taken to ensure the stability of the analytes to the hydrolysis conditions. Vigorous hydrolysis conditions often yield undesirable by-products or, if several compounds can be hydrolysed to a single entity, preclude accurate identification of the original compound present. For example, both the acid and the enzymatic hydrolysis of benzodiazepines remove glucuronide conjugates, but acid hydrolysis also converts two or three drugs to the same benzophenone compound (diazepam, temazepam and ketazolam are all converted into 2-methylamino-5-chlorobenzophenone). While this compound has good chromatography characteristics, the approach is unsuitable for those applications (such as forensic analysis) that require absolute identification of the drug ingested.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:7
652
Gas Chromatography
Derivative formation Derivatisation enables the analysis of compounds that otherwise could not be monitored readily by GC. To some extent the availability of stable polar stationary phases in capillary columns and the use of temperature programming has negated the requirement for derivatisation, although it is still widely used. Choice of reagent is based on the functional group that requires derivatisation, the presence of other functional groups in the molecule and the reason for performing the reaction. Although the retention characteristics are changed, the order of elution of a series of derivatives will be the same as that for the parent compounds. The preparation of derivatives modifies the functionality of the solute molecule to increase (or sometimes decrease) volatility, and thereby shortens or lengthens the retention time of a substance, or to speed up the analysis. Another common reason for derivatisation is to improve resolution and reduce tailing of polar compounds (hydroxyl, carboxylic acids, hydrazines, primary amines and sulfhydryl groups). For instance, hydroxylated compounds often have long retention times and column adsorption causes tailing, which results in low sensitivity. However, they readily form silyl ethers and these derivatives show excellent chromatography, and sensitivity can often be improved by a factor of 10 or more. Derivatisation can also help to remove the substance peak away from interfering material. For example, the reaction of amfetamine with acetone enables successful differentiation from methyl ethyl ketone on most stationary phases. Derivatives may also be used to make the molecule amenable to detection by selective detectors, or can be used to improve the fragmentation pattern of the compound in the mass spectrometer. The reaction may be carried out during extraction (e.g. extractive alkylation), on the dry residue after solvent extraction (e.g. silylation) or during injection (e.g. methylation). In choosing a suitable reagent, certain criteria must be used. A good reagent produces stable derivatives without harmful by-products that interact with the analytical column, in a reaction that is almost 100% complete. Poor reagents cause rearrangements or structural alterations during formation, and contribute to loss of sample during reaction. Most manufacturers of derivatising reagents provide information on the potential uses of each product, along with standard operating instructions. Entire texts, such as that by Blau and Halket (1993), are devoted to this topic. Chiral separations
Chiral compounds can be derivatised to improve their chromatographic characteristics, and the enantiomers separated on a chiral stationary phase. Both enantiomers behave similarly, provided that steric hindrance does not preclude a reaction with one enantiomer. An alternative approach is to use a chiral derivatising reagent which, when reacted with enantiomers, produces diastereoisomers that can then be separated on a conventional stationary phase. As with enantiomers, diastereoisomers still produce similar mass spectra, but are resolved in time by the chromatography column. This approach is less expensive and also less restrictive, since a dedicated column is not required. Care should be taken to ensure the enantiomeric purity of the derivatising reagent, and to guard against racemisation during the reaction. n-Trifluoroacetyl-Lpropyl chloride (TPC) in triethylamine and chloroform (or ethyl acetate) is a commonly used chiral reagent that couples with enantiomeric amines. Excess reagent is washed off with 6 mol/L HCl and the organic phase is dried over magnesium sulfate. For chiral alcohols, (1R,2S,5R)(–)-menthylchloroformate (MCF) reacts well if pyridine is used as a catalyst.
Quantitative determinations Quantitative work usually requires some form of sample preparation to isolate the drug from the bulk of the sample and some degree of concentration or, more rarely, dilution. These processes inevitably introduce a degree of analytical error. A further difficulty is caused by the non-reproducibility of injected volumes. To compensate for these errors, it is usual to compare the response of the unknown with the response of an added internal standard. The internal standard
should be added as early as possible in the assay process and should have chromatographic properties matching the drug’s as closely as possible, preferably with a longer retention time. It is often possible to obtain unmarketed analogues of drugs, or compounds specially synthesised for use as internal standards (e.g. a methyl addition or a halogen substitution). However, the internal standard usually does not behave exactly as the drug and careful control of variables, such as pH, is necessary. If a derivative is to be prepared, the internal standard should also be amenable to derivatisation. Use of an inappropriate internal standard can seriously affect precision (Dudley 1980). If a mass spectrometer is being used as the detector, then the ideal internal standard is a 3H- or 13C-substituted analogue of the drug, a number of which are readily available at reasonable cost. Calibration should include points of higher and lower concentrations than the sample, and quality assurance samples should be included at appropriate concentrations in frequently run assays. Peak measurement may be by peak height or by the peak area obtained by integration. If the peaks show even a modest degree of tailing, use of peak area usually provides a more accurate quantitative result. A plot of the ratio of peak height (or area) of the drug to internal standard versus concentration is a straight line with most detectors. Care should be taken in the preparation of standards to match the matrix to that of the specimens, and to allow for any associated salt or water of crystallisation in the calculation of the concentration. The best results are obtained when the amount of internal standard used produces a peak response ratio of 1 at the mid-point of the calibration range.
Optimising operation conditions to customise applications Additional sensitivity can be achieved by increasing sample size, using a concentration step, derivatisation, injecting a larger sample volume, selecting a different stationary phase or using the detector at a higher sensitivity level. When attempting a new analysis, it is advisable first to review published literature for a method that can be copied or for a method that involves a similar type of compound and can be adapted. Column manufacturers’ catalogues are a useful source of information and invariably show examples of separations performed with their columns. Data on boiling points and RI (see monographs in Volume 2) are also useful indicators. If the review is not helpful, a start can be made with a standard column, such as a 100% methyl-PSX capillary column (25 m with a 0.5 mm film) and using standard flow conditions (1–2 mL/min helium). The oven temperature should be taken from 80 C to 300 C at 10 /min (or started at 200 C or 250 C if only an isothermal oven is available). A solution of the compounds of interest in ethanol or methanol should be injected with the injector temperature set at 250 C. If a peak tails, derivatisation or use of a more polar stationary phase should be considered. Fine-tuning is carried out once some peaks have been obtained. Having established the chromatography, the extraction and concentration steps can be determined. Manufacturers’ catalogues are again a useful source for both derivatisation and solid-phase extraction procedures. Good preventive maintenance is essential. The injector (or liner) should be cleaned periodically, and any glass wool changed regularly (approximately every 100 to 1000 injections, depending on the quality of the extracts). For capillary columns, the performance is improved by periodically removing the first 5–10 cm of capillary tubing, or a retention gap could be considered for dirty samples. It is advisable to monitor performance by selecting certain performance criteria (e.g. a certain response size or amount of acceptable separation between two closely eluting components) to indicate when maintenance is required. The manufacturers’ instructions for cleaning detectors should be followed. The presence of traces of contaminants in the carrier gas supply shortens the column life drastically, and also causes detector deterioration. In-line filters (to remove oxygen, hydrocarbons, etc.) and molecular sieves (to remove water vapour) are strongly recommended, and the use of stainless-steel gas tubing minimises further contamination.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:8
Optimising operation conditions to customise applications Carrier gas flow should be optimised for a particular column and a particular carrier gas. This is most important for capillary columns. Fig. 40.6 shows the relationship between efficiency expressed as the HETP versus carrier gas velocity (van Deemter plot) for a 28 m 0.25 mm i.d. WCOT OV-101 column. Modifying the mobile phase in GC has very little effect compared with that observed with HPLC or thin-layer chromatography (TLC) and, in general, affects efficiency rather than selectivity. Nitrogen gives higher efficiency but at the expense of longer analysis time, while the less dense, but more hazardous, hydrogen gives lower efficiency but faster analysis. In practice, nitrogen is usually used for packed columns and helium for capillary columns. Certain detectors impose restrictions on the choice of carrier gas, but an additional supply of gas can be added to the column effluent to purge the detector. Experimenting with higher flow and a lower operating temperature (or vice versa) can give rewarding results for the separation of compounds that elute closely. This effect is particularly noticeable for two compounds that have different polarities, as the retention of the more polar compound is influenced to a greater extent the longer it resides in the column (non-polar compounds elute in boiling point sequence). Conditions of constant flow improve the efficiency of late-eluting peaks and produce faster chromatography than do constant pressure conditions. For a particular separation, the lowest temperature compatible with a reasonable analysis time should be used. In general, retention times double with each 20 C decrease in temperature. If the time is excessive, it is generally better to reduce the stationary phase loading or use a shorter column than to increase the column operating temperature. There is a maximum temperature at which a column can be operated and there is also a minimum temperature below which efficiency drops sharply. Manufacturers give the temperature operating ranges for each of their stationary phases (see Table 40.3). The stationary phase must be a liquid at the temperature of operation, and if a column is run at too low a temperature to obtain longer retention times the stationary phase may still be in the solid or semi-solid form. When using temperature programming, experimentation with a faster initial ramp followed by a slower subsequent ramp or an isothermal period can help resolve problematic separations. Efficiency can also be improved by decreasing the column diameter or increasing the column length. The resultant increase in analysis time (particularly if the flow must be reduced to accommodate the increased pressure demand imposed by a narrower column) can usually be offset by using a slightly higher operating temperature (temperature increases
affect retention time much more than do increases in gas flow). As shown in Table 40.7, reducing the diameter of a capillary column markedly increases efficiency, but the retention time remains constant only as long as the same phase ratio is maintained. Therefore, unless there is a simultaneous reduction in film thickness, retention increases in direct proportion to the phase ratio. The solvent used for the sample can sometimes produce unexpected derivatives that give different retention times (traces of acetic anhydride that remain in butyl acetate avidly derivatise primary amines at room temperature). An inert non-polar solvent should be used if possible to minimise the co-extraction of unwanted contaminants. Acetone, other ketones, ethyl acetate and carbon disulfide readily form derivatives with primary amines and should be avoided. The choice of injector type and injection solvent also play an important part in the chromatography. A solvent volume should be chosen that does not expand to exceed the capacity of the injector (see Table 40.5), otherwise backflush and irreproducible results are obtained. Split injection significantly reduces the amount of solvent and associated contaminants that enter the column and, although the analyte response is reduced, the improvement in the signal-to-noise ratio often results in enhanced sensitivity. The use of a selective detector, such as an ECD (with the preparation of a strongly responsive derivative if appropriate), can improve sensitivity typically up to 100-fold. Similarly, switching from full scan to SIM in MS improves the sensitivity, usually by a factor of 10. However, selective detectors should not be used as a substitute for cleaning up of sample extracts, as loading contaminants onto the column affects the chromatography adversely, even if the selective detector does not respond to the compounds. Increasing the detector temperature may also improve sensitivity. Fronting or splitting of peaks indicates column overload. If the detector sensitivity permits, the best option here is to inject a smaller sample volume (or a more dilute sample), rather than to increase the column loading or diameter, otherwise efficiency is also affected. If trace impurities are sought in the presence of a preponderant component, a number of stationary phases of differing polarities should be tried. Trace impurities are seen easily if they emerge before the main component of a mixture, while they may be lost completely in the tail if they elute just after the large peak. Early peaks are also sharper and thus, for the same peak area, higher – an effect that can contribute enormously to the successful detection of trace substances.
Table 40.7 Relationship of film thickness, phase ratio (b)(a), efficiency (N)(b) and column diameter Film thickness d (mm)
Column internal diameter (mm) 0.10
0.10
0.18
250(a)
450
0.20 500
0.25 625
0.32 800
0.45
0.53
1125
1325
0.18
139
250
278
347
444
625
736
0.25
100
180
200
313
400
450
663
0.40
63
113
125
156
200
282
331
0.42
—
107
119
149
190
265
315
0.50
—
90
100
125
160
225
265
0.83
—
—
60
75
96
136
160
0.85
—
—
59
74
94
133
156
1.00
—
—
50
63
80
113
133
1.27
—
—
—
49
63
88
104
1.50
—
—
—
42
53
75
88
2.55
—
—
—
25
31
44
52
3.00
—
—
—
21
27
38
44
5.00
—
—
—
13
16
23
27
12 500
6600
5940
4750
3710
2640
2240
Efficiency N (b)
Phase ratio b = r/2d, where r = column radius (mm), d = film thickness (mm). N, theoretical plates per metre; maximum efficiency calculated for a solute with k = 5.
(a)
(b)
653
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:23:8
654
Gas Chromatography
Two-dimensional GC
For most quantitative applications in drug analysis the chromatogram contains only two or three compounds, while some qualitative applications may contain 20 or more peaks of interest. Using an efficient column maximises the probability that a peak in a given time window consists of only one compound and that it is indeed the compound of interest. Other separations are far more complex, and the compound of interest may be present at minute concentrations relative to the background (e.g. flavours in foods or trace residue analysis in foods or groundwater). While these analyses can be fine-tuned to a limited extent by the use of element-specific detectors, the problem of obtaining a clean peak for positive identification and quantitation often remains. Consideration of equation (40.5c) shows column resolution to be related to two terms that can be varied by the analyst. The first of these is N, the number of theoretical plates, which in a typical capillary column is a few more than 100 000 plates. However, since R increases with the square root of N, a substantial increase in resolution can be obtained only by a very large increase in column length, and with a correspondingly large increase in analysis time. Some 500 million theoretical plates would be needed to separate 99 compounds out of a 100 component mixture. The other term is a, which describes the selectivity of a stationary phase for a particular pair of analytes. A modest increase in a can have a significant impact on resolution. This same philosophy was applied by analysts in the 1970s and 1980s who used two or more complementary stationary phases in parallel housed in different packed columns to make positive peak identifications (Moffat et al. 1974b). These columns were selected to have chemical properties as different as possible, but were limited by temperature compatibility since they were often placed in the same GC oven, and sometimes were split off from the same injector to allow reproducible temperature programming. However, recent advances in electronic pressure control and electronic proportional back-pressure regulators with pressure sensing, the manufacture of inert connector fittings and improvements in cryogenic focusing devices are enabling analysts to contemplate using two different analytical columns in series to achieve a satisfactory result. Two main multidimensional approaches are receiving attention for routine use: two-dimensional gas chromatography (GC GC) and 2DGC with heartcutting. Instruments are now commercially available with two independently operated and controlled column ovens in a variety of injector and detector configurations. 2D-GC techniques began with heartcutting, in which only a timed portion of the chromatographed effluent from the first column was diverted from the detector or waste line into a second column of different polarity. A connector (or modulator) was employed to trap and focus the first eluent into narrow bands and transfer it to the second column at a rate that preserved the separation already achieved. In GC GC the entire chromatography effluent from the first column is introduced to the second one. Difficulties arise when the retention of a solute on the second column exceeds the modulation cycle and ‘wraparound peaks’ appear with the solutes in subsequent cycles. A more recent modification, aimed at preventing this phenomenon, is called stopped-flow GC GC. Here, the flow through the first column is stopped for a brief period, typically a few seconds, during each modulation cycle. This allows not only for better preservation of the separation of the first column, but also for longer separation times on the second column because subsequent bands are held up. This renders the secondary separation time independent of the modulation cycle, and increases the options for varying the chromatography conditions. A disadvantage is the longer run time and associated larger data file. Time-of-flight (TOF)-MS is almost mandatory to de-convolute the rapid analysis in the second column. These techniques have been demonstrated across many areas of industry, for example for identification of flavours in liquors, pesticide residues in foods and essential oils, and oxygenates in gasoline. The topic was recently reviewed by Pierce et al. (2008), and detailed examples showing hardware configurations can be found at www.chem.agilent.com/cag/prod/GC/Simplified_2DGC.
Specific applications The systems given below are applicable to the routine screening, separation and identification of groups of drugs and chemicals. They are not
exhaustive lists and references to specific systems for individual drugs and chemicals are given in the relevant monographs. Some of these systems use columns that are identical or very similar in terms of discriminating power (see Table 40.3), but are operated with different temperature programmes for specific groups of compounds. Moreover, some groups of substances are chromatographed as derivatives rather than as the parent compounds. The most commonly used general screening system is a 100% dimethyl-PSX (methyl-PSX or X-1) capillary column (for packed columns, SE-30, OV-1 or OV-101 is equivalent). This should always be used for screening purposes, since it has the best chance of eluting any compound of interest. Analysts have collaborated to compile comprehensive lists of retention indices using this system (De Zeeuw 2002), some of which are included in the Index of Gas Chromatographic Data. Most of the data are for the drugs themselves, but thermal decomposition may occur and the peak observed may be for the decomposition product (referred to as ‘artefact’) rather than the original drug. Where the drug is known to chromatograph badly, or to decompose, data are given for suitable derivatives (e.g. methyl or ethyl esters for the sulfonamides, and TMS derivatives for hydroxides). Wherever possible, the RI of the drug is given, since this is a more reproducible parameter than retention time or relative retention (see discussion above). However, if a laboratory prefers routinely to use the latter parameters, the RI data can be converted easily after chromatography of a few representative drugs and using a regression analysis of RI against either retention time or relative retention. RIs for some additional non-drug substances that might interfere with toxicological analyses, but are not included in the monographs. A nitrogen–phosphorus (alkali flame ionisation) detector is the best detector for nitrogenous drugs and phosphorus-containing pesticides, but an FID should also be used, since some drugs do not contain nitrogen (e.g. some anti-inflammatory agents). ECDs are excellent for benzodiazepines and halogen-containing compounds, such as some phenothiazines and herbicides. Extra selectivity can always be obtained by using element-specific detectors (e.g. those for phosphorus and sulfur for compounds that contain these elements). Additional specificity or confirmation of identity can be obtained by using a mass-selective detector, such as MS or an ion-trap detector. Where improved fragmentation can result from the use of derivatisation, data for suitable derivatives have been included. As mass spectrometry has matured as a technique, significant improvements in detection of higher-mass fragments have enabled the use of larger derivatising reagents such as heptafluorobutyrate (HFB). In any analysis for an unknown compound, the data obtained from complementary techniques, such as TLC, HPLC or colour tests, should always be assessed for compatibility with the GC result. (In the tables of retention indices given here, a dash indicates that no value is available for the compound, not that it does not elute.) General screen, systems GA and GB Both systems use standard columns that are able to chromatograph a wide variety of drugs and chemicals. System GB uses a slightly more polar column, which gives better peak shapes for hydroxylated compounds (many drug metabolites are hydroxylated), better resolution between structural isomers and improved peak shape for primary amines over the less polar GA. However, the retention indices are very similar for GA and GB, and can be interconverted using the equations: GB RI ¼ 1:079 ðGA RIÞ 66 or GA RI ¼ ðGB RIþ66Þ=1:079 As the stated values for drugs are retention indices, the operating conditions for the columns may be varied to suit particular laboratory situations.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:24:1
Specific applications n
System GA
Details are taken from the TIAFT book (De Zeeuw 2002) and the PMW Spectral Library (Pfleger et al. 2004). Chromatography details are given for both systems below. n
n
n n n n n n n n
Column: 3% SE-30 or OV-1 on 80 to 100 mesh Chromosorb G HP (acid washed and dimethyldichlorosilane treated) glass (2 m 2 mm i.d.); it is essential that the support be fully deactivated. Temperature: Normally between 100 C and 300 C; for isothermal conditions, an approximate guide to temperature is to use the RI divided by 10. Carrier gas: N2, 45 mL/min. Capillary column: 100%-dimethyl-PSX (X-1) (10–15 m 0.32 or 0.53 mm i.d., 1.5–3 mm). Carrier gas: He. Temperature programme: 135 C for 4 min to 200 C at 13 /min to 312 at 6 /min for 6 min. Column: HP1 (100%-dimethyl-PSX) fused-silica capillary (12 m 0.2 mm i.d., 0.33 mm). Injector: 280 C splitless mode. Temperature programme: 100 C for 2 min to 310 C at 30 /min for 8 min. Carrier gas: He, 1 mL/min.
System GB
Data generated by the author. n n n n
Column: 5% phenyl–95% dimethyl-PSX (X-5) capillary (20–30 m 0.2 or 0.25 mm i.d., 0.5–1 mm). Carrier gas: He, 1 mL/min. Temperature programme: 90 C for 0.7 min to 240 C at 35 /min to 290 C at 8 /min to 325 C at 25 /min for 6 min. Reference compounds: n-Alkanes with an even number of carbon atoms, or a reference drug mix that contains amfetamine (1125), ephedrine (1365), benzocaine (1545), methylphenidate (1725), diphenhydramine (1870), tripelennamine (1976), methaqualone (2135), trimipramine (2215), codeine (2375), nordazepam (2490), prazepam (2648), papaverine (2825), haloperidol (2930) and strychnine (3116). (RI values for system GA are given in parentheses for the drug mix.)
655
Retention indices: Values for drugs in these systems are found in drug monographs and in the Indexes of Analytical Data; they are also included in the systems for specific groups of drugs that follow. The search window should be 50 RI units if hydrocarbons are used to calculate RI, or 30 RI units if a reference drug mixture is used for the RI calculation.
Amfetamines and other stimulants Amfetamines are basic drugs that require strongly alkaline conditions to be extracted from aqueous solution. The conditions are too basic to extract the phenolic metabolites, but these can be recovered at pH 8 or 9 and the extracts combined prior to chromatography. For high sensitivity, back extraction into dilute sulfuric acid (0.05 mol/L) is a useful clean-up procedure. When using packed columns, derivatives are almost always required for the primary and secondary amines, since the peaks tail badly. Suitable derivatives are acetyl, trifluoroacetyl, pentafluoropropionate or TMS (see Derivative formation). With capillary columns, derivatives are used most often to improve mass spectral patterns or to modify the separation of compounds that elute closely. For hydroxylated metabolites, derivatisation is invariably required to achieve acceptable chromatography. Data for the most commonly used derivatives are given in Table 40.8. Care must be taken to avoid drug loss during solvent evaporation, which can be obviated by adding a small amount of concentrated aqueous acid (20 mL 6 mol/L HCl) to the organic solvent. Unless otherwise stated, GC retention data and mass spectral data are identical for both D- and L- (þ and –) enantiomers. To differentiate enantiomers (such as D- and L-metamfetamine or amfetamine), a chiral column or chiral derivatising reagent is required (Cody, Schwarzhoff 1993). At present, all amfetamine- or metamfetamine-producing drugs (aminorex, amfetaminil, clobenorex, ethylamfetamine, fencamine, fenethylline, fenproporex, mefenorex, prenylamine, benzfetamine, dimethylamfetamine, famprofazone, furfenorex) are racemates (with the exception of L-selegiline, L-metamfetamine and dexamfetamine). Stereo-inversion does not occur in humans (Nagai, Kamiyama 1991). Drugs that are metabolised to amfetamines, but are not themselves classified as such, are also listed. System GA or GB, previously described, may be used as well as system GC.
Table 40.8 GC retention data and mass spectral data for the amfetamines and derivatives (reference compounds are n-alkanes with an even number of carbon atoms; AC, acetyl; ET, ethyl; HFB, heptafluorobutyrate; PFP, pentafluoropropionate; TFA, trifluoroacetyl; TMS, trimethylsilyl) Compound
System
Principal ions (m/z)
GA
GB
GC
Amfetaminil (metabolised to amfetamine, see below)
1755
—
—
132
105
133
89
77
65
Amfetamine (D or L)
1125
1150
—
91
65
51
63
89
120
Amfetamine-TFA
1095
—
1536
140
118
91
69
65
117
Amfetamine-PFP
1330
—
—
118
190
91
119
65
117
Amfetamine-TMS
1190
—
—
116
73
100
91
117
192
Amfetamine-AC
1501
—
—
44
86
118
91
117
65
Art (formyl)
1100
1142
—
56
91
125
146
147
132
M (3OH-)-PFP2
1520
—
—
190
280
119
253
69
M (3OH-)-TMS2
1850
—
—
116
73
100
280
117
— 179
M (3OH-)-AC2
1930
—
—
86
134
176
107
77
235
M (4OH-)
1480
—
—
56
107
77
108
91
151
M (4OH-)-AC
1890
—
—
134
107
86
77
133
193
M (4OH-)-AC2
1900
—
—
134
86
176
107
77
133
M (3,4-di-OH-)-AC3
2150
—
—
86
150
234
192
137
123
M (OH-methoxy-)
1465
—
—
138
137
122
123
94
181
M (OH-methoxy-)-AC2
2065
—
—
164
86
206
137
165
265
M (desamino-oxo-OH-)-AC
1520
—
—
107
149
150
176
192
—
M (desamino-oxo-OH-methoxy-)
1510
—
—
137
180
94
122
138
107
M (desamino-oxo-OH-methoxy-)-AC
1600
—
—
137
180
138
109
122
222 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:24:22
656
Gas Chromatography
Table 40.8 continued Compound
System
Principal ions (m/z)
GA
GB
GC
1735
—
—
123
166
208
150
124
250
Aminorex (metabolised to amfetamine)
2065
—
—
56
118
162
91
119
145
Amiphenazole-AC2
2575
—
—
191
233
121
275
149
257
3,4-Benzodioxazol butanamine (BDB)
1570
1622
—
58
136
77
135
164
193
BDB-TFA
1705
—
—
135
176
154
77
161
289
BDB-PFP
1700
—
—
135
176
119
204
126
339
BDB-AC
1950
—
—
58
176
162
100
135
235
M (desamino-oxo-di-OH-)-AC2
Art (formyl)
1585
—
—
70
135
77
205
176
92
M (desmethylenyl-methyl-)-AC2
2140
—
—
58
178
220
100
137
279
2235
—
—
58
100
164
248
123
307
2,3-Benzodioxazol butanamine (2,3-BDB)
1550
1602
—
58
77
135
83
164
193
2,3-BDB-TFA
1705
—
—
176
154
135
77
136
289
2,3-BDB-PFP
1615
—
—
135
176
119
204
136
339
2,3-BDB-TMS
1670
—
—
130
73
135
236
250
77
M (desmethylenyl-)-AC3
1895
—
—
58
176
100
135
235
131
1575
—
—
70
77
135
205
176
105
Bemegride
—
—
1253
—
—
—
Benzfetamine (metabolised to metamfetamine and amfetamine)
1855
1899
2172
2,3-BDB-AC Art (formyl)
— 91
—
—
148
65
149
92
56
N-Benzylpiperazine (BZP)
—
1530
—
91
134
176
56
120
146
BZP-AC
1920
—
—
91
146
85
134
132
218
BZP-HFB
1730
—
—
91
281
372
175
146
295
BZP-TFA
1665
—
—
91
181
272
195
146
132
BZP-TMS
1860
—
—
102
248
157
86
116
233
M (4-OH-) isomer 1-AC2
2275
—
—
107
85
149
192
204
276
M (4-OH-) isomer 2-AC2
2245
—
—
107
149
204
85
190
276
M (OH-methoxy-)-AC2
306
2380
—
—
137
85
127
179
234
4-Bromo-2,5-dimethoxyamfetamine (DOB)
1804
1875
—
44
77
230
232
105
91
DOB-TFA
1935
—
—
229
231
256
258
69
369
DOB-PFP
1905
—
—
229
231
119
256
258
419
DOB-TMS
1920
—
—
116
73
117
229
272
201
2150
—
—
86
256
258
162
315
317
1790
—
—
56
254
256
285
229
199
4-Bromo-2,5-dimethoxyphenethylamine (2C-B, BDMPEA)
1785
1867
—
230
232
215
217
259
261
2C-B-AC
303
DOB-AC Art (formyl)
2180
—
—
242
244
229
148
301
Art (formyl)
1840
1860
—
242
240
229
231
271
273
M (O-desmethyl-) isomer 1-AC2
2410
—
—
228
230
287
289
215
329
M (O-desmethyl-) isomer 2-AC2
2440
—
—
228
230
287
289
215
329
M (O-desmethyl deamino-OH-)-AC2
2160
—
—
228
230
288
290
213
329
Cathinone
—
—
—
—
—
—
—
—
—
Cathinone-TFA
1350
—
—
105
77
69
106
140
78
Cathinone-PFP
1335
—
—
190
119
105
280
253
225
Cathinone-TMS
1590
—
—
116
73
77
117
191
206
Cathinone-AC
1610
—
—
86
77
105
191
134
132
1-(3-Chlorophenyl)-piperazine (mCPP)
—
1806
—
154
196
138
111
156
75
mCPP-AC
2265
—
—
166
238
138
154
168
195
M (pOH-) isomer 1-AC2
2515
—
—
182
254
169
184
211
296
M (pOH-) isomer 2-AC2
2525
—
—
182
254
169
184
296
211
M (desethylene-)-AC2
2080
—
—
140
195
153
142
111
169
M (chloroaniline)-AC
1580
—
—
127
129
169
171
99
100
Cathine (see [D+]-norpseudoephedrine)
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:24:41
Specific applications
657
Table 40.8 continued Compound
System
Principal ions (m/z)
GA
GB
GC
M (OH-chloroaniline) isomer 1-AC2
1980
—
—
143
145
167
185
227
169
M (OH-chloroaniline) isomer 2-AC2
2020
—
—
143
185
145
187
78
114
Chlorphentermine
1355
1393
1725
58
107
108
125
168
89
Chlorphentermine-AC
1730
—
—
58
100
86
166
167
125
Chlorphentermine-HFB
1560
—
—
254
125
166
214
169
255
Chlorphentermine-PFP
1515
—
—
204
166
164
154
125
119
Chlorphentermine-TFA
1520
—
—
154
125
114
166
89
69
Chlorphentermine-TMS
1520
—
—
130
73
114
125
89
240
Clobenzorex (metabolised to amfetamine and norephedrine)
1940
—
—
168
125
170
127
89
244
Clobenzorex-AC
2290
—
—
168
125
210
170
91
266
Clobenzorex-PFP
2040
—
—
125
127
118
91
314
316
Clobenzorex-TFA
2075
—
—
125
127
91
118
264
266
M (chlorobenzyl-OH-)-AC2
2565
—
—
226
141
183
200
268
324
M (OH-) isomer 1-AC2
2585
—
—
168
125
210
107
272
364
M (OH-) isomer 2-AC2
2630
—
—
168
125
210
170
134
176
M (OH-methoxy-)-AC2
2690
—
—
168
125
164
137
206
210
M (OH-chlorobenzyl-OH-) isomer 1-AC3
2705
—
—
226
141
183
161
215
268
M (OH-chlorobenzyl-OH-) isomer 2-AC3
2725
—
—
141
226
268
183
125
150
M (OH-chlorobenzyl-OH-) isomer 3-AC3
2775
—
—
226
141
183
161
215
268
M (OH-chlorobenzyl-OH-) isomer 4-AC3
2795
—
—
141
226
183
134
107
268
M (OH-alkyl-OH-)-AC3
2725
—
—
168
125
210
192
150
220
M (di-OH-)-AC3
2765
—
—
168
125
210
192
150
234
Diethylpropion (amfepramone)
1486
1532
1715
100
44
72
101
77
56
M (phenylpropanolamine)
1360
1352
—
44
77
79
51
45
M (diethylnorephedrine)
—
1599
—
Dexamfetamine (see Amfetamine)
—
—
—
—
—
42 —
M (ethylnorephedrine)
—
1457
—
—
—
—
—
—
—
M (N-desethyl-)
—
1423
—
—
—
—
—
—
—
M (N-didesethyl-)
—
1338
—
—
—
—
—
—
—
M (norephedrine)
—
—
1383
—
—
—
—
—
—
2,5-Dimethoxyamfetamine (DMA)
1546
1601
—
44
152
137
121
195
91
DMA-AC
1870
—
—
44
178
86
121
237
91
1550
—
—
56
176
151
121
207
91
2C-E-AC
2000
—
—
192
177
149
179
91
251
2C-E-TFA
1770
—
—
179
192
305
177
149
193
M (O-desmethyl-) isomer 1-AC2
2210
—
—
178
237
165
163
179
279
M (O-desmethyl-) isomer 2-AC2
2240
—
—
178
237
165
163
179
279
M (OH-) isomer 1-AC2
2340
—
—
250
191
207
309
175
237
M (OH-) isomer 2-AC2
2420
—
—
190
191
250
309
164
295
M (OH-) isomer 3-AC2
2500
—
—
196
195
250
208
309
212
M (desamino-OH-)-AC
1850
—
—
192
177
149
91
252
179
M (O-desmethyl-desamino-OH-) isomer 1-AC2
1990
—
—
178
163
145
165
238
280
M (O-desmethyl-desamino-OH-) isomer 2-AC2
2000
—
—
178
163
220
238
154
280
M (O-desmethyl-OH-) isomer 1-AC3
2430
—
—
176
235
177
277
309
337
M (O-desmethyl-OH-) isomer 2-AC3
2460
—
—
176
177
235
277
161
337
2,5-Dimethoxy-4-ethylthio-b-phenethylamine (2C-T-2)
1980
—
—
212
211
183
241
153
197
2C-T-2-AC
2310
—
—
224
211
283
209
153
181
Art (formyl) 2,5-Dimethoxy-4-ethyl-b-phenethylamine (2C-E)
2C-T-2-TFA M (N-acetyl-)
2210
—
—
211
337
224
181
151
222
2310
—
—
224
211
283
209
153
181 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:24:49
658
Gas Chromatography
Table 40.8 continued Compound
System
Principal ions (m/z)
GA
GB
GC
M (N-acetyl-)-AC
2400
—
—
224
211
209
181
153
M (desamino-OH-)-AC
2050
—
—
224
284
209
167
225
100
M (O-desmethyl-)-AC2 or (O-desmethyl-N-acetyl-)-AC 2120
—
—
269
210
197
252
311
297
M (OH-N-acetyl-)-TFA
2270
—
—
259
427
260
428
367
M (O-desmethyl-)-TFA2
1980
—
—
306
293
419
209
294
325
307
2290
—
—
306
323
293
307
355
197
2,5-Dimethoxy-4-iodo-b-phenethylamine (2C-I)
2330
—
—
278
263
307
247
279
232
2C-I-AC
2260
—
—
290
349
275
277
148
247
2C-I-TFA
2100
—
—
290
277
247
275
231
403
M (O-desmethyl-) isomer 1-AC2
2480
—
—
276
335
233
263
259
377
M (O-desmethyl-) isomer 2-AC2
2500
—
—
276
335
263
261
377
358
M (desamino-OH-)-AC
2150
—
—
290
275
148
350
247
277
M (O-desmethyl-desamino-OH-) isomer 1-AC2
2240
—
—
276
336
134
261
191
378
M (O-desmethyl-desamino-OH-) isomer 2-AC2
2275
—
—
276
261
336
263
150
378
—
—
M (O-desmethyl-N-acetyl-)-TFA
2,5-Dimethoxy-4-methyl-b-phenethylamine (2C-D) 1940
—
—
178
135
163
165
179
237
M (O-desmethyl-) isomer 1-AC2
2130
—
—
164
223
151
149
265
165
M (O-desmethyl-) isomer 2-AC2
2200
—
—
164
223
151
149
265
206
M (OH-)-AC2
2390
—
—
236
295
193
177
235
223
M (desamino-OH-)-AC
1740
—
—
178
163
135
238
79
104
M (O-desmethyl-desamino-OH-) isomer 1-AC2
1880
—
—
164
149
72
224
182
266
M (O-desmethyl-desamino-OH-) isomer 2-AC2
2C-D-AC
1890
—
—
164
149
121
224
266
206
2,5-Dimethoxy-4-propylthio b-phenethylamine (2C-T-7) 2470
—
—
226
183
225
153
169
255
2C-T-7-AC
2410
—
—
238
255
181
297
153
183
2C-T-7-TFA
2170
—
—
225
351
181
153
238
183
M (OH-N-acetyl-)-AC and (OH-)-AC2
2590
—
—
296
236
101
355
356
283
M (desamino-OH-)-AC
2080
—
—
238
298
181
239
255
299
M (OH-)-TFA2
2105
—
—
337
463
350
181
2350
—
—
350
409
351
337
181
2,5-Dimethoxy-4-methylamfetamine (STP or DOM)
1612
1652
—
44
166
151
135
91
209
DOM-PFP
1730
—
—
165
192
135
119
91
355
DOM-AC
2020
—
—
44
192
86
165
166
251
DOM-AC2
2090
—
—
192
165
86
135
177
293
Art (formyl)
1565
—
—
56
190
165
135
221
91
M (O-desmethyl-)-PFP2
1780
—
—
324
297
190
119
325
487
M (OH-)-PFP2
1830
—
—
354
327
190
119
355
517
M (OH-)-AC2
2260
—
—
250
309
86
164
191
91
M (desamino-oxo-OH-)-PFP2
2045
—
—
353
326
516
233
206
396
M (desamino-oxo-OH-)-AC2
2560
—
—
164
249
206
233
308
91
1235
—
1429
72
91
73
44
42
56
M (OH-N-acetyl-)-TFA
Dimethylamfetamine (trimethylbenzeneethenanamine; metabolised to metamfetamine) Ephedrine (D or L)
1365
1410
—
58
77
105
146
131
—
Ephedrine-TFA2
1345
—
—
154
110
69
115
244
338
Ephedrine-PFP2
1370
—
—
204
119
95
160
294
338
Ephedrine-TMS2
1620
—
—
130
73
147
149
163
294
Ephedrine-AC2
1795
—
—
58
100
148
117
249
122
M (nor-)
1360
1356
—
77
79
91
107
118
132
M (nor-)-TFA2
1355
—
—
140
69
230
203
105
175
M (nor-)-PFP2
1380
—
—
190
119
105
117
280
253
M (nor-)-TMS2
1555
—
—
116
73
117
147
280
163
M (nor-)-AC2
1805
—
—
86
87
107
134
176
235
M (OH-)
1875
—
—
58
71
77
95
148
107
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:25:9
Specific applications
659
Table 40.8 continued Compound
System
Principal ions (m/z)
GA
GB
GC
2145
—
—
58
100
205
247
123
Etafedrine
1519
1510
1737
86
58
87
42
56
Ethylamfetamine (etilamfetamine; also metabolised to amfetamine)
1230
—
—
72
91
117
148
162
—
Ethylamfetamine-AC
1675
—
—
72
114
91
205
148
119
Ethylamfetamine-HFB
1485
—
—
268
240
91
118
269
169
Ethylamfetamine-PFP
1450
—
—
218
190
118
91
119
117
Ethylamfetamine-TFA
1450
—
—
168
69
140
118
91
83
M (OH-methoxy-)
1640
—
—
72
94
137
122
77
209 263
M (OH-)-AC3
307 77
M (4OH)-AC2/(PHEA)-AC2
1995
—
—
72
114
134
176
107
M (OH-methoxy-)-AC
2000
—
—
72
164
114
137
251
—
M (OH-methoxy-)-AC2
2080
—
—
72
114
164
206
137
293
2200
—
—
72
114
150
234
192
321
2965
—
—
286
229
91
135
377
—
M (3-OH-methylpropylphenazone)
2410
—
—
231
246
232
215
77
M (3-OH-methylpropylphenazone)-AC
2240
—
—
245
232
273
288
190
274
Fencamfamin (also metabolized to amfetamine)
1675
1723
—
98
58
84
215
71
186
Fencamfamin-AC
2085
—
—
170
142
58
97
91
84
Fencamfamin-HFB
1795
—
—
170
142
67
117
129
280
Fencamfamin-TFA
1970
—
2180
142
170
91
180
115
242
Fencamfamin-TMS
1780
—
—
170
258
287
272
259
130
Fencamfamin-PFP
1755
—
—
170
142
91
230
105
292
M (di-OH-)-AC3 Famprofazone (also metabolised to metamfetamine)
154
M (desethyl-)-AC
2005
—
—
170
142
91
171
115
229
M (desethyl-OH-)-AC2
2305
—
—
142
168
228
91
119
287 148
Fenethylline (fenetylline; also metabolised to amfetamine) 2830
2900
—
250
70
207
91
119
Fenethylline-AC
3110
—
—
250
207
91
292
180
383
Fenethylline-HFB
2815
—
—
91
446
419
266
180
118
Fenethylline-PFP
2790
—
—
91
396
369
207
339
217
Fenethylline-TFA
2840
—
—
91
166
346
319
207
170
M (N-desalkyl)-AC
2480
—
—
206
180
193
265
122
86
M (etophylline)
2125
—
—
180
95
224
109
122
194
M (etophylline)-TMS
2160
—
—
180
73
281
296
252
123
M (etophylline)-AC
2200
—
—
87
266
206
180
122
223
Fenfluramine
1230
1252
—
72
44
159
73
58
42
Fenfluramine-TFA
1455
—
1621
168
140
159
169
186
308
Fenfluramine-PFP
1455
—
—
218
190
159
119
168
358
Fenfluramine-AC
1580
—
—
72
114
159
216
58
254
M (desethyl-, norfenfluramine)
1133
1157
—
44
42
159
43
45
184
M (desethyl-)-AC
1510
—
—
86
159
186
109
226
245
Fenproporex (metabolised to amfetamine)
1585
1648
—
97
56
91
68
132
173
Fenproporex-TFA
1705
—
—
193
118
140
91
56
152
Fenproporex-PFP
1685
—
—
243
118
190
56
91
202
Fenproporex-AC
1915
—
—
97
56
139
91
118
65
4-Hydroxyamfetamine (PHA)
1480
—
—
56
107
77
108
91
151
4-Hydroxyamfetamine-AC
1890
—
—
134
107
86
77
133
193
4-Hydroxyamfetamine-AC2
1900
—
—
134
86
176
107
77
133
M (methoxy-)
1465
—
—
138
137
122
123
94
181
M (methoxy-)-AC2
2065
—
—
164
86
206
137
165
265
M (1-OH-)/(4-OH-norephedrine)-AC3
2150
—
—
86
150
234
192
137
123 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:25:14
660
Gas Chromatography
Table 40.8 continued Compound
System
Principal ions (m/z)
GA
GB
GC
Mebeverine (metabolised to PMEA)
3045
—
—
Meclofenoxate
1770
1804
2200
Mefenorex (also metabolised to Amfetamine)
1719
1602
Mefenorex-TFA
1715
Mefenorex-PFP Mefenorex-AC
Levamfetamine (see Amfetamine) 308
165
309
121
55
58
111
71
42
75
98 59
—
120
122
91
56
65
121
—
—
216
218
140
118
91
154
1710
—
—
266
190
91
118
268
119
1935
—
—
120
122
162
164
91
147
M (OH-dechloro-)-AC2
2060
—
—
144
186
101
91
84
118
M (OH-) isomer 1-AC2
2300
—
—
120
121
58
162
107
77
M (OH-) isomer 2-AC2
2230
—
—
120
121
58
162
107
77
M (OH-methoxy-)
2145
—
—
120
84
122
137
107
256
M (OH-methoxy-)-AC
2360
—
—
120
122
162
164
137
257
M (OH-methoxy-)-AC2
2410
—
—
120
164
162
206
137
341
M (dechloro-di-OH-)-AC3
2400
—
—
144
84
101
107
134
186
M (dechloro-tri-OH-)-AC4
2630
—
—
144
150
186
192
234
84
M (di-OH-)-AC3
2510
—
—
120
122
162
150
192
234
M (dechloro-di-OH-methoxy-)-AC3
2520
—
—
144
164
186
206
101
137
Mephentermine (metabolised to phentermine)
1240
1250
—
72
91
56
65
115
148
Mephentermine-TFA
1335
—
—
168
110
91
56
117
122
Mephentermine-AC
1501
—
—
72
114
91
132
148
117
Mescaline
1680
1737
—
182
167
211
151
148
—
Mescaline-TFA
1830
—
—
181
194
179
307
148
151
Mescaline-PFP
1835
—
—
181
194
357
179
119
151
Mescaline-TMS
1745
—
—
102
73
181
182
268
283
Mescaline-TMS2
1990
—
—
174
73
175
86
340
100
Mescaline-AC
2160
—
—
194
179
181
253
151
148
Art (formyl)
1700
—
—
181
223
182
148
167
77
4-Methoxyamfetamine (PMA)
1412
1410
—
122
121
77
78
91
107
PMA-AC
1720
—
—
44
148
121
86
77
207
4-Methoxyethylamfetamine (PMEA; metabolised to PMA 1660 and 4OH-ethylamfetamine)
1512
—
72
121
91
149
77
192
PMEA-TFA
1775
—
—
168
148
121
140
149
289
PMEA-PFP
1765
—
—
218
148
121
190
149
339
PMEA-TMS
2065
—
—
144
73
145
250
121
264
PMEA-AC
1855
—
—
72
148
141
121
77
235
Methoxyphenamine
1361
1416
—
58
91
59
56
42
121 134
Methylamfetamine (see Metamfetamine) Metamfetamine (also metabolised to amfetamine)
1175
1200
—
58
91
65
56
77
Metamfetamine-AC
1575
—
—
58
100
91
117
191
Metamfetamine-HFB
1460
—
—
254
210
118
91
169
Metamfetamine-PFP
1415
—
—
204
160
118
119
91
69
Metamfetamine-TFA
1300
—
1722
154
110
118
91
69
245
Metamfetamine-TMS
206
— 69
1325
—
—
130
73
59
91
131
M (4-OH-, pholedrine)
1885
—
—
58
77
107
135
150
—
M (4-OH-, pholedrine)-TFA2
1585
—
—
154
110
69
230
155
357
M (4-OH-, pholedrine)-PFP2
1605
—
—
204
160
119
280
154
253
M (4-OH-, pholedrine)-TMS2
1620
—
—
179
206
73
154
110
309
M (4-OH-, pholedrine)-AC2
1995
—
—
58
100
134
176
107
—
M (OH-methoxy-)
1810
—
—
58
137
94
122
65
195
M (OH-methoxy-)-AC2
2115
—
—
58
100
164
206
136
279
M (di-OH-)-AC3
2190
—
—
58
100
150
123
234
307
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:25:23
Specific applications
661
Table 40.8 continued Compound
System GA
Principal ions (m/z) GB
GC
4-Methoxy-metamfetamine (PMMA) (also metabolized to PMA, MDMA and MDA) 1820
—
—
58
100
148
121
77
221
M (O-desmethyl-, pholedrine)-AC2
1995
—
—
58
100
134
176
249
107
M (1-OH-pholedrine)-AC2
2095
—
—
58
100
164
206
137
279
5-Methoxy-N,N-diisopropyltryptamine (5-MeO-DIPT)
1965
—
—
114
72
160
174
144
274
Methyl-3,4-benzodioxazol butanamine (MBDB; also metabolised to BDB, see above)
1630
1690
—
72
57
135
77
178
207
PMMA-AC
MBDB-AC
1995
—
—
72
176
114
135
77
249
MBDB-PFP
1785
—
—
218
176
135
160
119
353
MBDB-TFA
1800
—
—
168
176
135
110
303
140
2170
—
—
72
114
178
220
137
293
M (desmethylenylmethyl-)-AC2
2295
—
—
72
114
164
248
123
321
Methyl-2,3-benzodioxazol butanamine (2,3-MBDB; metabolised to 2,3-BDB)
1610
1660
—
72
57
89
135
178
120
2,3-MBDB-PFP
1710
—
—
218
176
160
135
129
353
2,3-MBDB-TFA
1725
—
—
168
176
110
135
303
140
2,3-MBDB-AC
1965
—
—
72
114
176
135
249
77
2,3-MBDB-TMS
1730
—
—
144
73
135
145
250
264 161
M (desmethylenyl-)-AC3
2,3-Methylenedioxyamfetamine (2,3-MDA)
1470
—
—
44
77
51
135
179
2,3-MDA-AC
1770
—
—
162
77
135
105
86
51
2,3-MDA-HFB
1595
—
—
162
135
240
77
163
375
2,3-MDA-PFP
1545
—
—
162
135
119
190
77
325
2,3-MDA-TFA
1585
—
—
162
135
140
275
77
136
2,3-MDA-TMS
1655
—
—
116
73
77
135
236
251
1490
—
—
56
135
77
191
105
176
1480
1512
—
44
136
135
51
77
179
Art (formyl) 3,4-Methylenedioxyamfetamine (MDA; metabolised to amfetamine metabolites) MDA-AC
1860
—
—
44
162
135
77
86
221
MDA-HFB
1650
—
—
135
162
169
77
240
375
MDA-PFP
1605
—
—
135
162
119
190
136
325
MDA-TFA
1615
—
—
135
162
77
105
136
275
1520
1689
—
56
77
135
191
136
105
3,4-Methylenedioxyethylamfetamine (MDEA; metabolised 1560 to MDA and ethylamfetamine metabolites)
1630
—
72
77
135
105
163
207
MDEA-TFA
1770
—
—
168
162
140
135
125
303
MDEA-PFP
1755
—
—
218
190
162
135
119
353
MDEA-TMS
1825
—
—
144
73
135
264
100
77
MDEA-AC
1985
—
—
72
162
114
135
77
249
3,4-Methylenedioxymetamfetamine (MDMA; metabolised 1585 to MDA, and amfetamine metabolites)
1572
—
58
135
77
177
105
193
MDMA-HFB
1770
—
—
254
162
135
210
77
389
MDMA-PFP
1830
—
—
204
162
160
135
119
339
MDMA-TFA
1720
—
—
154
162
135
110
77
289
MDMA-TMS
1710
—
—
58
100
162
77
135
235
—
—
Art (formyl)
—
1735
—
—
—
—
Methylephedrine (metabolised to ephedrine)
1405
1451
—
72
77
105
115
161
—
Methylephedrine-AC
1495
—
—
72
77
91
105
117
162
Methylephedrine-TFA
1185
—
—
72
134
91
162
115
117
Art (formyl)
—
Methylephedrine-TMS
1485
—
—
72
149
163
236
117
251
2,3-Dimethylbenzodioxazolbutanamine (MMBDB; also metabolised to MBDB)
1660
1700
—
86
71
96
135
192
105
1890
—
—
86
87
123
180
222
264
M (desmethylenyl-methyl-)-AC
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:25:26
662
Gas Chromatography
Table 40.8 continued Compound
System GA
GB
GC
3,4-Methylenedioxy-5-methoxyamfetamine (MMDA)
1690
1743
—
44
166
165
77
65
MMDA-AC
2050
—
—
44
192
165
86
166
77
1685
—
—
56
165
221
120
77
166
4-Methylthioamfetamine (MTA)
1300
1610
—
44
138
137
122
91
78
MTA-TFA
1750
—
—
137
164
122
69
277
140
MTA-PFP
1760
—
—
137
164
122
190
327
91
MTA-TMS
1750
—
—
116
73
117
100
137
238
MTA-AC
Art (formyl)
Principal ions (m/z)
209
1760
—
—
164
86
137
122
117
265
Art (formyl)
1560
—
—
56
137
193
122
78
91
Methylphenidate
1725
1793
2200
84
56
91
115
77
Methylphenidate-AC
2085
—
—
84
126
112
56
275
—
Methylphenidate-TFA
1730
—
—
180
67
150
91
181
126
—
—
—
84
91
56
55
136
175
2328
—
—
—
—
M (ritalinic acid) Methylpiperidyl benzilate 4-Methoxy-a-pyrrolidinopropiophenone (MOPPP) M (desmethyl-)-ET
—
—
—
—
85
1705
—
—
98
77
92
135
233
107
1955
—
—
98
99
121
69
149
247
M (desmethyl-)-TMS
2005
—
—
98
73
56
135
193
276
M (desmethyl-3-OH-)-ET2
2165
—
—
98
99
56
137
165
193
M (desmethyl-3-methoxy-)-ET
2135
—
—
98
99
56
151
123
179
M (desamino-oxo-)
1440
—
—
135
77
92
107
136
178
M (desmethyl-desamino-oxo-)-ET
1530
—
—
149
121
93
65
150
192
M (desmethyl-3-methyoxy-desamino-oxo-)-ET
1680
—
—
179
151
123
108
73
222
M (oxo-)
2120
—
—
112
121
135
164
150
246
M (dihydro-)-TMS
1880
—
—
98
121
135
209
218
292
4-Methyl-a-pyrrolidinopropiophenone (MPPP) M (carboxy-)-ET
1725
—
—
98
56
65
91
119
216
2320
—
—
98
177
149
230
104
275
M (carboxy-)-TMS
2195
—
—
98
290
135
99
—
—
M (oxo-)
1920
—
—
112
69
119
84
113
231
M (OH-)-TMS
2095
—
—
98
290
135
90
99
M (dihydro-)-TMS
1730
—
—
98
73
163
193
276
202
M (desmethyl-3-methoxy-desamino-oxo)-ET
1680
—
—
179
151
123
108
73
222
105
202
—
1960
—
—
98
223
306
321
1595
—
—
98
56
77
69
M (oxo-)
1820
—
—
112
69
77
84
105
217
M (4-OH-)-ET
1955
—
—
98
99
121
69
149
247
M (desmethyl-3-methoxy-)-TMS a-Pyrrolidinopropiophenone (PPP) (also metabolized to cathinone and norephedrine)
M (4-OH-)-TMS
2005
—
—
98
73
56
135
193
276
M (dihydro-)-TMS
1665
—
—
98
73
105
56
188
262
1995
—
—
98
56
99
121
149
178
3,4-Methylenedioxy-a-pyrrolidinopropiophenone (MDPPP) M (desmethylene-)-ET2
2165
—
—
98
99
56
137
165
193
M (desmethylene-3-methyl-)-ET
2135
—
—
98
99
56
151
123
179
321
M (desmethylene-3-methyl-)-TMS
1960
—
—
98
223
306
M (desamino-oxo-)
1525
—
—
149
121
192
65
91
150
M (desmethylene-desamino-oxo-)-ET2
1720
—
—
193
165
137
109
194
136
M (desmethylene-3-methyl-desamino-oxo-)-ET
1680
—
—
179
151
123
108
73
222
M (desmethylene-3-methyl-oxo-)-ET
2290
—
—
112
179
151
208
123
290
M (oxo-)
2290
—
—
112
149
178
121
175
261
M (desmethylene-oxo-)-ET2
2325
—
—
112
151
69
193
222
305
1965
—
—
98
121
149
232
306
223
4 0 -Methyl-a-pyrrolidinohexanophenone (MPHP)
1965
—
—
140
141
91
119
84
202
a-Methyltryptamine (AMT)
1740
—
—
44
131
77
103
174
M (dihydro-)-TMS
—
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:25:30
Specific applications
663
Table 40.8 continued Compound
System
Principal ions (m/z)
GA
GB
GC
Phendimetrazine (metabolised to phenmetrazine)
1334
1504
1735
57
85
191
77
91
Phenmetrazine
1432
1483
—
71
56
177
77
91
105
Phenmetrazine-TFA
1530
—
1873
70
167
98
105
134
273
Phenmetrazine-TMS
1620
—
—
100
73
114
115
143
249
Phenmetrazine-AC
1810
—
—
71
113
86
85
176
219
105
M (OH-) isomer 1
1830
—
—
71
56
193
121
107
105
M (OH-) isomer 2
1865
—
—
71
56
193
106
163
121
M (OH-methoxy-)
1900
—
—
71
56
223
151
107
137
M (OH-) isomer 1-AC2
2150
—
—
71
70
113
85
234
277
M (OH-) isomer 2-AC2
2200
—
—
71
70
113
85
234
277
M (OH-methoxy)-AC2
2320
—
—
71
70
113
86
265
307
Phentermine
1155
1191
—
—
—
—
—
Phentermine-TFA
1100
—
1450
154
59
91
132
114
230
Phentermine-PFP
1305
—
—
204
91
132
164
129
280
Phentermine-TMS
1195
—
—
130
73
91
114
206
221
Phentermine-AC
1510
—
—
58
100
91
117
134
191
N-(1-Phenylcyclohexyl)-2-ethoxyethenamine (PCEEA)
1825
—
—
159
91
204
188
247
218
N-(1-Phenylcyclohexyl)-2-methoxyethenamine (PCMEA) 1770
—
—
91
190
159
283
188
218
—
—
N-(1-Phenylcyclohexyl)-3-ethoxypropanamine (PCEPA)
1915
—
—
218
91
117
261
232
174
N-(1-Phenylcyclohexyl)-propanamine (PCPR)
1630
—
—
174
91
58
217
159
188
Phenylephrine
1606
0000
—
—
—
—
—
—
—
Phenylephrine-TFA
1755
—
—
95
141
123
77
140
136 359
Phenylephrine-TFA2
1755
—
—
140
69
232
121
219
Phenylephrine-TMS3
2110
—
—
116
73
368
146
267
383
Phenylephrine-AC3
2110
—
—
86
87
115
129
165
220
Phenyl-1-ethylamine
—
1078
—
—
—
—
—
—
—
Phenyl-2-ethylamine
1111
1122
—
—
—
—
—
—
— 167
Phenylpropanolamine (see Norephedrine) Prenylamine (also metabolised to amphetamine)
2555
—
—
58
238
91
45
239
Prenylamine-AC
2925
—
—
58
91
100
280
238
164
Prolintane
1634
1660
1849
—
—
—
—
—
M (oxo-)
1895
—
—
140
98
91
86
188
231
M (OH-phenyl-)
2135
—
—
126
127
96
107
190
232
M (OH-phenyl-)-AC
2110
—
—
126
127
107
190
232
274
M (OH-methoxy-phenyl-)-AC
2115
—
—
126
127
137
55
262
304
M (oxo-OH-alkyl-)
2200
—
—
86
71
156
91
188
—
M (oxo-OH-alkyl-)-AC
2255
—
—
138
86
198
156
91
71
M (oxo-OH-methoxy-phenyl-)
2240
—
—
140
98
192
86
163
277
—
M (oxo-OH-methoxy-phenyl-)-AC
2360
—
—
140
192
98
77
234
319
M (oxo-di-OH-)-AC2
2485
—
—
198
156
128
162
279
107
M (oxo-di-OH-phenyl-)
2475
—
—
140
98
86
178
123
263
M (oxo-di-OH-phenyl)-AC2
2450
—
—
140
98
77
141
178
220
M (oxo-OH-phenyl)-AC
2275
—
—
140
98
86
162
204
289
M (oxo-di-OH-methoxy-)AC2
2560
—
—
198
192
156
234
128
377
M (di-OH-phenyl-)-AC2
2295
—
—
126
123
248
150
290
232
M (tri-OH-)-AC3
2630
—
—
198
156
128
178
151
123
Propylhexedrine
1175
1192
—
—
—
—
—
—
—
Propylhexedrine-TFA
1385
—
—
154
182
69
110
155
251
Propylhexedrine-PFP
1385
—
—
204
182
160
119
124
205
M (OH-)
1475
—
—
58
156
81
171
138
108
M (OH-)-AC2
1915
—
—
58
100
74
240
195
255 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:25:31
664
Gas Chromatography
Table 40.8 continued Compound
Propylhexedrine-AC
System
Principal ions (m/z)
GA
GB
GC
1570
—
—
100
1450
1453
—
—
58
182
140
101
114
—
—
—
—
Pseudoephedrine (see Ephedrine) Selegiline (also metabolised to L-metamfetamine and L-amfetamine)
—
M (OH-)
1580
—
—
96
56
97
107
76
171
M (OH-)-AC
1860
—
—
96
56
97
107
77
—
M (nor-)
1350
—
—
82
91
65
67
115
128
M (nor-)-AC
1735
—
—
82
124
91
65
118
214
M (nor-OH-)
1550
—
—
82
107
77
67
135
—
M (nor-OH-)-AC2
2030
—
—
44
182
167
151
107
225
1890
—
—
200
56
174
188
172
272
M (OH-)-AC2
2275
—
—
216
288
56
203
188
330
M (desethylene-)-AC2
1865
—
—
174
187
229
73
175
145
M (OH-desethylene-)-AC3
2275
—
—
190
203
245
287
191
232
M (trifluoroaniline)-AC
1400
—
—
161
203
184
142
114
111
M (OH-trifluoromethylaniline)-AC2
1840
—
—
157
177
219
117
129
261
1,(3-Trifluoromethylphenyl)piperazine (TFMPP) TFMPP-AC
3,4,5-Trimethoxyamfetamine (3,4,5-TMA) 3,4,5-TMA-AC Art (formyl) 2,3,5-Trimethoxyamfetamine (2,3,5-TMA)
1740 2020
—
—
44
208
193
86
181
267
1680
1745
—
56
181
237
148
77
222
2040
—
—
182
167
181
151
107
225
2,3,5-TMA-AC
2285
—
—
208
181
193
86
267
167
2,3,5-Trimethoxymethamfetamine (TMMA)-AC
2310
—
—
58
208
100
224
281
177
1-(1,3-Benzodioxol-5-yl)-butan-2-one
1525
—
—
135
57
77
192
136
105
1-(1,3-Benzodioxol-5-yl)-butan-1-ol
1560
—
—
151
93
65
123
194
Benzylmethylketone (BMK)
1110
1153
—
—
—
—
—
2,5-Dimethoxybenzaldehyde
1345
1381
—
166
63
95
120
151
123
2,5-Dimethoxyphenyl-2-nitroethene
1900
—
—
209
77
133
147
148
162
2,5-Dimethoxyphenylethylamine
1630
1689
—
152
44
137
121
162
181
2,5-Dimethoxytoluene
1020
—
—
137
152
77
109
65
Dimethylphenylethylamine
—
1954
—
—
—
—
—
Isosafrole
1215
—
—
162
104
103
131
3,4-Methylene dioxymethylbenzylamine
—
1423
—
—
—
—
—
—
—
Methylene dioxyphenylacetone
—
1530
—
—
—
—
—
—
—
Methylphenylethylamine acetate
—
1593
—
—
—
—
—
—
—
2-Methylhydroquinone
1210
—
—
124
123
67
95
77
107
Piperonal
1302
1373
—
149
150
63
121
91
—
Piperonylacetone
1315
1357
—
135
77
178
79
105
3,4,5-Trimethoxybenzaldehyde
1550
1630
—
196
181
125
118
93
95
3,4,5-Trimethoxyphenylacetonitrile
1610
—
—
192
207
164
78
149
124
Precursors and intermediates of synthesis of illicit amfetamines
System GC
DA Cowan, personal communication (2003). In this system, the drugs are chromatographed as tertiary bases or trifluoroacetyl derivatives. n n n n
Column: 3% OV-17 on 80–100 mesh Chromosorb W HP glass (2 m 3 mm i.d). Temperature programme: 170 C for 2 min to 270 C at 16 /min for 8 min. Carrier gas: N2, 30 mL/min. Reference compounds: n-Alkanes with an even number of carbon atoms.
n
—
—
— 77
77
94 — 65
136
Retention indices: The retention indices given for system GC are those of the tertiary bases or of trifluoroacetyl derivatives.
Analgesics (non-narcotic) and non-steroidal antiinflammatory drugs Analgesics (non-narcotic) and non-steroidal anti-inflammatory drugs (NSAIDs) are acidic and/or neutral drugs and, although water soluble, are readily extracted at pH 5 (sodium acetate or phosphate buffer) into polar solvents, such as ethyl acetate or diethyl ether. Recovery can be improved by ‘salting out’ using excess solid sodium chloride. Many are arylacetic (indometacin), arylpropionic (ibuprofen), salicylic
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:26:3
Specific applications (diflunisal) or fenamic (mefenamic acid) acid derivatives, and thus require the formation of suitable derivatives prior to GC (data for methyl derivatives are given in Table 40.9; the values given for system GD are retention times of methyl derivatives relative to n-C16H34). This is most pertinent at low concentrations or with packed columns, while at higher concentrations peaks may tail, and there is thus a tendency for retention time to increase with concentration. Capillary columns, especially those with higher phase ratios, give better peaks for underivatised phenols. The arylpropionic acid derivatives are chiral, with the nonsteroidal anti-inflammatory (NSAI) activity usually residing in the S-enantiomer, but these are usually marketed as racemates and undergo enantiomeric inversion in vivo. Separation of NSAI enantiomers has been reviewed (Davies 1997). Systems GA or GB, described above, may be used, or systems GD and GL. System GD
In this system, the substances are chromatographed as their methyl derivatives. n n n n
Column: 3% SE-30 on 80–100 mesh Chromosorb G (acid-washed and dimethyldichlorosilane-treated) glass (2 m 3 mm i.d). Temperature programme: 120 C for 2 min to 260 C at 10 /min for 5 min. Carrier gas: N2, 40 mL/min. Reference compound: Hexadecane (n-C16H34). Note: Free carboxylic acids and phenols will not generally give peaks, although large quantities may give tailing peaks.
n
665
Retention indices and relative retention times: The values given for system GD are retention times of methyl derivatives relative to nC16H34.
System GL
In this system (Maurer et al. 2001), the substances are chromatographed as their methyl derivatives after extractive methylation. Equal volumes of urine and phase-transfer reagent (0.02 mol/L tetrahexylammonium hydrogensulfate in 1 mol/L phosphate buffer, pH 12) are incubated with three volumes of 1 mol/L methyl iodide in toluene on a shaker at 50 C for 30 min. The organic phase is eluted on a diol solid-phase cartridge (conditioned with methanol, then toluene) using diethyl ether–ethyl acetate (95 : 5 v/v). After evaporation to dryness at 60 C, the residue is reconstituted in ethyl acetate for injection. n n n n n
Column: HP1 (methyl-PSX) fused-silica capillary (12 m 0.2 mm i.d. 0.33 mm). Injector: 280 C splitless mode. Temperature programme: 100 C for 2 min to 310 C at 30 /min for 8 min. Carrier gas: He, 1 mL/min. Retention indices are given in Table 40.9.
Anticholinergics The anticholinergics comprise a chemically diverse group of drugs, although most can be extracted successfully from biological specimens under mildly alkaline conditions. Some (atropine, hyoscine) are
Table 40.9 GC retention data and mass spectral data for analgesics (non-narcotic) and NSAIDs (ET, ethyl; Me, methyl) Compound
System GA
Principal ions (m/z) GB
GD
GL
Acemetacin (metabolised to indometacin)
Not eluted
—
—
—
Acemetacin-Me
3150
—
—
—
139
429
141
431
312
430
Acemetacin-ET
3220
—
—
—
139
443
141
312
445
442 131
Art-Me2
2390
Acetanilide (also metabolised to paracetamol) 1368 Alclofenac
—
—
—
—
174
233
291
175
159
1400
—
—
93
135
43
66
65
39
—
—
—
41
226
77
143
181
141
Aletamine
1293
—
—
—
70
120
43
91
39
103
Amidopyrine/aminophenazoine
1895
1992
—
—
56
231
97
111
112
42
M (nor-)
1980
—
—
—
—
—
—
—
—
—
M (bis-nor-)
1955
—
—
—
—
—
—
—
—
—
—
—
—
—
—
M (desamino OH-) Aspirin (metabolised to salicylic acid and salicylamide)
1855
—
—
—
—
1545
—
—
—
120
43
138
92
121
64
Aspirin-Me
1394
1430
—
—
135
194
179
136
91
76
Azapropazone (metabolised to paracetamol and aspirin)
2461
—
—
—
160
300
189
145
188
301
1804
1779
—
—
—
—
—
—
—
—
1840
—
—
—
121
163
151
109
43
122 65
Art Benorilate (metabolised to paracetamol and aspirin) Benoxaprofen
2550
Not eluted —
—
256
301
91
258
119
Benoxaprofen-Me
2485
—
1.98
—
256
315
91
119
258
65
Benzydamine
2380
—
—
—
85
58
86
91
84
70
Bufexamac
—
—
—
—
—
—
—
—
—
Celecoxib
—
—
—
—
381
300
382
301
281
140
Clonixin-Me
—
—
1.61
—
—
—
—
—
—
—
Dexketoprofen (see Ketoprofen)
—
—
—
—
—
—
—
—
—
—
Diclofenac
2271
2231
—
—
214
216
242
295
215
297
Diclofenac-Me
2195
—
1.42
2200
214
242
309
216
311
179
Diclofenac-Me2
2220
—
—
—
228
323
229
325
214
264
2322
2418
—
—
320
355
357
322
228
292
Art
—
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:26:7
666
Gas Chromatography
Table 40.9 continued Compound
System
Principal ions (m/z)
GA
GB
GD
GL
Art (-H2O)
2135
2231
—
—
214
277
242
279
179
Art (-H2O)-Me
2300
2436
—
—
228
230
200
263
291
164
M (OH-)-Me2
2460
—
—
2460
244
339
272
341
201
246
M
—
2592
—
—
214
216
242
277
179
294
M (OH-) isomer 1
—
2600
—
—
230
293
232
295
258
195
M (OH-) isomer 2
—
2941
—
—
230
293
232
271
158
310
Diflunisal
2095
—
—
—
232
250
175
204
176
233
Diflunisal-Me
2050
—
1.20
—
—
—
—
—
—
—
278
247
245
175
188
204
56
42
83
57
77
Diflunisal-Me2
1990
—
—
1990
Dipyrone
—
2069
—
—
M (bisdesalkyl-)
1955
—
—
—
M (desalkyl-)-AC
2395
Etenzamide (also metabolised to salicylamide) — M (desethyl-)-AC
—
—
—
—
—
216
51 —
—
—
—
—
—
—
—
—
—
—
—
—
—
—
—
—
120
92
105
148
150
121
—
228
258
287
198
57
229
1660
—
Etodolac
2333
—
—
Etodolac-Me
—
—
—
2225
228
272
301
198
229
115
Famprofazone (also metabolised to metamfetamine)
2965
—
—
—
286
229
91
81
287
77 229
2410
2850
—
—
244
273
44
302
214
Fenbufen
3078
—
—
—
181
152
153
254
182
151
Fenbufen-Me
2315
—
1.79
1975
181
152
153
182
268
237
2190
M (OH-)
—
—
—
197
256
154
128
152
198
Fenclofenac-Me
—
—
1.55 and — 1.26
—
—
—
—
—
—
Fenoprofen
2016
2040
—
—
197
241
198
77
242
104
Fenoprofen-Me
1906
—
1.31
1970
197
256
198
257
91
103
—
—
—
2130
286
227
287
123
91
152
Feprazone
2380
—
—
—
183
77
252
320
184
41
Feprazone-Me
—
—
1.81
Floctafenine
3132
—
—
—
—
—
—
—
—
M (acetic acid OH-)-Me2
M (OH)-Me2
—
—
Floctafenine-Me
2433
—
—
—
—
—
—
—
—
Flufenamic acid
1950
—
1.26
—
263
281
166
92
145
167
Flufenamic acid-Me
—
—
—
1875
263
295
235
166
264
92
—
—
—
2115
325
293
278
250
223
202 —
M (OH-)-Me2 Flunixin-Me
—
—
1.39
—
—
—
—
—
—
Flupirtene
2603
—
—
—
109
304
231
124
258
110
Flurbiprofen
1900
—
—
—
199
244
200
178
179
184
Flurbiprofen-Me
1885
—
1.3
1880
199
178
183
258
170
200
M (OH)-Me2
2180
—
—
2180
229
288
230
289
214
183
2310
—
—
—
318
259
319
260
215
303
2770
—
—
—
—
—
—
—
—
—
Ibuprofen
1615
1637
—
—
163
161
119
91
206
117
Ibuprofen-ME
1510
—
0.89
1505
161
177
220
119
91
117
—
2096
—
—
177
117
119
91
118
221
M (OH-methoxy-)-Me2 Glafenine-ME
M (2OH-) M (2OH-)-Me
1750
—
—
—
177
117
159
145
131
236
M (3OH-)-Me
1630
—
—
1680
119
118
91
59
178
162
1765
—
—
—
205
145
177
117
121
264
Indometacin
2550
—
—
—
139
141
357
111
359
140
Indometacin-Me
2770
—
1.55 and 2770 0.49
139
141
111
371
312
114
M (HOOC-)-Me2
—
—
—
139
141
140
401
111
262
Indoprofen-Me
2708
—
2.27 and — 2.07
236
295
237
296
218
206
Isopropylaminophenazone
2033
—
—
—
—
—
—
—
—
M (OH)-Me2
2880
—
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:26:12
Specific applications
667
Table 40.9 continued Compound
System
Principal ions (m/z)
GA
GB
GD
GL
—
—
—
2510
77
336
105
183
266
117
—
—
—
2690
77
107
366
367
213
296
Ketoprofen
2245
—
—
—
105
177
77
209
254
210
Ketoprofen-Me
2090
—
1.45
2090
209
105
77
268
191
210
—
—
—
2250
239
298
135
191
107
103
—
2686
—
—
105
210
77
44
132
254 328
Kebuzone-Me M (OH)-Me2
M (OH)-Me2 Ketorolac
—
—
—
2685
267
326
77
232
269
—
—
—
2875
356
297
262
358
299
247
Meclofenamic acid
2420
—
—
—
—
—
—
—
—
—
Meclofenamic acid-Me
—
—
1.62
2240
242
244
309
311
277
214
Mefenamic acid
2201
2370
—
—
223
241
208
222
194
180
Mefenamic acid-Me
2069
—
1.45
2110
223
255
208
180
194
222
—
—
—
2400
209
271
221
224
180
194
Methyl salicylate (metabolised to salicylic acid 1195 and salicylamide)
1228
—
1210
92
120
152
65
121
93
Methylsalicylate-Me
1200
—
—
—
135
133
77
92
166
104
Mofebutazone-Me2
—
—
—
1955
121
77
204
260
83
105
—
—
—
2075
77
276
121
71
128
220
Morazone (metabolised to phenazone and phenmetrazine)
3130
—
—
—
201
56
176
202
70
258
Nabumetone
—
2084
—
—
171
228
172
185
128
115
M (6-methoxy-2-naphthyl acetic acid, 6-MNA)
—
2322
—
—
171
215
128
172
44
102
M (O-desmethyl-)
Lonazolac-Me M (OH)-Me2
M (OH)-Me
M (OH)-Me2
—
2385
—
—
157
201
128
158
127
44
Naproxen
2045
2337
—
—
185
230
141
186
184
115
Naproxen-Me
1980
—
1.37 and 2120 1.18
185
244
170
141
115
186
M (ET)
1830
2115
—
—
185
258
186
170
153
141
M (O-desmethyl-)
—
2396
—
—
171
215
115
141
153
130
M (O-desmethyl-)-Me2
1980
—
—
2120
185
244
170
141
115
186
M (OH)-Me2 Nifenazone
1800
—
—
1800
274
215
259
171
184
275
3080
—
—
—
—
—
—
—
—
—
1955
—
—
—
—
—
—
—
—
—
Niflumic acid
2085
—
1.35
—
282
236
237
281
263
145
Niflumic acid-Me
1955
—
—
1960
236
295
296
263
237
145
—
—
—
2140
326
325
251
293
294
266
Not eluted
Not eluted —
—
199
324
93
77
65
55
Art (phenyldiazophenol)
2070
—
—
—
93
77
65
198
121
51
Art (phenyldiazophenol)-Me
2020
—
—
—
77
107
212
135
64
51
Oxyphenbutazone-Me2 (isomer 1)
2545
—
2.11
—
352
213
77
107
118
135
Oxyphenbutazone-Me2 (isomer 2)
2720
—
—
—
352
77
160
190
309
278
Paracetamol (acetaminophen)
1665
1722
—
—
109
51
43
80
108
81
1253
1280
—
—
109
52
53
80
81
108 122
M (desacyl-)
M (OH-) Me2 Oxyphenbutazone
Art (p-aminophenol) Paracetamol-Me Art (p-aminophenol)-Me2
1512
—
—
1630
108
123
165
80
95
1220
—
—
—
136
137
121
120
94
65
1730
—
—
108
109
179
137
43
81
Phenacetin (also metabolised to paracetamol) 1675 Phenacetin-Me M (hydroquinone)
—
—
—
—
122
193
151
56
123
94
1240
—
—
—
110
81
55
53
82
39
M (p-phenetidine)
1275
—
—
—
108
137
109
80
53
65
Phenazone/antipyrine
1835
1951
—
—
188
96
77
56
105
189
M (4-OH) Phenazopyridine (also metabolised to paracetamol) M (aniline)
1855
—
—
—
2245
2370
—
—
1158
—
—
—
85 — 93
56 — 66
84 — 65
204 — 92
77 — 46
120 — 41
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:26:13
668
Gas Chromatography
Table 40.9 continued Compound
Phenylbutazone (also metabolised to oxyphenbutazone) Art Phenylbutazone-Me
System
Principal ions (m/z)
GA
GB
GD
2367
2472
2.05 and — 1.81
GL —
2435
2550
—
—
183
2290
—
—
2290
183
—
—
—
—
—
77
184
324
325
119
77
322
266
118
323
266
184
—
—
—
—
—
2500
183
338
162
Phenyramidol
1960
—
—
—
—
—
—
Piroxicam
1413
—
—
—
173
117
145
78
104
94
Propyphenazone
1920
2030
—
—
215
230
56
77
216
96
M (OH-alkyl)-Me
77 —
M (nor-)
1772
—
—
—
174
216
77
173
129
145
M (OH-methyl-)
2410
—
—
—
231
246
232
77
215
154
M (OH-phenyl-)
2300
—
—
—
231
56
246
96
122
217
M (OH-propyl-)
2210
—
—
—
215
56
246
124
77
231
M (isopropanolyl-)
2020
—
—
—
231
246
213
56
232
61
M (isopropenyl-)
1970
—
—
—
136
228
95
77
108
106
M (nor-OH-)
1780
—
—
—
93
232
77
190
120
174 161
M (nor-OH-phenyl-)
2080
—
—
—
190
232
93
121
65
M (nor-di-OH-)
2090
—
—
—
248
109
136
206
121
232
M (nor)-Me
1735
—
—
—
215
77
230
51
200
185
M (OH-phenyl)-Me
2310
—
—
—
56
246
260
122
96
77
M (COOH)-Me
2160
—
—
—
215
274
56
77
105
165
M (nor-OH-phenyl isomer 1)-Me2
2030
—
—
—
245
230
260
215
77
92
M (nor-OH-phenyl isomer 2)-Me2
2060
—
—
—
245
230
260
77
92
215
M (nor-di-OH)-Me3
2240
—
—
—
275
290
260
252
236
276
—
3119
—
—
257
314
178
131
176
165
Rofecoxib Salicylamide
1414
1489
—
—
120
92
137
65
121
64
Salicylic acid
1307
1340
—
—
120
92
138
64
63
121
120
43
138
92
121
64
92
120
152
65
121
93
Salicylic acid-AC
1545
—
—
—
Salicylic acid-Me
1195
1228
—
1210
Salicylic acid-Me2
1200
—
—
—
135
133
77
92
166
104
—
—
—
1530
196
165
163
181
107
151 149
M (5-OH)-Me3 M (glycine conj)
1825
—
—
—
120
121
92
65
195
M (glycine conj)-Me
1810
—
—
—
121
120
209
119
65
92
M (glycine conj)-Me2
1845
—
—
—
135
90
77
105
223
121
Salsalate (metabolised to salicylic acid)
—
Not eluted —
—
121
120
92
65
138
258
Sulindac
2890
—
0.49
—
341
233
356
246
247
281
Sulindac-Me
3220
—
—
—
233
354
355
370
248
247
M (sulfide-)
2896
2959
—
—
328
233
234
313
159
247
M (sulfone)
—
3029
—
—
328
233
234
329
220
246
—
2715
—
—
58
143
142
115
156
295
Tenoxicam-Me2
2690
—
—
—
—
—
—
—
—
—
Tiaprofenic acid
1976
—
—
—
216
139
201
77
173
105
Tiaprofenic acid-Me2
2180
—
—
2175
229
288
230
77
105
201
Tolfenamic acid
—
—
—
—
—
—
—
—
—
—
Tolfenamic acid-Me
—
—
—
2255
208
243
275
180
89
Tolmetin
1890
—
—
—
212
213
122
198
44
91
Tolmetin-Me
2247
—
1.77 and 2235 1.36
212
271
256
119
270
91
M (COOH)
—
2615
—
—
212
91
256
119
44
65
Sumatriptan
245
—
—
—
2600
256
315
242
197
135
314
Zomepirac-Me
2343
—
—
—
246
305
248
304
139
111
M (-CO2)
2025
—
—
—
246
247
248
211
230
136
M (COOH)-Me2
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:26:16
Specific applications relatively unstable and produce several artefacts, either from hydrolysis during extraction or from thermal degradation in the GC. The quaternary ammonium compounds (e.g. emepromium bromide) are not amenable to gas chromatography. Systems GA or GB, previously described, may be used. Retention indices and relative retention times are given in Table 40.10.
Anticonvulsants and barbiturates Most barbiturates and anticonvulsants are acidic or mildly basic drugs and are extracted readily from aqueous medium into organic solvents. Although phenobarbital requires pH 7 or less for good recovery in liquid–liquid extractions, the other barbiturates and anticonvulsants (such as carbamazepine and phenytoin) may be extracted from aqueous
Table 40.10 GC retention data and mass spectral data for anticholinergics (AC, acetyl) System GA
Principal ions (m/z) GB
Adiphenine
2200
—
86
167
99
87
58
Atropine
2190
2293
124
82
94
83
42
96
Art (-CH2O)
1980
2051
124
259
140
94
221
178
Art (-H2O)
165
2085
2250
124
271
96
82
140
94
Benzatropine (benztropine)
2302
2423
83
140
82
124
96
42
Biperiden
2276
—
98
218
99
55
41
77
2645
—
98
218
114
327
284
85
Caramiphen
—
—
86
99
91
144
58
56
Chlorphenoxamine
2080
2190
58
59
179
42
178
72
2470
—
58
152
165
181
195
231
M (OH-)
M (OH-) M (nor-)
2094
2205
—
—
—
—
—
—
M (OH-methoxy-carbinol)-H2O
2220
—
260
262
210
245
227
181
Cyclopentolate
2022
2092
58
71
72
207
42
91
Cyclopentolate-H2O
2000
1551
58
71
91
115
129
273
Cycrimine
2114
—
98
41
42
55
99
77
Dicycloverine (dicyclomine)
2111
2175
86
71
99
58
55
56
Diethazine
2377
—
86
298
87
58
299
212
Eucatropine
2026
—
124
276
58
140
56
72
Homatropine
2072
2165
124
107
82
83
42
77
Hyoscine (scopolamine)
2300
2427
94
138
42
108
136
41
Art (H2O)
2230
2255
94
103
138
154
108
285
M (desacyl-)
1210
—
96
94
155
126
110
70
Metixine (methixine)
2480
2596
99
197
44
58
112
309
Orphenadrine
Hyoscyamine (see Atropine) 1935
2014
58
73
44
45
165
181
M (nor-, tofenacin)
1900
2007
180
179
86
255
165
240
M (methylbenzophenone)
1700
1827
195
196
77
105
119
165
M
1560
1630
167
182
107
108
165
119
1661 and 2250
—
105
129
112
77
42
313
111
96
167
112
165
71
84
204
205
85
42
—
—
Oxyphencyclimine Piperidolate
2318
—
Procyclidine
2156
2261
—
2487
M (OH-isomer 1)
—
—
—
55 —
M (OH-Isomer 2)
—
2517
—
—
—
—
—
—
M
—
2548
—
—
—
—
—
—
M
—
2603
—
—
—
—
—
—
M (oxo-) art-H 2O
2490
2669
200
115
86
98
198
283
Tigloidine
1687
—
124
82
83
94
55
42
Trihexyphenidyl (benzhexol)
2245
2354
98
105
55
99
77
218
2500
2618
Scopolamine (see Hyoscine)
M (OH-) Tropicamide Art (-CH2O)
—
669
—
—
—
—
—
2335
2442
92
91
65
103
93
163
2230
—
92
91
163
65
254
107
Art (-H2O)
2250
—
103
266
92
251
77
265
Tropicamide-AC
2410
—
92
104
266
65
163
326
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:26:53
670
Gas Chromatography
solutions at pH values as high as 11. There is little to be gained in terms of sensitivity by using NPD over FID, especially for barbiturates, but the former is more specific and excludes fatty acids, which can be problematic in decomposing samples. For GC, some investigators prefer to methylate, either during the extraction (iodomethane–tetramethylammonium hydroxide in dimethyl sulfoxide) (Liu et al. 1994), or by flash methylation in the injection port (trimethylphenylammonium hydroxide in ethyl acetate) (Brugmann 1981). This method is sometimes considered unreliable for quantitative analysis, and is arguably only necessary when using packed chromatography columns. Data for the methyl derivatives are therefore given where they are available. When using packed columns for GC, the medium- and higher-polarity phases, such as *-1310, *-2100, DEGS and waxes, are useful, although methylation allows the use of some more non-polar phases such as *-1 (Stern, Caron 1977). Systems GA and GB, previously described, may be used, or systems GE, GF and GAJ. System GE
System GF
(Flanagan, Berry 1977). n n n n
System GAJ
Data generated by the author. n n n n n n
Quantitative analysis of underivatised antiepileptic drugs (Supelco 1979). n n n n n
Column: 2% SP-2110 and 1% SP-2510-DA on 100–120 mesh Supelcoport glass (1 m 2 mm i.d.). Temperature programme: 120 C to 250 C at 16 /min. Carrier gas: N2, 50 mL/min. Reference compound: Phenytoin. Note: This system separates cholesterol from all drugs in the group. Retention indices and relative retention times: The values given for system GE are retention times relative to phenytoin.
Column: 3% Poly A103 on 80–100 mesh Chromosorb W HP glass (1 m 4 mm i.d.). Temperature: 200 C. Carrier gas: N2, 60 mL/min. Reference compounds: n-Alkanes with an even number of carbon atoms.
Column: DB1301 capillary (25 m 0.32 mm i.d., 0.25 mm). Temperature programme: 100 C to 235 C at 35 /min for 3.6 min to 290 C at 8 /min for 3.5 min. Carrier gas: He, 1.5 mL/min. Reference compound: Methylphenobarbital. Detection by FID and NPD split. Retention indices and relative retention times are given in Table 40.11. The values given for system GAJ are retention times relative to methylphenobarbital.
Antidepressants Antidepressants (tricyclics, selective serotonin reuptake inhibitors (SSRIs), monoamine oxidase inhibitors (MAOIs)) can be extracted readily under mildly basic conditions (pH 10) into many solvents, such as ethyl acetate, hexane, diethyl ether. Less polar solvents, such as hexane, limit the extraction of hydroxylated metabolites. An acidified (0.05 mol/L H2SO4) back extraction is a useful clean-up procedure
Table 40.11 GC retention data and mass spectral data for anticonvulsants and barbiturates (Me, methyl; ET, ethyl) Compound
Allobarbital
System
Principal ions (m/z)
GA
GB
GE
GF
GAJ
1600
1636
—
2340
—
41
167
124
80
53
68 236
Me2
1505
—
—
—
—
195
138
194
110
221
M
1785
—
—
—
—
—
—
—
—
—
1710
1742
—
2430
0.794
156
141
157
41
55
142 239
Amobarbital (amylobarbital)
—
Aobarbital-Me2
1593
—
—
—
—
184
169
170
185
226
M (3OH-)
1915
2015
—
—
1.138
156
157
141
227
214
195
M (3OH)-Me2
1750
—
—
—
—
137
184
169
185
255
270
M (COOH-)
1960
—
—
—
0.775
156
141
157
183
212
155
M (COOH)-Me3
1850
—
—
—
—
169
184
137
185
240
211
Aprobarbital
1618
—
—
—
—
167
41
124
168
97
39
M (OH-)
1815
—
—
—
—
183
154
184
122
165
226
Aprobarbital-Me2
1540
—
—
—
—
195
196
138
181
111
220
Barbital
1489
—
—
2230
0.612
156
141
55
155
98
82
Barbital-Me2
1420
—
—
—
—
184
169
126
112
183
83
Barbituric acid
0000
—
—
—
—
42
128
85
44
70
69
Barbituric acid-Me3
1645
—
—
—
—
170
55
82
98
113
155
Beclamide
1720
1778
—
—
—
91
106
197
162
107
148
1680
1539
—
—
—
55
161
106
116
79
77
1853
—
—
2765
1.000
207
41
39
124
91
165
Art Brallobarbital Brallobarbital-Me2 M (2OH-) M (desbromo-OH-) Butalbital Butalbital-Me2 M (OHButobarbital (butobarbitone)
1725
—
—
—
—
235
193
136
194
236
121
2040
—
—
—
—
223
165
180
136
152
122
167
124
141
98
181
224
41
167
168
124
97
141
1795
—
—
—
—
1665
1698
—
2395
0.778
1655
—
—
—
—
196
195
138
209
169
237
1940
2016
—
—
—
168
167
153
141
222
240
1660
—
—
2390
0.732
141
156
41
55
98
142
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:27:6
Specific applications
671
Table 40.11 continued Compound
Butobarbital-Me2 M (3 0 -OH-)
System
Principal ions (m/z)
GA
GB
GE
GF
1565
—
—
—
GAJ —
169
184
112
170
183
211
1920
—
—
—
1.053
156
141
157
199
181
213
M (3 0 -oxo-)
1880
—
—
—
—
156
141
198
157
199
211
Carbamazepine
2285
2435
0.83
—
1.716
193
192
236
191
194
165
—
—
1905
—
—
—
—
—
—
—
—
M (10,11-epoxide)
2220
—
—
—
1.188
180
179
178
152
44
181
M (iminostilbene)
1998
2064
—
—
0.732
193
192
194
165
179
191
Carbamazepine-Me
M (acridine)
1800
1880
—
—
0.591
179
178
151
152
180
177
M (formylacridine)
2025
2158
—
—
0.813
179
207
178
180
151
152
M (methylacridine)
—
2054
—
—
0.699
193
192
165
191
194
167
M
—
2316
—
—
—
180
209
181
210
152
190
M
—
2332
—
—
—
283
180
208
284
192
266
M
—
2387
—
—
—
209
180
208
210
152
167
M
—
2402
—
—
—
180
210
227
209
181
208
M (10,11-di-OH-)
—
2738
—
—
—
180
208
196
270
253
167
Cyclobarbital
1955
—
—
2825
1.142
207
141
81
79
67
80
Cyclobarbital-Me2
1845
—
—
—
—
235
169
236
178
121
264
M (oxo-)
2190
—
—
—
—
221
193
260
179
222
178
M (oxo)-Me2
2050
—
—
—
—
249
221
250
164
192
278
Cyclopentobarbital
1865
—
—
—
—
67
193
66
41
169
39
Cyclopentobarbital-Me2
1775
—
—
—
—
221
196
164
181
111
107
Dimethadione
1060
—
—
—
—
43
59
42
41
58
129
Enallylpropymal-Me2
1520
—
—
—
—
181
41
182
124
53
138
Clobazam (see Benzodiazepine section) Clonazepam (see Benzodiazepine section)
Ethosuximide
1205
1258
0.18
—
0.453
113
70
55
42
41
39
Ethosuximide-Me
1130
—
—
—
—
55
127
70
112
140
155
1370
1436
—
—
—
113
85
98
69
71
142 157
M (OH-ET-) M (3OH-)
1322
1395
—
—
—
71
86
129
139
142
M (oxo-)
1270
—
—
—
—
70
155
55
113
69
98
1800
1751
0.57
—
0.940
104
105
204
77
78
133
Ethotoin
1450
1475
—
—
—
104
103
91
77
121
134
Art (-C2H3NO2)
1890
1854
—
—
—
104
103
91
77
121
134
Art (-CH3NO2)
2210
2212
—
—
—
134
104
103
91
77
177
1750
1633
—
—
—
81
153
152
87
110
96
1560
—
—
—
—
81
67
167
166
124
110
Heptabarb
2055
2110
—
2940
1.282
221
43
78
93
80
141
Heptabarb-Me2
1915
—
—
—
—
249
169
250
133
183
192
2275
—
—
—
—
219
93
141
115
237
157
—
—
—
—
Felbamate(H2O)
Fosphenytoin (see Phenytoin) Gabapentin Art (-H2O) Art (-H2O)-Me
M (OH-) M (3 0 -oxo-)
2320
—
—
—
—
—
—
Hexethal
1850
—
—
—
—
156
141
55
41
157
98
Hexethal-Me2
1745
—
—
—
—
169
184
112
185
170
209
Hexobarbital
1855
—
—
2380
0.940
221
81
157
80
79
155
Hexobarbital-Me
1800
—
—
—
—
235
81
169
171
170
236
M (3 0 -oxo-)
2055
—
—
—
—
95
235
250
156
193
123
M (nor-)
1980
—
—
—
—
81
143
207
123
139
222
M (oxo)-ME
2020
—
—
—
—
249
264
95
221
207
170
M (3 0 -OH-)
1970
—
—
—
—
156
79
219
234
233
191 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:27:12
672
Gas Chromatography
Table 40.11 continued Compound
Ibomal (propallylonal; also metabolised to aprobarbital)
System
Principal ions (m/z)
GA
GB
GE
GF
GAJ
1880
—
—
—
—
167
209
43
124
39
41
1745
—
—
—
—
237
195
138
110
238
196
M (desbromo-OH-)
1770
—
—
—
—
169
141
142
98
184
226
M (desbromo-oxo)-Me2
1720
—
—
—
—
169
197
112
212
140
170
M (desbromo-OH)-Me2
1730
—
—
—
—
169
183
170
112
198
214
1700
—
—
—
—
167
41
168
124
97
141 223
Ibomal-Me2
Idobutal Isobutal-Me2
1610
—
—
—
—
195
196
138
181
169
Lamotrigine
2635
2562
—
—
1.941
185
187
255
257
123
124
Levetiracetam
—
1629
—
—
—
126
41
69
98
44
127
Metharbital (metabolised to barbital)
1470
—
—
—
—
155
170
112
169
55
82
Methohexital
1770
1827
—
—
0.798
41
81
53
221
79
39
Methohexital-Me
1735
1797
—
—
—
235
178
195
247
261
275
M (4 0 -OH-)
1880
—
—
—
—
219
124
181
245
261
278
1785
—
0.55
—
0.918
189
104
190
77
44
105
Methoin (mephenytoin) M (p-OH-)
2400
—
—
—
—
205
120
109
152
176
234
M (OH-methoxy-)
2380
—
—
—
—
235
150
135
137
247
264
M (nor-)
1950
—
—
—
—
104
175
77
132
163
204
1705
—
0.35
—
0.689
181
117
203
103
77
78
1750
—
—
—
0.779
118
117
103
77
189
155
Mesuximide (methsuximide) M (nor-) M (OH-)
2220
—
—
—
—
134
219
119
107
91
204
M (nor-OH-)
2300
—
—
—
—
134
205
119
133
103
165
Methylphenobarbital
1890
2222
—
—
1.000
218
117
118
146
103
77
Methylphenobarbital-Me
1855
—
—
—
—
232
118
117
146
175
260
M (OH-, MHD)
2370
—
—
—
—
134
233
234
262
162
133
M (OH-methoxy-)
2310
—
—
—
—
231
292
263
164
188
174
Nealbarbital
1720
—
—
2460
0.789
57
41
141
167
39
83
Nealbarbital-Me2
1620
—
—
—
—
169
195
209
112
138
250
Oxcarbazepine
—
2266
—
—
—
209
180
208
153
181
210
M (formylacridine)
2025
2158
—
—
—
179
207
178
180
151
152
M (methylacridine)
—
2054
—
—
—
193
192
165
191
194
167
M
—
2204
—
—
—
211
180
194
182
167
152
M
—
2296
—
—
—
193
192
180
165
191
237
M (10-OH-, MHD)
—
2580
—
—
—
193
180
194
210
254
167
M (carbamazepine)
2285
2435
—
—
1.716
193
192
236
191
194
165
Paramethadione
1115
—
0.06
—
—
43
129
57
56
41
72
Pentobarbital
1735
1776
—
2465
0.803
141
156
43
41
157
55
Pentobarbital-Me2
1630
—
—
—
—
184
169
112
225
185
126
M (3 -OH-)
1955
2039
—
—
—
156
141
157
197
195
227
M (3 0 -OH)-Me2
1820
—
—
—
—
169
184
185
223
225
241
Phenacemide
1473
—
—
—
—
91
92
118
44
43
135
Pheneturide
1465
—
—
—
—
91
146
44
119
206
41
Phenobarbital (phenobarbitone)
1953
2031
0.74
2960
1.150
204
117
146
161
77
103
0
Phenobarbital-Me2 M (4-OH-)
1855
—
—
—
—
232
118
117
146
175
260
2295
2378
—
—
—
219
248
148
220
176
204
2200
—
—
—
—
290
261
148
233
262
176
Phensuximide
1634
—
0.39
—
—
104
189
103
78
51
77
Phenylmethylbarbituric acid (heptobarbital)
1880
—
—
—
1.087
104
132
218
51
103
77
M (4-OH)-Me3
Phenylmethylbarbituric acid-Me2
1790
—
—
—
—
132
104
246
103
79
189
Phenytoin
2320
2435
1.00
—
1.773
180
104
223
77
209
252
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:27:15
Specific applications
673
Table 40.11 continued Compound
System
Principal ions (m/z)
GA
GB
GE
GF
GAJ
2795
2910
—
—
—
268
239
196
225
120
180
2245
—
—
—
—
180
266
237
209
189
165
M (p-OH)-Me2
2720
—
—
—
—
296
267
219
210
180
134
M (p-OH-methoxy-)
2770
—
—
—
—
298
269
226
196
211
254
M (OH-) Phenytoin-Me
2740
—
—
—
—
326
397
249
282
210
196
Primidone (also metabolised to phenobarbital)
2250
2384
0.89
—
1.674
146
190
117
118
161
189
Primidone-Me2
246
M (p-OH-methoxy)-Me2
—
2161
—
—
—
146
218
117
118
217
M (phenylethylmalondiamide)
1884
1996
—
—
1.074
148
163
103
120
91
117
M (diamide)
1935
—
—
—
—
163
148
103
118
120
133
M (AC) Secbutabarbital
2115
2189
—
—
—
146
232
117
118
189
218
1655
—
—
—
—
141
156
41
57
39
98
1565
—
—
—
—
—
—
—
—
—
—
1926
—
—
—
—
—
—
—
—
—
—
Secobarbital (quinalbarbitone)
1786
1827
—
—
0.865
167
168
41
43
97
Secobarbital-Me2
1690
—
—
—
—
196
195
138
181
224
237
M (3 0 -OH-)
1865
2029
—
—
1.206
168
167
169
153
209
195
M (3 0 -keto)
—
—
—
—
—
43
168
69
85
167
124
M (desallyl-)
1665
—
—
—
—
129
128
85
86
154
169
M (2,3 0 -diOH-)
—
—
—
—
—
171
143
128
159
198
241
—
—
—
—
—
28
99
56
27
26
55
Secbutabarbital-Me2 M (2 0 -OH-)
Succinimide
124
Sulthiame-Me
2880
—
—
—
—
304
274
226
198
210
211
Sulthiame-Me2
2815
—
—
—
—
318
274
226
210
211
104 168
Sultiame
3000
—
—
—
—
290
184
185
104
77
Talbutal
1703
—
—
—
—
167
168
41
97
124
39
Talbutal-Me2
1600
—
—
—
—
195
196
138
181
111
211
Thialbarbital
2116
—
—
—
—
81
223
79
80
157
185
Thiamylal
1899
—
—
—
—
43
41
184
168
167
97
Thiopental (thiopentone; also metabolised to pentobarbital)
1857
1923
—
2600
0.948
172
157
173
43
41
55
Thiopental-Me2 M (OH-)
1825
—
—
—
—
200
185
201
127
157
167
—
2134
—
—
—
172
173
157
97
258
229
156
157
113
111
358
96
—
2253
—
—
—
324
43
80
110
189
206
Taigabine-Me Topiramate Art (fructopyranose) Troxidone (trimethadione; metabolised to dimethadione) Valproate (valproic acid) M
—
1621
—
—
—
43
245
69
59
127
85
1090
—
0.04
—
—
43
58
143
42
41
128
1064
1098
0.09
—
0.350
73
102
41
57
43
55
—
1195
—
—
—
100
55
41
69
127
113 113
M
1200
—
—
—
—
100
55
41
69
127
M
—
1267
—
—
—
72
101
114
100
55
44
M
—
1312
—
—
—
100
55
41
113
99
69
Vigabatrin
—
Not eluted—
—
Not eluted 56
84
111
69
82
54
Vinbarbital
1753
—
2495
—
41
141
69
152
135
—
195
1670
—
—
—
—
223
224
166
169
138
135
2070
—
—
—
—
167
169
85
211
193
155
Vinylbital
1729
—
—
—
0.798
154
83
71
55
155
67
Vinylbital-Me2
1655
—
—
—
—
182
181
183
97
125
154
83
Vinabarbital-Me2 M (OH-)
M (3 0 -OH-)
1995
—
—
—
—
154
155
M (desvinyl-)
1665
—
—
—
—
—
—
—
2042
—
—
—
132
Zonisamide
77
112
139
195
—
—
—
—
133
104
51
64
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:27:37
674
Gas Chromatography
where sensitivity is important. Chromatography of primary and secondary amines is poor on packed columns, but is adequate on wellmaintained capillary columns, particularly those of low–medium polarity such as PSX-5 (see Table 40.3). Some authors prefer to chromatograph the secondary amines and hydroxylated metabolites as acetylated derivatives, prepared by heating the dried residue with acetic anhydride and pyridine (3 : 2, v/v) (Maurer, Bickeboeller-Friedrich 2000). Others employ an enzymatic hydrolysis procedure to improve recovery of both parent drug and metabolites, although the additional sensitivity gained is often negated by the increased analytical time in the emergency setting. Acid hydrolysis is quicker, but some relevant compounds are destroyed under these conditions. System GA, GF or GB, described above, may be used, or system GM. System GM
System GM (Dawling et al. 1990) is ideal for plasma samples, since the isothermal conditions allow high throughput and the limited resolution of hydroxylated metabolites is not important as these do not constitute a significant fraction of the extract from plasma. Conditions are given for both packed and capillary column systems.
n n n n n n n n n
Column: 3% SP2250 on Supelcoport 80–100 mesh glass (2.1 m 2 mm i.d.). Temperature: 265 C. Carrier gas: He, 25 mL/min. Column: HP-50 þ fused silica capillary (25 m 0.53 mm i.d., 1 mm). Temperature: 250 C. Carrier gas: He, 7 mL/min. Reference compound: Iprindole. Quantification: NPD. Retention indices, relative retention to iprindole, are given in Table 40.12. Only the metabolites known to occur in urine and/ or plasma specimens are included in this list; the list in De Zeeuw (2002) is more extensive.
Antihistamines Antihistamines are a diverse group of drugs that includes the ethanolamines (diphenhydramine), ethylenediamines (pyrilamine), alkylamines (hydroxyzine), phenothiazines (promethazine) and piperidines (chlorphenamine). Many share common metabolites with other members of
Table 40.12 GC retention data and mass spectral data for antidepressants (AC, acetyl) Compound
System
Principal ions (m/z)
GA
GB
GF
GM
Amitriptyline (also metabolised to nortriptyline)
2194
2284
2510
0.723
M (cis-10-OH-)
2348
2454
—
1.149
M (cis-10-OH-N-oxide)
—
2215
—
—
M (trans-10-OH-)
2348
2466
—
1.168
M (trans-10-OH-N-oxide)
—
2239
—
—
M (cyclobenzaprine)
2235
2330
—
0.850
58
59
202
42
203
214
58
202
215
178
189
165
215
229
230
207
248
178
58
202
215
178
189
165
215
229
230
207
248
178
58
215
202
189
176
163
Amitriptyline N-oxide
1975
2051
—
—
232
217
215
202
117
189
Amoxapine
2638
2746
—
2.831
245
257
247
193
56
246
M (7-OH-)
2951
3525
—
—
261
209
273
263
244
329
M (8-OH-)
2959
3546
—
—
261
209
273
263
244
329
Atomoxetine Bupropion M
—
1645
—
—
44
100
111
139
224
57
—
1746
—
—
44
100
77
57
208
113
M
—
1764
—
—
44
100
77
57
208
113
M (OH-)
—
1898
—
—
44
100
116
139
224
110
M
—
1916
—
—
44
100
224
157
57
M
—
2107
—
—
44
100
57
84
260
2181
2288
2465
0.683
58
293
—
2330
—
0.761
Butriptyline M (nor-)
—
—
45 —
59 —
65 —
193
100
—
—
Citalopram
2525
2499
—
1.121
58
238
208
42
324
190
M (nor-)
2500
2526
—
1.232
44
238
208
138
310
190
M
—
2846
—
—
238
81
136
192
265
221
M
—
2987
—
—
238
207
163
254
265
282
Clomipramine
2415
2511
2795
1.172
58
85
269
268
270
271
M (nor-)
2432
2540
—
1.374
268
269
229
227
242
300
M (N-oxide)
2146
2246
—
—
228
193
192
269
230
165
M (ring)
2230
2335
—
—
229
194
193
214
228
231
M (8-OH-)
2727
2843
—
—
58
285
243
209
284
330
M (8-OH-nor-)
2762
2880
—
—
M (2OH-)
2569
2735
—
—
M (10OH-)
2574
2698
—
—
44 — 58
245
243
284
258
316
—
—
—
—
—
268
329
313
85
86
Clorgiline
1883
—
—
—
—
—
—
Desipramine
2235
2338
—
0.896
235
195
208
— 44
—
—
234
193
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:27:52
Specific applications
675
Table 40.12 continued Compound
System
Principal ions (m/z)
GA
GB
GF
M (2-OH-)
2553
2669
—
GM —
44
211
224
250
180
282
M (10-OH-)
—
2521
—
—
44
180
194
206
251
282
M (ring)
1930
2014
—
—
195
194
180
167
97
89
M (ring-OH-)
2240
2335
—
—
211
210
196
180
167
212
M (ring di-OH-)
2600
—
—
—
227
226
157
196
228
183
M (di-OH-)
—
2995
—
—
44
266
240
227
298
225 222
M (AC-)
2670
2811
—
—
208
114
308
193
194
M (OH-methoxy-)
—
2749
—
—
227
241
254
280
312
44
M (ring OH-methoxy-)
2390
—
—
—
241
240
226
180
210
198
Dibenzepin
2450
2566
2885
1.735
M (nor-)
2449
—
—
—
58 —
324
209
—
—
71 —
225 —
72 —
M (di-nor-)
2406
—
—
—
235
234
207
206
179
192
M (ter-nor-)
2680
—
—
—
235
234
206
207
179
192
M (N5-desmethyl-)
2455
—
—
—
Dosulepin (dothiepin)
2380
2486
2770
1.259
58 —
210
211
167
195
223
—
—
—
—
—
M (nor-)
2421
2507
—
1.450
204
281
221
263
238
165
M (N-oxide)
2100
—
—
—
217
235
250
202
221
240
M (OH-N-oxide)
2130
—
—
—
266
165
251
233
237
215
M (sulfoxide)
2392
2533
—
—
—
—
—
—
—
—
—
—
—
—
—
—
220
219
191
189
M (norsulfoxide)
2421
2839
—
—
Doxepin cis-isomer
2220
2301
2570(a)
0.788
Doxepin trans-isomer
58
59
2220
2321
—
0.823
58
220
219
59
191
189
M (cis-N-oxide)
1970
2077
—
—
234
219
165
178
202
189
M (trans-N-oxide)
—
2081
—
—
234
219
165
178
202
189
M (cis-nor-)
2245
2333
—
0.830
44
165
178
189
202
219
M (trans-nor-)
2245
2339
—
0.933
44
165
178
189
202
219
M (cis-OH-)
2535
2528
—
—
58
165
295
152
178
220
M (trans-OH-)
2560
2544
—
—
58
165
295
152
178
220
M (cis-nor-OH-)
2540
2644
—
—
44
220
238
165
152
281
M (trans-nor-OH-)
—
2671
—
—
44
220
238
165
152
281
—
2750
—
—
44
297
265
240
181
115
Duloxetine Escitalopram (see Citalopram) Fluoxetine
1859
1903
—
0.304
44
309
183
104
251
91
M (nor-)
1851
1888
—
0.284
104
134
103
77
162
191 115
M (AC-)
2250
2319
—
—
44
86
190
117
104
M (nor-AC-)
2190
2278
—
—
117
176
72
104
115
91
1885
1911
—
0.295
187
71
45
276
172
145
Fluvoxamine Art (ketone)
1525
—
—
—
173
228
145
159
188
241
Art
1560
1602
—
—
187
172
200
228
244
259
M (AC-)
2240
2284
—
—
86
102
187
258
341
360
Art
1895
1921
—
—
258
71
226
242
311
329
M
—
2200
—
—
71
145
172
198
226
258
M
—
1791
—
—
241
172
212
145
144
198
M
-
Imipramine (also metabolised to desipramine) 2230
1687
—
—
86
257
198
145
281
341
2314
2540
0.784
58
235
85
234
236
195 224
M (2-OH-)
2565
2636
—
—
58
250
251
211
296
M (10-OH-)
—
2494
—
—
58
193
180
232
251
296
M (di-OH-)
—
2962
—
—
58
266
267
227
312
252
—
2715
—
—
58
280
241
326
254
266
Iprindole
2335
2437
—
1.000
58
170
284
213
145
212
Iproniazid
1593
1609
—
—
123
58
106
79
43
78
M (OH-methoxy-)
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:28:1
676
Gas Chromatography
Table 40.12 continued Compound
System
Principal ions (m/z)
GA
GB
GF
GM
Isocarboxazid
1949
0000
—
—
Lofepramine (metabolised to desipramine)
Not elutedNot eluted—
Maprotiline
2390
2440
—
1.086
44
70
59
277
71
191
M (nor-)
2293
2404
—
1.107
56
202
203
178
263
189
M (desamino-di-OH-)
2570
2620
—
—
252
207
280
253
219
195
44
70
203
189
187
293
—
—
—
—
M (OH-)
—
—
—
—
—
Not eluted —
—
—
—
—
2622 1240
—
Mianserin
2210
2302
2595
0.879
193
264
43
72
71
M (nor-)
2235
2348
—
1.105
193
208
250
165
178
220
M (8-OH-)
2495
2628
—
—
280
209
72
236
265
180
M (OH-methoxy-) M (CM30488) Mirtazapine M (nor-)
—
—
—
—
Mebanazine
Minaprine
—
—
220
2530
—
—
—
310
239
266
295
224
72
2855
3023
—
—
100
113
186
56
198
77
3040
3222
—
—
—
—
—
—
—
—
2250
2361
—
—
195
194
208
180
167
265
—
2414
—
—
195
194
209
180
167
251 208
M (oxo-)
—
2665
—
—
195
250
279
180
194
Moclobemide
—
2333
—
0.967
100
56
113
139
111
—
2578
—
2.191
—
—
—
—
—
— 317
M Nefazodone
42
4510
Not eluted—
Not eluted 303
274
260
304
454
M (m-chlorophenylpiperazine, mCPP)
—
1806
—
—
154
196
138
111
156
75
M (mCPP)-AC
2265
—
—
—
166
238
138
154
168
195
M (N-desalkyl-OH-) isomer 2-AC2
2525
—
—
—
182
254
169
184
296
211
M (desamino-OH-)-
2340
120
198
291
127
171
140
M (desamino-OH-)-AC
2500
—
120
240
91
333
77
126 238
—
—
2650
—
—
—
120
298
391
91
101
Nialamide
1500
—
—
—
91
177
44
106
45
78
Nomifensine
2130
2239
2670
0.850
194
195
238
193
72
178
M (OH-ethyl-desamino-OH)-AC2
M (4-OH-)
2450
—
—
—
86
210
211
194
254
228
M (OH,MeO-) isomer 1
2505
—
—
—
284
86
241
210
209
224
M (OH,MeO-) isomer 2
2590
—
—
—
284
86
241
210
209
224 215
2215
2304
—
0.816
44
202
45
220
218
M (cis-10-OH-)
2375
2480
—
1.261
44
218
203
202
178
165
M (trans-10-OH-)
2375
2494
—
1.323
44
218
203
202
178
165
Nortriptyline
M (norcyclobenzaprine)
—
2343
—
0.880
44
215
218
202
189
163
M (AC-)
2660
2774
—
—
44
232
202
217
86
203 178
—
2949
—
—
44
230
215
202
86
2270
—
—
—
58
71
208
72
59
42
M (dibenzocycloheptanone)
1850
—
—
—
208
180
179
178
165
152
M (OH-dibenzocycloheptanone)-H 2O
2200
—
—
—
178
206
176
152
76
89
M (norcyclobenzaprine-AC) Noxiptyline
Opipramol
3050
3219
—
—
363
206
143
M (ring)
1985
—
—
—
—
—
—
42 —
70
193
—
— 109
—
2691
—
2.047
44
329
192
70
138
M (desmethylenyl-3-methyl-)
—
2734
—
—
44
192
140
331
177
70
M
—
2687
—
—
105
210
77
254
132
44
Paroxetine
Phenelzine
1335
1278
—
—
31
45
46
29
59
74
Protriptyline
2253
2329
2590
0.878
70
44
191
192
188
59
M (nor-)
—
2343
—
—
M (10-OH-)
—
2406
—
—
2472
—
—
—
2481
—
1.166
M (10,11-di-OH-) Sertraline
— 70
— 44
—
—
—
—
207
178
279
249
44
70
179
178
207
280
274
276
159
262
239
304
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:28:4
Specific applications
677
Table 40.12 continued Compound
System
Principal ions (m/z)
GA
GB
GF
M (nor-)
—
2468
—
GM 1.218
119
145
274
246
130
290
M (ketone)
—
2496
—
—
227
290
292
199
163
248
M
—
2279
—
—
274
276
128
202
239
259
M
—
2333
—
—
202
272
274
200
236
100
M
—
2619
—
—
131
290
292
189
220
254
M
—
2786
—
—
287
289
217
251
189
108
250
252
305
307
263
214
44
59
165
166
181
179
—
2802
—
—
Tofenacin
1920
2013
—
0.420
Tranylcypromine
1220
1252
1455
—
133
132
56
115
30
117
Trazodone
3330
3564
—
Not eluted 205
70
231
78
135
166
M (mCPP)
—
1806
—
—
154
196
138
111
156
75
M (mCPP)-AC
2265
—
—
—
166
238
138
154
168
195
M (OH-AC-)
3380
3640
—
—
205
336
414
429
231
176
M (desalkyl AC-)
2265
2261
—
—
166
238
56
195
140
153
M (N-desalkyl-OH-) isomer 2-AC2
2525
—
—
—
182
254
169
184
296
211
2215
2302
2505
0.734
58
249
208
99
193
234
M
Trimipramine M (nor-)
—
2335
—
0.858
208
193
44
249
234
280
M (OH-)
2575
2631
—
—
58
265
224
250
209
310
M (nor-OH-)
—
2662
—
—
224
44
209
265
250
296
M (OH-methoxy-)
2590
2715
—
—
58
295
280
254
340
241
M (nor ring) Venlafaxine
1930
2107
—
—
194
249
208
193
167
179
—
2163
—
0.544
58
134
179
119
91
277
M (N-desmethyl-)
—
2196
—
0.570
44
202
134
121
91
263
M (O-desmethyl-)
—
2230
—
0.625
58
120
165
107
91
263
M (N,O-didesmethyl-)
—
2264
—
0.687
44
188
120
107
145
249
M (nor-OH-)
—
2450
—
—
44
134
218
200
121
185
M (O-desmethyl-OH-) isomer 1
—
2373
—
—
58
134
91
179
121
77
M (O-desmethyl-OH-) isomer 2
—
2408
—
—
58
134
91
179
121
77
1855
1923
—
—
56
100
138
110
57
237
2325
—
—
—
56
100
110
138
265
128
Viloxazine M (di-oxo-) Zimeldine
2270
—
—
0.820
58
70
318
316
317
193
M (nor-)
2223
—
—
0.941
302
304
224
193
260
272
(a)
Racemate.
their class, which may compromise the identification of the parent drug ingested. Some authors advocate the preparation of acetylated derivatives, particularly to analyse the hydroxylated metabolites, but to identify them in biological fluids this is an unnecessary additional step. Systems GA, GB, GC or GF, described above, may be used. The retention indices and principal ions are given in Table 40.13. Benzodiazepines The analysis of benzodiazepines in biological specimens is hampered by their high potency and resultant low plasma concentrations, and by their inter-connected metabolic pathways. Several benzodiazepines appear in urine almost exclusively as glucuronide-conjugated metabolites, and these can be hydrolysed with glucuronidase (1000 U glucurase/mL of urine at 60 C for 1–2 h), although some can degrade with prolonged heating. Extraction can be performed at any pH between 3 and 12, but basic extracts (pH 9–11) give cleaner chromatograms. The extraction solvent should be moderately polar (ethyl acetate is appropriate), and TMS derivatives form easily in 20–30 min at 60 C using 50% bis(trimethylsilyl)trifluoroacetamide (BSTFA) with 1% trimethylchlorosilane (TMCS) in acetonitrile. These derivatives markedly improve peak shape
and sensitivity. All compounds except 7-aminonitrazepam show electron-capture responses with high sensitivity. However, quantitation by ECD is problematic as it has a narrow linear range, and a multiple point calibration is essential. Alternatively, for most compounds a nitrogen detector (NPD) gives adequate sensitivity with a much improved linear range, although it is not advisable to make TMS derivatives if using this detector. MS detection is required to confirm the identity. System GA or GB, described above, may be used or system GG. The retention indices and principal ions are given in Table 40.14. System GG
M M€ oller, personal communication n n n
Column: 2.5% OV-17 on 80–100 mesh Chromosorb G, treatment and dimensions as for system GA. Column temperature, carrier gas, reference compounds: As for system GA. Retention indices: The retention indices of benzodiazepines have been shown to be dependent on column temperature (Schuetz, Westenberger 1978, 1979). The values given by these authors are about 50 RI units above those generally quoted. The values given below should therefore be checked before use by chromatographing a few sample compounds.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:28:29
678
Gas Chromatography
Table 40.13 GC retention data and mass spectral data for antihistamines (BP, benzophenone; CBPH, chlorobenzophenone) Compound
Alimemazine (trimeprazine) M (OH-)
System
Principal ions (m/z)
GA
GB
GC
GF
2315
2402
2646
2715
58
198
298
100
180
84
2650
—
—
—
58
314
100
255
196
281
M (OH-)-AC
2600
—
—
—
58
100
356
269
214
196
M (bis-nor-)-AC
2765
—
—
—
212
312
114
198
199
180 252
M (nor-)
2335
2432
—
—
199
284
212
198
180
M (nor-OH-)-AC 2
2930
—
—
—
128
384
228
214
270
196
M (phenothiazine)
2120
2130
—
—
199
167
198
166
154
139
M (sulfoxide)
2665
2805
—
—
M (norsulfoxide)
—
2829
—
—
M (nor-OH-) Antazoline Astemizole M (N-desalkyl)
58 —
212
199
180
298
299
—
—
—
—
—
—
2845
—
—
—
—
—
—
—
—
2318
2421
2749
—
—
—
—
—
—
—
3900
Not eluted
—
—
96
337
109
338
294
458
2470
—
—
—
109
241
132
83
242
111
2377
2499
—
—
246
280
232
247
291
217
M (OH-alkyl-)-H 2O
2410
—
—
—
244
255
288
230
216
229
M (nor-)-AC
2710
—
—
—
128
326
212
198
180
86 182
Azatadine
2250
—
—
—
97
91
96
70
77
M (OH-)
2580
—
—
—
97
91
296
198
98
96
M (OH-)-AC
2620
97
70
98
96
338
240 183
Bamipine
M (nor-)-AC
2675
91
182
77
217
308
M (nor-OH-)-AC 2
3020
91
366
240
56
199
275
Benzophenone (BPH)
1610
105
77
51
182
106
183
1673
—
—
M (OH-BPH) isomer 1
2065
—
—
—
198
121
77
199
192
151
M (OH-BPH) isomer 2
2080
—
—
—
121
198
77
105
199
122
M (OH-methoxy-BPH) isomer 1
2050
—
—
—
105
151
198
228
77
121
M (OH-methoxy-BPH) isomer 2
2070
—
—
—
105
151
198
228
77
121
M (carbinol)
1670
1722
—
—
79
219
77
218
78
108
Bromodiphenhydramine (Bromazine)
2155
2231
—
2480
58
73
45
165
59
166
Brompheniramine
2092
2184
2457
2470
247
249
58
72
167
168
M (nor-)
—
2219
—
—
247
249
167
44
246
168
M (bis-nor-)
—
2203
—
—
247
249
167
106
260
180
3360
3461
—
—
231
147
285
201
132
165
2520
2355
—
—
85
165
201
241
230
286
Buclizine (also metabolised to chlorobenzophenone) M (desalkyl-) (norchlorcyclizine) Carbinoxamine M (chlorobenzoylpyridine) M (nor-) Chlorcyclizine (also metabolised to chlorobenzophenone) M (nor-) Chlorobenzophenone (CBPH) M (4OH-CBPH)
2080
2147
2430
—
58
71
54
167
72
202
1645
—
—
—
139
189
111
216
217
191
203
167
202
220
205
204
99
56
72
165
300
228
2150
—
—
—
2220
2316
—
2560
2520
2355
—
—
85
165
201
241
230
286
1850
—
—
—
105
139
77
216
218
141
2300
—
—
—
121
232
111
139
234
197
M (4-chloromethylbiphenyl)
—
1688
—
—
167
165
202
152
82
166
M (4-chlorobiphenylmethanone)
—
1862
—
—
105
139
216
111
141
181
M (carbinol)
1750
—
—
—
105
77
139
218
165
111
Chloropyriline
2133
—
—
—
58
131
72
71
79
42 204
Chlorphenamine (see Chlorpheniramine) Chlorpheniramine
1996
2079
2586
2355
203
58
44
205
54
M (nor-)
2014
2115
—
—
203
167
44
205
202
204
M (bis-nor-)
—
2065
—
—
203
167
205
202
204
216
M (nor-AC-)
2530
2563
—
—
203
216
167
205
202
302
M (OH-)-AC
2405
—
—
—
—
—
—
—
—
—
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:28:42
Specific applications
679
Table 40.13 continued Compound
System
Principal ions (m/z)
GA
GB
GC
GF
M (bis-nor-)-AC
2535
—
—
—
—
—
—
—
—
—
M (desamino-OH-)-AC
2530
—
—
—
—
—
—
—
—
—
3233
—
—
201
117
167
251
165
202
2120
2128
—
—
167
165
207
85
152
252
2425
2521
—
2710
84
128
179
85
214
2220
—
—
—
—
—
—
Cinnarizine (also metabolised to benzophenone) 3050 M (desalkyl-, norcyclizine) Clemastine M (OH-methoxy-carbinol)-H 2O
—
98
—
— 152
2440
—
—
—
246
248
288
247
330
2620
—
—
—
131
256
125
42
255
89
2965
—
—
—
255
256
214
339
131
186
2025
2104
2348
2320
99
56
167
207
194
266
2120
2128
—
—
167
165
207
85
152
252
2355
2460
2307
2710
287
96
286
215
70
58
M (OH-)
3060
—
—
—
303
203
202
217
304
205
M (nor-)
2400
—
—
—
273
215
216
231
229
272
M (oxo-)
2960
—
—
—
229
215
202
242
301
258
M (nor-OH)-H2O
2450
2608
—
—
271
272
270
165
193
241
2615
—
—
—
83
140
82
124
96
193
M (di-OH-)-H2O Clemizole M (oxo-) Cyclizine (also metabolised to benzophenone) M (nor-) Cyproheptadine
Deptropine Dimenhydrinate (see Diphenhydramine) Dimetindene
2275
2376
2669
—
58
59
72
45
292
218
Dimetotiazine
3060
3096
—
—
72
73
230
56
210
198
M (nor-)
3150
—
—
—
72
320
306
58
198
210
M (OH-)-AC
3200
—
—
—
72
245
59
198
398
263
M (bis-nor-)-AC
3380
—
—
—
319
405
346
211
210
320
M (nor-)-AC Diphenhydramine (also metabolised to benzophenone)
3360
—
—
—
58
114
319
346
72
419
1873
1928
2387
2105
58
73
167
165
166
152
M (nor-)
1520
1922
—
—
44
165
59
167
152
166
M (nor-acetyl-)
2265
2360
—
—
44
101
167
86
165
152
M (di-nor-acetyl-)
2240
2318
—
—
167
165
87
183
72
152
M (desamino-)
—
1883
—
—
214
181
183
153
152
167
M (methoxy-)
2010
2239
—
—
58
73
165
181
152
153
M
—
2190
—
—
183
165
167
105
152
166 115
M
—
2631
—
—
167
165
152
168
253
M (diphenylmethane)
—
1465
—
—
167
168
165
152
153
91
M (diphenylmethanol)
1645
1644
—
—
167
165
152
162
168
115
—
1780
—
—
184
183
165
107
152
115
Diphenylpyraline (metabolised to benzophenone)
2100
2128
2447
2405
99
114
98
167
70
165
Doxylamine
1910
1970
—
2170
58
71
167
182
180
72
M (4-phenylmethylphenol)
M (nor-)
—
1974
—
—
182
183
167
200
149
44
M
1520
1623
—
—
182
173
167
168
94
106
M (carbinol)-H 2O
1560
1670
—
—
180
181
152
77
90
102
M (OH-)-AC
2300
—
—
—
58
71
183
198
72
182
M (OH-carbinol-)-AC
2980
—
—
—
137
257
78
79
106
200
M (OH-methoxy-)-AC
2320
—
—
—
58
71
72
183
198
196
M (bis-nor-)-AC
2280
—
—
—
182
86
167
183
181
198
M (desamino-OH-)-AC
1960
—
—
—
198
87
182
183
167
180
M (nor-)-AC
2340
—
—
—
182
183
167
100
181
58
Histapyrrodine
2240
—
—
—
84
91
196
280
197
65
M (OH-)
1650
—
—
—
84
91
212
296
213
297
M (oxo-)
2570
—
—
—
91
196
209
197
275
294
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:28:49
680
Gas Chromatography
Table 40.13 continued Compound
System
Principal ions (m/z)
GA
GB
GC
M (N-desbenzyl-)
1800
—
—
—
84
190
106
111
122
77
M (N-desbenzyl-oxo-)
2120
—
—
—
106
119
118
98
77
204
M (N-desphenyl-oxo)-H2O Hydroxyzine (also metabolised to chlorobenzophenone)
GF
2100
—
—
—
91
159
216
160
215
84
2880
3000
—
—
201
203
165
299
166
202 286
2520
2355
—
—
85
165
201
241
230
Isothipendyl
2225
—
—
—
72
214
200
285
86
56
M (nor-)
2220
—
—
—
58
214
213
181
199
271
M (desalkyl-) (norchlorcyclizine)
M (OH-)
2450
—
—
—
72
301
218
197
178
228
M (bis-nor-)
2230
—
—
—
214
257
213
58
181
215
382
265
245
280
292
294
91
233
232
276
275
65
189
105
201
285
165
190
—
—
Loratadine
—
3236
—
—
Mebhydrolin
2450
2575
2739
2920
Meclozine (also metabolised to chlorcyclizine)
3035
3193
—
—
2520
—
—
—
—
2220
2328
—
2560
121
M (N-desalkyl-) Mepyramine
58
72
—
—
—
214
122
215
M (N-desalkyl-)
2120
—
—
—
121
214
78
165
136
154
M (N-desalkyl-)-AC
2150
—
—
—
107
214
78
256
163
197
M (N-desmethoxybenzyl-) Mequitazine
1580
—
—
—
58
78
107
60
95
119
2765
2939
—
—
124
322
198
125
212
180
M (phenothiazine)
2120
2130
—
—
199
167
198
166
154
139
M (sulfoxide)
3120
—
—
—
124
198
338
321
180
—
M (sulfone)
3250
—
—
—
124
125
354
180
152
—
Methapyrilene
1981
—
—
2305
58
97
72
71
191
261
Methdilazine
2462
—
—
2920
97
98
296
199
55
212
Phenindamine
2165
2245
2926
2515
260
261
42
57
184
215
M (OH-)
2300
—
—
—
276
277
275
233
234
200
M (N-oxide)
2230
—
—
—
260
277
259
276
215
202
M (nor-)
2210
—
—
—
246
247
202
217
168
215
M (nor-OH-)
2590
—
—
—
262
263
261
184
233
228
1805
1874
—
2100
169
58
168
170
72
167 226
Pheniramine M (nor-) Phenyltoloxamine M (N-oxide)-(CH3) 2NOH
2080
1890
—
—
169
168
167
182
184
1940
2030
—
—
58
255
42
71
59
44
1500
1580
—
—
165
210
181
167
195
152
M (nor-)
2140
2002
—
—
44
58
210
165
241
181
M (O-desalkyl-)
1680
1724
—
—
184
165
78
77
106
183
M (O-desalkyl-OH-)
2220
—
—
—
200
107
94
122
152
181
M (OH-) isomer 1
2280
—
—
—
58
72
271
152
226
197
M (OH-) isomer 2
2300
—
—
—
58
72
271
152
226
197
M (nor-OH-) isomer 1
2320
2398
—
—
58
257
226
107
152
197
M (nor-OH-) isomer 2
2340
2402
—
—
58
257
226
152
197
91
M (OH-methoxy-)
2320
—
—
—
58
72
301
271
152
107
228
183
165
184
106
181
72
284
198
213
199
180
M (desamino-OH-) Promethazine
1830
1928
—
—
2339
2383
2546
2675
M (nor-)
2250
2333
—
—
58
213
180
198
152
270
M (phenothiazine)
2120
2130
—
—
199
167
198
166
154
139
M (sulfoxide)
2710
2797
—
—
72
198
180
152
213
229
M (norsulfoxide)
—
2732
—
—
58
212
180
198
229
152
M (nor-OH-)
2580
2717
—
—
212
58
180
229
198
286
M (OH-)
2590
—
—
—
72
196
300
229
214
288
Propiomazine
2738
—
—
3225
72
73
340
269
197
56
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:29:19
Specific applications
681
Table 40.13 continued Compound
System
Principal ions (m/z)
GA
GB
GC
GF
2419
—
—
2815
205
240
91
84
125
242
2920
—
—
—
205
115
98
240
200
123
Thenalidine
2318
—
—
—
97
70
99
43
188
44
Thenyldiamine
1999
—
2300
2340
58
97
72
71
203
191
Pyrrobutamine M (oxo-)
Thiazinamium methysulfate Art (promethazine) 2339
2383
2546
2675
72
284
198
213
199
180
Thonzylamine
2203
—
2576
—
58
121
72
71
216
215
Tolpropamine
1900
—
—
—
58
253
165
193
178
117
M (OH-)
2150
—
—
—
58
269
165
115
178
193
M (nor-)
2100
—
—
—
193
239
165
178
208
117
M (nor-OH-)
2200
—
—
—
255
193
167
165
178
115
M (N-oxide)
1750
—
—
—
115
193
208
178
165
116
Trimethobenzamide
3281
—
—
—
58
195
59
72
388
89
Tripelennamine
1976
—
—
—
58
91
72
71
197
185
2420
—
—
—
91
129
197
58
147
241
Trimeprazine (see Alimemazine)
M (nor-) M (OH-)
2400
—
—
—
58
91
72
213
200
271
M (benzylpyridylamine)
1650
—
—
—
184
106
79
183
78
107
2253
2340
2954
2600
208
209
278
207
193
200
Triprolidine
Table 40.14 GC retention data and mass spectral data for benzodiazepines (ET, ethyl; TMS, trimethylsilyl) Compound
System
Principal ions (m/z)
GA
GB
GG
2955
—
—
308
307
309
310
58
280
Alprazolam(a)
3100
3108
—
308
279
204
273
77
307
M (aOH-)
3245
0000
—
287
322
321
323
288
324
M (aOH)-TMS
—
3183
—
381
396
382
383
398
397
M (4OH-)
3045
—
—
—
—
—
—
—
Bromazepam
2665
2760
3280
236
317
318
288
316
286
Bromazepam-TMS
—
2702
—
388
386
387
389
372
374
M (3OH-)
2470
—
—
304
314
331
305
303
302
M (3OH)-TMS2
—
2650
—
388
386
477
475
179
360
M (aminohydroxybromazepam)-TMS
—
2590
—
249
247
366
364
338
336
3070
—
—
394
245
316
210
291
176
Adinazolam
(a)
Brotizolam(a)
3050
—
—
380
378
379
299
301
245
Camazepam(a) (metabolised to oxazepam and temazepam)
2945
3162
—
58
72
43
78
271
44
Chlordiazepoxide (metabolised to nordazepam and oxazepam)
2795
2981 thermally unstable
3065
282
299
284
283
241
253
M (nor-)
2452
2679
—
268
269
270
233
271
205
M (demoxepam)
2529
2806
—
120
285
286
269
241
287
Clobazam(a)
2558
2683
3174
300
258
77
259
283
231
M (nor-)
2747
2759
—
286
218
215
217
216
244
M (OH-)
3000
—
—
316
318
274
271
299
247
M (OH-MeO-)
3255
—
—
346
316
301
348
274
271
Clonazepam
2823
3000
3600
280
314
315
285
234
288
Clonazepam-TMS
M (OH-) Art (-CH2O)
—
2781
—
387
352
306
372
386
388
M (7-amino-)
2890
2996
—
285
256
257
287
250
111
M (7-amino)-TMS2
—
2742
—
429
394
414
430
431
314
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:29:23
682
Gas Chromatography
Table 40.14 continued Compound
System
Principal ions (m/z)
GA
GB
2935
—
—
283
220
225
285
254
284
Clorazepic acid (hydrolysed in vivo and absorbed as diazepam)
2457
2618
3125
242
43
270
269
241
103
Clotiazepam(a)
2532
—
—
289
318
291
320
275
290
M (OH-)
2705
—
—
287
316
318
289
288
317
M (OH-)-AC
2870
—
—
271
316
256
300
273
241
M (di-OH-)-AC2
2995
—
—
332
374
319
291
303
434
M (amino-OH-)
GG
Demoxepam
2529
2806
3043
285
286
269
287
241
242
Diazepam(a) (metabolised to nordazepam, oxazepam and temazepam)
2428
2556
2940
256
283
284
287
257
255
Estazolam(a)
3070
3050
—
259
294
293
205
239
101
Etizolam(a)
2980
—
—
342
266
313
224
239
45
Flunitrazepam(a)
2600
2744
3190
285
312
313
286
266
238
M (nor-)
2720
2816
—
224
299
298
372
271
252
M (nor-)-TMS
—
2622
—
371
370
352
356
324
372
M (7-amino-)
2723
2804
—
283
255
254
282
264
240
M (7-amino-)-TMS
—
2836
—
355
327
326
354
356
312
M (nor-amino-)
2825
—
—
269
240
241
268
270
213
2780
2896
3220
86
87
99
58
84
387
M (desalkyl-)
2470
2559
—
288
260
259
287
261
289
M (desalkyl-)-TMS
—
2350
—
359
360
341
361
345
362
M (2-OH-ET-)
2675
2805
—
288
273
287
332
331
304
M (2-OH-ET-)-TMS
—
2778
—
288
287
273
389
273
360
M (bis-desethyl-)
2694
2739
—
313
315
314
312
250
259
M (desalkyl-OH-)
2255
2373
—
223
286
258
75
257
251
Flutazolam
2460
—
—
289
245
246
210
259
211
Halazepam(a) (metabolised to nordazepam, oxazepam)
2285
—
—
324
352
323
325
351
353
Flurazepam(a)
Ketazolam (hydrolyses to diazepam) Loprazolam(a)
Not eluted Not eluted —
Lorazepam
2410
2528
2910
70
464
42
43
465
394
291
239
274
293
75
302
Lorazepam-TMS2
—
2566
—
429
431
430
347
449
432
Lormetazepam
2660
2770
—
305
307
306
309
308
334
Lormetazepam-TMS (metabolised to lorazepam)
—
2799
—
377
379
391
291
317
406
Medazepam(a) (metabolised to diazepam, nordazepam, oxazepam and temazepam)
2235
2340
2620
242
207
244
270
243
271
2280
—
—
228
193
256
257
165
110
2640
—
—
349
347
321
351
350
394
M (O-desmethyl-)
2730
—
—
321
319
380
378
349
347
M (nor-)
2690
—
—
335
333
349
347
378
380
(a)
Mexazolam
2600
—
—
251
70
253
41
42
139
Midazolam(a)
2575
2722
—
310
312
311
163
325
75
M (a-OH-)
2830
2901
—
310
311
312
341
283
313
M (a-OH-)-TMS
—
2866
—
310
398
413
312
400
415
M (4-OH-)-TMS
—
2775
—
268
269
297
412
298
397
2740
2915
3450
280
253
281
206
234
252
M (nor-) Metaclazepam(a)
Nitrazepam
—
2642
—
352
353
306
338
354
307
M (7-amino-)(b)
2785
2878
—
251
223
222
250
252
235
M (7-amino-)-TMS3
—
2634
—
394
395
396
380
280
322
43
222
Nitrazepam-TMS
—
Not eluted —
293
265
264
292
Nordazepam
2490
2625
3041
—
—
—
—
—
—
Nordazepam-TMS
—
2367
—
341
342
343
327
344
329
Oxazepam
2803
2325
2438
257
77
268
239
205
267
M (7-acetamido-)
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:29:34
Specific applications
683
Table 40.14 continued Compound
System GA
GB
GG
Oxazepam-TMS2
—
2468
—
429
430
431
313
415
401
Oxazolam(a) (metabolised to oxazepam)
2540
—
—
251
70
253
241
105
252
Prazepam(a) (metabolised to nordazepam and oxazepam)
2648
2783
3145
91
269
324
55
296
295
2860
—
—
257
55
311
259
313
340
2440
2576
—
386
359
323
388
245
303
2255
—
—
342
341
343
370
369
259
Temazepam (metabolised to oxazepam)
2595
2727
3125
271
273
300
272
256
77
Temazepam-TMS
—
2713
—
343
344
257
283
357
372
Tetrazepam(a)
2430
—
—
253
288
287
289
225
259
M (nor-)
2530
—
—
239
274
273
275
245
240
M (OH-) isomer 1
2570
—
—
235
375
304
237
261
247
M (OH-) isomer 2
2580
—
—
235
375
304
237
261
247
M (oxo-)
2430
—
—
285
287
267
245
302
247
M (3-OH-) Quazepam(a) (also metabolised to desalkylflurazepam) M (2-oxo-)
Principal ions (m/z)
3080
3219
—
313
238
342
315
75
344
M (a-OH-)
3000
0000
—
328
330
293
265
239
357
M (a-OH)-TMS
—
3308
—
415
417
430
432
416
380
—
2426
—
257
285
267
286
258
145
(a)
Triazolam
Zolazepam(a) (a)
Does not form a TMS derivative. No electron-capture response.
(b)
Hydrolysis of benzodiazepines (preparation of benzophenones)
Boil an aqueous solution (or urine) with concentrated hydrochloric acid (1 part to 10 parts urine or solution) for 30–60 min. Cool, and neutralise with solid KHCO3 or adjust the pH to 8–9 with 10 mol/L KOH. Mix with an equal volume of petroleum ether for 10 min. Centrifuge and evaporate the upper organic phase to dryness at 60 C. The reconstituted extract can be used for GC or other analytical procedures such as TLC (Chapter 39). Data are also presented here for some acetylated hydrolysis products (treatment of the dried residue with acetic anhydride and pyridine (3 : 2) for 30 min at 60 C (Maurer, Pfleger 1987). Not all benzodiazepines make benzophenones when hydrolysed by acid, and a number of other degradation products are furnished. The a-OH-metabolites of alprazolam, brotizolam and triazolam are partly altered by the elimination of formaldehyde. Hydrolysis products of bis-desethylflurazepam and di-OH-tetrazepam are dehydrated; OH-bromazepam, lorazepam and oxazepam form artefacts by rearrangement; the nor-metabolites of
clobazam are cleaved and rearranged to benzimidazole derivatives; tetrazepam, and its two hydroxylated metabolites, are transformed into a pair of cis- and trans-isomeric hexahydroacridone derivatives. Since the metabolism of benzodiazepines is complex, assays that convert drugs and metabolites into hydrolysis products are not ideal, because they do not permit unequivocal identification of the parent compound. After acid hydrolysis, care must be taken to ensure that the acid is neutralised prior to extraction or before injecting the solvent onto the chromatograph, otherwise the column deteriorates rapidly. The retention indices and principal ions are given in Table 40.15. Cardioactive drugs Cardioactive drugs (beta-blockers, calcium channel antagonists, angiotensin-converting enzyme (ACE) inhibitors, etc.) are a diverse group of
Table 40.15 GC retention data and mass spectral data for benzophenones, hydrolysis products (HY) and some acetylated derivatives (AC) of benzodiazepines and their metabolites Abbreviation
Structure
ABP
2-(2-Amino-5-bromobenzoyl)pyridine 2245
ABP-AC
ACB
System GA
Parent compound (in-vivo by metabolism)
Principal ions (m/z)
3-OH-Bromazepam, bromazepam
247
249
276
278
246
248
121
247
249
318
320
289
2490 3OH-bromazepam HY Art 1
2255
(Bromazepam)
285
287
206
286
284
179
3OH-bromazepam HY Art 2
2265
(Bromazepam)
299
301
220
300
298
179
2-Amino-5-chlorobenzophenone
2039
Nordazepam, oxazepam (camazepam, chlordiazepoxide, clorazepic acid, diazepam, halazepam, ketazolam, medazepam, oxazolam, prazepam)
230
77
231
232
233
195
ACB-AC
2245
230
231
232
273
77
105
ACB Art 1
2060
239
205
240
241
163
177
ACB Art 2
2070
253
219
254
255
110
238
ACDP
2-Amino5-chlorodiphenylamine
2210
Norclobazam (clobazam)
242
241
77
166
206
243
Nor-OH-MeO-clobazam HY
2405
(Clobazam)
288
290
289
272
245
281
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:29:36
684
Gas Chromatography
Table 40.15 continued Abbreviation
ACFB
Structure
System GA
Nor-OH-MeO-clobazam HY-AC
2615
Nor-OH-clobazam HY
2650
Nor-OH-clobazam HY-AC
3000
OH-MeO-clobazam HY
2905
2-Amino-5-chloro-2 0 fluorobenzophenone
2030
ACFB-AC
Parent compound (in-vivo by metabolism)
Principal ions (m/z) 288
290
330
332
290
235
258
257
259
260
224
246
258
300
260
259
257
302
(Clobazam)
320
322
206
240
321
207
Desalkylflurazepam, N-desalkylflutoprazepam, oxo-quazepam (flurazepam, flutoprazepam, quazepam)
249
248
250
251
123
124
(Clobazam)
2195
249
248
291
123
95
250
Didesethylflurazepam Art (-H2O)-AC
2460
Didesethylflurazepam (flurazepam) 246
316
245
211
273
275
ACNB
2-Amino-2 0 -chloro5-nitrobenzophenone
2470
Clonazepam, loprazolam
241
276
139
111
195
165
ADCB
2-Amino-5,2 0 -dichlorobenzophenone 2120
Lorazepam, mexazolam (lormetazepam)
230
265
267
232
154
195
ADCB-AC AFMAB
239
265
307
287
232
309
Lorazepam HY Art
2300 2170
Lorazepam (lormetazepam)
239
241
274
273
275
276
5-Amino-2 0 -fluoro-2methylaminobenzophenone
2753
7-Acetamidoflunitrazepam, 7-aminoflunitrazepam (flunitrazepam)
244
227
243
245
211
123
205
286
328
244
243
269
AFNB
2-Amino-2 0 -fluoro-5nitrobenzophenone
2870 2330
Desmethylflunitrazepam (flunitrazepam)
260
259
123
165
213
241
ANB
2-Amino-5-nitrobenzophenone
2388
Nitrazepam
242
241
77
105
195
165
CPMACB
2-Cyclopropylmethylamino-5chlorobenzophenone
2385
3-Hydroxyprazepam, prazepam
56
77
105
285
270
165
AFMAB-AC2
CPMACB-AC
257
256
259
241
283
343
CTFEAB
5-Chloro-2-(2,2,2-trifluoro)ethylaminobenzophenone
2595 2380
Halazepam
313
312
314
315
244
296
DAB
2,5-Diaminobenzophenone
2175
7-Acetamidonitrazepam, 7-aminonitrazepam (nitrazepam)
211
212
77
107
195
183
296
212
211
254
253
297
246
211
245
248
107
247
330
288
246
211
139
332
230
229
211
210
107
231
230
314
272
229
123
201
244
279
229
281
111
75
86
87
57
348
350
109
262
109
166
264
293
275
262
109
166
275
335
264
77
245
244
228
105
246
DAB-AC2 DACB
2985 2,5-Diamino2 0 -chlorobenzophenone
7-Acetamidoclonazepam, 7-aminoclonazepam (clonazepam)
2845
DACB-AC2 DAFB
2305
2,5-Diamino2 0 -fluorobenzophenone
DAFB-AC2
2175
7-Aminodesmethyl flunitrazepam (flunitrazepam)
2715
DCMAB
2 0 ,5-Dichloro-2-(methylamino) benzophenone
2220
Lormetazepam
DEACFB
2-Diethylaminoethylamino5-chloro-2 0 fluorobenzophenone
2505
Flurazepam
HEACFB
2-Hydroxyethylamino5-chloro2 0 -fluorobenzophenone
2400
Hydroxyethylflurazepam, flutazolam (flurazepam, quazepam)
HEACFB-AC
2470
MACB
2-Methylamino-5chlorobenzophenone
2105
Diazepam, temazepam (camazepam, chlordiazepoxide, clorazepate, ketazolam, tetrazepam)
MACDP
2-Methylamino-5chlorodiphenylamine
2220
Clobazam
257
259
77
274
215
231
MANFB
2-Methylamino-5-nitro2 0 -fluorobenzophenone
2385
Flunitrazepam
274
273
257
211
123
275
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:30:23
Specific applications
685
Table 40.15 continued Abbreviation
Structure
System GA
Parent compound (in-vivo by metabolism)
Principal ions (m/z)
a-OH-alprazolam HY Art (-CH2O)
3070
(Alprazolam)
259
294
293
205
239
265
a-OH-alprazolam-AC
3180
(Alprazolam)
323
324
325
366
271
295
OH-brotizolam HY Art (-CH2O-)
3050
(Brotizolam)
380
378
379
299
301
245
OH-brotizolam-AC
3140
(Brotizolam)
409
407
410
450
452
408
a-OH-midazolam-AC
2820
(Midazolam)
310
340
383
312
342
385
a,4-Di-OH-midazolam-AC2
3020
(Midazolam)
310
326
399
340
383
441
Normedazepam-AC
2470
(Medazepam)
228
297
193
256
298
255
Tetrazepam HY (isomer 1)
2220
Tetrazepam
207
249
220
206
209
234
Tetrazepam HY (isomer 2)
2260
Tetrazepam
207
249
220
206
209
234
Tetrazepam M (OH-) HY-AC (isomer 1) 2380
(Tetrazepam)
307
248
234
264
309
220
Tetrazepam M (OH-) HY-AC (isomer 2) 2470
(Tetrazepam)
307
248
234
264
309
220
Tetrazepam M (OH-) HY-AC (isomer 3) 2535
(Tetrazepam)
307
206
309
248
218
220
Tetrazepam M (OH-) HY-AC (isomer 4) 2560
(Tetrazepam)
307
218
220
206
248
264
Tetrazepam M (nor-OH-) HY-AC2
2500
(Tetrazepam)
233
232
196
154
275
335
a-OH-triazolam HY Art (-CH2O)
3000
(Triazolam)
328
293
330
265
239
329
a-OH-triazolam-AC
3200
(Triazolam)
357
359
358
400
402
329
chemicals that require different analytical strategies. The calcium channel antagonists of the phenylalkylamine class (e.g. verapamil) and the benzothiazines (e.g. diltiazem) chromatograph well on standard GC phases (X-1 or X-5) after solvent extraction under mildly basic conditions (pH 10–12). As a rule, the beta-blockers (those with names that end in ‘-olol’) are more water soluble and often require ‘salting out’ of aqueous solution at mildly basic conditions (pH 10–12) with an excess of solid sodium chloride. When subjected to GC they have a tendency to produce artefacts by loss of water and/or their amino-alkyl side-chain. Although they chromatograph reasonably well on capillary columns such as X-1 or X-5, they often give tailing peaks on packed columns, and various derivatisation strategies have been employed to overcome this. Simultaneous preparation of N-TFA and O-TMS derivatives have been described (Leloux et al. 1989; Lho et al. 1990), and cyclic boronates may be formed from phenyl or n-butylboronic acids using either triethylamine or pyridine catalysts (Lee et al. 1998). Acetylation of acid-hydrolysed sample extracts is described using acetic anhydride with a pyridine catalyst; although this process results in the complete destruction of some of the parent compounds, data for many metabolites are given (Maurer, Pfleger 1986). The ACE inhibitors (those with names that end in ‘-pril’) have a free carboxylic acid group, and acquire a second such group by enzymatic hydrolysis of their ethyl ester link (‘prilates’). Neither of these groups of compounds, nor the dihydropyridine calcium channel antagonists (e.g. nifedipine), chromatographs in their native
state, but GC has been applied successfully to their analysis following extractive methylation. Here, equal volumes of urine and phase-transfer reagent (0.02 mol/L tetrahexylammonium hydrogensulfate in 1 mol/L phosphate buffer pH 12) are incubated with three volumes of 1 mol/L methyliodide in toluene on a shaker at 50 C for 30 min. The organic phase is eluted on a diol solid-phase cartridge (conditioned with methanol, then toluene) using diethyl ether–ethyl acetate (92.5 : 7.5, v/v). After evaporation to dryness at 60 C, the residue is reconstituted in ethyl acetate for injection. Systems GA and GB, described above, can be used, and system GP chromatographs many of the drug metabolites as their methyl derivatives (Maurer et al. 1998; Maurer, Arlt 1999). System GP n n n n n
Column: HP1 (methyl-PSX) fused silica capillary (12 m 0.2 mm i.d., 0.33 mm) Injector: 280 splitless mode. Temperature programme: 100 C for 3 min to 310 C at 30 /min for 8 min. Carrier gas: He, 1 mL/min. Retention indices and principal ions are given in Table 40.16.
Coumarins and other anticoagulants Coumarins are extracted fairly readily from acidic solution, especially when ‘salted out’ with solid sodium chloride; the coumarin structure is
Table 40.16 GC retention data and mass spectral data for cardioactive drugs (Me, methyl) Compound
Acebutolol Art
System
Principal ions (m/z)
GA
GB
GP
2811
2926
—
72
43
56
151
221
98
2910
3014
—
151
221
333
98
86
348
Art
—
2761
—
72
193
43
151
136
122
Art (-H2O)
2850
2569
—
303
98
140
318
82
233
M (phenol-)
2450
2056
—
151
221
136
108
43
132
Art (desacetyl-)
—
2089
—
194
209
264
279
166
234 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:30:34
686
Gas Chromatography
Table 40.16 continued Compound
System
Principal ions (m/z)
GA
GB
GP
Acecainide (NAPA)
2550
2724
—
Acetyldigitoxin
Not eluted Not eluted —
162
120
205
—
86
—
99
—
58
—
—
—
Ajmaline
2880
—
—
144
326
173
198
297
237
Alprenolol
1820
—
—
72
56
73
249
98
234
Amiodarone
3335
Not eluted —
2800
—
—
Amlodipine
—
2982
—
297
208
44
254
298
347
Amlodipine-Me
2820
—
—
311
254
88
312
208
238
Art (O-desalkyl)
86
87
84
58
56
44
142
121
294
265
251
237
2815
—
—
72
58
325
165
208
347
M (dehydro-2-HOOC-)-Me
—
—
2430
356
296
224
268
357
391
M (dehydro-desamino-HOOC-)-Me
—
—
2635
260
347
318
316
349
400
2462
—
—
113
84
116
98
117
115
M (N-desalkyl-)
1920
—
—
104
209
77
116
115
94
M (p-aminophenol)
1253
1280
—
109
52
53
80
81
108
Amlodipine-Me2
Amyl nitrite (see Chapter 12) Aprindine
2385
2469
—
72
56
98
43
107
41
Art (-H2O)
2150
2090
—
98
56
107
248
72
190
Art
2400
2648
—
46
86
107
127
72
263
Art (HOOC)-Me
2140
—
—
72
107
116
237
56
266
2175
—
—
278
127
112
56
293
292
—
Not eluted —
—
—
—
—
—
Atenolol
Art (HOOC)-Me Benazepril
—
—
—
3030
365
204
91
366
392
347
—
—
2985
379
204
380
91
144
438
Benziodarone
—
Not eluted —
518
173
264
519
373
376
Betaxolol
2370
2420
—
72
253
292
307
55
107
Art
2410
2508
—
304
319
318
290
127
55
Art (-H2O)
2400
2519
—
72
98
53
56
107
158
Benazepril-Me M (benazeprilate)-Me3
Bethanidine
1925
—
—
71
91
106
177
57
72
Bisoprolol
2378
2427
—
72
116
107
100
281
73
Art
2595
2680
—
127
112
86
322
337
224
Art (-H2O)
2400
2480
—
98
56
307
107
204
220
M (phenol)
1690
—
—
107
123
77
167
103
210 221
Bunazosin
3330
—
—
247
260
233
373
234
Bunitrolol
1960
—
—
86
233
70
71
58
204
1980
—
—
245
70
56
119
158
260
Art Captopril
Not eluted Not eluted —
70
41
69
75
114
217
Captopril-Me
1730
—
—
70
128
172
199
231
198
Captopril-Me2
—
—
—
70
128
89
245
203
130
Carazolol
2810
—
—
183
72
298
154
184
116
2830
—
—
183
127
112
310
222
295
Carazolol-Me
2815
—
—
86
183
154
312
298
268
Carteolol
2588
—
—
86
57
277
70
292
87
Art
2690
—
—
289
141
126
202
304
290
Carvedilol Art 1
—
2056
—
222
223
11
151
98
77
Carvedilol Art 2
—
2224
—
183
154
155
127
77
184
Art
Celiprolol
2610
—
—
86
58
250
44
291
307
Art 1
2350
—
—
333
86
56
96
112
216
Art 2
2650
—
—
86
151
291
277
114
265
2740
—
—
86
209
323
294
114
56
Cilazapril
—
Not eluted —
211
143
91
283
197
344
Cilazapril-Me
—
—
157
225
297
344
91
358
Art 3
3010
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:30:55
Specific applications
687
Table 40.16 continued Compound
System
Principal ions (m/z)
GA
GB
GP
—
—
2960
157
225
297
245
361
2100
—
—
86
57
127
190
243
90
Art (-H2O)
1895
—
—
57
174
175
176
202
258
Art
2160
—
—
99
243
245
187
174
176
M (cilazaprilate)-Me3 Clenbuterol
372
2090
2165
—
229
231
172
194
174
200
M
—
2046
—
192
157
227
194
229
193
M
—
2264
—
243
188
245
186
194
236
Art (dichlorophenylisocyanate)
1350
—
—
187
189
124
159
126
161
Art (dichlorophenylmethylcarbamate)
1500
—
—
184
186
174
133
160
219
Art 5
2110
—
—
248
194
250
229
243
283
44
117
Clonidine
0000
0000
—
132
104
175
103
Debrisoquine-acetylhydrazone deriv.
—
2100
—
—
—
—
—
—
—
M (4OH)-acetylhydrazone deriv.
—
2450
—
—
—
—
—
—
—
—
—
578
195
577
367
351
579
125
189
230
127
191
63
Debrisoquine
Deserpidine
—
Deslanoside (metabolised to digoxin)
Not eluted Not eluted —
Diazoxide
Not eluted Not eluted —
Digitoxin
Not eluted Not eluted —
Digoxin
Not eluted Not eluted —
Diltiazem
2949
3076
—
58
71
72
121
150
136
M (desacetyl-)
2990
3092
—
58
71
121
136
150
109
M (O-desmethyl-)
3050
3147
—
58
71
159
283
207
253
M (N-desmethyl-)
—
3114
—
44
150
283
161
121
136
72
194
2505
2608
—
195
212
114
167
M (N-desalkyl-)
—
2286
—
—
—
—
—
—
—
M
—
2264
—
194
196
167
180
280
252
—
Disopyramide
Doxazosin
—
3054
—
—
—
—
Enalapril-H2O
2770
2864
—
208
254
117
70
—
—
169
358 317
2650
—
2675
234
91
70
130
160
M (enalaprilate)-Me3
—
—
2680
234
130
174
235
331
91
M (enalaprilate-H2O)-Me
2730
—
2735
208
240
91
70
117
344
Enalapril-Me
Encainide
3016
—
—
98
135
70
99
77
352
Enoximone
—
Not eluted —
248
247
151
201
249
200
Esmolol
—
2311
—
72
56
107
116
73
91
Art
—
2395
—
292
306
307
293
278
234
Felodipine
2670
2793
—
238
210
239
354
383
338
Felodipine-Me
2725
—
—
252
224
324
326
164
338
M (dehydro-desethyl)-Me
—
—
2235
322
324
323
300
258
173
M (dehydro-)
—
—
2280
346
318
348
320
286
173
2250
2351
—
84
56
97
301
219
209
Flecainide
2500
2240
—
301
125
42
97
218
343
Heptaminol
1120
—
—
59
56
69
113
95
127
Hexobendine
Not eluted —
—
296
195
58
253
297
Hydralazine
1528
—
—
—
—
—
—
Art (formyl)
1914
Hydroquinidine
2810
—
—
—
Imidapril-Me
2700
—
—
234
M (desethyl-)-Me2
2695
—
—
M (desethyl-)-Me3
2710
—
—
2177
—
—
Indoramin
—
—
—
Isradipine
2680
—
—
Imolamine
—
84 —
—
—
—
—
91
346
159
160
117
220
56
117
70
159
346
234
360
130
235
56
117
—
—
—
—
217
174
105
218
143
130
210
252
77
150
178
284
—
—
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:31:5
688
Gas Chromatography
Table 40.16 continued Compound
System
Principal ions (m/z)
GA
GB
GP
2670
—
—
298
224
268
56
385
M (dehydro-desiospropyl)-Me
2270
—
2270
309
264
341
310
294
279
M (dehydro-)
2360
—
—
295
327
265
251
369
310
1320
1270
—
44
91
132
117
65
78
Isradipine-Me
Labetalol Art
284
Lacidipine
2955
—
—
252
196
326
169
119
382
Lanatoside c (hydrolyses to digoxin)
—
Not eluted —
41
43
55
57
73
81
Lidocaine
1870
1947
—
86
87
58
44
72
42
M (MEGX)
1800
1870
—
58
121
120
163
91
77
M (GX)
—
1776
—
121
178
120
106
148
77
M (2,6-xylidine)
1180
1195
—
106
121
120
91
77
65
M (3OH-)
2350
—
—
86
58
250
194
110
120
3870
Not eluted —
343
70
344
109
42
113
2230
—
—
—
—
—
—
—
—
2125
—
—
—
—
—
Lidoflazine M (desaminocarboxy-)
—
—
—
Lisinopril
Not eluted Not eluted —
70
91
84
113
245
224
Lorcainide
2810
2923
—
82
56
110
355
124
251
M (N-desmethyl-)
2660
2789
—
—
—
—
—
—
M (desacyl-)
2100
—
—
110
56
125
252
180
254 435
M (desaminocarboxy)-Me
—
Losartan-Me2
—
—
3555
192
201
249
165
450
Mecamylamine
—
—
—
98
84
71
56
99
124
Methoserpidine
—
—
—
608
195
607
397
609
395
Methyldopa
—
Not eluted —
88
42
123
124
89
77
Metipranolol
2220
2320
—
72
152
56
116
102
194
Art
2240
—
—
127
114
112
86
152
306
Art
2190
—
—
72
152
116
56
137
223
2035
2090
—
72
107
56
73
223
100
Art
2120
2176
—
56
127
112
114
264
279
M
2200
2284
—
72
107
116
251
280
145
M (OH-) Art
2240
2355
—
128
127
250
280
295
294
1400
1431
—
58
44
83
77
69
85
M (desamino-oxo-)
1350
1395
—
105
178
135
134
133
121
M
—
1745
44
58
91
77
135
178
—
—
84
209
67
43
110
192
Metoprolol
Mexiletine
Minoxidil
—
—
—
3575
234
305
190
250
91
439
M (moexiprilate)-Me3
—
—
3580
234
305
190
91
220
453
M (moxeprilate-H2O)-Me3
Moexipril-Me
—
—
3775
190
466
449
91
164
290
Moracizine
—
—
—
100
286
142
56
239
70
Nadolol
2540
2658
—
86
57
294
71
310
70
Art
2560
2670
—
306
70
86
141
201
307
3900
Not eluted —
91
134
147
146
148
165
M (dehydro-desbenzylMeNH2)-Me
—
—
2300
327
297
313
312
344
252
M (dehydro-desamino-HOOC)-Me
—
—
2645
312
313
281
371
285
139
M (dehydro-desamino-OH-)
—
—
2665
312
313
299
281
252
374
M
2250
—
—
299
269
316
285
300
241 285
Nicardipine
2545
2708
—
239
284
224
268
330
M (dehydro-)
2250
2370
2255
298
299
252
267
313
344
M (dehydo-HOOC-)
2290
—
—
283
252
224
126
282
298
M (dehydro-2-HOOC)-Me
—
—
2695
342
343
139
195
357
388
Nifedipine
—
Not eluted —
—
—
—
—
—
—
M (dehydro-desisopropyl)-Me
—
—
2565
340
324
355
308
164
341
M (dehydro-)
—
—
2565
324
341
310
383
294
164
Nilvaldipine
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:31:25
Specific applications
689
Table 40.16 continued Compound
System
Principal ions (m/z)
GA
GB
2929
3096
—
—
—
—
—
—
—
M (dehydro-desisopropyldesmethoxyethyl)-Me2
—
—
2300
327
297
313
312
344
252
M (dehydro-desmethoxyethyl)-Me
—
—
2390
313
298
252
330
283
372
M (dehydro-desisopropyl-O-desmethyl- — HOOC)-Me
—
2645
312
313
281
371
285
139
Nimodipine
GP
—
—
2655
298
299
340
341
342
357
M (dehydro-desisopropyl-O-desmethyl)-— Me
—
2665
312
313
299
281
252
374
—
—
2740
298
340
281
299
341
371
2730
—
—
371
210
270
266
284
254
M (dehydro-desisobutyl)-Me
—
—
2255
298
299
252
267
313
344
M (dehydro-)
—
—
2450
284
340
57
285
236
303
M (dehydro-OH-)
—
—
2615
284
356
59
253
313
267
M (dehydro-desisobutyl-2-HOOC)-Me2 —
—
2695
342
343
139
195
357
388
M (dehydro-)
M (dehydro-O-desmethyl-HOOC)-Me Nisoldipine
2635
—
—
238
210
239
287
360
150
M (dehydro-desethyl-)-Me
—
—
2300
327
297
313
312
344
252
M (dehydro-)
—
—
2370
341
313
358
312
281
252
M (dehydro-desethyl-)-CO2
2275
—
—
269
329
255
286
139
180
Nitrendipine
M (dehydro-desmethyl-)-CO2
2330
—
—
251
139
253
255
283
300
M (dehydro-desethyl-OH-)-H2O
2650
—
—
311
281
328
297
312
250 326
M (dehydro-desethyl-OH-)-H2O Oxprenolol Art
2690
—
—
325
297
342
266
250
1870
1972
—
72
56
221
41
73
57
1985
2062
—
56
262
248
148
277
235
—
—
—
—
—
1700
—
—
Pargyline
1214
1257
—
82
68
91
159
42
Pempidine
—
—
—
140
84
51
41
72
69
Penbutolol
2139
2221
—
86
70
56
276
133
161
M (desamino-OH-desalkyl-)
—
158
M (OH-)
2425
—
—
86
304
56
178
319
292
Art (formyl)
2150
—
—
288
289
303
141
91
159
Perhexiline
2245
—
—
84
194
55
85
56
99
M (OH-)
2485
—
—
84
56
210
192
97
110
M (di-OH-)
2660
—
—
84
56
98
210
70
249
M (di-OH-)-H2O
2510
—
—
84
56
192
208
210
291
Perindopril
—
Not eluted —
—
—
—
—
—
Perindopril-Me
—
—
—
2450
172
98
309
173
124
382
M (perindoprilate)-Me3
—
—
2470
172
112
86
323
173
382
M (perindoprilate-H2O)-Me3
—
—
2560
222
249
277
336
133
294
Phenoxybenzamine
2235
2332
—
91
196
198
92
197
65
Phentolamine
Not eluted Not eluted —
281
120
91
122
280
160
Pindolol
2245
2335
—
72
133
116
248
134
56
Practolol
0000
2440
—
72
151
43
109
56
57
Prajamlium M (OH-) Art
2925
—
—
—
—
—
—
—
—
3130
—
—
224
126
196
384
313
356
M (methoxy-) Art
2895
—
—
254
370
398
255
126
297
M (OH-methoxy-) Art
3200
—
—
224
196
206
414
343
399
Prazosin
Not eluted Not eluted —
233
383
259
245
95
56
Prenylamine (also metabolised to amphetamine)
2555
—
—
58
238
91
45
239
167
Prenylamine-AC
2925
—
—
58
91
100
280
238
164
2320
—
—
73
165
167
193
253
152
M (N-desalkyl-)-AC
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:31:31
690
Gas Chromatography
Table 40.16 continued Compound
System
Principal ions (m/z)
GA
GB
GP
M (N-desalkyl-OH-)-AC2
2635
—
—
73
183
311
220
239
269
M (N-desalkyl-OH-methoxy-)-AC2
2700
—
—
73
213
299
152
240
341
M (OH-methoxy-)-AC2
3310
—
—
58
326
368
270
459
240
M (OH-)-AC2
3200
—
—
58
296
338
297
100
429
—
—
—
—
—
1940
—
—
2255
2332
—
86
99
120
92
87
58
M (N-acetyl-)
2550
2724
—
86
99
58
162
120
205
M
—
2245
—
120
58
71
92
137
149
M
—
2292
—
86
146
99
117
120
189
M
—
2642
—
58
71
120
162
92
191
M (desamino-OH-)-H2O Procainamide
—
—
2783
—
86
99
120
58
176
219
Propafenone
2730
—
—
72
91
98
297
131
312
Art (-H2O)
2300
—
—
91
98
105
294
323
230
72
M
M (OH-)-H2O
2720
—
—
230
137
339
310
M (O-desalkyl-)
1830
—
—
—
—
—
—
—
—
M (O-desalkyl-OH-) isomer 1
2345
—
—
—
—
—
—
—
—
M (O-desalkyl-OH-) isomer 2
2355
—
—
—
—
—
—
—
—
M (O-desalkyl-OH-methoxy-)
2400
—
—
—
—
—
—
—
—
2147
2234
—
72
56
98
115
144
116
M (4OH-)
—
2546
—
72
116
160
131
275
199
M (1-naphthol)
1505
1534
—
144
115
116
72
89
63
M (desamino-OH-)
2065
—
—
144
115
218
131
116
101
—
Propranolol
98
Protoveratrine
2465
—
—
—
—
Quinapril
—
—
—
316
270
3380
3467
—
91
316
270
130
117
104
—
—
3110
234
91
130
190
160
379
M (quinaprilate)-Me3
—
—
3080
234
91
130
148
174
235
M (quinaprilate-H2O)-Me3
—
—
3310
91
130
270
302
103
132
Art-H2O Quinapril-Me
91
—
—
—
130
117
104
2790
2979
—
136
81
322
188
55
172
M (N-oxide)
2950
3086
—
152
136
189
340
173
324
M
2940
3125
—
152
124
138
338
323
158
—
Not eluted —
294
248
91
110
117
209
2980
—
248
294
117
110
209
297
Quinidine
Ramipril M (-H2O)
—
—
—
2880
234
91
160
235
357
220
M (ramiprilate)-Me3
—
—
2865
234
91
235
130
371
148
M (ramiprilate-H2O)-Me3
Ramipril-ME
—
—
2925
280
248
91
110
193
284
Rescinnamine (Reserpinine)
2180
—
—
221
109
200
186
395
251
Reserpine
Not eluted Not eluted —
608
606
195
609
395
397
M (trimethoxybenzoic acid)
1780
—
—
212
197
141
154
169
111
M (trimethoxyhippuric acid)
2085
—
—
195
251
223
152
122
167
M (trimethoxybenzoic acid)-Me
1740
—
—
226
211
195
155
183
168
M (trimethyoxyhippuric acid)-Me
2350
—
—
283
195
284
152
268
252
Sotalol
2413
2520
—
72
43
122
73
106
121
Syrosingopine
—
Not eluted —
181
395
198
251
397
396
2310
—
—
86
166
251
280
57
151
2400
—
—
292
293
307
141
166
151
Terazosin
—
Not eluted —
233
71
316
245
43
387
Timolol
2266
2373
—
—
—
—
Art
Tertatolol Art
—
—
—
2275
2380
—
86
96
72
142
154
313
Tocainide
1714
1769
—
44
121
77
120
42
106
Trandolapril-H2O
3090
—
—
262
308
91
124
117
223
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:31:40
Specific applications
691
Table 40.16 continued Compound
System
Principal ions (m/z)
GA
GB
GP
Trandolapril-Me
—
—
2970
Trandolapril-Me2
2995
234
91
235
160
371
130
248
249
91
174
144
385
M (perindoprilate)-Me3
—
—
3005
234
235
91
385
130
174
M (perindoprilate-H2O)-Me3
—
—
3070
294
262
91
398
117
223
Trimetazidine
—
—
—
181
85
56
166
266
182
Valsartan-Me2
—
—
3420
264
378
192
320
249
164
Verapamil
3150
3305
—
303
58
43
304
151
44
M (nor-)
3180
3371
—
289
151
290
152
165
260
M (N-desalkyl-)
2100
2193
—
44
164
203
247
290
57
M (O-desmethyl-didesalkyl-)
—
2169
—
216
233
164
185
276
203
M (O-desmethyl-desalkyl-)
—
2246
—
44
257
212
171
247
290
M (didesalkyl-)
—
2300
—
247
248
275
233
216
290
M
—
2409
—
202
189
230
247
290
203
M (N-desalkyl-acetyl-)
2460
2546
—
247
289
332
216
248
290
M (N-didesalkyl-acetyl-)
2545
2579
—
275
318
233
276
234
170
not amenable to GC without prior derivatisation. Maurer and Arlt (1998) describe an extractive methylation procedure from aqueous alkali (5 mL) using iodomethane in toluene (5 mL of 0.5 mol/L) and 150 mL of a phase-transfer reagent (tetrahexylammonium hydrogensulfate (THA), 4.5 g in 50 mL of 0.5 mol/L NaOH). Removal of excess THA was achieved by passing the toluene layer through a solid-phase extraction (SPE) cartridge and eluting with diethyl ether–ethyl acetate (92.5 : 7.5, v/v), which enables GC with MS. System GA, described
above, is used, and the retention indices and principal ions are given in Table 40.17. Anticoagulants of the heparin family (e.g. enoxaparin) are not included as these peptides cannot be chromatographed. Diuretics As a group, the diuretics chromatograph poorly on packed columns, and only marginally better on capillaries because of the presence of one or
Table 40.17 GC retention data and mass spectral data for coumarins and other anticoagulants (Me, methyl) Compound
System GA
Principal ions (m/z)
Acenocoumarol-Me
3035
324
325
367
189
121
278
M (amino)-Me3
2985
308
365
292
309
293
249
M (acetamido)-Me2
3265
350
351
393
278
56
394
M (OH-) isomer 1-Me2
3350
354
355
397
151
308
219 219
M (OH-) isomer 2-Me2
3500
354
355
397
151
308
Anisindione
2273
252
237
253
181
238
77
Coumachlor-Me
2770
313
315
356
128
189
201
M (OH-) isomer 1-Me2
2990
343
345
386
125
151
231
M (OH-) isomer 2-Me2
3035
343
345
386
125
151
231
M (OH-dihydro)-Me2
3095
388
343
329
245
125
151
M (OH-methoxy)-Me2
3195
373
375
416
372
359
125
M (di-OH)-Me3
3195
373
375
416
372
359
125
2635
292
188
130
128
293
187 189
Coumatetralyl Coumatetralyl isomer 1-Me
2655
306
175
291
115
121
Coumatetralyl isomer 2-Me
2690
306
291
175
115
202
91
M (OH-) isomer 1-Me
2910
203
303
304
187
121
322
M (OH-) isomer 2-Me2
2925
336
205
217
232
302
321
M (OH-) isomer 3-Me2
2935
321
320
336
175
319
305
M (OH-) isomer 4-Me2
2990
336
205
232
217
321
337
3235
—
—
—
—
—
—
Dicoumarol-Me Diphenadione
2934
173
167
340
165
89
Phenindione
2055
222
165
223
194
76
152 90
Phenprocoumon isomer 1-Me
2375
203
279
265
294
249
121
Phenprocoumon isomer 2-Me
2395
91
265
294
203
279
221 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:31:51
692
Gas Chromatography
Table 40.17 continued Compound
System GA
Principal ions (m/z)
M (OH-) isomer 1-Me2
2655
295
324
233
91
309
251
M (OH-) isomer 2-Me2
2675
295
296
324
121
279
201
M (OH-) isomer 3-Me2
2705
295
324
296
91
233
151
M (OH-methoxy)-Me2
2770
325
354
326
151
279
201 201
2770
325
354
326
151
279
Pyranocoumarin/cyclocumarol
2670
322
72
265
249
275
148
M (O-desmethyl-) Art-Me (warfarin-Me)
2580
279
280
322
91
121
189
M (O-desmethyl-OH-) isomer 1 Art-Me2
2810
309
310
352
91
277
151
M (O-desmethyl-OH-) isomer 2 Art-Me2
2830
309
121
201
295
352
310
M (O-desmethyl-OH-) isomer 3 Art-Me2
2870
309
310
352
91
295
206
M (di-OH)-Me3
Warfarin-ME
2580
279
280
322
91
121
189
M (OH-) isomer 1-Me2
2810
309
310
352
91
277
151
M (OH-) isomer 2-Me2
2830
309
121
201
295
352
310
M (OH-) isomer 3-Me2
2870
309
310
352
91
295
206
more sulfonamide (SO2–NH2) or carboxylic acid groups. However, they can be methylated easily, and then systems GA or GB, described above, may be used effectively. The diuretics can be extractively alkylated from aqueous alkali (5 mL) using iodomethane in toluene (5 mL of 0.5 mol/L) and 150 mL of a phase-transfer reagent. Tetrahexylammonium hydrogensulfate (4.5 g in 50 mL 0.5 mol/L NaOH) is far superior to tetrabutylammonium hydroxide (TBAH) and tetrapentylammonium hydroxide (TPAH) (Carreras et al. 1994). Removal of excess THA was achieved by passing the toluene layer through an SPE cartridge and eluting with diethyl ether–ethyl acetate (92.5 : 7.5, v/v). Alternatively, diuretics can be extracted from aqueous acidic solution into ethyl acetate, and the evaporated residue heated with 10% methyl iodide in acetone and 100 mg solid K2CO3 for 6 h at 60 C, and the resultant extract applied directly to the chromatograph. The former method tends to produce more completely substituted derivatives than the latter, which gives a more varied pattern of substituted derivatives. Although good sensitivity for plasma samples can be obtained with an NPD, the use of a mass spectrometer is required to confirm the identity (Lisi et al. 1991; Yoon et al. 1990).
System GA or GB, described above, can be used, as can systems GX and GY. System GX
The details are taken from Carreras et al. (1994). n n n n
Column: 5%-phenyl-PSX (X-5) (25 m 0.2 mm i.d., 0.33 mm). Temperature programme: 230 C to 320 C at 35 /min (drugs elute isothermally). Carrier gas: He, 1 mL/min. Retention indices and times (min) are given in Table 40.18.
System GY
The details are taken from Lisi et al. (1991). n n n n n
Column: methyl-PSX (X-1) (25 m 0.22 mm i.d., 0.1 mm). Temperature programme: 130 C to 320 C at 40 /min for 3 min. Injection: Split 10 : 1. Carrier gas: H2, 1 mL/min. Retention indices and times (min) are given in Table 40.18.
Table 40.18 GC retention data and mass spectral data for diuretics (Me, methyl) Compound
System GA
Principal ions (m/z) GX
GY
Acetazolamide-N-Me3
1827
3.62
2.90
249
83
108
43
264
265
Acetazolamide-O-Me
1930
—
2.69
70
44
129
236
237
—
—
13.20
—
352
354
244
42
145
—
Art-Me
1840
—
—
202
144
171
116
204
101
Art-Me2
1860
—
—
187
170
189
142
116
101
—
10.70
5.62
386
278
91
42
387
145
Althiazide-Me4
Bendrofluazide-Me4
Bendroflumethiazide-Me3 3344
12.00
—
386
278
42
387
388
—
Benzbromarone
—
—
264
173
424
115
279
423
2760
Benzbromarone-Me
—
2730
—
278
438
173
440
439
—
Benzthiazide
2680
—
—
309
91
311
123
176
121
Bumetanide-Me3
2970
7.71
4.90
254
318
363
406
77
196
Chlorothiazide
1720
—
—
295
268
297
270
64
124
Chlorothiazide-Me3
—
6.55
4.34
337
245
42
339
230
293
Chlortalidone
2145
—
—
148
130
76
321
299
300
Chlortalidone-Me4
2630
7.67
4.81
176
287
363
365
364
289
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:31:54
Specific applications
Table 40.18 continued Compound
System
Principal ions (m/z)
GA
GX
Clopamide-Me2
2805
6.97
GY —
111
112
127
55
139
Clopamide-Me3
2600
—
—
372
374
387
373
264
245
Art(-SO2NH)
2195
—
—
111
127
139
83
96
251
Cyclopenthiazide-Me4
—
—
6.33
352
354
233
42
145
435
358
Dichlorphenamide-Me4
—
5.15
3.94
44
108
253
255
144
360
Ethacrynic acid-Me
2195
4.02
3.33
261
263
243
245
281
316
Furosemide (frusemide)
Not eluted
—
—
81
300
53
96
82
332
Furosemide-Me
2890
—
—
81
344
346
96
329
311
Furosemide-Me2
2850
—
—
81
358
360
96
325
343
Furosemide-Me3
2800
6.95
4.65
81
372
358
374
339
312
Art (-SO2NH)
2040
—
—
81
53
251
96
253
233
Art (-SO2NH)-Me
2020
—
—
81
265
53
96
232
250
Art (-SO2NH)-Me2
2050
—
—
81
232
279
250
234
204
M (N-desalkyl-)-Me
2750
—
—
264
232
266
234
248
200
M (N-desalkyl-)-Me2
2450
—
—
278
200
280
248
185
169
Not eluted
—
—
269
297
271
221
268
188
9.01
4.99
310
353
218
288
355
202
2170
—
—
139
232
127
63
167
189
Not eluted
—
—
303
239
331
255
266
158
Hydroflumethiazide-Me4 2653
6.30
4.38
387
236
215
344
252
322
Indapamide-Me3
3035
9.01
—
161
132
131
407
409
130
Mefruside-Me2
2860
7.43
4.74
85
43
86
110
325
367
—
9.80
5.14
99
325
327
218
282
326
Methazolamide
2187
—
—
221
43
83
236
223
221
Methazolamide-Me2
—
—
2.90
249
264
43
108
83
265
Hydrochlorothiazide
Hydrochlorothiazide-Me4 2966 Art (-SO2NH)-Me Hydroflumethiazide
M (5-oxo-)-Me2
Methyclothiazide
Not eluted
—
—
310
312
42
311
230
359
Methyclothiazide-Me3
—
9.90
—
352
354
244
246
—
—
Metolazone-Me3
3910
—
6.23
392
394
393
284
118
407
Piretanide-Me3
2965
8.40
—
295
296
404
266
297
—
Polythiazide
2380
—
—
310
312
42
129
311
230 —
Polythiazide-Me3
2985
11.01
—
352
354
244
42
246
Probenecid
2336
—
—
256
185
121
224
257
65
Probenecid-Me
2205
3.90
3.23
270
135
199
271
228
299
Quinethazone
Not eluted
—
—
260
262
180
289
261
145
Quinethazone-Me3
—
—
5.05
316
208
318
42
173
317
Spironolactone
3280
—
—
341
340
374
342
267
359
M (canrenone)
3250
—
5.57
340
267
107
91
341
325
M (canrenoic acid)
3100
—
—
358
84
85
329
359
274
M (canrenoic acid)-Me 3130
—
—
354
355
339
340
356
173
Triamterene
2010
—
—
253
252
43
104
254
235
Triamterene-Me6
2875
9.15
—
336
338
322
308
293
309
Trichlormethiazide-Me4
2810
10.72
—
352
354
244
Trometamol
1645
—
—
—
—
—
42 —
—
—
—
—
Xipamide-Me2
3350
—
—
262
264
382
263
168
223
Xipamide-Me3 isomer 1
2800
8.72
—
396
276
365
395
397
398
Xipamide-Me3 isomer 2
3320
—
—
276
396
278
277
168
233
Xipamide-Me4
2780
—
—
410
290
379
409
411
412
Art (-SO2NH)
2385
—
—
121
155
275
157
106
99
Art (-SO2NH)-Me
2480
—
—
169
170
289
126
290
291
Art (-SO2NH)-Me2
2115
—
—
183
303
272
257
302
304
M (OH-)-Me4
3000
—
—
426
428
395
396
275
262
693
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:31:59
694
Gas Chromatography
Essential oils, flavours and fragrances
System GN
Essential oils, flavours and fragrances are complex mixtures of many components, so Table 40.19 is representative of the most common ones only. For a more extensive list, the reader is referred to specialist texts (Adams 1995). In addition, many small esters and ketones are contributory and can be detected via the system described in the section below on volatile substances. Essential oils, once thought innocuous, are now recognised as a potential cause of serious poisonings, especially in children, and are encountered in highly concentrated forms with increasing frequency in everyday use as vehicles for medicines, aromatherapy and handicraft supplies. Since most natural flavours occur in predominantly one enantiomeric form, and the majority of synthetic flavours are racemates, chiral analysis of enantiomeric proportions is an effective way to determine the authenticity of flavours. The catalogues of chromatography column suppliers show many examples of chiral separations; some examples are found on-line at www.restek.com. Systems GA and GB, described above, can be used, as can systems GN and GO (Supelco 2000).
n n n n
Column: Supelcowax 10 (Wax10) (10 m 0.25 mm i.d., 0.25 mm). Temperature programme: 50 for 2 min, to 280 at 2 /min Carrier gas: He, 25 cm/s. Retention times (min) are given in Table 40.19.
System GO n n n n
Column: SPB-5 (5%-phenyl-PSX, X-5) (30 m 0.25 mm i.d. 0.25 mm). Temperature programme: 75 for 8 min to 200 at 4 /min. Carrier gas: He, 25 cm/s. Retention times (min) are given in Table 40.19.
Narcotic analgesics, opiates and opioids Many laboratories perform specific assays for opiates for federal or legal purposes; these are generally limited to codeine, morphine and more recently 6-monoacetyl morphine (MAM) (Paul et al. 1999).
Table 40.19 GC retention data for oils, flavours and fragrances Compound
System GA
GB
GN (min)
GO (min)
Anethole
1284
1316
—
—
Camphene
—
—
Camphor
1143
—
Carene
—
—
Carvone
—
1275
—
18.8
Cedrol
—
—
64.3
—
Cineole
—
1063
—
11.2
Citronellal
1265
—
—
14.4
Citronellol
—
—
45.8
—
Eugenol
1368
1380
—
—
Geranial
—
—
—
19.4
Geraniol
1192
—
51.7
18.8
Ionine
—
—
50.4
—
Jasmone
—
—
56
—
Lavandulol
—
—
40.5
—
Limonene
1053
1063
11.0
8.9
Linalool
1100
—
32.4
12.0
Menthol
1206
1194
—
—
Menthone
—
—
26.1
—
Methyl salicylate
1195
1228
—
—
Myrcene
—
—
Neral
—
—
Nerol
—
—
48.0
17.6
Nerolidol
—
—
59, 61
—
Patchouli
—
1691
—
—
a-Pinene
—
—
4.9
5.4
b-Pinene
—
—
7.1
6.7
Piperonal
—
1373
—
Sabinine
—
—
—
6.5
a-Terpinene
—
—
—
8.4
g-Terpinene
—
—
18.4
10.2
Terpinen-4-ol
—
—
—
15.5
a-Terpineol
1126, 1176
—
41.5
16.1
Thymol
—
1316
—
—
Vanillin
1630
1632
86.5
—
5.9 — 8.6
9.6 —
5.9 — 8.3
7.1 18.1
—
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:34:6
Specific applications However, for clinical purposes a wider range of analytes is desirable and can include codeine, dihydrocodeine, hydrocodone, hydromorphone, oxycodone and oxymorphone. All assays involve a hydrolysis step (acidic or enzymatic – see earlier discussion for an evaluation of these) to cleave the glucuronide conjugates, followed by a basic extraction (often using solid-phase or acidic back extraction for cleanliness). Derivatisation is possible with a number of reagents
695
(PFP, TMS, TFA or AC derivatives are the most common) (Chen et al. 1990; Grinstead 1991; Maurer, Pfleger 1984), and retention data for some of these are included in Table 40.20. The derivatising reagent is selected on the basis of personal preference for a desired separation or the formation of unique ions on MS fragmentation. Analysis of hydromorphone, oxycodone and oxymorphone is complicated by the possibility that several structurally different
Table 40.20 GC retention data and mass spectral data for narcotic analgesics, opiates and opiods (AC, acetyl; HFB, heptafluorobutyrate; PFP, pentafluoropropionate; TFA, trifluoroacetyl; TMS, trimethylsilyl) Compound
System GA
Principal ions (m/z) GB
GC
GF
GM
Acetorphine (hydrolyses to etorphine) Acetylcodeine
2503
2645
—
—
1.449
341
282
342
229
204
240
Alfentanil
2970
3108
—
—
Not eluted
289
268
290
140
222
170
—
—
—
Alphameprodine
1850
1927
—
—
—
—
—
Alphaprodine
1792
1862
—
—
—
172
187
84
57
42
— 188
Anileridine
2850
—
3469
—
—
246
247
218
120
277
106
Apomorphine
2715
0000
—
—
Not eluted
266
267
224
220
152
248
Apomorphine-AC 2
2830
—
—
—
—
351
350
266
308
309
292
Apomorphine-TMS2
2715
—
—
—
—
410
411
322
73
368
412
Benzylmorphine
3015
—
—
—
—
284
91
81
375
285
175
Buprenorphine
3360
3610
—
—
Not eluted
378
410
379
435
434
449
Buprenorphine-HFB
2960
—
—
—
—
574
606
575
562
548
607
Buprenorphine-HFB2
2820
—
—
—
—
55
562
83
630
646
604
Buprenorphine-PFP
3040
—
—
—
—
524
556
525
512
498
580
Buprenorphine-PFP2
2775
—
—
—
—
55
512
580
554
513
595
Buprenorphine-TFA
2920
—
—
—
—
55
474
506
475
448
507
Buprenorphine-TFA2
2800
—
—
—
—
55
462
530
463
504
546
Buprenorphine-TMS
3890
—
—
—
—
450
451
482
506
493
424
Art (-H2O)
3240
—
—
—
—
449
434
408
419
435
450
Art (-H2O)-AC
3320
—
—
—
—
491
476
450
434
477
492
Buprenorphine-AC
3410
—
—
—
—
420
452
421
408
394
509
Butorphanol
2761
2902
—
—
—
272
273
411
254
157
327
Butorphanol-TMS
—
2832
—
—
—
344
345
271
326
399
384
Butorphanol-TMS 2
—
2851
—
—
—
416
417
326
270
456
471
Cetobemidone
2045
—
—
—
—
70
71
190
247
119
57
Cetobemidone-AC
2095
—
—
—
—
70
71
232
289
190
247
Cetobemidone-HFB
1915
—
—
—
—
70
71
69
128
96
115
Cetobemidone-PFP
1865
—
—
—
—
70
57
128
336
393
129
Cetobemidone-TFA
1925
—
—
—
—
70
71
69
286
128
129
Cetobemidone-TMS
2070
—
—
—
—
70
71
262
319
191
304
M (nor-)-AC 2
2545
—
—
—
—
261
58
70
218
160
219
M (methoxy-)-AC
2265
—
—
—
—
70
71
188
319
220
262
2375
2511
2681
2860
1.519
299
162
229
300
124
59
Codeine (also metabolised to morphine, O-desmethylcodeine) Codeine-AC
2503
2645
—
—
1.449
341
282
229
342
204
298
Codeine-HFB
2320
—
—
—
—
282
283
169
225
495
266
Codeine-PFP
2430
—
—
—
—
282
445
446
388
266
283
Codeine-TFA
2280
—
—
—
—
282
395
283
225
266
396
Codeine-TMS
2520
2592
—
—
—
371
178
196
234
343
229
M (nor-)
2388
2535
—
—
—
285
215
148
164
200
242 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:34:22
696
Gas Chromatography
Table 40.20 continued Compound
System
Principal ions (m/z)
GA
GB
GC
GF
GM
M (nor)-AC
2945
—
—
—
—
87
223
224
369
209
195
M (nor)-PFP2
2440
—
—
—
—
563
355
209
400
327
387
M (nor)-TMS2
—
2631
—
—
—
429
254
250
292
284
414
2025
2104
—
—
0.514
194
98
165
167
207
208
M (nor-)
2120
2128
—
—
0.610
167
165
207
85
152
252
M (benzophenone) (BP)
1610
1673
—
—
—
105
77
51
182
106
183
M (carbinol)
1750
—
—
—
—
105
77
139
141
165
218
M (OH-BP) isomer 1
2065
—
—
—
—
198
121
77
199
192
151
M (OH-BP) isomer 2
2080
—
—
—
—
121
198
77
105
199
122
M (OH-methoxy-BP) isomer 1
2050
—
—
—
—
105
151
198
228
77
121
M (OH-methoxy-BP) isomer 2
2070
—
—
—
—
105
151
198
228
77
121
2940
3094
3625
—
—
100
128
265
56
165
266
M (OH)
3095
3310
—
—
—
100
128
281
165
194
322
M (OH)-AC
3210
—
—
—
—
100
128
194
323
325
365
M (methoxy-)
3269
—
—
—
—
100
128
194
323
422
423
2188
2268
2173
2370
1.220
58
91
105
115
59
208
M (nor-)
2214
2487
—
—
1.248
44
220
100
205
129
307
M (nor-amide)
2526
2673
—
—
1.969
234
100
105
220
129
94
M (nor-N-proponyl-)
2400
2514
—
—
1.300
44
220
100
205
129
91
M
—
2520
—
—
1.250
205
220
91
126
115
160
M
—
2526
—
—
1.255
205
220
91
126
115
160
M
—
2624
—
—
—
220
91
147
105
135
115
M (nor-)-AC
2365
—
—
—
—
220
205
86
293
129
191
Art
1621
1659
—
—
—
115
208
91
193
130
117
Art
—
1756
—
—
—
115
91
208
193
117
Art
1890
1957
—
—
—
58
91
191
178
130 ̀ 128
Art
—
1987
—
—
—
44
91
178
191
115
129
Art
—
2021
—
—
—
44
178
91
115
129
191
2230
2323
2230
—
—
257
59
150
256
200
157
Cyclizine
Dextromethorphan (see Methorphan) Dextromoramide
Dextropropoxyphene
Dextrorphan
115
Dextrorphan-AC
2280
—
—
—
—
59
150
299
198
231
256
Dextrorphan-PFP
2060
—
—
—
—
150
403
335
402
119
346
Dextrorphan-TFA
2015
—
—
—
—
150
285
353
352
128
296
Dextrorphan-TMS
2230
—
—
—
—
59
150
329
272
328
314
M (nor-)
2241
2328
—
—
—
243
157
136
198
200
242
M (nor)-AC2
2710
—
—
—
—
87
72
198
211
327
285
M (OH)-AC2
2555
—
—
—
—
357
231
356
355
298
315
2615
2769
—
—
—
327
369
310
268
204
215
M (6-MAM)
2525
2646
—
—
—
327
268
215
328
285
310
M (3-MAM)
2495
2625
—
—
—
327
285
162
215
268
310
M (6-MAM)-PFP
2650
—
—
—
—
414
473
361
204
430
454
M (3-MAM)-PFP
2490
—
—
—
—
268
310
431
473
267
211
M (6-MAM)-HFB
2425
—
—
—
—
464
465
480
677
407
411
M (6-MAM)-TMS
2590
2688
—
—
—
399
340
287
204
282
266
M (3-MAM)-TMS
2570
2668
—
—
—
399
357
234
196
164
329
Diethylthiambutene
2008
—
—
—
—
276
111
219
42
277
97
Dihydrocodeine (metabolised to dihydromorphine, hydrocodone and hydromorphone)
2390
2511
2702
2840
1.493
301
164
59
300
301
115
Diamorphine (heroin; metabolised to MAM, morphine and codeine)
Dihydrocodeine-AC
2445
—
—
—
—
343
300
284
344
226
328
Dihydrocodeine-HFB
2315
—
—
—
—
497
284
498
300
185
169
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:34:48
Specific applications
697
Table 40.20 continued Compound
System
Principal ions (m/z)
GA
GB
GC
GF
Dihydrocodeine-PFP
2360
—
—
—
GM —
447
448
284
300
392
432
Dihydrocodeine-TFA
2265
—
—
—
—
397
284
185
300
340
382
Dihydrocodeine-TMS
2480
2496
—
—
—
373
236
282
315
146
178
M (nor-)
—
2599
—
—
—
287
150
242
213
176
132
M (nor)-AC2
2750
—
—
—
—
243
371
225
224
285
285
M (nor)-TMS2
—
2559
—
—
—
431
316
226
294
416
340
Dihydromorphine
2400
2527
2504
—
—
287
70
164
286
230
288
Dihydromorphine-AC2
2545
—
—
—
—
329
371
286
270
212
310
Dihydromorphine-PFP2
2330
—
—
—
—
119
579
416
432
359
560
Dihydromorphine-TMS2
2520
2518
—
—
—
431
236
146
416
373
326
M (nor)-AC3
2790
—
—
—
—
357
399
229
211
272
315
Diphenoxylate
3514
3670
—
—
Not eluted
246
377
193
165
452
184
Dipipanone
2474
2586
2894
2710
1.309
112
113
91
165
334
223
Diprenorphine
—
3385
—
—
Not eluted
—
—
—
—
—
—
Ethoheptazine
1857
1923
1630
2110
—
—
—
—
—
—
—
Ethylmorphine (metabolised to morphine, see below)
2411
2530
—
—
—
—
—
—
—
—
—
Ethylmorphine-AC
2530
—
—
—
—
355
296
327
234
268
204
Ethylmorphine-PFP
2430
—
—
—
—
296
459
280
266
402
430
Ethylmorphine-TFA
2320
—
—
—
—
296
409
380
280
352
266
Ethylmorphine-TMS
2540
—
—
—
—
385
192
146
196
234
357
2930
—
—
—
—
87
209
237
383
341
181
3033
3211
—
—
Not eluted
44
215
411
324
164
216
—
—
—
—
272
396
250
162
354
483
2720
2833
—
—
—
146
245
189
105
202
158
M (nor-)
—
—
—
—
—
—
—
—
—
—
—
M (despropionyl)
—
—
—
—
—
146
189
44
118
132
280
Hydrocodone (also metabolised to hydromorphone, dihydromorphine and dihydrocodeine)
2440
2580
3028
2930
—
299
242
243
96
185
214
Hydrocodone-TMS
387
M (nor)-AC 2 Etorphine Etorphine-TMS Fentanyl
—
2674
—
—
—
297
386
371
329
298
M (nor)
—
2599
—
—
—
285
242
115
214
128
185
M (nor)-AC
2760
—
—
—
—
87
241
327
212
285
228
Hydromorphone (metabolised to dihydromorphine)
2445
2598
—
—
—
285
96
228
229
286
128
Hydromorphone-AC
2595
—
—
—
—
285
327
228
229
214
242
Hydromorphone-enol-AC2
2625
—
—
—
—
327
284
162
228
369
270
Hydromorphone-HFB
2385
—
—
—
—
481
425
424
410
482
452
Hydromorphone-HFB2
2325
—
—
—
—
481
425
424
410
482
452
Hydromorphone-PFP
2250
—
—
—
—
431
375
374
360
346
402
Hydromorphone-enol-PFP2
2320
—
—
—
—
430
308
414
577
372
520
Hydromorphone-enol-TFA2
2230
—
—
—
—
477
473
380
364
258
458
Hydromorphone-TMS
—
2621
—
—
—
357
300
243
342
314
286
Hydromorphone-enol-TMS2
—
2595
—
—
—
429
414
234
184
357
324
Hydromorphone oxime TMS2
—
2678
—
—
—
355
444
429
356
339
372
Ketamine
1840
1939
—
—
0.427
180
182
209
152
138
102
M (nor-)
1810
1907
—
—
0.423
166
168
195
131
138
223
M (nor-OH)-H2O
1960
2058
—
—
—
166
221
168
193
131
138
M (nor-OH)-NH3
1740
1840
—
—
—
187
222
117
159
131
224 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:34:59
698
Gas Chromatography
Table 40.20 continued Compound
System
Principal ions (m/z)
GA
GB
GC
GF
M (nor-di-OH)-2H2O
1920
2009
—
—
GM —
190
219
192
221
156
184
M (nor)-H2O
—
1931
—
—
—
153
138
221
118
155
192
Ketobemidone
2040
—
—
—
—
70
71
42
44
57
190
Levallorphan (metabolised to nordextrorphan)
2355
2460
—
—
—
283
157
282
176
256
84
Levallorphan-AC
2390
—
—
—
—
85
325
298
176
157
257
Levomethadyl acetate (LAAM)
—
2267
—
—
—
72
43
73
91
255
165
M (nor-)
—
2262
—
—
—
58
101
100
208
281
165
M (di-nor-)
—
2255
—
—
—
44
120
193
165
178
208
1910
—
—
—
—
243
67
95
223
245
97
Levopropoxyphene (see Dextropropoxyphene) Levorphanol (see Dextrorphan) Lofexidine Lofexidine-AC
2200
—
—
—
—
86
139
257
265
223
243
Meptazinol
1920
1980
—
—
0.429
58
84
98
71
85
233
M (nor-)
1995
2069
—
—
0.428
70
84
219
107
159
91
M (oxo-)
2410
2600
—
—
—
148
147
247
204
176
133
Methadone
2145
2228
2470
2370
0.606
72
73
294
57
223
91
Methadone-TMS
2260
—
—
—
—
72
73
296
85
165
178
2040
2120
—
—
0.520
277
276
262
220
165
115
M (dinor-;2-ethylidene-1-methyl-3,3- 2021 diphenylpyrrolidine, EMDP)
2069
—
—
—
262
263
248
221
186
165
M (nor-)
2095
—
—
—
—
58
72
224
165
115
178
M (methadol)
2185
—
—
—
—
72
91
165
105
253
193
M (normethadol)
—
—
—
—
—
58
91
115
165
178
193
2138
2237
—
—
—
59
150
271
270
214
171
2193
2244
—
—
—
257
212
171
136
213
214
M (2-ethylidene-1,5-dimethyl-3,3diphenylpyrrolidine, EDDP)
Methorphan (dextromethorphan/ racemethorphan/levomethorphan; also metabolised to dextrorphan see above) M (nor-methorphan/norracemethorphan) M (OH-)
—
2420
—
—
—
287
59
230
150
187
228
M (OH)-AC2
2555
—
—
—
—
357
231
356
355
298
315
2775
—
—
—
—
259
260
203
146
91
110
—
—
—
—
—
58
203
146
110
91
118
2445
2564
2542
—
Not eluted
285
162
215
284
124
268
Methylfentanyl (3- or a-) (china white) M (despropionyl-) Morphine Morphine-AC2 (diamorphine)
2615
2769
—
—
—
327
369
268
310
195
162
Morphine-PFP2
2360
—
—
—
—
414
577
415
578
558
430
Morphine-TMS2
2560
2602
—
—
—
429
236
357
414
401
196
Morphine-TFA2
2250
—
—
—
—
364
477
478
365
380
458
M (nor-)
2459
—
—
—
—
271
150
201
148
162
81
M (nor)-AC3
2955
—
—
—
—
87
209
210
355
397
181
M (nor)-PFP2
2440
—
—
—
—
563
355
387
373
400
544
M (nor)-PFP3
2405
—
—
—
—
355
709
367
382
533
546
M (nor)-TMS3
2605
—
—
—
—
222
416
487
472
192
355
Nalbuphine
2960
Not eluted
—
—
—
302
303
357
284
272
254
Nalbuphene-AC2
3110
—
—
—
—
386
387
441
344
296
326
Nalbuphene-AC3
3080
—
—
—
—
428
429
368
483
326
440
Nalbuphene-AC
3030
—
—
—
—
344
345
399
326
302
—
—
3093
—
—
—
573
518
428
468
574
410
M (N-desalkyl-)
2930
—
—
—
—
289
272
271
202
115
242
M (N-desalkyl)-AC2
2970
—
—
—
—
87
331
227
373
228
313
M (N-desalkyl)-AC3
3020
—
—
—
—
87
373
227
228
296
415
Nalbuphene-TMS3
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:35:16
Specific applications
699
Table 40.20 continued Compound
System
Principal ions (m/z)
GA
GB
GC
GF
Nalmefene
—
—
—
—
GM —
55
339
110
149
82
298
Nalorphine
2620
Not eluted
—
—
—
311
312
310
188
241
294
Nalorphine-AC2
2820
—
—
—
—
353
395
226
294
354
396
Nalorphine-AC
2800
—
—
—
—
353
294
354
241
310
230
Nalorphine-TFA
2403
—
—
—
—
—
—
—
—
—
—
Nalorphine-TMS2
—
2738
—
—
—
455
414
324
260
438
350
Naloxone
2715
Not eluted
—
—
—
327
328
242
96
286
229
Naloxone-AC
2840
—
—
—
—
327
369
328
286
244
310
Naloxone-AC 2
2750
—
—
—
—
369
411
285
310
326
352
Naloxone-enol-AC2
2810
—
—
—
—
411
369
330
270
228
244
Naloxone-enol-AC3
2770
—
—
—
—
327
369
328
411
286
453
Naloxone-enol-TMS3
2645
2787
—
—
—
438
528
543
355
371
461
Naloxone-enol-TMS2
2700
2843
—
—
—
471
456
366
390
229
398
Naloxone-TMS2
2680
2881
—
—
—
399
471
456
314
358
384
Naloxone-oxime-TMS3
—
2892
—
—
—
558
379
313
453
543
469
Naloxone-PFP
2530
—
—
—
—
473
388
432
375
348
446
Naloxone-enol-PFP2
2360
—
—
—
—
619
472
456
428
620
592
Naloxone-enol-PFP3
2270
—
—
—
—
765
602
618
738
519
454
Naloxone-PFP2
2470
—
—
—
—
82
119
619
472
592
456
M (dihydro)-AC2
2820
—
—
—
—
82
83
413
172
214
371
M (dihydro)-AC3
2855
—
—
—
—
82
413
455
327
254
372
Naltrexol
—
3033
—
—
—
343
55
302
110
98
288
Naltrexone
2880
Not eluted
—
—
—
341
55
300
342
110
243
Naltrexone-AC
2980
—
—
—
—
341
383
342
243
300
286
Naltrexone-AC 2
2870
—
—
—
—
383
425
341
324
340
366
Naltrexone-enol-AC2
3060
—
—
—
—
425
383
384
342
286
382
Naltrexone-enol-AC3
2960
—
—
—
—
425
467
408
382
366
324
Naltrexone-TMS2
—
3051
—
—
—
485
486
470
388
412
444
Naltrexone-TMS3
—
2975
—
—
—
540
555
500
288
272
450
Naltrexone-enol-TMS3
—
2945
—
—
—
557
542
355
242
484
452
Naltrexone-oxime-TMS3
—
3071
—
—
—
572
573
475
327
557
499
M (methoxy-)
2920
—
—
—
—
371
286
274
330
356
316
M (methoxy)-AC
3150
—
—
—
—
413
274
372
371
328
358
M (methoxy)-AC2
3130
—
—
—
—
455
412
396
456
413
273
M (methoxy)-enol-AC2
3300
—
—
—
—
455
414
456
384
400
440
M (methoxy)-enol-AC3
3180
—
—
—
—
497
454
498
396
440
412
413
427
469
384
370
426
58
179
180
225
178
165
—
2990
—
—
—
—
2035
2106
—
—
0.586
M (nor-)
—
2116
—
—
—
—
—
—
—
—
M (nor)-AC
2080
—
—
—
—
208
87
194
179
165
281
M (p-OH-)
—
2266
—
—
—
58
195
165
178
210
241
M (OH)-AC isomer 1
2250
—
—
—
—
195
194
238
165
224
311
M (OH)-AC isomer 2
2285
—
—
—
—
178
195
208
179
268
311
M (nor di-OH-)
—
2649
—
—
—
—
—
—
—
—
—
M (nor di-OH)-AC2 isomer 1
2610
—
—
—
—
87
266
337
295
224
252 178
M (dihydro)-AC3 Nefopam
2640
—
—
—
—
87
337
195
295
209
Neopine
2395
2532
—
—
—
299
162
229
123
59
42
Normethadone
2095
—
—
—
—
58
72
59
224
178
165
M (nor di-OH)-AC2 isomer 2
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:35:22
700
Gas Chromatography
Table 40.20 continued Compound
System
Principal ions (m/z)
GA
GB
GC
GF
GM
Norpipanone
2488
—
—
—
—
98
111
99
112
165
Noscapine
3145
3358
—
—
—
220
221
205
147
42
118
Oxycodone (metabolised to oxymorphone)
2524
2671
—
—
—
315
230
258
70
201
316 212
178
Oxycodone-AC
2555
—
—
—
—
357
314
298
240
230
Oxycodone-enol-AC2
2560
—
—
—
—
399
357
240
314
296
298
Oxycodone-TFA
2290
—
—
—
—
411
314
240
298
396
254
Oxycodone-TMS
—
2703
—
—
—
387
229
230
388
372
214
Oxycodone-enol-TMS2
—
2602
—
—
—
459
460
444
312
297
242
Oxycodone-oxime-TMS2
—
2740
—
—
—
474
229
214
459
295
385
M (nor-)
—
2703
—
—
—
313
187
314
115
214
229
M (nor)-enol-AC3
2680
—
—
—
—
385
427
343
281
326
368
M (nor)-TMS2
—
2763
—
—
—
373
445
288
258
226
240
M (nor)-enol-TMS2
—
2621
—
—
—
445
446
312
430
354
288
M (nor)-enol-TMS3
—
2746
—
—
—
517
518
502
312
342
428
M (dihydro-)
—
2666
—
—
—
317
230
115
242
260
216
M (dihydro)-AC2
2570
—
—
—
—
359
401
242
230
224
282
M (nor-dihydro)-AC3
2935
—
—
—
—
242
387
224
343
284
429
M (nor-dihydro)-AC2
2900
—
—
—
—
343
258
201
239
242
387
Oxymorphone
2538
2723
—
—
—
301
216
44
42
70
302
Oxymorphone-TMS
—
2715
—
—
—
373
288
259
374
316
358
Oxymorphone-TMS2
—
2728
—
—
—
445
446
430
287
331
372
Oxymorphone-TMS3
—
2641
—
—
—
517
502
355
412
518
503
Oxymorphone-oxime-TMS3
—
2748
—
—
—
532
533
517
287
443
459
M (nor-)
—
Not eluted
—
—
—
—
—
—
—
—
—
M (nor)-enol-TMS2
—
2788
—
—
—
431
259
316
346
432
416
M (nor)-enol-TMS3
—
2662
—
—
—
503
488
355
398
504
308
M (nor)-enol-TMS4
—
2773
—
—
—
575
503
355
560
486
242
M (dihydro-)
—
2690
—
—
—
303
286
115
216
315
256
2825
2973
—
—
—
338
324
339
340
308
325
2805
—
—
—
—
324
310
325
294
266
309
Pentazocine
2280
2356
2225
3030
0.870
70
217
110
69
285
202
Pentazocine-AC
2330
—
—
—
—
259
110
327
312
244
217
Pentazocine-PFP
2120
—
—
—
—
363
348
110
416
431
430
Papaverine M (O-desmethyl-)
Pentazocine-TFA
2075
—
—
—
—
69
313
110
298
366
381
Pentazocine-TMS
2320
—
—
—
—
289
244
245
274
342
357
M (desalkyl-)
—
2019
—
—
—
—
—
—
—
—
—
M (desalkyl)-AC 2
2380
—
—
—
—
87
88
301
172
217
218
202
268
301
110
71
70
172
247
246
218
72
73
2545
2649
—
—
—
1754
2025
1995
1809
0.319
M (nor-)
1885
1842
—
—
0.357
57
233
158
103
131
117
M (nor)-AC
2240
2256
—
—
—
187
57
275
158
232
202
M (OH-)
2045
2145
—
—
—
71
140
263
262
189
234
M (OH)-AC
2205
—
—
—
—
71
305
188
230
261
276
M (nor-OH)-AC2
2600
—
—
—
—
203
56
245
333
218
290
M (OH-) Pethidine (meperidine)
Phenazocine
2686
2833
—
—
—
230
231
58
105
158
173
Phenoperidine
2872
2983
—
—
—
246
247
367
91
158
172
Pholcodine
3070
3348
—
—
—
114
100
42
56
398
115
Pholcodine-AC
3260
—
—
—
—
114
100
56
70
115
440
Pholcodine-PFP
2980
—
—
—
—
114
100
277
354
380
544
Pholcodine-TFA
2800
—
—
—
—
114
100
277
354
380
494
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:35:25
Specific applications
701
Table 40.20 continued Compound
System
Principal ions (m/z)
GA
GB
GC
GF
GM
Pholcodine-TMS
3140
3410
—
—
—
114
100
115
470
196
356
M (nor)-PFP2
3010
—
—
—
—
100
114
380
70
513
676
M (nor)-TMS2
3260
—
—
—
—
114
100
73
468
528
456
M (nor)-PFP
3270
—
—
—
—
100
114
56
530
502
— 181
M (nor)-AC
3620
—
—
—
—
100
114
70
426
340
M (nor)-AC2
3650
—
—
—
—
100
114
56
468
382
70
Piminodine
2884
—
—
—
—
246
366
106
234
247
260
Pipazethate (pipazetate)
200
2037
—
—
—
—
98
111
99
199
288
M (alcohol)
1830
—
—
—
—
98
112
99
156
103
84
M (ring sulfone)
2720
—
—
—
—
232
200
168
184
156
140
—
—
—
2800
—
—
—
—
—
—
—
Piritramide
3560
—
—
—
—
386
138
387
Profadol
1748
—
—
—
—
—
—
—
—
—
—
2533
2559
—
—
—
341
298
299
242
284
162
M (OH-ring)
84
110
42
Propoxyphene (see Dextropropoxyphene) Racemorphan (see Methorphan) Thebacon (metabolised to dihydrocodeine) Thebacon-TMS
2475
—
—
—
—
371
234
356
184
370
313
Thebaine
2517
2672
—
—
—
311
296
312
297
242
139
Tilidine (tilidate)
1838
—
—
—
—
97
82
103
77
132
176
M (nor-)
1827
—
—
—
—
83
68
259
184
157
214
M (nor)-AC
2165
—
—
—
—
125
83
111
155
170
258
M (bis-nor-)
1830 — —
—
—
—
69
83
119
135
170
245
M (bis-nor)-AC
2100
—
—
—
—
69
111
155
170
244
287
1943
2021
—
—
—
58
263
135
77
264
92
M (nor-)
—
2049
—
—
—
44
188
249
135
150
159
M (nor)-AC
2295
—
—
—
—
86
200
58
273
172
184
M (nor-) carbamate Art
—
2065
—
—
—
189
121
135
202
261
188
M (O-desmethyl-)
1995
2093
—
—
—
58
249
121
93
107
131
M (O-desmethyl)-AC
1998
—
—
—
—
58
121
248
163
291
128
M (N,O-didesmethyl-)
—
2122
—
—
—
44
174
235
121
145
159
M (N,O-didesmethyl)-AC2
2464
—
—
—
—
86
186
301
228
107
113
Tramadol
M (didesmethyl-) carbamate Art
2148
—
—
—
—
73
173
174
188
145
247
M (OH-)
2200
2252
—
—
—
58
279
135
77
234
261
Trimeperidine
1808
1895
—
—
—
186
201
187
70
105
91
derivatives will form in non-reproducible proportions from the automerisation of the enol and keto forms. However, these compounds can be stabilised in their keto forms by incubating with hydroxylamine or methoxyamine–pyridine, and then yield only a single derivatised oxime product (Broussard et al. 1997; Meatherall 1999). Systems GA, GB, GC or GF, described previously may be used. For plasma, system GM, described previously, is a rapid isothermal packedcolumn method with good sensitivity on NPD. Table 40.20 gives retention indices, or relative retention times to iprindole for GF. Non-amfetamine stimulants and hallucinogens Non-amfetamine stimulants and hallucinogens have a variety of clinical and toxic actions. Extraction of cocaine is straightforward under basic conditions, and most metabolites, except benzoylecgonine, can be detected in the clinical setting without derivatisation. For
regulated testing, quantification of benzoylecgonine is required, and most laboratories use TMS as the derivatising reagent. As a result of the recent interest in other metabolites that may have clinical importance, data for these and their TMS derivatives are also included in Table 40.21. For analysis of cannabis metabolites, hydrolysis of conjugates with 10 mol/L potassium hydroxide is usually performed on urine prior to weakly acidic extraction (pH 6.5); TMS is the derivative of choice. Phencylidine (PCP) analysis is complicated by the low concentration present, although extraction is straightforward and derivatisation is required only for metabolite measurement (Nakahara et al. 1997). Chromatographic confirmation of lysergide (LSD) is hampered by the low concentrations and acidic nature of the metabolites, which necessitates both derivatisation (TMS) and tandem MS (Nelson, Foltz 1992). Systems GA or GB may be used. Table 40.21 gives the retention indices (reference compounds are the alkanes with an even number of carbon atoms).
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:35:40
702
Gas Chromatography
Table 40.21 GC retention data and mass spectral data for non-amfetamine stimulants and hallucinogens (AC, acetyl; PFP, pentafluoropropionate; TFA, trifluoroacetyl; TMS, trimethylsilyl) Compound
System GA
Principal ions (m/z) GB
Amiphenazole
2170
—
191
121
77
104
122
43
Bemegride
1367
—
55
83
82
113
70
69
Bufotenine
2057
—
58
204
146
59
160
42
Caffeine
1800
1904
94
109
55
67
82
195
M (1-nor-, theobromine)
1807
1920
180
55
67
109
82
137
M (7-nor-, theophylline)
1925
1990
180
95
68
53
181
96
Cannabidiol
2390
2480
231
232
245
174
314
187
Cannabidiol-TMS2
2330
2510
390
337
301
351
319
324
Cannabigerol
2500
—
193
123
231
316
247
136
Cannabigerol-TMS2
2440
2520
—
—
—
—
—
—
Cannabinol
2535
2644
295
296
310
238
251
223
Cannabinol-TMS
2485
2600
367
368
382
310
295
238
D9-Tetrahydrocannabinol (D9-THC)
2473
2578
299
314
231
271
243
258
D9-THC-TMS
2405
2499
386
371
315
303
343
330
2710
—
—
—
—
—
—
—
M (8a,11-di-OH-D9-THC)-TMS 9
M (8a,OH-D -THC)
2775
2975
271
295
297
311
312
214
M (8a,OH-D9-THC)-TMS
2580
—
—
—
—
—
—
—
M (11-OH-D9-THC)
2775
2975
299
300
330
217
231
193
M (11-OH-D9-THC)-TMS2
2630
2762
371
372
373
474
459
403
M (11-nor-D9-THC-9-carboxylic acid)
—
—
325
268
326
340
281
253
M (11-nor-D9-THC-9-carboxylic acid)-TMS2
2660
2820
371
473
488
372
398
417
Cinnamoylcocaine isomer 1
2345
2489
82
182
96
131
238
329
Cinnamoylcocaine isomer 2
2450
2625
82
182
96
131
238
329
Cocaine
2187
2289
82
182
94
77
83
303 185
M (ecgonine)
—
0000
82
96
83
97
124
M (ecgonine)-TMS2
1680
—
82
83
96
97
314
329
M (anhydroecgonine methyl ester)methylecgonidine
1280
1430
152
181
82
122
166
138
M (anhydroecgonine methyl ester)-TMS
1345
1472
210
239
224
211
122
183
M (norecgonine)
—
1472
82
156
126
96
171
116
M (methylecgonine)
1472
1530
82
96
83
199
168
182
M (methylecgonine)-TMS
1580
1585
82
96
212
271
182
240
M (ethylecgonine)
—
1602
82
96
83
97
168
213
M (ethylecgonine)-TMS
1485
1651
82
96
83
240
285
196
M (nor-)
2162
2259
168
136
105
77
68
289
M (nor-)-TMS
—
2378
105
240
140
152
179
346
M (benzoylecgonine)
2570
2663
124
168
82
105
94
289
M (benzoylecgonine)-TMS
2285
2365
82
240
105
122
256
361
M (benzoylnorecgonine)-TMS2
—
2400
404
140
298
—
—
—
M (m-OH-benzoylecgonine)-TMS2
2505
2600
82
240
193
210
256
449
M (p-OH-benzoylecgonine)-TMS2
—
2650
82
240
193
210
256
449
M (m-OH-)
2460
2608
82
182
94
121
198
319
M (m-OH-)-TMS
—
2550
182
82
94
193
391
198
M (p-OH-)
—
2650
82
182
94
121
198
319
M (p-OH-)-TMS
—
2610
182
82
94
193
391
198
M (OH-methoxy-)
2670
2729
82
182
151
349
18
168 223
M (OH-methoxy-)-TMS
2850
—
82
182
83
421
198
M (OH-dimethoxy-)-TMS
2970
—
82
182
83
94
451
240
M (cocaethylene)
2250
2345
196
82
317
272
94
105
M (norcocaethylene)
2115
2317
182
68
136
105
108
303
M (norcocaethylene)-TMS
—
2385
254
140
360
—
—
—
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:35:46
Specific applications
703
Table 40.21 continued Compound
System
Principal ions (m/z)
GA
GB
M (OH-cocaethylene)
—
2709
82
196
94
121
333
M (OH-methoxy-cocaethylene)
—
2779
82
196
151
212
318
363
M (methoxy-cocaethylene)
—
2663
82
196
121
94
267
333
1230
—
58
126
59
69
56
44
Cyclopentamine
288
Cyclopentamine-AC
1680
—
58
100
168
126
104
183
Ethamivan
1900
—
151
72
223
123
222
152
Ethamivan-AC
1970
—
151
222
223
194
195
265
Harmaline (makes harmine on heating)
2430
—
213
214
170
198
169
115
Harmaline-TFA
2525
—
241
310
121
169
184
198
Harmaline-PFP
2540
—
241
360
242
121
198
184 115
Harmaline-AC
2670
—
213
256
214
170
186
Harmaline-AC2
2800
—
255
298
256
241
212
141
Harmine
2291
2322
212
169
197
213
106
211
Harmine-AC
2545
—
212
254
197
169
213
140
2550
—
198
197
170
99
75
199
M (O-desmethyl-)
2600
—
198
240
169
197
199
115
Isometheptene
1052
—
58
55
128
44
59
56
Isoprenaline
1730
—
72
44
124
123
193
70
Isoprenaline-AC4
2460
—
84
193
235
277
319
365
Lobeline
1820
—
96
105
77
97
216
218
Lysergamide
—
—
267
221
207
180
223
154
Lysergic acid
—
—
268
224
154
180
207
223
Lysergide (lysergic acid diethylamide, LSD)
3445
3332
323
221
207
181
196
280 337
M (O-desmethyl)-AC
3595
—
395
253
293
268
279
M (nor-)
—
—
207
309
182
280
128
100
M (nor-)-TMS1
3705
—
381
279
254
100
265
205
LSD-TMS
M (nor-)-TMS2
3515
—
453
253
351
279
326
337
M (2-oxo-)
—
—
—
—
—
—
—
—
3430
—
309
499
235
325
397
409
Iso-LSD-TMS
3515
—
395
293
279
253
268
337
Mazindol
2325
2504
266
268
267
255
231
102
Mazindol-AC
2705
—
256
255
254
326
220
284
Meclofenoxate
1770
—
58
111
71
75
141
113
M (chlorophenoxyacetic acid)
1770
—
186
141
111
128
113
99
M (N-acetyl)
2160
—
194
179
181
253
151
148
Methoxamine
1726
1596
168
137
44
139
152
124
Myristic acid
1755
Not eluted
73
60
57
129
185
228
Myristic acid-Me
1715
—
74
87
143
129
199
242
Myristic acid-TMS
2280
—
73
117
285
149
101
300
M (2-oxo-3-OH-)-TMS2
Myristicin
1400
—
192
91
119
165
161
147
Naphazoline
2100
—
209
210
141
115
153
208
Nicotine
1350
1380
84
133
42
162
161
105
1715
1645
98
176
118
119
58
147
M (cotinine) Nikethamide
1525
1569
106
78
177
51
178
107
M (N-ethylnicotinamide)
1605
—
106
150
78
149
51
135
M (nicotinamide)
1341
1418
122
78
106
51
50
52
Oxymetazoline
2170
2254
245
260
44
217
218
246
Oxymetazoline-AC2
2760
—
302
344
287
320
203
245
Pemoline
1969
2081
107
176
90
77
70
105
Pemoline-Me2
1590
—
118
204
90
105
77
70 table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:35:56
704
Gas Chromatography
Table 40.21 continued Compound
System
Principal ions (m/z)
GA
GB
M (mandelic acid)
107
79
77
51
152
90
177
105
77
106
51
107
176
90
77
70
105
69
56
71
91
261
84
M (5-phenyloxazolidine-2,4-dione) Pentetrazole
1550
1579
Phenbutrazate (fenbutrazate)
2675
—
Phencyclidine (PCP)
125
1900
1981
200
242
243
91
84
186
M (4-phenyl-4 piperidinocyclohexanol, PPC)
—
—
216
258
259
91
202
182
M (PPC)-TMS
—
—
200
254
331
—
—
—
M (1(1-phenylcyclohexyl)-4-OH-piperidine)TMS
—
—
172
288
331
—
—
—
M (PCA) 5[N-(1 0 -phenylcyclohexyl)-amino]pentanoic acid
—
—
—
—
—
—
—
—
Pipradrol
2145
2242
Pipradrol-AC
2478
—
Psilocin
1985
Psilocin-AC
2270
Psilocin-AC2 Psilocybin
84
56
85
77
105
55
249
248
165
229
291
206
2080
58
204
150
146
205
155
—
58
246
146
130
202
117
2340
—
58
288
80
202
122
246
2046
—
—
—
—
—
—
—
Theobromine (see Caffeine) Theophylline (see Caffeine)
Oral hypoglycaemics
metabolites are not amenable to GC because of their acidity and polarity. Extractive methylation has been employed, but the use of standard conditions leads to the formation of unstable compounds, and in addition there may be thermal decomposition during chromatography. Thus, the formation of common fragments (superscripts a to f in Table 40.22; HH Maurer, personal communication, 2003) complicates
Oral hypoglycaemics constitute three chemical classes: biguanides (metformin), sulfonylureas (chlorpropamide) and thiazolidenediones (rosiglitazone). The latter two classes contain sulfur, and the sulfonylureas also share structural similarities with the thiazide diuretics (see above) and the sulfonamide antibiotics (see later). Sulfonylureas and their
Table 40.22 GC retention data and mass spectral data for oral hypoglycaemics (Me, methyl) Compound
System
Principal ions (m/z)
GA
GB
Acetohexamide
1859
Not eluted
210
56
43
184
211
Acetohexamide-Me
2250
—
183
198
98
119
115
Buformin-nitrobenzoyltriazine
—
3200
—
—
—
—
—
75 91 —
Carbutamide-Me
2300
—
109
156
92
285
—
—
Chlorpropamide
1791
1887
111
175
75
85
276
127
Chlorpropamide-Me
2165
—
58
115
111
175
290
127
Chlorpropamide-Me2
2250
—
109
156
92
304
—
—
Art 1-Me
1825
—
111
175
205
75
113
141
Art 1-Me2
1690
—
111
219
75
175
113
221
Art (chlorosulfonamide)-Me
1740
—
111
75
141
175
205
—
Art (chlorosulfonamide)-Me2
1655
—
111
75
219
175
155
—
Art (chloroamide)-Me
2135
—
111
75
125
248
175
—
Art (chloroamide)-Me2
2150
—
87
111
125
175
262
—
Glibenclamide-Me
3800
—
169
82
97
171
198
381
Glibenclamide-Me2
3840
—
169
289
171
291
353
126
Art 3-Me
3445
—
169
198
126
287
382
—
Art 3-Me2
3355
—
169
289
198
353
396
—
1730
1660
91
65
171
155
107
—
Art(e) (methylsulfonamide)-Me
1740
—
91
65
185
155
121
108
Art(f) (amide)
1620
1695
91
155
197
65
106
—
Glibornuride Art(d) (methylsulfonamide)
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:35:59
Specific applications
705
Table 40.22 continued Compound
System
Principal ions (m/z)
GA
GB
Art 5
1845
1995
95
109
134
164
195
—
Art 5-Me
1715
—
95
209
109
139
150
—
M (OH)-Art
2305
—
95
109
125
181
211
—
M (OH)-Art (sulfonamide)-Me(d)
2265
—
107
89
201
172
141
—
M (OH)-Art (sulfonamide)-Me2(e)
2030
—
107
89
215
171
151
—
M (COOH)-Art(sulfonamide)-Me3(f)
1955
—
135
243
103
199
212
—
1545
—
110
181
125
184
151
—
1620
—
91
155
197
65
106
— —
Gliclazide Art 1-Me Art(c) (amide) Art 3
1670
—
81
110
125
67
169
Art(a) (methylsulfonamide)
1730
1660
91
65
171
155
107
—
Art(b) (methylsulfonamide)-Me
1740
—
91
65
185
155
121
—
M (OH)-Art (sulfonamide)-Me(d)
2265
—
107
89
201
172
141
—
M (OH)-Art (sulfonamide)-Me2(e)
2030
—
107
89
215
171
151
—
1955
—
135
243
103
199
212
—
Glipizide-Me
3420
—
150
111
93
459
98
—
Glipizide-Me2
3455
—
150
121
93
334
392
Art 2-Me
3020
—
150
121
93
197
334
—
Art 2-Me2
3005
—
150
241
121
93
348
—
M (COOH)-Art (sulfonamide)-Me3
(f)
197
Gliquidone
2024
Not eluted
—
—
—
—
—
—
Gliquidone-Me
3850
—
323
220
204
175
176
430
Art 4-Me
3460
—
204
219
176
321
416
—
Art 4-Me2
3415
—
204
219
176
321
416
—
Glymidine
1632
2750
244
59
77
43
245
168
Glymidine-Me
—
—
—
—
—
—
—
—
Metformin-nitrobenzoyltriazine
—
3050
—
—
—
—
—
—
Tolazamide
1651
1720
91
155
114
65
197
Tolazamide-Me
2630
—
113
155
170
241
325
42 —
2540
—
91
155
339
184
229
114
Art 1-Me
1315
—
98
113
59
68
85
172
Art (methylsulfonamide)
1730
1660
91
65
171
155
107
Tolazamide-Me2
—
Art (methylsulfonamide)-Me
1740
—
91
65
185
155
121
—
M (OH)-Art (sulfonamide)-Me(d)
2265
—
107
89
201
172
141
—
M (OH)-Art (sulfonamide)-Me2(e)
2030
—
107
89
215
171
151
—
M (COOH)-Art(sulfonamide)-Me3(f)
1955
—
135
243
103
199
212
—
1683
—
91
30
155
108
65
Art(c) (amide)
1620
—
91
155
197
65
106
Tolbutamide-Me
2320
—
91
129
155
284
269
87
Tolbutamide-Me2
2170
—
91
155
113
121
220
184
Art (methylsulfonamide)
1730
1660
91
65
171
155
107
—
Art (methylsulfonamide)-Me
1740
—
91
65
185
155
121
—
M (OH)-Me
2645
—
129
171
300
285
200
—
M (OH)-Me2
2740
—
134
215
107
197
314
—
M (COOH)-Me3
2590
—
129
135
199
297
328
—
M (OH)-Art (sulfonamide)-Me(b)
2265
—
107
89
201
172
141
—
M (OH)-Art (sulfonamide)-Me2(a)
2030
—
107
89
215
171
151
—
M (COOH)-Art(sulfonamide)-Me3(c)
1955
—
135
243
103
199
212
—
Tolbutamide
(a)–(f)
Hydrolysis artefacts common to several sulfonylureas.
197 —
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:37:0
706
Gas Chromatography
identification of the parent compound and limits the application as a screening tool. For this purpose, HPLC may be more applicable (Maurer et al. 2002). However, stable N-methyl (sulfonamide nitrogen) derivatives can be formed by maintaining the extraction pH below 7, and by using TBAH as the ion-pairing reagent. With the alkylation pH above 10, and using TPAH as the counter-ion, there is almost complete hydrolysis to sulfonamide and amide artefacts (Hartvig et al. 1980). Thermal stability can also be achieved by the judicious choice of pairs of derivatising reagents: methyl iodide plus trifluoroacetic anhydride (TFAA) for parent compounds, and methyliodide plus heptafluorobutyric anhydride (HFBA) for the hydroxy and carboxy metabolites (Braselton et al. 1977). This strategy offers the additional advantage of an improved ECD response. The biguanides also present analytical difficulties, although successful chromatography is achieved by forming a triazine derivative by them reacting with p-nitrobenzoyl chloride, for which the retention data are given in Table 40.22 (Brohon, N€ oel 1978; Paroni et al. 2000). Screening methods are not yet developed for the newer classes of drugs such as the alpha-glucosidase inhibitors, glinides, glitazones and gliptins, although LC-MS methods have been published for some of the individual compounds. Systems GA and GB may be used, and the reference compounds are n-alkanes with an even number of carbon atoms. Pesticides A comprehensive method for screening pesticides using systems GA and GK can be found in Chapter 16, Table 16.1. Systems GKA, GKB, GKC and GKD cater for a smaller set of compounds and these can be found in the Indexes of Analytical Data. System GKA
(Osselton, Snelling 1986). n n n n
Column: Chromosorb W HP 3% SE-30 on 80–100 mesh silanised glass (2 m 4 mm i.d.). Carrier gas: O2-free N2, 50 mL/min. Detector: Flame ionisation and nitrogen–phosphorus. Reference compound: Straight-chain hydrocarbons
System GKB
(Osselton, Snelling 1986). n n n
Column: Chromosorb W HP 3% OV-7 on 80–100 mesh silanised glass (2 m 4 mm i.d.). Carrier gas: O2-free N2, 50 mL/min. Detector: Flame ionisation and nitrogen–phosphorus.
n
Reference compound: Straight-chain hydrocarbons.
System GKC
(Osselton, Snelling 1986). n n n n
Column: Chromosorb W HP 3% OV-17 on 80–100 mesh silanised glass (2 m 4 mm i.d.). Carrier gas: O2-free N2, 50 mL/min. Detector: Flame ionisation and nitrogen–phosphorus. Reference compound: Straight-chain hydrocarbons.
System GKD
(Junting, Chuichang 1991). n n n n
Column: Fused silica HP-1, methyl silica gum (5 m 0.53 mm i.d., 2.65 mm). Temperature programme: 190 C to 235 C at 10 /min. Carrier gas: N2, 20 mL/min flow rate. Detector: Flame ionisation.
Phenothiazines and other tranquillisers Phenothiazines and other tranquillisers can be extracted readily under mildly basic conditions (pH 10) into solvents such as ethyl acetate, hexane, butyl chloride and diethyl ether. An acidified (0.05 mol/L H2SO4) back extraction is a useful clean-up procedure where sensitivity is important. Chromatography of the primary and secondary amines is poor on packed columns, but is adequate on well-maintained capillary columns, particularly those of low-to-medium polarity, such as PSX-5 (see Table 40.3). Meprobamate is unstable in basic solution, and benefits from the use of mildly acidic (pH 5) extraction conditions. Some authors prefer to chromatograph the secondary amines and hydroxylated metabolites as acetylated derivatives, prepared by heating the dried residue with acetic anhydride and pyridine (3 : 2, v/v) (Maurer, Bickeboeller-Friedrich 2000). Others employ an enzymatic hydrolysis procedure to improve the recovery of both parent drug and metabolites, although the additional sensitivity gained is often negated by the increased analytical time in the emergency setting. Acid hydrolysis is quicker, but some relevant compounds are destroyed under these conditions. System GA or GB may be used (Table 40.23), and the reference compounds are n-alkanes with an even number of carbon atoms. Laxatives Table 40.24 lists the stimulant laxatives. Other types of laxatives, such as bulkers (bran), osmotic (PEG and lactulose), stool softeners and saline
Table 40.23 GC retention data and mass spectral data for phenothiazines and other tranquillisers Compound
System
Principal ions (m/z)
GA
GB
Acepromazine
2735
2844
100
72
240
340
44
197
M (dihydro)-H2O
2720
2824
58
86
310
225
224
251
M (7-OH-)
—
3160
58
86
342
326
296
257
2650
—
72
73
255
326
56
240
2920
—
72
56
84
238
270
340
Acetophenazine
—
Not eluted
254
143
42
70
411
113
Alimemazine (trimeprazine)
Aceprometazine M (methoxy-dihydro)-H2O
2305
2402
58
298
212
198
100
299
M (nor-)
2335
2432
199
284
212
198
180
252
M (sulfoxide)
2665
2805
M (norsulfoxide)
—
2817
58 —
199
180
298
297
—
—
—
—
M (OH-)
2650
2829
314
100
255
196
281
M (nor-OH-)
—
2845
—
—
—
—
—
—
—
—
—
—
—
Alpidem
—
3313
Amisulpride
3260
—
M (O-desmethyl-) Apronal
58
212 —
98
99
44
242
—
70
111
2960
—
98
135
182
228
99
107
1331
—
55
44
142
141
61
81
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:37:21
Specific applications
707
Table 40.23 continued Compound
System GA
Principal ions (m/z) GB
Aripiprazole M
—
2108
174
176
218
220
75
44
M
—
2258
213
215
242
244
98
172
Azacyclonol
2243
2361
Azaperone
2705
—
Benactyzine
2255
—
Benperidol
3433
3667
M
1490
M (N-desalkyl-)
2415
Benzoctamine Bromisoval
85
84
183
105
56
107
165
123
95
121
77 —
86
105
77
87
182
99
230
109
82
187
243
363
—
56
125
123
180
136
95
—
134
79
51
106
217
161
2078
2172
218
44
191
221
219
178
1540
—
55
70
163
165
83
222
M (Br-isovaleric acid)
1190
—
136
140
101
59
120
122
M (OH-isovaleric acid)
1140
—
76
73
55
58
57
74
M (iso-valeric acid carbide)
1850
—
102
59
85
57
70
61
Bromperidol
3037
—
42
268
270
281
283
123
Art (-H2O)
3020
—
236
238
252
250
253
265
M
1890
—
233
235
56
94
127
154
M (N-desalkyl-oxo)-2H2O Buspirone
1850
—
233
235
127
154
63
101
3300
3468
177
277
265
122
148
108
—
1558
122
108
96
80
164
134
Butaperazine
3190
—
113
70
409
141
283
127
Captodiame
2774
—
58
165
255
359
166
73
M (1-pyrimidinyl piperazine)
Carbromal M (OH-carbromide)
1513
—
44
69
208
210
55
71
1340
—
150
152
165
167
183
194
M (carbromide)
1215
—
69
165
167
114
71
150
M (desbromo-)
1380
—
87
113
71
130
86
115
Art
1450
—
69
58
70
105
179
97
Art
1470
—
69
70
140
151
193
191
Carphenazine
3590
—
268
143
245
70
269
394
Chlormezanone
2199
2346
98
152
154
42
69
174
Art
1235
1245
152
153
154
89
111
59
M (4-chlorobenzoic acid)
1400
—
139
156
111
75
141
113
M (N-methyl-4-chlorobenzamide)
1555
1596
139
111
75
169
141
168
2495
2618
58
86
318
85
320
272
M (nor-)
2480
2656
44
232
233
196
214
304
M (didesmethyl-)
2480
2646
232
290
233
246
272
214
M (sulfoxide)
2809
3003
58
246
214
232
272
318
M (norsulfoxide)
2900
3046
44
246
232
302
214
196
M (7-OH-)
—
2939
58
86
334
248
288
262
M (N-oxide)
2100
2355
233
198
201
154
166
171
Chlorprothixene
2492
2608
58
59
221
42
222
255
2750
—
58
333
335
247
334
215
Chlorpromazine
M (OH-dihydro-) isomer 1 M (OH-dihydro-) isomer 2
2790
—
58
333
247
335
249
334
M (OH-methoxy-dihydro-)
2810
—
58
363
277
173
262
249
M (N-oxide)-(CH 3)2NOH
2410
—
234
235
270
269
202
255
M (N-oxide sulfoxide)- (CH3)2NOH
2560
—
203
234
202
251
286
269
M (sulfoxide)
2720
—
58
221
189
255
176
331
Art (dihydro-)
2490
—
58
317
231
73
152
195
Art (Cl-thioxanthenone)
2260
—
246
218
248
139
220
183
—
1269
112
161
85
45
163
113
Clomethiazole (chlormethiazole) Clopenthixol (see Zuclopenthixol)
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:37:37
708
Gas Chromatography
Table 40.23 continued Compound
System
Principal ions (m/z)
GA
GB
2712
2833
—
2882
—
Cloral betaine (see Chapter 14) Cloral hydrate (see Chapter 14) Clothiapine (clotiapine) M (nor-)
83
70 —
273
244
209
—
—
—
71 —
3030
—
357
209
244
285
273
291
2895
3024
—
—
—
—
—
—
M (nor-)
3105
3092
192
243
256
56
227
312
M (nor-acetyl-)
3490
3609
396
310
298
192
227
256
M
—
2833
58
300
256
243
299
160
M (oxo-) Clozapine
M
—
2972
243
286
44
256
244
270
M
—
3150
225
238
294
209
250
264
M
—
3264
255
268
192
239
338
280
M
—
3320
255
192
268
324
239
280 256
—
3527
340
192
339
228
243
Dichloralphenazone
1855
—
188
47
82
96
77
84
M (phenazone)
1835
1951
188
96
77
56
105
189
M (4-OH-phenazone; metabolised to chloral hydrate)
1855
—
85
56
84
204
77
120
Diethazine
2377
—
86
298
87
58
299
212
Dimetotiazine
3060
3096
72
73
320
56
210
198
M (nor-)
3150
—
72
58
320
306
198
210
M
Dixyrazine M (phenothiazine) Droperidol
3220
—
212
42
187
70
180
56
2120
2130
199
167
198
166
154
139 247
3430
Not eluted
246
165
42
123
199
1950
—
134
79
106
121
105
67
Emylcamate
1105
—
73
43
84
55
69
44
Ethchlorvynol
1015
1060
115
117
89
53
109
51
Ethinamate
1365
—
91
81
106
78
95
68
M (benzimidazolone)
Ethomoxane
1975
—
86
44
265
180
87
Fluanisone
2785
—
205
218
123
356
219
162
2715
—
194
165
123
342
338
134
M (O-desmethyl-)
—
Fluopromazine (see Triflupromazine) Flupentixol (flupenthixol) cis isomer
3058
3199
143
70
100
144
98
58
Flupentixol trans isomer
—
3217
143
70
100
144
98
58
M (ring)
2190
—
267
235
247
222
198
216
M (N-oxide)
2120
—
304
303
234
235
289
283
M (desalkyl-) cis
—
2832
—
—
—
—
—
—
M (desalkyl-) trans
—
2855
—
—
—
—
—
—
3050
3194
280
143
437
406
113
Fluphenazine
70
M (ring)
2190
—
280
143
113
248
M (7-OH-)
—
3572
—
—
—
—
—
—
—
—
—
M (sulfoxide) Fluspirilene M (N-desalkyl-oxo-)
70
56
—
3752
—
—
—
1017
—
244
42
72
475
109
245
2405
—
57
56
245
68
228
206
M (desamino-OH-)
2120
—
203
201
262
183
216
244
M (desamino-carboxy-)
2230
—
203
201
183
216
276
167
Glutethimide
1830
1910
189
132
117
160
91
115
M (OH-ethyl-)
1865
1958
146
104
233
103
133
117
M (OH-phenyl-)
1875
2040
133
204
233
176
77
205
M (2-phenylglutarimide)
2235
—
104
189
103
117
78
M (desethylphenylgutarimide)
2370
—
—
—
—
—
—
91 —
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:37:57
Specific applications
709
Table 40.23 continued Compound
System
Principal ions (m/z)
GA
GB
2930
3094
224
42
237
226
123
M (reduced)
3152
3152
224
206
226
193
377
139
M (N-desalkyl-oxo)-2H2O
1650
1707
189
154
127
191
126
190
M
1750
1872
56
139
84
223
206
111
Haloperidol
206
M (N-desalkyl)
1800
—
56
84
139
111
133
211
Hydroxyphenamate
1724
—
135
57
91
77
119
105
Hydroxyzine
166
2849
3000
201
203
165
45
299
M (norchlorcyclizine)
—
2355
165
201
166
85
241
230
M (4-chlorobenzophenone)
1850
1862
105
139
216
77
111
218
M (4-chloromethylbiphenyl)
1600
1688
167
165
202
152
82
204
M (OH-chlorobenzophenone)
2300
2230
121
139
95
234
111
152
M
—
2704
201
165
166
228
242
299
M
—
2847
85
165
166
201
242
256
Levomepromazine (methotrimeprazine) M (norsulfoxide)
2514
2641
328
100
228
185
329
—
3088
—
58
—
—
—
—
—
—
—
—
—
—
3114
—
—
2555
2717
70
83
257
193
56
M (8-OH-)
2931
3077
70
83
273
209
260
343
M (7-OH-)
—
3068
70
273
260
209
244
343
M (amoxapine)
2638
2746
245
193
257
247
228
164
M (7-OH-amoxapine)
2951
3525
261
209
273
263
244
329
M (8-OH-amoxapine)
2959
3546
261
209
273
263
244
329
M (sulfoxide) Loxapine
228
Mebutamate
1889
—
97
55
69
72
71
98
Mecloqualone
2255
—
235
111
75
76
236
50
1854
83
84
55
56
43
71
Meprobamate (also carisoprodol metabolite) 1785 Art
1535
1487
84
55
56
44
83
75
M
1720
1763
104
43
45
62
148
86
M
—
1932
104
43
45
71
119
204
M
—
2079
43
111
104
132
172
62
Mesoridazine
3380
3629
98
70
99
386
126
55
Methaqualone
2135
2256
235
250
91
233
236
65
M (2-formyl-)
2240
2370
235
132
264
206
248
192
M (2-OH-methyl-)
2360
2437
235
266
251
175
132
160
M (2-carboxy-)
2400
—
235
146
77
221
252
280
M (2 0 -OH-methyl-)
2410
2500
160
266
235
251
77
247
M (3 0 -OH-)
2490
—
251
266
249
77
148
252
M (4 0 -OH-)
2510
—
251
266
249
77
143
235
M (6-OH-)
2525
—
251
266
249
132
65
92
M (OH-methoxy-)
2560
2698
296
281
143
249
279
266
Methdilazine
2462
—
97
98
296
199
212
198
Methyprylon
1527
1581
155
140
83
98
55
41
M (OH)-H2O
1540
1601
83
55
153
166
84
98
M (oxo-)
1870
1834
83
98
55
168
151
182
Molindone
2465
—
100
56
176
98
120
70
Olanzapine
—
2861
242
229
231
198
312
169
—
2911
229
213
242
298
198
254
Oxypertine
2355
—
175
70
176
132
379
204
Pecazine
2540
2669
310
58
199
112
111
212
Penfluridol
3360
—
42
292
56
294
109
203
M (nor-)
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:38:2
710
Gas Chromatography
Table 40.23 continued Compound
System
Principal ions (m/z)
GA
GB
M (N-desalkyl-oxo)-2H2O
1920
—
257
259
258
222
167
M (N-desalkyl-)
2210
—
56
261
279
179
260
114
M (desamino-OH-)
2120
—
—
—
—
—
—
—
M (desamino-carboxy-)
2230
—
—
—
—
—
—
—
202
2798
—
113
339
141
340
M (OH-)
3175
—
—
—
—
—
—
—
M (phenothiazine)
Perazine
44
70
2120
2130
199
167
198
166
154
139
Pericyazine
3260
3486
114
44
142
365
223
115
M (ring)
2555
—
224
192
223
120
112
179
Perphenazine
3380
3594
246
143
403
70
404
248
2100
—
—
—
—
—
—
—
Phenothiazine
2120
2130
199
167
198
166
154
139
Phenprobamate
1520
—
118
117
91
92
119
65
Pimozide
42
217
83
461
M (ring)
3870
Not eluted
230
187
M (N-desalkyl-)
2415
—
—
—
—
—
—
—
M (benzimidazolone)
1950
—
—
—
—
—
—
—
M (desamino-OH-)
2120
—
—
—
—
—
—
—
M (desamino-carboxy-)
2130
—
—
—
—
—
—
—
Pipamperone
3040
—
165
138
331
123
110
194
M (OH-)
3250
—
165
154
347
123
292
194
Piperacetazine
0000
—
142
170
44
410
143
42
Pipotiazine (pipothiazine)
2932
Not eluted
142
44
140
198
170
96
Prochlorperazine
2954
3129
113
70
373
141
43
72
M (N-oxide)
2100
2356
233
198
235
218
272
201
M (norsulfoxide)
—
3571
70
113
373
246
319
232
M (sulfoxide)
—
3758
70
113
246
319
373
232
Promazine
2315
2425
58
284
86
238
198
199
M (nor-)
2405
2452
199
270
198
213
238
212
M (sulfoxide)
2705
2840
212
199
300
284
180
M (norsulfoxide)
—
2875
—
—
—
—
—
—
M (phenothiazine)
2120
2130
199
167
198
166
154
139
M (OH-)
2685
2781
58
86
300
215
254
228
M (nor-OH-)
—
2797
—
—
—
—
—
Promethazine
2339
2383
72
284
198
213
199
180
M (nor-)
2250
2333
58
213
180
198
152
270
M (phenothiazine)
2120
2130
199
167
198
166
154
139
M (sulfoxide)
2710
2797
72
198
180
152
213
229
M (norsulfoxide)
—
2732
58
212
180
198
229
152
M (nor-OH-)
2580
2717
212
58
180
229
198
286 288
58
—
M (OH-)
2590
—
72
196
300
229
214
Propiomazine
2738
—
72
340
269
197
73
71
Prothipendyl
2345
—
58
285
214
200
86
227
M (OH-)
2720
—
58
301
86
216
230
243
M (OH-ring)
2800
—
216
187
168
188
200
161
M (ring)
2045
—
200
168
199
156
201
155
M (sulfoxide)
2750
—
58
86
216
179
155
200
Quetiapine M M Remoxipride M
—
3400
210
239
144
251
321
226
—
2745
227
210
239
251
265
295 233
—
2709
195
207
178
151
219
2520
2588
98
99
70
228
230
243
—
2981
—
—
—
—
—
—
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:38:4
Specific applications
711
Table 40.23 continued Compound
System GA
Principal ions (m/z) GB
M
—
3022
—
—
—
—
—
—
M
—
3313
—
—
—
—
—
—
Risperidone Art Sulforidazine (also thioridazine and mesoridazine metabolite) M (ring) Sulpiride Art (-SO2NH) Tetrabenazine M (O-desmethyl-OH-)
—
1877
220
191
204
178
192
221
2063
—
—
—
—
—
—
—
3415
3690
197
198
290
98
402
70
3180
—
277
198
154
127
278
263
3102
Not eluted
—
—
—
—
—
—
2295
—
98
70
77
135
99
111
2490
2579
191
261
260
274
316
176
2500
2638
205
191
274
318
319
232
Thalidomide
2440
Not eluted
173
104
76
111
148
170
Thiethylperazine
3226
—
70
113
141
399
72
259
M (ring)
2750
—
259
230
198
186
260
167
M (sulfone)
3400
—
70
113
127
305
431
212
Thiopropazate (metabolised to perphenazine) 3467
—
246
70
185
98
87
213
Thioproperazine
—
70
113
127
212
320
141 154
3552
M (ring)
3200
—
306
198
199
197
277
Thioridazine
3115
3292
98
370
126
99
371
250
—
3275
84
56
112
356
245
185
M (nor-) M (sulfoxide) mesoridazine
3380
3629
98
70
99
386
126
55
M (ring)
2570
2639
245
198
186
230
154
166
384
385
244
112
258
245
98
370
402
244
258
290
M (oxo-)
3500
—
M (ring sulfone)
3420
3626
M (side chain sulfone)
3800
Not eluted
416
112
290
277
276
417
3060
—
—
—
—
—
—
—
Thiothixene Trichloroethanol (see Chapter 14) Triclofos (metabolised to trichloroethanol)
—
1952
31
49
77
113
51
Trifluomeprazine
2250
—
58
366
100
266
248
84
Trifluoperazine
2683
2798
113
70
407
43
141
127
M (phenothiazine)
2120
2130
199
167
198
166
154
139
M (sulfoxide)
2990
3145
113
141
248
266
306
280
M (norsulfoxide)
—
3191
—
—
—
—
—
—
115
2675
—
42
271
258
123
83
240
M (N-desalkyl-oxo-)-2H 2O
1570
—
223
224
154
170
183
204
M (N-desalkyl-)
1970
—
56
227
226
245
223
198
2230
2318
58
352
86
353
306
266
M (phenothiazine)
2120
2130
199
167
198
166
154
139
M (OH-)
2700
—
58
368
86
322
282
323 265
Trifluperidol
Triflupromazine
2730
—
58
398
86
312
313
Trimetozine
2253
—
195
281
196
152
280
81
Tybamate
1725
—
55
72
97
158
118
56
M (OH-methoxy-)
Zolpidem
2715
2941
235
236
307
219
92
65
Zopiclone
2950
3263
143
245
112
99
139
217
1200
1261
128
101
130
73
98
93
Zotepine
Art (amino-chloropyridine)
2660
—
208
199
221
163
231
147
Zuclopenthixol (clopenthixol) cis isomer
3360
3557
143
70
144
100
42
56
Zuclopenthixol trans isomer
3400
3680
143
70
144
100
42
56
M (N-oxide)-C6H14N2O2
2410
—
—
—
—
—
—
—
Art (Cl-thioxanthenone)
2260
—
—
—
—
—
—
—
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:38:12
712
Gas Chromatography
Table 40.24 GC retention data and mass spectral data for laxatives (AC, acetyl; Me, methyl; TMS, trimethylsilyl) Compound
System GA
Principal ions (m/z) GB
Aloe-emodin
2660
—
—
—
—
—
—
—
Aloe-emodin-AC
2735
—
270
241
312
271
242
225
Aloe-emodin-AC2
3000
—
270
354
312
241
271
224
Aloe-emodin-Me
2900
—
284
266
238
225
209
237
Aloe-emodin-Me2
2705
—
298
267
239
299
240
291
Aloe-emodin-TMS
2685
—
311
312
342
225
296
268
Aloe-emodin-TMS2
2785
—
399
400
184
310
383
325
Aloe-emodin-TMS3
2900
—
471
472
472
399
367
281
Aloin
0000
0000
—
—
—
—
—
—
Arecoline
1195
—
155
96
140
43
81
94
Bisacodyl
2818
2956
361
277
319
276
199
318
M (bismethoxybisdesacetyl)
2820
—
337
322
336
338
259
307
M (bismethoxydesacetyl-)
2890
—
379
322
364
336
378
380
M (desacetyl-)
2750
2876
319
276
277
199
318
246
M (bisdesacetyl-)
2655
2793
277
276
199
183
278
246
M (methoxybisdesacetyl-)
2680
—
307
306
229
292
275
198
M (methoxydesacetyl-)
2810
—
349
306
307
229
292
348
M (bismethoxybisdesacetyl)-AC2
2950
—
379
421
364
322
336
378
M (methoxybisdesacetyl)-AC2
2870
—
349
391
307
306
229
348
M (trimethoxybisdesacetyl)-AC2
3060
—
409
367
451
329
352
203
M (desacetyl)-TMS
—
2830
391
348
349
271
390
392
M (bisdesacetyl)-TMS2
—
2728
421
343
420
422
256
240
Dantron
2330
2450
240
212
241
184
138
92
Dantron-Me
2435
—
254
208
236
225
139
168
Dantron-Me2
2475
—
253
268
139
152
209
180
Dantron-AC
2460
—
240
282
241
212
184
155
Dantron-AC2
2595
—
240
282
241
212
184
155
Dantron-TMS
2465
2574
297
298
253
240
210
312
Dantron-TMS2
2530
2611
369
370
297
371
268
210
Emetine
2505
Not eluted
192
206
272
480
288
246
Emetine-Me
4010
—
206
207
190
272
288
494
Frangula-emodin
2620
—
—
—
—
—
—
—
Frangula-emodin-AC
2740
—
270
312
271
242
213
241
Frangula-emodin-Me2
2775
—
298
252
280
269
237
281
Frangula-emodin-Me3
2845
—
297
312
295
283
266
251
Phenolphthalein
—
3292
274
225
318
273
275
226
Phenolphthalein-AC2
3375
3351
360
318
274
225
257
402
Phenolphthalein-Me2
3060
—
271
302
346
301
239
287
Phenolphthalein-TMS2
—
3205
418
417
419
297
253
329
3395
—
390
273
272
348
304
391
Physcion
2660
2732
284
285
255
128
283
241
Physcion-AC2
2920
—
284
326
285
255
227
184
M (methoxy)-AC2
Physcion-Me2
2845
—
—
—
—
—
—
—
Physcion-Me
2775
—
—
—
—
—
—
—
Physcion-TMS
2150
2247
341
117
129
132
145
356
Picosulfate (hydrolysed to bisacodyl metabolites, see above) Rhein
2675
—
284
285
255
128
241
139
Rhein-Me
2660
—
298
267
239
155
126
284
Rhein-Me2
2740
—
312
266
294
251
235
126
Rhein-Me3
2855
—
311
326
312
309
235
295
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:38:18
Specific applications
713
Table 40.25 GC retention data for steroids (AC, acetyl; TMS, trimethylsilyl) GA
GAG
GAI
GAR
—
—
—
—
dihydrotestosterone
2510
—
—
—
Androstanolone dihydrotestosterone-AC
2630
—
—
—
dihydrotestosterone-TMS
2485
—
—
—
dihydrotestosterone enol-TMS2
2450
—
—
—
5a-dihydrotestosterone
—
—
0.95
—
2475
—
—
11.9
Androsterone -AC
2580
—
—
—
enol-TMS2
2500
—
—
— 12.8
—
1.05
0.961
undecylenate
—
2.62
—
22.4
5b-androst-1-en-17b-ol-3-one
—
—
0.96
—
acetate
—
—
—
13.6
benzoate
—
—
—
18.7
undecylenate
—
—
—
22.4
2530
—
—
11.8
DHEA-H2O
2595
—
—
—
DHEA enol-TMS2
2580
—
—
—
2985
—
0.974
—
drostanolone
2555
—
—
—
drostanolone-AC
2700
—
—
—
drostanolone-TMS
2575
—
—
—
drostanolone-enol-TMS2
2625
—
—
—
drostanolone propionate
2985
—
0.974
—
2835
1.5
1.155
14.6
2672
1.2
—
13.2
—
—
0.925
—
Boldenone
DHEA
Drostanolone propionate
Fluoxymesterone Methandienone 17a-methyl-5b 17a-androstan-3a,17b-diol
—
—
0.925
—
17a-methyl-5b-androst-1en-3a,17b-diol
—
—
0.921
—
17a-methyl-1,4-androstadien-6b,17b-diol-3-one
—
—
1.117
—
—
0.89
—
12.3
dipropionate
—
1.70
—
—
17a-methyl-5b-androstan-3a,17b-diol
—
—
0.925
—
Methandriol
2645
1.05
—
13.1
-AC
2770
—
—
—
-TMS
2590
—
—
—
enol-TMS2
2665
—
—
—
17a-methyl-5b
—
—
0.925
—
2395
0.91
—
12.5
2760
—
—
—
—
1.17
1.111
13.7
Methyltestosterone
Nandrolone -TMS Oxandrolone Oxymetholone
3005
1.28
—
13.7
enol-TMS3
2870
—
—
—
17a-methyl-5a-androstan-3a,17b-diol
—
—
0.925
—
2-hydroxymethyl-17a-methyl5-androstan-3,17-diol
—
—
1.106
—
2-hydroxymethyl-17a-methyl5-androstan-3,6,17-triol
—
—
1.180
—
Stanozolol
3085
—
1.31
15.4
-TMS2
3025
—
—
—
-AC
2120
—
—
—
3 0 -hydroxystanozolol
—
—
1.38
—
4b-hydroxystanozolol
—
—
1.393
— table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:38:18
714
Gas Chromatography
Table 40.25 continued GA
GAG
GAI
GAR
2620
—
0.97
12.9
-AC
2750
—
—
—
enol-TMS2
2690
—
—
—
propionate
2815
1.43
—
14.2
dipropionate
3350
—
—
—
methyltestosterone
—
1.05
—
13.1
acetate
—
1.21
—
13.5
isobutyrate
—
1.54
—
—
cipionate
—
2.19
—
18.7
enantate
—
1.92
—
16.7
undecylate
—
2.56
—
—
isocaproate
—
1.77
—
15.9
decanoate
—
2.36
—
19.8
benzoate
—
—
—
18.0
phenylpropionate
—
—
—
20.2
Testosterone
(magnesium and other salts), are detected by other means (Duncan 2000) and are not discussed here. Stools may be analysed after homogenisation, or alternatively purgatives may be detected in urine, both after enzymatic hydrolysis to release conjugated metabolites. Extraction of hydrolysis products with chloroform–isopropanol (9 : 1) or other moderately polar solvents at the hydrolysis pH yields good recovery, and derivatisation (e.g. silylation, methylation or acetylation) improves chromatography, particularly of the more polar compounds such as rhein. Rhein is a product of many vegetable glycoside laxatives, including sennosides, aloes and cascara (except frangula, which is metabolised to emodin), which are hydrolysed by colonic bacteria to active aglycones prior to absorption (this is important to remember when analysing pharmaceutical products). System GA or GB may be used, and the reference compounds are n-alkanes with an even number of carbon atoms.
n n
Column: Fused silica capillary with methylsilicone (12 m 0.25 mm i.d., 0.25 mm). Temperature programme: 70 C to 150 C at 15 /min to 250 C at 25 / min.
System GAG
(Lurie et al. 1994). n n n n
Column: Bonded DB-1 fused silica capillary, cross-linked (30 m 0.25 mm i.d., 0.25 mm). Split ratio of 30 : 1. Temperature programme: 180 C to 230 C at 10 /min to 245 C at 1 / min to 295 C at 30 /min for 15 min. Carrier gas: H2. Detector: Flame ionisation.
System GAI
Solvents and other volatile compounds Methods for the analysis and analytical data for solvents and other volatile substances are described in detail in Chapter 16. An ECD run in parallel with an FID produces the optimal detection rates, since many of the compounds of interest are halogenated. The low boiling point of these compounds requires their careful isolation from biological samples by headspace analysis, and the GC may benefit from cryogenic cooling (Flanagan et al. 1997; Sharp 2001). Standard capillary columns can be used (e.g. dimethyl-PSX), but chromatography and durability benefit from the use of a column with a high-phase ratio (3–5 mm film thickness). Other columns, such as an X-wax phase (a 0.25 mm film thickness is adequate here), or those specifically designed for volatiles, such as X-624 (3 mm film thickness), are used commonly (see Table 40.3 for details of these stationary phases). System GA or GI may be used and a more comprehensive method for screening volatiles can be found in Chapter 16, Table 16.3. System GA, previously described, may be used, or system GI, below. System GI
(Ramsey, Flanagan 1982). n n n
Column: 0.3% Carbowax 20M on 80-100 mesh Carbopak C glass, (2 m 2 mm i.d.). Temperature programme: 35 C for 2 min to 175 C at 5 /min for at least 8 min. Carrier gas: N2, 30 mL/min.
Steroids (Table 40.25) System GAR
(CND Analytical, 1989).
(Ayotte et al. 1996). n n n n n
Column: HP-5 5% phenyl polymethyl siloxane capillary (25 m 0.25 mm i.d., 0.33 mm). Temperature programme: 100 for 1 min to 220 at 16 /min to 301 at 20 /min for 5.5 min. Carrier gas: He. Detection: Mass-selective detector. Retention time: Relative to 17a-methyl-5a-androstan-3b,17b-diol.
Sulfonamides Gas chromatography of sulfonamides is only possible after extractive N-methylation of the secondary amino group (Gyllenhaal et al. 1978) and some authors additionally prepare HFB or PFP derivatives of any primary amino groups (Tarbin et al. 1999). These latter derivatives may require the use of positive chemical ionisation MS (Reeves 1999). As with the oral hypoglycaemics, differences in methylation conditions may lead to hydrolysis, and a number of sulfonamides may yield products or metabolites that correspond to sulfanilamide derivatives. The main metabolites are the N4-acetylacted derivatives, which are hydrolysed relatively easily back to the parent compound under acidic conditions. System GA or GJ (Gyllenhaal et al. 1978) may be used. System GJ n n n n
Column: 5% OV-17 on 80 to 100 mesh Gas-Chrom Q glass (1.5 m 2 mm i.d.). Temperature: 250 . Carrier gas: N2, 30 mL/min. Relative retention times are given in Table 40.26 (retention times of methyl derivatives are relative to griseofulvin).
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:38:24
Specific applications
715
Table 40.26 GC retention data and mass spectral data for the sulfonamides (AC, acetyl; Me, methyl) Compound
System
Principal ions (m/z)
GA
GJ
Mafenide
2340
—
106
77
185
105
104
89
Mafenide-Me2
1920
—
58
214
213
74
89
133
Mafenide-Me3
1900
—
58
89
228
227
133
214
Mafenide-Me4
1870
—
58
242
89
107
134
117
M (AC-)
2425
—
105
106
147
228
185
160
M (AC)-Me
2300
—
119
161
185
242
89
199
Sulfabenzamide-Me2
2770
0.09
118
105
77
304
170
240
Sulfabenzamide-Me (metabolised to sulfanilamide)
2700
—
118
105
77
92
226
290
Sulfacetamide
2132
—
92
109
156
180
65
214
—
—
Phenylbenzenesulfonamide (see Sulfabenzamide) Phthalylsulfacetamide (metabolised to sulfacetamide) Phthalylsulfathiazole (metabolised to sulfathiazole) Succinylsulfathiazole (metabolised to sulfathiazole)
Sulfacetamide-Me
—
0.16
—
—
Sulfadiazine
2502
—
185
186
—
—
92
65
108
170
Sulfadiazine-Me
2625
0.66
199
M (AC)-Me
3710
1.69
241
200
92
108
156
184
242
199
108
92
Sulfadimidine
2613
—
266
—
—
—
—
—
Sulfadimidine-Me
—
0.71
—
—
—
—
—
—
—
Sulfaethidole (also metabolised to sulfanilamide)
2620
—
108
156
220
Sulfadoxine
—
—
—
—
—
—
—
—
Sulfaethidole (also metabolised to sulfanilamide)
2620
—
—
—
—
—
—
—
Sulfaethidole-Me
3060
—
298
92
83
190
234
156
Sulfaethidole-Me 2
2840
—
106
92
65
161
156
234
2490
—
213
108
80
326
136
283
M (AC-) M (AC)-Me2 Sulfafurazole (sulfisoxazole)
92
284
65
3410
—
148
106
203
276
302
354
1212
—
156
92
108
140
65
267
Sulfafurazole-Me
—
0.42
Sulfaguanidine
0000
—
108
214
92
65
148
156
Sulfaguanole-Me (metabolised to sulfanilamide)
2905
—
203
57
323
322
249
204
Sulfamerazine
2566
—
199
200
92
65
108
100
Sulfamerazine-Me
2625
0.69
199
200
65
92
108
140
Sulfamethizole-Me (metabolised to sulfanilamide)
2660
0.98
92
284
108
156
176
220
Sulfamethoxazole-Me (also metabolised to sulfanilamide)
2500
—
92
108
119
162
156
203
Sulfamethoxazole-Me2
2460
—
92
108
119
62
156
188
Sulfamethoxazole-Me
2500
0.40
92
108
119
162
156
188
3255
0.91
161
134
230
245
205
199
Sulfamethoxydiazine-Me3 (also metabolised to sulfanilamide) 2925
1.38
229
230
92
108
138
156
3620
—
271
272
229
65
92
—
0.93
—
—
—
M (AC)-Me M (AC)-Me Sulfamethoxypyridazine-Me
—
—
139 —
Sulfametopyrazine/sulfalene-Me
—
0.69
—
—
—
—
—
—
Sulfamoxole-Me
—
0.40
—
—
—
—
—
—
Sulfanilamide
2185
—
65
92
156
172
108
80
Sulfanilamide-Me
2135
—
92
186
56
65
108
122
Sulfanilamide-Me3
—
—
122
214
170
106
77
79
Sulfanilamide-Me4
2095
—
136
120
228
184
77
105
M (AC-)
2690
—
172
156
92
108
125
214
M (AC)-Me
2600
—
186
156
228
92
108
65
1795
—
213
214
65
92
198
306
3420
—
255
256
65
93
122
213
—
1.71
—
—
—
—
Sulfaperin-Me3 (also metabolised to sulfanilamide) M (AC)-Me2 Sulfaphenazole-Me
—
—
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:38:24
716
Gas Chromatography
Table 40.26 continued Compound
System GA
Principal ions (m/z) GJ
Sulfapyridine
2600
—
184
185
65
92
108
66
Sulfapyridine-Me
—
0.47
198
199
92
65
78
108
M (AC)-Me
—
1.16
—
—
0.50
—
—
—
—
Sulfasalazine (metabolised to sulfapyridine) Sulfasomidine/sulfaisomidine-Me
—
—
—
—
—
—
—
Sulfathiazole
Not eluted —
—
—
—
—
—
—
Sulfathiazole-Me
—
—
—
—
—
—
—
0.49
Sulfaurea (metabolised to sulfanilamide)
References Adams RP (1995). Identification of Essential Oil Components by Gas Chromatography/Mass Spectrometry. Carol Stream, IL: Allured Publications. Ardrey RE, Moffat AC (1981). Gas–liquid chromatographic retention indices of 1318 substances of toxicological interest on SE-30 or OV-1 stationary phase. J Chromatogr 220: 195–252. Ayotte C et al. (1996). Testing for natural and synthetic anabolic agents in human urine. J Chromatogr B Biomed Appl 687: 3–25. Blau K, Halket J (1993). Handbook of Derivatives for Chromatography, 2 edn. New York: Wiley. Braselton WE Jr et al. (1977). Measurement of antidiabetic sulfonylureas in serum by gas chromatography with electron-capture detection. Diabetes 26: 50–57. Brohon J, N€ oel M (1978). Determination of metformin in plasma therapeutic levels by gas–liquid chromatography using a nitrogen detector. J Chromatogr 146: 148–151. Broussard LA et al. (1997). Simultaneous identification and quantitation of codeine, morphine, hydrocodone, and hydromorphone in urine as trimethylsilyl and oxime derivatives by gas chromatography–mass spectrometry. Clin Chem 43: 1029–1032. Brugmann G (1981). Gas chromatographic determination of phenytoin, phenobarbital and primidone: flash-methylation after direct addition of trimethyl-phenyl-ammonium hydroxide to the ethyl acetate extract (author’s transl.). J Clin Chem Clin Biochem 19: 305–306. Carreras D et al. (1994). Comparison of derivatization procedures for the determination of diuretics in urine by gas chromatography–mass spectrometry. J Chromatogr A 683: 195–202. Chen BH et al. (1990). Comparison of derivatives for determination of codeine and morphine by gas chromatography/mass spectrometry. J Anal Toxicol 14: 12–17. CND Analytical (1989). Analytical Profile of the Anabolic Steroids. Auburn, AL: CND Analytical Inc. Cody JT, Schwarzhoff R (1993). Interpretation of methamphetamine and amphetamine enantiomer data. J Anal Toxicol 17: 321–326. Davies NM (1997). Methods of analysis of chiral non-steroidal anti-inflammatory drugs. J Chromatogr B Biomed Sci Appl 691: 229–261. Dawling S et al. (1990). Rapid measurement of basic drugs in blood applied to clinical and forensic toxicology. Ann Clin Biochem 27(Pt5): 473–477. De Zeeuw RA (2002). Gas Chromatographic Retention Indices of Toxicologically Relevant Substances on Packed or Capillary Columns with Dimethylsilicone Stationary Phases, 3rd edn. New York: Wiley. Dudley KH (1980). Trace organic sample handling. In: Reid E, ed. Methodological Surveys Sub-series (A). Chichester: Ellis Horwood, 336. Duncan A (2000). Screening for surreptitious laxative abuse. Ann Clin Biochem 37 (Pt1): 1–8. Flanagan RJ, Berry DJ (1977). Routine analysis of barbiturates and some other hypnotic drugs in the blood plasma as an aid to the diagnosis of acute poisoning. J Chromatogr 131: 131–146. Flanagan RJ et al. (1997). Volatile Substance Abuse. United Nations International Drug Control Programme Technical Series Number 5. Vienna: UNIDCP. Franke JP et al. (1993). An overview on the standardization of chromatographic methods for screening analysis in toxicology by means of retention indices and secondary standards. Fresenius J Anal Chem 347: 67–72. Gorecki T, Poerschmann J (2001). In-column pyrolysis: a new approach to an old problem. Anal Chem 73: 2012–2017. Grinstead GF (1991). A closer look at acetyl and pentafluoropropionyl derivatives for quantitative analysis of morphine and codeine by gas chromatography/mass spectrometry. J Anal Toxicol 15: 293–298. Gyllenhaal O et al. (1978). Electron-capture gas chromatography of sulphonylureas after extractive alkylation. J Chromatogr 156: 275–283. Hartvig P et al. (1980). Electron-capture gas chromatography of plasma sulphonylureas after extractive methylation. J Chromatogr 181: 17–24.
Junting L, Chuichang F (1991). Solid phase extraction method for rapid isolation and clean-up of some synthetic pyrethroid insecticides from human urine and plasma. Forensic Sci Int 51: 89–93. Kovats E (1961). Zusammenh€ange zwischen strucktur und gaschromatographischen daten organischer verbindungen. Fresenius Z Anal Chem 181: 351–366. Lee J et al. (1998). The effect of organic solvents on the determination of cyclic boronates of some beta-blockers by gas chromatography/mass spectrometry. Rapid Commun Mass Spectrom 12: 1150–1160. Leloux MS et al. (1989). Improved screening method for beta-blockers in urine using solid-phase extraction and capillary gas chromatography-mass spectrometry. J Chromatogr 488: 357–367. Lho DS et al. (1990). Determination of phenolalkylamines, narcotic analgesics, and beta-blockers by gas chromatography/mass spectrometry. J Anal Toxicol 14: 77–83. Lisi AM et al. (1991). Screening for diuretics in human urine by gas chromatography-mass spectrometry with derivatisation by direct extractive alkylation. J Chromatogr 563: 257–270. Liu RH et al. (1994). Improved gas chromatography/mass spectrometry analysis of barbiturates in urine using centrifuge-based solid-phase extraction, methylation, with d5-pentobarbital as internal standard. J Forensic Sci 39: 1504–1514. Lurie IS et al. (1994). The determination of anabolic steroids by MECC, gradient HPLC, and capillary GC. J Forensic Sci 39: 74–85. Maurer HH, Arlt JW (1998). Detection of 4-hydroxycoumarin anticoagulants and their metabolites in urine as part of a systematic toxicological analysis procedure for acidic drugs and poisons by gas chromatography–mass spectrometry after extractive methylation. J Chromatogr B Biomed Sci Appl 714: 181–195. Maurer HH, Arlt JW (1999). Screening procedure for detection of dihydropyridine calcium channel blocker metabolites in urine as part of a systematic toxicological analysis procedure for acidic compounds by gas chromatography–mass spectrometry after extractive methylation. J Anal Toxicol 23: 73–80. Maurer HH, Bickeboeller-Friedrich J (2000). Screening procedure for detection of antidepressants of the selective serotonin reuptake inhibitor type and their metabolites in urine as part of a modified systematic toxicological analysis procedure using gas chromatography–mass spectrometry. J Anal Toxicol 24: 340–347. Maurer HH, Pfleger K (1984). Screening procedure for the detection of opioids, other potent analgesics and their metabolites in urine using a computerized gas chromatographic–mass spectrometric technique. Fresenius Z Anal Chem 317: 42–52. Maurer HH, Pfleger K (1986). Identification and differentiation of beta-blockers and their metabolites in urine by computerized gas chromatography–mass spectrometry. J Chromatogr 382: 147–165. Maurer HH, Pfleger K (1987). Identification and differentiation of benzodiazepines and their metabolites in urine by computerized gas chromatography–mass spectrometry. J Chromatogr 422: 85–101. Maurer HH et al. (1998). Screening for the detection of angiotensin-converting enzyme inhibitors, their metabolites, and AT II receptor antagonists. Ther Drug Monit 20: 706–713. Maurer HH et al. (2001). Screening procedure for detection of non-steroidal anti-inflammatory drugs and their metabolites in urine as part of a systematic toxicological analysis procedure for acidic drugs and poisons by gas chromatography-mass spectrometry after extractive methylation. J Anal Toxicol 25: 237–244. Maurer HH et al. (2002). Screening, library-assisted identification and validated quantification of oral antidiabetics of the sulfonylurea-type in plasma by atmospheric pressure chemical ionization liquid chromatography–mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 773: 63–73. McReynolds WO (1970). Characterization of some liquid phases. J Chromatogr Sci 8: 685–691.
Clarke's Analysis of Drugs and Poisons Chapter No. 40 Dated: 10/3/2011 At Time: 15:38:25
Further reading Meatherall R (1999). GC-MS confirmation of codeine, morphine, 6-acetylmorphine, hydrocodone, hydromorphone, oxycodone, and oxymorphone in urine. J Anal Toxicol 23: 177–186. Moffat AC et al. (1974). Otimum use of paper, thin-layer and gas-liquid chromatography for the identification of basic drugs. I. Determination of effectiveness for a series of chromatographic systems. J Chromatogr 90: 1–7. Moffat AC et al. (1974). Optimum use of paper, thin-layer and gas–liquid chromatography for the identification of basic drugs. III. Gas–liquid chromatography. J Chromatogr 90: 19–33. Nagai T, Kamiyama S (1991). Simultaneous HPLC analysis of optical isomers of methamphetamine and its metabolites, and stereoselective metabolism of racemic methamphetamine in rat urine. J Anal Toxicol 15: 299–304. Nakahara Y et al. (1997). Hair analysis for drugs of abuse. XVII. Simultaneous detection of PCP, PCHP, and PCPdiol in human hair for confirmation of PCP use. J Anal Toxicol 21: 356–362. Nelson CC, Foltz RL (1992). Determination of lysergic acid diethylamide (LSD), iso-LSD, and N-demethyl-LSD in body fluids by gas chromatography/tandem mass spectrometry. Anal Chem 64: 1578–1585. Osselton MD, Snelling RD (1986). Chromatographic identification of pesticides. J Chromatogr 368: 265–271. Paroni R et al. (2000). Comparison of capillary electrophoresis with HPLC for diagnosis of factitious hypoglycemia. Clin Chem 46: 1773–1780. Paul BD et al. (1999). A practical approach to determine cutoff concentrations for opiate testing with simultaneous detection of codeine, morphine, and 6-acetylmorphine in urine. Clin Chem 45: 510–519. Pfleger K et al. (2004). Mass Spectral and GC Data of Drugs, Poisons, Pesticides, Pollutants and their Metabolites, Part 5, 2 edn. Weinheim: Wiley-VCH. Pierce KM et al. (2008). Recent advancements in comprehensive two-dimensional separations with chemometrics. J Chromatogr A 1184: 341–352. Ramsey JD, Flanagan RJ (1982). Detection and identification of volatile organic compounds in blood by headspace gas chromatography as an aid to the diagnosis of solvent abuse. J Chromatogr 240: 423–444. Reeves VB (1999). Confirmation of multiple sulfonamide residues in bovine milk by gas chromatography-positive chemical ionization mass spectrometry. J Chromatogr B Biomed Sci Appl 723: 127–137. Rorschneider L (1966). Eine methode zur charakterisierung von gaschromatographischen trennflussigkeiten. J Chromatogr 22: 6–22. Schuetz H, Westenberger V (1978). GLC-data of 19 hydrolysis-derivatives rised from 12 important benzodiazepines and 17 main-metabolites (author’s transl.). Z Rechtsmed 82: 43–53.
717
Schuetz H, Westenberger V (1979). Gas chromatographic data of 31 benzodiazepines and metabolites. J Chromatogr 169: 409–411. Scott RPW (1996). Chromatographic Detectors: Design, function, and operation. Chromatographic Science Series, 73, Cazes J, ed. New York: Marcel Dekker. Sharp ME (2001). A comprehensive screen for volatile organic compounds in biological fluids. J Anal Toxicol 25: 631–636. Stern EL, Caron GP (1977). Measuring barbiturates, sedatives, and anticonvulsants in serum by gas–liquid chromatography. Am J Med Technol 43: 834–842. Supelco (1979). Supelco Bulletin. Bellefonte, PA: Supelco Inc, p. 779. Supelco (2000). Supelco Catalogue test chromatograms. Supelco Catalogue 2000, pp. 506-508. Tarbin JA et al. (1999). Screening of sulphonamides in egg using gas chromatography–mass-selective detection and liquid chromatography–mass spectrometry. J Chromatogr B Biomed Sci Appl 729: 127–138. Yoon CN et al. (1990). Mass spectrometry of methyl and methyl-d3 derivatives of diuretic agents. J Anal Toxicol 14: 96–101.
Further reading Blau K, Halket J (1993). Handbook of Derivatives for Chromatography, 2nd edn. New York: Wiley. Grob K (1993a). On-Column Injection in Capillary Gas Chromatography. Heidelberg: H€ uthig. Grob K (1993b). Split and Splitless Injection in Capillary Gas Chromatography. Heidelberg: H€ uthig. Hill HH, McMinn DG (1992). Detectors for Capillary Chromatography. New York: Wiley. Jennings W et al. (1997) Analytical Gas Chromatography, 2nd edn. London: Academic Press. Jinno K (1997). Chromatographic Separations Based on Molecular Recognition. New York: Wiley. Rood D (1995). A Practical Guide to the Care, Maintenance, and Troubleshooting of Capillary Gas Chromatographic Systems, 2nd edn. Heidelberg: H€ uthig. Scott RPW (1996). Chromatographic Detectors: Design, Function, and Operation. Chromatographic Science Series, 73, Cazes J, ed. New York: Marcel Dekker. Stevenson D, Wilson ID (1994) Sample Preparation for Biomedical and Environmental Analysis. New York: Plenum Press.
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:1
CHAPTER
41
High Performance Liquid Chromatography T Kupiec and P Kemp
Introduction The ability to separate and analyse complex samples, both small and large molecules, is important to the biological and medical sciences. Classic column chromatography has evolved over the years, with chromatographic innovations introduced at roughly decade intervals. These techniques offered major improvements in speed, resolving power, detection, quantification, convenience and applicability to new sample types. The most notable of these modifications was high performance liquid chromatography (HPLC). Modern HPLC techniques became available in 1969; however, they were not widely accepted in the pharmaceutical industry until several years later. Once HPLC systems capable of quantitative analysis became commercially available, their usefulness in pharmaceutical analysis was fully appreciated. By the 1990s, HPLC had begun an explosive growth that made it a popular analytical method judged by sales of instruments and also scientific importance. During the last decade developments in chromatographic supports and instrumentation for liquid chromatography have continued to evolve. The use of silica-based monolithic supports, elevated mobile phase temperatures and columns packed with particles 50 C) are used for variation and single-nucleotide polymorphism (SNP) discovery and detection are based on DNA fragment sequence as described above. Fully denaturing temperatures (>70 C) can be used for the separation and analysis of oligonucleotides and nucleic acids as illustrated in Figure 41.9. On-line sample preparation The preparation of samples typically demands a large amount of time, work and cost in an analytical laboratory. The innovation of on-line sample preparation makes the process more efficient and reduces the cost. On-line sample preparation techniques usually involve direct elution of the extract from a solid-phase extraction cartridge into the system by the mobile phase. The on-line method gives superior analytical results and can be automated fully. Another benefit is that the sample preparation is reliable, reproducible and robust. This sample preparation method is also discussed in the column-switching section of this chapter. Rapid screening The need for high throughput in a laboratory environment is constantly increasing. The use of short, highly efficient, analytical columns, rapid gradients and column-switching apparatus in HPLC systems is helping to facilitate this. Sample turnaround time can often be reduced to a few minutes or less in highly automated and optimised systems. Other information on this topic is given earlier in this chapter in the sections on gradients and column switching.
Systems for drug analysis Eluent systems A large number of eluent and/or packing material combinations have been used for drug analysis. However, currently most are performed on silica or one of the hydrocarbon-bonded silicas (usually ODS). Other types of packing are employed when these conventional materials fail. The majority of drug analyses can be carried out with the four types of system described next. Silica with non-polar eluents
With silica normal-phase systems the principal mechanism is adsorption chromatography. Separation is controlled by the competition between solute molecules and molecules of the mobile phase for the adsorption sites on the silica surface. Polar groups are attracted most strongly to these sites and hence polar compounds are retained more strongly than non-polar ones. Retention can be decreased by increasing the polarity of the eluent. Adsorption energies of numerous solvents on alumina (e values are given in Table 41.1) have been measured and this scale can be used as a good guide to the elution strengths of eluents on silica as well as alumina (Snyder 1968). Mixtures of solvents can be employed to give elution strengths between those of the pure solvents. Furthermore, different solvent mixtures that have the same e value often give different separations of a group of compounds. Water is strongly bound to silica and thus the water content of the eluent must be controlled strictly to maintain constant activity of the silica surface and hence reproducible retention times. This is most critical when the eluent is of very low polarity. However, because anhydrous systems are difficult to maintain, a low concentration of water can be used in the eluent, sufficient to deactivate the most active sites without deactivating the whole surface. Typical water concentrations range from 0.01% to 0.2% (v/v). The most satisfactory method used to prepare a solvent of known water content is to mix anhydrous and water-saturated solvents in known proportions. Anhydrous hydrocarbon or halo hydrocarbon solvents can be prepared by passing them through a bed of activated silica or alumina (200 mm) in a glass column. The problems associated with the control of water concentration mean
Table 41.1 e values for numerous solvents on alumina (Snyder 1968)
70-80°C Heat
(a) EP
+C
(b)
+T +G
+A
Time
Figure 41.9 Principle of completely denaturing HPLC. (a) At column temperatures >70 C, double-stranded DNA fragments will denature completely. (b) The single-stranded components can then be resolved from each other even if they differ only in sequence and not in size. The chromatogram depicts the separation of an extension primer (EP) and the four possible isomeric products generated by single nucleotide extension sequencing. Resolution for any given pair of alleles can be optimised by varying column temperature. The order of elution of the alleles depends on the stationary phase used. On poly(styrene-divinylbenzene) monoliths, as depicted, extension products elute in the order C < G < T < A. On micropellicular alkylated poly(styrene-divinylbenzene) particles, in contrast, the elution order is G < C < A < T.
731
Solvent
e
Pentane
0.00
Hexane
0.01
Isooctane
0.01
Cyclohexane
0.04
Toluene
0.29
1-Chlorobutane
0.30
Ether
0.38
Chloroform
0.40
Methylene chloride
0.42
Tetrahydrofuran
0.45
Acetone
0.56
Ethyl acetate
0.58
Diethylamine
0.63
Acetonitrile
0.65
Isopropyl alcohol
0.82
Ethanol
0.88
Methanol
0.95
Acetic acid
Large
Water
Large
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:6
732
High Performance Liquid Chromatography
that commonly alcohols, such as methanol (0.01–0.5% v/v), are employed to moderate the silica surface (Engelhard 1977). Silica with polar eluents
Several systems have been described that involve the use of silica with eluents of moderate-to-high polarity containing alcohols and/or water as major components. With such eluents, adsorption chromatography is most probably not the principal mechanism. The mechanisms are poorly understood, which makes the prediction of retention behaviour difficult; nevertheless, many of these systems are very useful for drug analysis. An eluent that consists of methanol–ammonium nitrate buffer (90 : 10) is suitable for a wide range of basic drugs (e.g. amfetamines and opiates). Retention can be controlled by changes to the pH, ionic strength or methanol : water ratio, or by the addition of other organic solvents such as methylene chloride. With these alkaline eluents the silica surface must bear a negative charge and the principal mechanism is probably cation exchange. Benzodiazepines can be chromatographed with methanol eluents that contain perchloric acid (typically 0.001 mol/L). Retention can be modified by the addition of other organic solvents (e.g. ether) or by changes to the acid concentration. Both acidic and basic drugs can be chromatographed on silica using aqueous methanolic eluents that contain cetyltrimethylammonium bromide (Hansen 1981). Hydrophobic quaternary ammonium ions are strongly adsorbed on silica to give a dynamically coated stationary phase. Retention may be controlled by varying the concentration or nature of the quaternary ammonium ion, changing the ionic strength or pH of the buffer, or changing the concentration or nature of the organic component. ODS with polar eluents
Eluents for RPC on ODS are usually mixtures of methanol or acetonitrile with an aqueous buffer solution. Retention is controlled mainly by the hydrophobic interactions between the drugs and the alkyl chains on the packing material. Retention increases as the analytes decrease in polarity (i.e. polar species are eluted first). Hence, the elution time is increased by increasing the polarity of the eluent (i.e. increasing the water content). The pH of the eluent and the pKa of the drug are also important, since non-ionised species show greater retention. Thus, acids show an increase in retention as the pH is reduced, while bases show a decrease. It is important to use a buffer of sufficient capacity to cope with any injected sample size, otherwise tailing peaks can arise from changes in ionic form during chromatography. Phosphate buffers (0.05–0.2 mol/L) are widely used as they have a good pH range and low UV absorbance. Drugs that contain basic nitrogen atoms sometimes show poor efficiencies and give tailing peaks caused by interactions with residual silanol groups on the packing material. This can often be improved by the addition of an amine or quaternary ammonium compound to the eluent, which competes with the analytes for adsorption sites on the silica. Amines of small molecular weight (e.g. diethylamine) can be used as part of the buffer system. Alternatively, low concentrations (0.001 mol/L) of long-chain hydrophobic modifiers (e.g. N,N-dimethyloctylamine) can be added to eluents together with conventional buffers. Other hydrocarbon-bonded packing materials can be used in RPC. A decrease in retention is associated with a decrease in the alkyl chain length. ODS with polar eluents that contain hydrophobic cations or anions
Drugs that bear positive or negative charges are retained poorly in reversed-phase systems. If the pH of the eluent cannot be changed to convert the drug into its non-ionised form, a hydrophobic ion of opposite charge can be added to form a neutral ion-pair and increase retention. Hence, for a basic drug an acidic eluent is chosen and a hydrophobic anion is added. This technique is referred to as reversedphase ion-pair chromatography. The sodium salts of alkylsulfonic acids (RSO3 Naþ, where R ¼ pentyl, hexyl, heptyl or octyl) are used widely as ion-pair reagents for basic drugs, while quaternary ammonium compounds (e.g. tetrabutylammonium salts) are used for acidic drugs. Ion-pair reagents are generally added to eluents in the concentration range 0.001–0.005 mol/ L, and within this range an increase in concentration leads to an increase
in retention. When detergents such as sodium lauryl sulfate or cetyltrimethylammonium bromide are used as the ion-pair reagents, the method is sometimes referred to as ‘soap chromatography’. With these salts, ions build up on the surface of the packing material and produce a stationary phase, which behaves like an ion exchanger. This type of mechanism has been described as ‘dynamic ion exchange’ and probably also occurs with less hydrophobic ion-pair reagents. It is virtually impossible to remove an ion-pair reagent completely from a hydrocarbon-bonded phase, and such columns should therefore not be re-used with other reversed-phase eluents. Selection of chromatographic systems Many different combinations of packing material and eluent may be suitable for the analysis of a particular compound or group of compounds and the final choice can be influenced by many factors. The time required to develop a new system can be shortened if it is possible to predict the way in which changes in eluent composition influence chromatographic retention. Systems that use hydrocarbon-bonded phases are particularly attractive from this viewpoint as a large range of parameters can be adjusted (pH, organic solvent, ionic strength, ion-pair reagents) with largely foreseeable consequences. Predictions for silica are generally less reliable. Silica is good for separating drugs that belong to different chemical classes, while hydrocarbon-bonded silicas are preferred for separations of drugs with closely related structures (e.g. barbiturates). Most of the endogenous materials in biological extracts that can interfere with the analysis of a drug are fairly polar. In reversed-phase systems this material generally elutes before the drug and can obscure the drug peak. In these circumstances, reversed-phase ion-pair chromatography can be valuable to increase selectively the retention of the drug relative to the interfering peaks. Normal-phase systems that use silica do not generally suffer from this problem, as most of the endogenous material usually elutes after the drug. However, these slow-eluting compounds can lead to a noisy baseline or may remain adsorbed to the packing material and thus eventually lead to a loss in column performance. The vast majority of compounds are separated using a silica-based column with C18, and fine-tuning of the separation can be done by selecting a column with a shorter bonded phase, such as C8 (see later). Specially end-capped columns are available that are designed to minimise the tailing that is common with nitrogen-containing weak bases. These are often marketed as a ‘basic’ column (e.g. Metachem’s MetaSil Basic). There are also specially end-capped columns designed to withstand extremely high concentrations of aqueous mobile phase (95–100%). These columns are end-capped with a hydrophilic moiety that ensures proper ‘wetting’ of the silica to prevent bonded-phase collapse. The columns are typically marketed as ‘AQ’ for aqueous (e.g. YMC’s ODS-AQ). Analysis of drugs in pharmaceutical preparations HPLC has found widespread use for the quantitative analysis of drugs in preparations of pharmaceutical and illicit manufacture. Drug concentrations are generally high enough to allow dissolution of the sample (tablet, powder, ointment, etc.) in a suitable solvent followed by injection. UV, visible, FL, RI or mass spectrometric detection methods are used often. These techniques are well suited to provide specific data regarding the chemical composition of the sample in question (e.g. a UV spectrum, mass spectrum). Within the pharmaceutical industry, HPLC is used at various stages of drug development, such as the optimisation of synthetic reactions and stability testing. Furthermore, it is used extensively for quality control during production to monitor the purity of drugs and excipients. HPLC systems can be automated easily (including injection and data handling), which allows large numbers of samples to be analysed rapidly and economically. HPLC is particularly valuable for the analysis of drugs that are polar (e.g. aspirin), thermally unstable (e.g. benzodiazepines) or present in oil-based formulations for which analysis by GC can be very difficult. Similarly, HPLC can be used for the forensic analysis of illicit preparations to aid the identification of an unknown drug by the measurement of retention times and UV spectra and
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:6
Systems for drug analysis comparison to spectral libraries. Furthermore, as the technique can be non-destructive, depending on the detection system used, the eluted compounds can be collected for further analysis. Example of a drug analysis system
Opiates have been separated by many methods in the past, and the system described here was developed for this purpose. The three opiates separated were morphine sulfate, hydrocodone bitartrate and oxycodone hydrochloride. The column used was a Phenomenex Luna C18 (2), 150 4.60 mm 5 mm. The mobile phase was 39 mmol/L dipotassium hydrogenphosphate (K2HPO4) and methanol in a 40 : 60 ratio. The final pH was 10 and the mobile phase flow rate was 1.0 mL/min. The retention times obtained (Figure 41.10) for morphine sulfate, hydrocodone bitartrate and oxycodone hydrochloride were 2.799, 4.696 and 6.143 min, respectively. Analysis of drugs in biological fluids and tissues Several factors determine the ability of HPLC to detect a drug among the endogenous compounds present in biological material. Clearly, selective detection of the drug relative to the endogenous material is advantageous. In addition, the stationary phase and/or mobile phase can be altered to separate the drug peak from interfering peaks (e.g. using ionpair reagents). Finally, the sample may be extracted before HPLC to concentrate the drug relative to the endogenous material. The chromatographic system and detector should always be chosen to minimise the time needed for sample preparation. The complexity of the sample preparation procedure is controlled by several factors, which include the nature of the sample (urine, blood, liver, etc.), the condition of the sample and the concentration of the drug. Interference from endogenous compounds is most acute when drug concentrations are low (e.g. in therapeutic drug monitoring), so more extensive sample preparation and more sensitive and specific detectors are often required. Such assays can be very susceptible to changes in the condition of the sample (e.g. a method developed for fresh blood may not be satisfactory for urine or hair samples), which can present severe difficulties in forensic toxicology. Thus, methods should be tested and validated with the most difficult samples that may be encountered. In contrast, the analysis by HPLC of biological samples that contain high drug concentrations (e.g. in fatal drug overdose) may require much less sample preparation and is less susceptible to changes in sample condition. Sample preparation for HPLC is essentially the same as for other methods of drug analysis. A drug that is physically trapped within solid tissue (e.g. liver), or chemically bound to the surface of proteins, must be
733
released; then the protein is precipitated to leave the drug in aqueous solution. The protein may be degraded by strong acids or enzymes, precipitated by various chemicals (e.g. tungstic acid, ammonium sulfate) or removed by ultrafiltration. Some drugs are destroyed by protein degradation methods, while ultrafiltration and precipitation can lead to drug losses through protein binding. No single procedure works well for all drugs and the method should be selected to give the maximum recovery of the drug being analysed. When drug concentrations are high (typically mg/mL) and systems with polar mobile phases are used, the direct injection of deproteinised solutions may be acceptable. Proteins must be removed to protect the column from irreversible contamination. A rapid procedure is to: mix the biological fluid with at least two volumes of methanol or acetonitrile; centrifuge to remove the precipitated protein; evaporate the organic supernatant; and reconstitute the sample in a volume of mobile phase. Urine can be treated similarly to guard against the precipitation of salts on the column. Great care and consideration should be afforded when injecting minimally prepared biological samples on to a HPLC system. Particulates are more likely to become trapped in the system plumbing and a more rapid degradation of column performance may be observed from build-up of contaminants on the head of the column. To help maximise column performance and lifetime, it is good policy to use a guard column between the injector and analytical column. This is packed with the same material as the analytical column and replaced at frequent intervals. The configurations of guard columns range from easily replaceable and relatively inexpensive frit-like filters and/or cartridges to shorter versions of the analytical column itself. All are designed to protect the analytical column by acting as a trap for components that would otherwise irreversibly bind to the analytical column and thus decrease the usable life of the column. Extraction of drugs and other analytes away from endogenous materials prior to analysis is a common procedure for all types of biological samples. This may also entail a concentration step, which increases the sensitivity of the method. Solvent extraction remains the most popular approach, as many factors can be modified to optimise the extraction. These modifications include changing the polarity of the organic solvent, the pH and ionic strength of the aqueous phase, and the use of ion-pairing agents. It is generally recommended that the collected organic phase be evaporated to dryness and the residue dissolved in a suitable solvent, typically something greater than or equal to the polarity and composition of the initial mobile phase before injection. Care must be taken that volatile drugs are not lost by evaporation and that lipid material in the residue does not prevent the drug from dissolving in the new solvent.
Figure 41.10 Separation of opiates by HPLC. Conditions for separation are described in the text.
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:6
734
High Performance Liquid Chromatography
Example protocol for the extraction of a wide variety of weak bases n n n n n
n
To 1 mL of plasma, urine or other homogenised matrix add 100 mL concentrated ammonium hydroxide. Extract the sample with 4 mL of a mixture of n-butyl chloride– acetonitrile (4 : 1) for 20 min. Centrifuge at high speed for 20–30 min to partition the phases. Carefully collect the organic phase into a clean tube. Evaporate the organic phase under a stream of air or nitrogen at 25– 40 C, depending on the volatility of the analytes (a small volume of acidified methanol can be added to prevent the loss of amfetaminetype analytes). Reconstitute the residue in an HPLC mobile phase that is more polar than the LC mobile phase to be used for analysis (e.g. if the HPLC elution ratio is 60% aqueous, reconstitute the sample in a phase that is >60% aqueous). This ensures that, when injected, the sample is focused on the front end of the column and minimises band (peak) broadening.
An example of a chromatogram that utilises this extraction technique is shown in Figure 41.11. The urine was fortified with analytes and deuterated internal standards for amfetamine and methamfetamine (dashed chromatograms) and extracted as described above. The sample was eluted using a MetaSil Basic 3 100 mm 3 mm column. The mobile phase was 85% (0.1% formic acid in water), 15% methanol, pumped isocratically at 0.2 mL/min. The instrument used was an Agilent 1100 LC/MSD with ESI. SPE columns are also widely used to extract drugs from biological samples. The column is washed with suitable solvents to remove endogenous material before the drug is removed by passing through a solvent of higher elution strength. Such columns are usually attached to extraction manifolds utilising either positive or negative pressure to draw the liquids through the sorbent beds. Extraction selectivity can be controlled by adjustments to the biological fluid before extraction (e.g. pH, ionic strength) and the choice of washing solvents. Most, if not all, manufacturers of SPE columns offer methods and columns optimised for a particular drug class and/or matrix. As less traditional biological matrices are used for drug analysis (e.g. sweat, hair, oral fluids), some modifications of the sample preparation scheme are needed. Hair requires solubilisation prior to extraction; oral fluids and sweat may need to be isolated from their respective collection devices. Consideration of the pH and solubility may be needed prior to sample preparation, but in general the principles in
place for the extraction of blood, urine, etc. apply to these alternative matrices. Some important issues unique to these matrices are: n n
Sample volume is typically much less than blood or urine. The amount of drug extracted from a particular matrix may be much less than from traditional matrices, so that much more sensitive detectors (e.g. MS or MS–MS) are required.
Identification of drugs by HPLC with photodiode array detection and UV spectra library search HPLC with DAD in combination with a UV spectra library has proved to be a very successful ‘systematic toxicological analysis’ (STA) technique for use in clinical and forensic toxicology (see Chapter 1). Any drugs or other poisons in the sample are identified by coincidence of the UV spectrum and of the retention time or another chromatographic retention parameter with the library data; one system and its use are described below (F Pragst and M Herzler, personal communication). Chromatographic conditions
Since the method is used in combination with a database of UV spectra and retention parameters, the chromatographic conditions must be reproducible and the same as used to generate the database. The mobile phase must be suitable for the separation of a large variety of organic substances and must be transparent in the wavelength range used. These prerequisites are best met by reversed-phase columns (RP8 or RP18) and acidic acetonitrile–buffer mixtures as mobile phases. Systems described in the literature generally use either a gradient elution or two isocratic runs with different buffer : acetonitrile ratios. Gradient elution has the advantages that strongly polar and non-polar substances can be analysed in one run, that peaks are not broadened with increasing retention time, and that the retention times of the toxicologically relevant compounds are distributed more evenly over the run time, but it has some disadvantages (see above). A system of HPLC retention indices was introduced by Bogusz et al. (1993) analogous to the Kovats indices used in GC and based on the retention times of the nitroalkanes. Isocratic HPLC has the advantage of higher reproducibility of the retention times, greater ruggedness and a more economic use of the mobile phase by recycling. Disadvantages are an unfavourable distribution of the retention times of toxicologically relevant compounds with an increased number at the beginning of the chromatogram, and the need for a second mobile phase for non-polar compounds. Nevertheless, isocratic HPLC–DAD procedures are used successfully in many toxicological laboratories for screening purposes. Suitable experimental
Figure 41.11 Separation of amfetamines by HPLC–MS. Conditions for separation are described in the text.
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:8
Systems for drug analysis conditions, also used in the recording of an extensive UV spectra library, were as follows (Pragst et al. 2001): n n n n
HPLC column: RP8, end-capped, 5 mm, 250 4.0 mm Mobile phase A: 0.1 mol/L phosphate buffer pH 2.3–acetonitrile (67 : 33 v/v) Mobile phase B: 0.1 mol/L phosphate buffer pH 2.3–acetonitrile (33 : 67 v/v) Flow rate: 1 mL/min.
Standard compounds are histamine hydrochloride to measure the time of an unretained peak t0 (dead time), 5-(4-methylphenyl)-5-phenylhydantoin (MPPH) to calculate relative retention times (RRTs) in mobile phase A, and 4-phenylbenzophenone to calculate RRTs in mobile phase B. The UV spectra of a large number of compounds listed in this book were measured under these conditions. An overview of HPLC–DAD conditions used for STA is given in Pragst et al. (2001). Retention parameters
Absolute retention times are not suitable for peak identification purposes, since they depend strongly on the configuration and experimental conditions of the HPLC device. Moreover, the capacity ratio kA (see above) is sensitive to small fluctuations of the experimental conditions and is not suitable for an identification system used in different laboratories. Therefore, for gradient elution, retention indices are preferred (Bogusz et al. 1993). Under isocratic conditions RRTs related to a standard compound are more reproducible (Equation 41.6). RRTx ¼
tx t0 ts t0
ð41:6Þ
where RRTx is the RRT of compound x, tx is the absolute retention time of compound x, t0 is the retention time of an unretained peak, and ts is the retention time of the standard compound. The relatively small peak resolution of HPLC and the differences between charges of the reversed-phase material mean that the value of retention indices or of RRTs in the identification of a compound from a large number of candidates is rather limited. However, it is very useful for distinguishing between compounds with very similar UV spectra. In this way an RRT window can be chosen as a pre-selection parameter for the spectra library search. UV spectral library search and specificity of UV spectra
Before peak identification a ‘peak purity check’ should be carried out. A pure peak means that it originates only from one compound and that the UV spectrum does not change over the whole peak width. A UV spectral library search is based on the comparison of the spectrum of the unknown peak with all spectra of the library. This comparison is not confined to UV maxima and minima, but can comprise all absorbance–wavelength pairs measured by DAD. Mathematical models to assess spectral similarity use the description of the spectrum as a vector in n-dimensional space, where n is the number of absorbance–wavelength pairs measured. For the complete identity of two spectra, both vectors point in exactly the same direction, that is the angle between them is u ¼ 0 . Different concentrations have an effect on vector length, but not on its direction in space. The similarity index (SI) is defined as cos u and is calculated with Equation (29.7): SIx ¼ cosus1 s2 ¼
s1 s2 js1 j js2 j
ð41:7Þ
where Si is the vectorised spectrum of compound i. UV spectra can be measured with extremely high reproducibility. Therefore, small differences between spectra measured under identical conditions indicate that they originate from different compounds. SI is 1.000 for completely identical spectra. However, in practice two spectra with SI >0.9990 can be regarded as identical. At small concentrations, and in the case of partly overlapping peaks, SI >0.990 may be a sufficient criterion for identity.
735
It was shown in a systematic study on the selectivity of an HPLC– DAD method (Herzler et al. 2003) that, from 2888 toxicologically relevant compounds, 2682 (93%) exhibited UV absorption above 195 nm. Out of these, 1619 (60.4%) had a unique UV spectrum and could be identified unambiguously. By inclusion of the retention time this proportion was increased to 84.2%. Large UV spectra libraries can be divided into sub-libraries, according to the retention parameter or the effect or use of the substance, to facilitate a faster and more specific library search. The result can also be supported by the presence of metabolites, while in doubtful cases complementary methods may be used for confirmation (e.g. MS). As an example, in Figure 41.12 the results of the library search for a peak with RRT ¼ 0.0811 in an intoxication case are shown. In this case a sub-library of all compounds with RRT ¼ 0.601–0.900 was used. Hit 1 was promethazine with SI ¼ 0.9992; hit 2 (promazine, SI ¼ 0.9964) and hit 3 (dixyrazine, SI ¼ 0.9961) also originated from compounds of the phenothiazine type. The small difference between the spectra of hits 1 and 2 may arise because in these two compounds the amino group of the side chain is separated from the phenothiazine ring by two and three saturated carbon atoms, respectively. Dixyrazine could clearly be excluded by the much smaller retention time. However, promethazine and promazine could not be distinguished by the RRT values stored in the database. Therefore, to confirm the library search result, promazine and promethazine standards were measured immediately after the sample, which resulted in an exact agreement with promethazine. As a prerequisite for the optimal use of a commercially available UV spectral library, the same mobile phase must be used and the technical parameters of the DAD (wavelength accuracy and resolution) need to be (and stay) sufficient. This can be controlled by daily measurement of a compound with a vibration fine structure of the UV spectrum, such as benzene. UV spectra and retention times of metabolites
The use of HPLC–DAD has the advantage that in many cases, metabolites can be attributed easily to the parent drug by the UV spectrum. Depending on the site of metabolism, the UV spectrum may be altered significantly (change of the UV-absorbing unsaturated part of the molecule, the chromophore) or it may be the same as (or very similar to) that of the parent drug (reaction at the aliphatic part of the molecule). As an example, in Figure 41.13 the spectrum of flunitrazepam is compared with that of its metabolites, 7-aminoflunitrazepam (strong change of the chromophore by transformation of the aromatically bound nitro group into the amino group) and 3-hydroxyflunitrazepam (no essential change of the chromophore by hydroxylation at the aliphatic carbon atom 3). The retention times of drugs on reversed-phase columns are shifted in a typical way by metabolism. Metabolism to more hydrophilic products (e.g. hydroxylation, reduction of the nitro to an amino group; Figure 41.13) leads to a decrease in retention time, whereas deamination strongly increases retention time, particularly in an acidic mobile phase (removal of the strongly hydrophilic, protonated amino group). For many drugs, the chromatograms obtained from blood or urine extracts have a typical metabolite pattern that supports identification in the context of STA. Sample pretreatment
Tablets, powders or residues in syringes can simply be dissolved in the mobile phase and analysed by HPLC-DAD without further treatment. The investigation of biological samples, such as whole blood (serum, plasma), stomach contents, urine or tissue samples, is more complicated. In these cases the drug must be separated from the biological matrix. Although SPE has been much improved in the past decade, liquid– liquid extraction (LLE) is still preferred if HPLC-DAD is used for toxicological screening, since it is less susceptible to interferences, more reproducible and easier to handle for single samples. An important advantage of UV detection is that cholesterol and fatty acids, coextracted to a high extent from human samples by lipophilic solvents, show no UV absorption and therefore, in contrast to GC-MS, do not interfere with the analysis. Moreover, derivatisation is not necessary. A sample pretreatment method by extraction with n-butyl chloride–acetonitrile (4 : 1), which can be used for a wide variety of basic compounds,
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:8
736
High Performance Liquid Chromatography
Figure 41.12 Results of the HPLC–DAD library search for a peak in the chromatogram of an alkaline extract of a lethal trimipramine–promethazine intoxication. Hit 1 (promethazine) was confirmed by exact agreement of the retention time with the reference compound measured immediately after the sample. sa, sample; li, library.
Figure 41.13 Change of the UV spectrum and the relative retention time (RRT) of flunitrazepam by metabolism.
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:9
Recommended HPLC systems is given above. For systematic toxicological screening of blood (serum, plasma) samples by HPLC-DAD, the measurement of two extracts obtained at pH 2 and pH 9 with dichloromethane and of the supernatant of a protein precipitation by acetonitrile has proved to be very useful (Pragst et al. 2002). Preparation of a basic and an acidic methylene chloride extract
1. Dispense 500 mL of whole blood, serum or plasma into two 1.5-mL vials. 2. To vial 1 add 100 mL of a 0.2 mol/L solution of tri(hydroxymethyl) amine (basic extract). 3. To vial 2 add 100 mL of 0.1 mol/L hydrochloric acid (acidic extract). 4. To both vials add 400 mL of dichloromethane. 5. Vortex mix the vials for 1 min and centrifuge. 6. Withdraw 200 mL of the dichloromethane extract and evaporate the solvent at room temperature under a stream of nitrogen. 7. Dissolve the residue in 100 mL of mobile phase. 8. Analyse 50 mL of each extract (basic extract in mobile phase A and acidic extract in mobile phase B).
General screens System HA
Jane I et al. (1985). J Chromatogr 323: 191–225. n n
n
Protein precipitation is particularly useful for hydrophilic drugs, which are extracted poorly by the procedure mentioned above. These include paracetamol, salicylic acid and lamotrigine. The limits of detection are between 0.01 and 0.1 mg/mL for dichloromethane extraction (depending on the extinction coefficient and on the extraction yield) and between 0.1 and 1 mg/mL for protein precipitation. Application example
In STA, the library search must be applied to all peaks of the HPLC-DAD chromatogram. As an example, the chromatogram at 225 nm of the basic extract from the blood sample of a lethal drug poisoning case and the UV spectra of the highest peaks are shown in Figure 41.14. To determine RRT, the standard compound (MPPH, peak no. 10, RRT ¼ 1.000) was added. From the remaining 11 peaks of the chromatogram, 7 could be identified by both UV spectrum and RRT. As the result, a high overdose of trimipramine and promethazine was found to be the cause of death. The extensive metabolism indicated that there had been a long survival time after drug ingestion. The similarities between the UV spectra of the parent drugs (peaks 9 and 12) and some of their metabolites (peak 8, and peaks 6 and 11, respectively) are also demonstrated in this case. On the other hand, the sulfoxides of promethazine (peak 3) and desmethylpromethazine (peak 2) show completely changed spectra because of the transformation that takes place directly at the UV-absorbing phenothiazine ring. Caffeine (peak 1) is found in almost all samples. The poor separation of peaks 4, 5 and 7 meant that the UV spectra were not suitable for a library search.
Recommended HPLC systems There are general screening methods based on gradient elution and retention indices that have proven value by many laboratories, and data from these are listed below (systems HA, HX, HZ, HY and HAA). Another (system HBK) is based on a combination of isocratic systems. The tabulated data are derived from systems in which groups of compounds have been chromatographed either as part of a general screening procedure or from systems that have been used specifically for that group of compounds. Other systems for the chromatography of individual compounds, especially those used for quantification, are given in the monographs. Chromatographic retention data are presented as k values as well as retention times (RTs), retention indices (RIs) and relative retention times (RRTs). Note In the tables, a dash indicates that no value is available for the compound, not that it does not elute.
Column: Silica Spherisorb S5W (125 4.9 mm i.d., 5 mm). Mobile phase: Solution containing 1.175 g (0.01 mol/L) ammonium perchlorate in 1 L methanol; adjust to pH 6.7 by the addition of 1 mL 0.1 mol/L sodium hydroxide in methanol. k values: Values for drugs in this system will be found in drug monographs and in the Indexes to Analytical Data; they are also included in the systems for specific groups of drugs that follow.
System HX
J Hartstra, JP Franke, RA de Zeeuw, personal communication. n n n
Protein precipitation by acetonitrile
1. To 500 mL of whole blood, serum or plasma add 500 mL of acetonitrile. 2. Vortex the mixture for 2 min and centrifuge. 3. Separate off the supernatant. 4. Analyse 50 mL in mobile phase A.
737
n n n n
Column: Lichrospher 60 RP-Select B (125 4.0 mm i.d., 5 mm) with precolumn Lichrospher 60 RP-Select B (4 4.0 mm i.d., 5 mm). Mobile phase: (A:B) triethylammonium phosphate buffer (25 mmol/ L, pH 3.0)–acetonitrile. Elution programme: (A:B) (100 : 0) to (30 : 70) in 30 min, hold 10 min, back to initial conditions in 3 min with equilibration for 10 min before next injection. Flow rate: 1 mL/min. Detection: DAD. Standards: Nitro-n-alkanes (C1 to C11) 10 mL in 10 mL acetonitrile. RI values: Values for drugs in this system will be found in the monographs and in the Indexes to Analytical Data; they are also included in the systems for specific groups of drugs that follow.
System HY
RK Watt, RA Waters, AC Moffat, unpublished information. n n n n
n n n
Column: C18 symmetry (250 4.6 mm i.d., 5 mm). Column temperature: 40 C. Mobile phase: (A:B) sulfuric acid (0.5 mL 2.5 mol/L) in water (500 mL)–sulfuric acid (0.5 mL 2.5 mol/L) in acetonitrile (500 mL). Elution programme: (98 : 2) for 3 min to (2 : 98) over 23 min, hold for 10 min, back to initial conditions over 2 min with equilibration of 8 min before next injection. Detection: DAD. Standards: Nitro-n-alkanes (C1 to C16) 10 mL in 10 mL acetonitrile. RI values: Values for drugs in this system will be found in the monographs and in the Indexes to Analytical Data; they are also included in the systems for specific groups of drugs that follow.
System HZ
Conemans JMH et al. http://www.zanob.nl/pages/LSShowElements Page_v2.asp?ListID¼1650&elemid¼29275&articleid¼133751&token¼ (accessed 14 December 2010). n n
n n n
Column: C18 end-capped LiChrospher 100 RP-18e (125 4.0 mm i.d., 5 mm), with precolumn LiChrocart 124-4. Mobile phase: Add 146 mL triethylamine and about 750 mL phosphoric acid to 530 mL water. Adjust pH to 3.3 using a 10% potassium hydroxide solution and finally add 470 mL acetonitrile. Flow rate: 0.6 mL/min. Detection: DAD. Retention times: Values for drugs in this system will be found in the monographs and in the Indexes to Analytical Data; they are also included in the systems for specific groups of drugs that follow.
System HAA
Gaillard Y, Pepin G. (1997). J Chromatogr A 763: 149–163. n n n n
Column: C8 Symmetry (250 4.6 mm i.d., 5 mm) with Symmetry C18 precolumn (20 mm). Column temperature: 30 C. Mobile phase: (A:B) phosphate buffer (pH 3.8)–acetonitrile. Elution programme: (85 : 15) for 6.5 min to (65 : 35) until 25 min to (20 : 80) for 3 min, and back to initial conditions for equilibration for 7 min.
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:9
738
High Performance Liquid Chromatography
Figure 41.14 HPLC–DAD investigation of a combined trimipramine–promethazine poisoning. Chromatogram of a basic extract of a venous blood sample, UV spectra of the highest peaks, results of the library search and semiquantitatively determined concentrations.
n
n
Flow rate: 1 mL/min for 6.5 min, then linear increase to 1.5 mL/min for 6.5–25 min and hold for 3 min (re-equilibration is made at 1.5 mL/min). Detection: DAD.
n
Retention times: Values for drugs in this system will be found in the monographs and in the Indexes to Analytical Data; they are also included in the systems for specific groups of drugs that follow.
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:13
Recommended HPLC systems Amfetamines, other stimulants and anorectics
System HBK
Pragst F et al. (2001) UV Spectra of Toxic Compounds. Heppenheim: Verlag Dr Dieter Helm. n n
n n n
739
Column: Lichrospher RP-8ec (250 4.0 mm i.d., 5 mm). Mobile phase: Three different composition are used: A: acetonitrile–phosphate buffer pH 2.3 (33 : 67). Internal standard: 5-(4methylphenyl)-5-phenylhydantoin (for compounds eluting within 30 min). B: acetonitrile–phosphate buffer pH 2.3 (67 : 33). Internal standard: 4-phenylbenzophenone (for compounds eluting after 30 min). C: acetonitrile–phosphate buffer pH 2.3 (20 : 80). Internal standard: salicylamide (for compounds with RRTs below 0.2). Flow rate: 1 mL/min. Detection: DAD. Note: The phosphate buffer is prepared by dissolving 4.8 g phosphoric acid (85%) and 6.66 g potassium dihydrogenphosphate in 1 L water, adjust pH to 2.3. Values for drugs in this system will only be found in the Indexes to Analytical Data.
Systems HA, HX or HY previously described may be used, or Systems HB or HC below. System HB
Gill R et al. (1981). J Chromatogr 218: 639–646. n n
Column: ODS Hypersil (250 5 mm i.d., 5 mm). Mobile phase: Solution containing 19.60 g (0.2 mol/L) phosphoric acid and 7.314 g (0.1 mol/L) diethylamine in 1 L of a 10% v/v solution of methanol; adjust the pH to 3.15 by the addition of sodium hydroxide solution.
System HC
Law B et al. (1984). J Chromatogr 301: 165–172. n n
Column: Silica Spherisorb (250 5 mm i.d., 5 mm). Mobile phase: Methanol–ammonium nitrate buffer solution (90 : 10). To prepare the buffer solution add 94 mL strong ammonia solution and 21.5 mL nitric acid to 884 mL water and adjust to pH 10 by the addition of strong ammonia solution.
Amfetamines, other stimulants and anorectics
Adrenaline
HA
HB
HC
HX
HY
k
k
k
RI
RI
–
–
0.63
–
–
0.98
244
–
Amfetamine
0.9
8.48
1.2
–
0.15
–
–
11.1
–
–
312
267
Caffeine
0.2
–
Cathine
1
Benzfetamine Brucine
4.39
0.26
–
–
0.83
–
–
Chlorphentermine
0.9
–
0.82
–
–
Diethylpropion
1.7
–
0.16
–
230
Dimethylamfetamine
–
11.08
1.89
–
–
DOM
–
–
1.13
–
–
1.79
–
–
–
0.72
354
309
–
0.27
–
–
1.3
–
0.88
371
315
1
–
–
–
–
–
–
–
226 –
Ephedrine
1.0
Fencamfamin
1.3
Fenethylline
–
Fenfluramine Norfenfluramine Fenproporex
–
5.68
Hordenine
–
2.00
–
–
Hydroxyamfetamine
–
2.24
1.11
–
–
Hydroxyephedrine
–
0.73
–
–
–
Mazindol
1.8
–
0.2
357
286
Mephentermine
1.5
–
2.48
–
–
Mescaline
1.3
16.82
2.17
–
–
2
10.52
2.07
262
216
14.95
–
–
–
32.17
–
–
– –
Metamfetamine Methoxyamfetamine
–
Methoxyphenamine
1.7
Methylamfetamine Methylenedioxymethamfetamine
10.52
2.07
–
–
–
278
252
2.3
–
1.83
–
–
1.7
–
2.0 –
Methylephedrine Methylphenidate Noradrenaline
–
Normetanephrine
–
Oxedrine
–
0.10 – 0.27 0.2
–
Phendimetrazine
0.9
–
Phenelzine
1.0
Pemoline
5.91
0.36
–
277
–
–
–
1.08
–
–
–
–
–
0.1
307
271
0.3
263
218
0.37
–
– table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:16
740
High Performance Liquid Chromatography
Amfetamines, other stimulants and anorectics, continued HA
HB
HC
k
k
k
RI
RI
1.31
–
–
3.64
HX
HY
Phenethylamine
1.2
Phenmetrazine
1.7
–
–
258
241
Phentermine
0.6
19.46
0.86
–
245
Phenylephrine
1.3
–
1.64
–
–
Phenylpropanolamine
0.9
0.70
–
–
Pipradrol
1.2
–
0.69
355
–
Prolintane
2
–
1.3
370
–
Pseudoephedrine
1.2
1.77
–
–
3.87
5.90
Tranylcypromine
1.0
–
0.26
–
–
Trimethoxyamfetamine
–
–
1.48
–
–
Tyramine
1.2
1.47
–
–
0.81
Analgesics, non-steroidal anti-inflammatory drugs
Antifungals
System HD
The general screening systems, previously described, may be used.
HM Stevens, R Gill, unpublished data. n n
Column: ODS Hypersil (160 5 mm i.d., 5 mm). Mobile phase: Isopropyl alcohol–formic acid–0.1 mol/L potassium dihydrogenphosphate (13.61 g/L; 540 : 1 : 1000).
Antifungals HX
HY
HZ
HAA
RI
RI
RT
RT
Econazole
526
385
–
20.1
Fluconazole
340
289
–
11.4
Flucytosine
72
–
1.5
3.1
System HW
Griseofulvin
–
488
–
18.4
HM Stevens, R Gill, unpublished data.
Ketoconazole
439
464
5.2
15.7
System HV n n n
n n
Column: ODS Spherisorb (200 4.6 mm i.d., 5 mm). Mobile phase: Acetronitrile–acetic acid (45 : 55) for 2 min, to (75 : 25) at 3%/min, for 6 min. Flow rate: 1.7 mL/min.
Column: As for System HD, above. Mobile phase: Isopropyl alcohol–formic acid–0.1 mol/L potassium dihydrogenphosphate (13.61 g/L; 176 : 1 : 1000).
Analgesics, NSAIDs HD
HV
HW
HX
HY
HZ
HAA
k
RRT
k
RI
RI
RT
RT
0.5
–
2.3
–
281
–
–
0.1
–
0.32
–
–
–
–
Alclofenac
2.6
0.61
–
–
–
Aminophenazone
0.2
–
0.32
262
204
Aspirin
0.5
–
2.7
350
318
0.7
–
4.6
–
–
–
–
0.7
–
22.4
–
–
–
–
Aspirin
0.5
–
2.7
–
–
–
–
Paracetamol
0.1
–
0.32
–
–
–
– –
Acetanilide Paracetamol
Salicylic acid Benorilate
–
– 2.1
–
2.7
–
Benoxaprofen
11.3
0.98
–
–
–
–
Clonixin
–
0.87
–
–
345
–
–
Diclofenac
11.5
0.85
–
616
592
14.8
22.1
Diflunisal
4.1
0.77
–
508
583
5.4
–
Dipyrone
0.1
–
0.45
316
194
1.4
–
Etenzamide
0.55
–
4.6
–
303
–
–
Fenbufen
4
0.81
–
520
461
–
19.3
Fenoprofen
7.9
–
–
574
524
10.9
21.2
Floctafenine
–
–
–
–
–
Flufenamic acid
19.7
1
–
671
667
–
–
Flunixin
–
0.99
–
–
414
–
–
4.4
17.2
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:18
Recommended HPLC systems
Analgesics, NSAIDs, continued HD
Flurbiprofen
HV
HW
HX
HY
HZ
HAA
k
RRT
k
RI
RI
RT
RT
–
0.89
–
585
–
11.8
21.3
Glafenine
–
–
–
372
276
2.3
Ibuprofen
15.1
–
–
616
598
16.5
23.8
–
Indometacin
6.95
0.87
–
607
590
14.4
21.7
Indoprofen
1.2
0.52
–
–
406
–
–
0.66
–
495
–
6.4
Ketorolac
–
–
–
–
–
4.1
Meclofenamic acid
–
–
–
653
690
–
–
Ketoprofen
2.4
19.6 –
Mefenamic acid
21.1
0.95
–
661
686
–
–
Methyl salicylate
3.9
–
–
480
449
–
–
Salicylic acid
0.7
–
–
–
–
–
–
Morazone
0.4
–
–
294
–
Naproxen
3.3
–
–
501
468
–
–
–
313
–
12.7 –
Nefopam
–
Nifenazone Niflumic acid
2.05
–
0.1 –
– –
6.8
310
–
–
0.93
–
595
530
–
–
501
459
6.7
–
0.45
22
Oxyphenbutazone
1.95
0.69
Paracetamol
0.1
–
0.32
264
241
1.9
5.6
Phenacetin
0.6
–
4.4
377
335
3.0
–
Paracetamol
0.1
–
0.3
264
241
1.9
–
Phenazone
0.1
–
0.95
333
299
2.1
–
Phenylbutazone
6.5
0.95
–
672
643
19.5
1.95
0.7
–
501
459
6.7
Oxyphenbutazone Piroxicam M (5-hydroxy)
–
0.6 –
7.7
–
– 11
431
382
–
446
441
370
327
289
– –
4.9
1.3
–
0.4
–
Salsalate
3.6
0.69
–
–
–
Sulindac
1.25
0.78
–
488
462
3.9
–
–
–
–
–
7.2
–
–
–
366
–
Tenoxicam Tiaprofenic acid
–
–
–
484
452
Tolfenamic acid
–
–
–
690
–
Tolmetin
2.05
0.60 and 0.99
–
470
434
Zomepirac
3.7
–
–
–
495
16.6 –
Salicylamide
Sulindac sulfoxide
–
–
Propyphenazone
2.5
24.1
–
4.7
– – 16.6 –
–
12.7 5.8
17.6 –
37.9
–
5.4 –
–
Antibacterials The general screening systems, previously described, may be used.
Antibacterials, continued
HY
HAA
RI
RI
RT
Amoxicillin
–
226
3.1
Ampicillin
–
250
3.8
Azithromycin
–
–
Ceftriaxone
239
–
Chloramphenicol
390
336
14.1
Ciprofloxacin
318
260
9.1
Clarithromycin
–
–
–
Clindamycin
354
291
12
Furazolidone
336
–
12.2
– 5.3
table continued
HY
HAA
RI
RI
RT
Isoniazid
–
246
–
Metronidazole
257
226
6.8
Minocycline
–
240
22.6
Nalidixic acid
–
380
16
Nitrofurantoin
319
288
–
Ofloxacin
314
260
Oxytetracycline dehydrate
299
260
–
Rifampicin
–
417
16.2
Roxithromycin
–
–
15.8
Tetracycline
314
265
9.9
Trimethoprim
299
254
8.3
Antibacterials HX
HX
8.6
741
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:20
742
High Performance Liquid Chromatography
Anticholinergics
Anticonvulsants and barbiturates
The general screening systems, previously described, may be used or systems HAX and HAY below.
System HE
System HAX
n n
Koves EM (1995). J Chromatogr A 692: 103–119. n n n n n n
Column: Supelcosil LC-DP (250 4.6 mm i.d., 5 mm). Eluent: (A : B : C) Acetonitrile–phosphoric acid (0.025% v/v)– triethylamine buffer. Isocratic elution: (25 : 10 : 5). Flow rate: 0.6 mL/min. Detection: DAD (l 229 nm). Note: The triethylamine (TEA) buffer is prepared by adding 9 mL concentrated phosphoric acid and 10 mLTEA to 900 mLwater, adjusted to pH 3.4 with diluted phosphoric acid and made up to 1 L with water.
System HAY
Column: Alkyl-silica SAS-Hypersil (125 4.5 mm i.d., 5 mm). Mobile phase: Acetonitrile–tetrabutylammonium phosphate, 0.005 mol/L, pH 7.5 (20 : 80).
System HG
Gill R et al. (1981). J Chromatogr 204: 275–284. n n
Column: ODS Hypersil (150 4.6 mm i.d., 5 mm). Mobile phase: Methanol–0.1 mol/L sodium dihydrogenphosphate (11.998 g/L) (40 : 60); adjust to pH 3.5 by the addition of phosphoric acid.
System HH
Koves EM (1995). J Chromatogr A 692: 103–119. n n n n n
Christofides JA, Fry DE (1980). Clin Chem 26: 499–501.
Gill R et al. (1981). J Chromatogr 226; Biomed Appl 15: 117–123.
Column: LiChrospher 100 RP-8 (250 4.0 mm i.d., 5 mm). Eluent: (A : B : C) as for System HAX. Isocratic elution: (60 : 25 : 15). Flow rate: 0.6 mL/min. Detection: DAD (l 229 nm).
n n
Column: As for System HG, above. Mobile phase: As for System HG except that the mixture is adjusted to pH 8.5 by the addition of sodium hydroxide solution.
Anticholinergics HA
HX
HY
HZ
HAA
HAX
HAY
k
RI
RI
RT
RT
RT
RT
Adiphenine
1.8
422
–
–
–
–
Atropine
3.9
306
251
2.2
10.4
– 7
3.8
Biperiden
–
–
–
6.4
14.8
–
–
Chlorphenoxamine
2.9
–
346
–
–
–
–
Clidinium
–
379
–
–
–
–
–
Clidinium bromide
–
–
–
–
13.3
–
–
Cyclopentolate
1.6
353
287
3.2
–
–
–
Dicycloverine
1.1
–
575
–
–
–
–
Diethazine
3.4
–
–
–
–
15.1
7.4
Emepronium bromide
5.2
420
–
–
–
–
–
–
Homatropine
4.2
272
223
–
Hyoscine
1.1
270
253
–
Hyoscyamine
3.7
–
–
–
Isopropamide Iodide
2.4
379
–
–
7.4 9.7 –
6.8
3.6
7
3.7
–
–
–
–
Metixene
3.6
451
–
–
–
–
–
Orphenadrine
3
418
323
6
–
–
–
1.7
–
–
–
–
–
–
N-Monodesmethylorphenadrine N-Oxide Oxyphencyclimine
1.1
–
–
–
–
–
–
2.8
424
–
–
–
–
–
Oxyphenonium bromide
2.6
424
–
–
–
–
–
Piperidolate
1.7
429
–
–
–
–
–
Procyclidine
2
406
–
6.2
–
>20
4.7
Profenamine
2.4
444
338
–
–
16.6
8.3
Propantheline bromide
4.4
454
–
–
–
–
–
–
499
–
–
–
–
–
1.8
429
381
7.6
15.3
–
–
Xanthanoic acid Trihexyphenidyl
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:22
Recommended HPLC systems
743
Anticonvulsants, barbiturates and antiepileptics HG
HH
k
k
Allobarbital
2.46
1.33
HX
HY
HZ
RI
RI
RT
346
–
2.7
Amobarbital
10.91
7.05
424
374
4
Aprobarbital
3.42
2.22
357
319
2.8
Barbital Benactyzine
1.11 –
0.63 –
308
258
2.2
382
–
–
Brallobarbital
3.09
1.72
371
336
3
Butalbital
6.17
3.48
394
342
3.4
390
–
–
3.42
384
355
3.2
Butetamate
–
Butobarbital
– 5.43
Carbamazepine
–
–
418
368
–
Clonazepam
–
–
465
403
4.6
Cyclobarbital
5.25
2.61
384
352
3.2
Cyclopentobarbital
6
3.84
391
352
–
Enallylpropymal
8.65
Ethosuximide
–
Flavoxate
–
Heptabarb Hexethal
6.96 – –
9.9 34.28
4.93 20.39
–
394
–
301
276
2.3
–
–
–
416
377
3.9
–
451
–
Hexobarbital
7.37
5.67
419
242
4.3
Ibomal
4.01
2.58
379
352
–
Idobutal Mebeverine
8.12 –
4.77 –
–
357
–
448
–
7.1
Mephenytoin
–
–
–
366
3.7
Mesuximide
–
–
–
387
4.8
Metharbital
2.69
1.99
435
324
–
Barbital
1.11
0.63
–
–
–
7.27
3.84
435
395
4.6
Nealbarbital
Methylphenobarbital
10.22
6.19
417
382
–
Papaverine
–
363
295
–
–
Pentobarbital
10.96
Phenacemide
–
Phenobarbital
8.07 –
3.09
Phenytoin
–
Primidone
–
Secbutabarbital
1.23 – –
4.9
3.3
424
383
4.1
339
266
–
379
335
3
431
381
3.7
322
288
2.1
377
331
–
Secobarbital
16.28
11.47
437
407
4.7
Sultiame
–
–
344
275
–
403
370
–
Talbutal
7.2
4.7
Thiamylal
–
–
516
476
–
Thiopental
–
–
485
433
6.9
Vinbarbital Vinylbital
4.83 –
2.32 –
Antidepressants The general screening systems, previously described, may be used or systems HF and HAZ below. System HF
R Gill, unpublished data, after Kabra PM et al. (1981). Clin Chim Acta 111: 123–132.
379
363
–
424
–
4.1
n n
Column: ODS Hypersil (160 5 mm i.d., 5 mm). Mobile phase: Acetonitrile–phosphate buffer (pH 3.0; 30 : 70). To prepare the phosphate buffer, add 0.6 mL nonylamine to 1 L 0.01 mol/L sodium dihydrogenphosphate (1.1998 g/L) and adjust the pH to 3.0 by the addition of phosphoric acid.
System HAZ
Chiba K et al. (1995). J Chromatogr B 668: 77–84.
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:26
744 n n
High Performance Liquid Chromatography
Column: C18 (250 4.0 mm i.d., 5 mm). Mobile phase: (A : B : C) Water–methanol–triethylamine adjusted to pH 5.5 with phosphoric acid.
n n n
Isocratic elution: (70 : 30 : 0.1). Flow rate: 0.7 mL/min. Detection: UV (l ¼ 240 nm).
Antidepressants and antipsychotics HA
HF
HX
HY
HZ
HAA
HAX
HAZ
k
k
RI
RI
RT
RT
RT
k
440
375
15.9
15.8
1.76
10-Hydroxyamitriptyline
2.9
–
–
–
–
–
–
–
10-Hydroxynortriptyline
1.8
–
–
–
–
–
–
–
Nortriptyline
2
–
–
–
–
–
1.71
398
–
–
14.2
–
–
Amitriptyline
3.3
5.42
4.58
–
–
Benperidol
1.1
–
Butriptyline
2.7
Amoxapine
Norbutriptyline Citalopram Desmethylcitalopram Clomipramine Monodesmethylclomipramine Desipramine
7.33
7.5
393
324
–
–
–
–
369
–
–
–
–
–
–
–
– –
3.6
1.7
–
–
–
–
–
403
–
4.5
–
–
–
–
–
–
3.7
–
–
–
9.92
462
405
10.2
16.4
–
–
–
–
–
3.6
424
361
3.4 2
–
2.1
5.9
–
–
–
14.9
13
1.52
Didesmethylimipramine
1.3
–
–
–
–
–
–
–
2-Hydroxydesipramine
1.2
–
–
–
–
–
–
–
M (2-OH-)
–
–
–
–
–
–
–
0.39
0.5
361
300
–
–
–
–
3.6
Dibenzepin
2.8
Dosulepin
3.2
428
367
–
–
–
M (sulfoxide)
4.6
–
–
–
–
–
–
–
M (nor-)
2.2
–
–
–
–
–
–
–
404
316
14.1
12.9
–
Doxepin M (nor-) Fluoxetine
3.7
2.27
5.7
5
2.2
–
–
–
4.6
–
–
–
–
–
–
400
7.6
16.2
12.2
–
–
–
–
–
6.7
–
–
–
Fluvoxamine
–
–
430
363
5.6
15.3
10
–
Imipramine
6.7
Desmethylfluoxetine
4.2
4.17
437
335
15.1
14.7
1.62
Desipramine
2.1
3.6
–
–
–
–
–
–
2-Hydroxydesipramine
1.2
–
–
–
–
–
–
–
2-Hydroxyimipramine
3.1
–
–
–
–
–
–
–
M (10-OH-)
–
–
–
–
–
–
–
0.39
M (2-OH-)
–
–
–
–
–
–
–
0.39
M (N-oxide)
–
–
–
–
–
–
–
1.85
Iprindole
4.1
10.83
–
–
–
–
–
–
Isocarboxazid
–
–
392
353
–
–
–
–
Maprotiline
2.2
4.92
15.5
–
1.44
–
–
–
13.8
–
1.18
–
–
–
–
–
–
0.88
–
–
–
–
0.53
–
–
–
438
389
1.1
–
–
–
Mianserin
1.8
–
391
342
M(nor-)
2.4
–
–
–
–
M (nor-)
–
–
–
–
M (N-oxide)
–
–
–
M (8-OH-)
Desmethylmaprotiline
6.6 – 4.6
–
–
–
–
Moclobemide
–
–
295
–
Nialamide
1.2
–
334
–
– –
2.4
10.2 –
0.19 6.9
–
– –
–
–
–
15.6
13.7
1.71
Nomifensine
0.9
0.42
349
296
Nortriptyline
2
4.58
–
338
–
–
–
–
–
–
1.63
–
330
–
–
–
–
377
340
3.9
14.2
–
–
10-Hydroxynortriptyline
1.8
Noxiptiline
–
Opipramol
2.2
– 1.63
6.6
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:30
Recommended HPLC systems
745
Antidepressants and antipsychotics, continued HA
Paroxetine
HF
HX
HY
HZ
HAA
HAX
HAZ
k
k
RI
RI
RT
RT
RT
k
–
–
426
337
5.6
15.3
11.1
–
Phenelzine
1
–
184
–
–
–
–
–
Protriptyline
2.1
3.6
418
362
–
–
–
–
Remoxipride
–
–
334
–
3
–
M (FLA-838)
–
–
316
–
–
–
–
8.8 –
–
M (NCM-001)
–
–
364
–
–
–
–
–
M (NCM-009)
–
–
341
–
–
–
–
–
Sertraline
–
–
460
–
8.2
–
14.5
–
–
–
–
–
7.0
–
–
–
1.7
–
–
–
5.3
–
–
–
(desmethylsertraline) Tofenacin Trazodone
0.6
–
378
305
3.3
12.7
–
–
Trimipramine
2.7
6.17
454
345
8.3
15.9
15.5
–
1.8
–
–
–
–
–
–
–
Viloxazine
–
2.7
325
273
–
11
–
–
Zimeldine
3.2
0.67
–
270
–
–
–
–
M (nor-)
2.9
–
–
–
–
–
–
–
M (nor-)
Antihistamines
Antimalarials
The general screening systems, previously described, may be used.
The general screening systems, previously described, may be used.
Antihistamines HA
HX
HY
HZ
HAA
HAX
HAY RT
k
RI
RI
RT
RT
RT
Alimemazine
3.1
420
–
–
–
14.9
Antazoline
1.8
383
294
–
–
–
–
7.1
–
–
286
13.2
–
–
(astemizole)
–
383
–
–
–
–
–
(M-nor)
Astemizole
3.9
–
361
–
–
–
–
–
Bromazine
2.7
444
–
–
–
–
–
Brompheniramine
4.1
–
267
–
13.9
–
–
Buclizine
0.7
–
454
–
–
–
–
Carbinoxamine
4.7
359
–
–
12.8
–
Cetirizine
–
–
–
Chlorcyclizine
2.3
–
340
Chlorphenamine
3.9
356
264
Cinnarizine
0.8
560
–
Clemastine
3.7
501
–
Clemizole
4.8
420
–
Cyclizine
2.9
405
–
Norcyclizine
2.2
–
–
Cyproheptadine
3.2
–
354
Deptropine
5
471
–
10.3
Dimetindene
5.1
338
288
Diphenhydramine
3.3
393
Diphenylpyraline
3.3
Doxylamine
4.4
Hydroxyzine
3.6 –
15.7
– 8.89
5.29
–
–
12.9
10.8
22
19.3
–
–
14
–
–
–
–
–
–
–
–
12.4
–
–
–
15
–
–
–
–
–
–
–
–
–
336
–
–
12.2
401
–
–
–
–
–
–
259
–
11.1
–
–
1.4
437
326
15.3
11.4
Isothipendyl
3.8
390
–
–
13.5
–
–
Loratadine
–
523
362
14.6
22.9
10.9
13.3
3.5
4.8 – 6.5
5.7
– 5.3
5.8
6
6.3
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:32
746
High Performance Liquid Chromatography
Antihistamines, continued HA
HX
HY
HZ
HAA
HAX
HAY
k
RI
RI
RT
RT
RT
RT
Mebhydrolin
3
411
–
5.3
–
–
–
Meclozine
0.7
587
398
–
20
–
–
Mepyramine
3.9
448
257
–
–
–
–
Methapyrilene
4.1
342
197
–
–
–
–
Methdilazine
6
–
–
–
–
15.2
6.7
Phenindamine
2.5
397
–
–
–
–
–
Pheniramine
4.1
283
206
–
–
Phenyltoloxamine
3.1
415
–
–
–
–
–
Pizotifen
3.4
435
–
6.6
15.2
–
–
Promethazine
5
409
324
5.7
14.5
13.2
6.4
Propiomazine
2.1
440
359
–
–
14.1
7.1
Pyrrobutamine
2.8
477
–
–
–
–
–
Thenyldiamine
4
317
–
–
–
–
–
Thiazinamium metilsulfate
–
–
–
6.4
–
–
–
Trimethobenzamide
4.7
347
–
–
–
–
–
Tripelennamine
3.6
336
265
–
–
–
–
Triprolidine
3.2
388
270
–
13.1
–
–
9.5
4.5
Antimalarials HA
HX
HY
HZ
HAA
HAX
HAY
RT
RT
RT
k
RI
RI
RT
Chloroquine
15.2
282
246
2.1
Cinchonidine
3.1
306
214
–
5.4
12.7
3.6
–
–
– –
Cinchonine
–
304
209
–
10.2
–
Halofantrine
–
800
–
–
23
–
Hydroxychloroquine
–
280
–
1.9
–
1.4
–
276
–
–
379
–
3.8
13.6
–
–
Pyrimethamine
1
–
289
–
12.5
–
–
Quinine
2.4
327
246
2.6
11.3
Primaquine Proguanil
–
– 9.6
3.2
–
–
8.3
4.5
Antitussives
Antineoplastics
HA
HX
HY
HAA
The general screening systems, previously described, may be used.
k
RI
RI
RI
Antineoplastics
Bromhexine
0.4
417
334
–
Dextromethorphan
5.6
377
298
13.3
HX
HAA
4.7
–
–
–
RI
RT
Dextrorphan
–
325
–
–
Dextrorphan
Diethylstilbestrol
592
20.9
Dropropizine
–
240
–
Doxorubicin
370
12.1
Guaifenesin
–
328
262
11.4
3.4
Noscapine
0.3
368
289
12.8
Pholcodine
6
65
92
2.7
Pipazetate
5.4
385
–
Fluorouracil
70
Methotrexate
292
Vinblastine
–
– 8.4
7.2
–
Antivirals Antitussives The general screening systems, previously described, may be used.
The general screening systems, previously described, may be used or systems HAB and HAC below.
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:37
Recommended HPLC systems System HAB
Sparidans RW et al. (2000). J Chromatogr B Biomed Sci Appl 742: 185–192. n n n n n
Column: C18 Symmetry (100 4.6 mm i.d., 3.5 mm) with Symmetry C18 precolumn (20 3.8 mm, 5 mm). Mobile phase: Acetonitrile–sodium phosphate buffer (25 mmol/L, pH 6.8) (40 : 60). Flow rate: 1.5 mL/min. Detection: Fluorescence (lex ¼ 270 nm, lem ¼ 340 nm). Note: 8 min after each injection, flush column for 5 min at 1.5 mL/ min with acetonitrile–water (30 : 70). Equilibrate for about 8 min with the original eluent before injecting the next sample.
System HAC
Aymard G et al. (2000). J Chromatogr B Biomed Sci Appl 744: 227–240. n n n n n n
Column: C18 Symmetry (250 4.6 mm i.d., 5 mm) with C18 precolumn (Guard-Pak, mBondapak). Column temperature: 37 C. Mobile phase: (A : B) Disodium hydrogenphosphate (0.04 mol/L) with 4% (v/v) octane sulfonic acid (0.25 mol/L)–acetonitrile. Isocratic elution: (50 : 50). Flow rate: 1.3 mL/min. Detection: DAD (l ¼ 261 nm between time 0 and 9 min; l ¼ 241 nm between time 9 and 20 min; l ¼ 254 nm between time 20 and end of the run (32 min).
Cannabinoids System HL
Baker PB et al. (1980). J Anal Toxicol 4: 145–152. n n
Column: ODS Spherisorb (250 4.6 mm i.d., 5 mm). Mobile phase: 0.01 mol/L sulfuric acid–methanol–acetonitrile (7 : 8 : 9).
Cannabinoids System HL k Cannabichromene
19.09
Cannabicyclol
14.78
Cannabidiol
7.47
Cannabidiolic acid
8.76
Cannabigerol
8.18
Cannabinol
11.77
Cannabivarin
7.47
D8-Tetrahydrocannabinol
14.07
D9-Tetrahydrocannabinol
13.35
Tetrahydrocannabinolic acid
25.83
Tetrahydrocannabivaric acid
14.64
Tetrahydrocannabivarin Antivirals HAB
HAC
RT
k
Abacavir
1
–
Amprenavir
4
2.5
Efavirenz
–
8.5
Indinavir
4.2
2
747
8.18
Cardiac glycosides System HM
Cobb PH (1976). Analyst (Lond) 101: 768–776. n n
Column: Silica LiChrosorb SI60 (250 4 mm i.d., 10 mm). Mobile phase: Cyclohexane–ethanol–acetic acid (60 : 9 : 1).
Cardiac glycosides System HM k
Benzodiazepines System HI
Digitoxigenin
2.0
R Gill, unpublished data.
Digitoxigenin bisdigitoxoside
3.9
Digitoxigenin monodigitoxoside
2.8
Digitoxin
5.4
Digoxigenin
4.5
Digoxigenin bisdigitoxoside
8.2
Digoxigenin monodigitoxoside
5.5
n n
Column: ODS Hypersil (200 5 mm i.d., 5 mm). Mobile phase: Methanol–water–phosphate buffer (55 : 25 : 20). To prepare the phosphate buffer, dissolve 11.038 g (0.092 mol/L) sodium dihydrogenphosphate and 1.136 g (0.008 mol/L) disodium hydrogenphosphate in sufficient water to produce 1 L.
System HJ
R Gill, unpublished data.
Digoxin
11.3
n n
Gitaloxin
6.8
Gitoxigenin
3.7
Gitoxigenin bisdigitoxoside
6.5
System HK
Gitoxigenin monodigitoxoside
4.5
R Gill, unpublished data, after RJ Flanagan et al. (1980). J Chromatogr 187: 391–398.
Gitoxin
n n
Column: As for System HI, above. Mobile phase: Methanol–water–phosphate buffer (as in System HI) (70 : 10 : 20).
Column: Silica Spherisorb (250 5 mm i.d., 5 mm). Mobile phase: Methanol to which has been added 100 mL perchloric acid per litre.
8.6
Lanatoside A
17.9
Lanatoside B
31.8
Lanatoside C
39.5
Benzodiazepines HI
HJ
HK
HX
HY
HZ
HAA
HAX
HAY
k
k
k
RI
RI
RT
RT
RT
RT
Acecarbromal
–
–
–
429
374
–
–
–
–
Alprazolam
–
–
2.79
–
–
–
–
–
–
Bromazepam
–
–
2.99
–
–
–
–
–
– table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:42
748
High Performance Liquid Chromatography
Benzodiazepines, continued HI
HJ
HK
HX
HY
HZ
HAA
HAX
HAY
RT
k
k
k
RI
RI
RT
RT
RT
Bromisoval
–
–
–
365
307
2.9
–
–
–
Brotizolam
–
–
–
484
–
4.6
–
Carbromal
–
–
–
410
377
3.9
–
–
–
Chlordiazepoxide
–
–
2.87
–
–
–
–
–
–
Clobazam
–
–
0.03
–
–
–
–
–
–
7.4
7.9
Clomethiazole
–
–
–
395
292
–
16
–
–
Clonazepam
–
–
0.35
–
–
–
–
–
–
Clorazepic acid
–
–
2.00
–
–
–
–
–
–
Demoxepam
–
–
0.03
–
–
–
–
–
–
Diazepam
–
–
2.49
–
–
–
–
–
–
Flumazenil
–
–
–
387
327
2.6
–
–
–
Flunitrazepam
3.15
–
0.47
483
305
5.6
18.6
–
–
Flurazepam
–
3.19
6.5
397
305
4.2
–
10.5
5.5
Glutethimide
–
–
–
436
401
4.8
–
6.6
6.2
Ketazolam
–
–
0.04
–
–
–
–
–
–
Loprazolam
–
–
–
388
–
–
13.4
–
–
Lorazepam
–
–
0.14
–
–
–
–
–
–
Lormetazepam
6.32
–
0.08
487
463
–
–
–
Medazepam
–
–
4.44
–
–
–
–
Methaqualone
–
–
–
459
400
Methyprylon
–
–
–
347
302
Midazolam
9.75
2.1
5.9
399
306
Nitrazepam
2.96
–
1.49
448
370
6.2 – 5.4 –
–
– 6.8
–
–
4.2
14.9
10.2
4.2
16.9
6.3
7.4 – 6.3 6
Nordazepam
–
–
1.99
–
–
–
–
–
–
Oxazepam
4.62
–
0.73
–
–
–
–
–
–
Prazepam
–
–
2.19
–
–
–
–
–
–
Quazepam
–
–
–
–
766
37.5
–
11.9
Temazepam
5.68
–
0.6
472
438
5.5
–
–
0.73
–
–
Oxazepam
4.38
–
1.83
476
390
–
–
–
–
–
Zolpidem
–
–
–
–
291
Zopiclone
–
–
–
331
269
Triazolam Not detected
–
18.6 –
4.2 –
17.7
8.9
6.7
–
17.4
– 6.4
6.7
–
–
–
3.2
11.9
–
–
2.3
–
7.5
3.8
Cardioactive drugs The general screening systems, previously described, may be used.
Cardioactive drugs, continued
Cardioactive drugs
HA HA
HX
HY
HZ
HAA
k
RI
RI
RT
RT
Ajmaline
2.8
–
277
Alfuzosin
–
–
–
Amiodarone
2.4
683
476
90.4
–
1.8
–
–
–
–
–
433
–
–
17
0.9
250
–
–
415
–
Monodesethylamiodarone Aprindine Bamethan Benzthiazide
–
–
– 2.4
Betahistine
3.1
–
–
–
Bretylium tosilate
4.3
–
275
–
Buphenine
HX
HY
HZ
k
RI
RI
RT
0.9
370
–
–
HAA RT –
–
Captopril
–
316
283
2.1
9.7
10.4
Cilazapril
–
420
–
4.5
14.4 –
–
–
1.7
1.2
258
194
2.5
Clopamide
–
377
310
–
–
Debrisoquine
1.2
–
245
–
–
Diltiazem
–
–
361
4.5
14
Deacetyldiltiazem
–
–
–
–
–
Desmethyldiltiazem
–
–
–
–
–
Cilazaprilate
5.9 – 3.2 –
–
Clonidine
6.1
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:46
Recommended HPLC systems
Cardioactive drugs, continued
Desacetyldiltiazem Disopyramide N-Monodesisopropyldisopyramide
HA
HX
HY
HZ
HAA
k
RI
RI
RT
RT
–
–
–
–
–
2.4
345
281
1.8
–
–
n n
Column: ODS Hypersil (160 5 mm i.d., 5 mm). Mobile phase: Acetonitrile–water containing 10 mL/L acetic acid (30 : 70).
Diuretics HN
3 –
11.4
–
201
–
Encainide
–
363
–
– 25.8
HX
HY
HAA RT
k
RI
RI
Acetazolamide
–
268
226
–
Enalapril
749
6.9
Amiloride
–
257
190
–
Bendroflumethiazide
15.35
508
–
18.6
1.5
3.4
3.6
Felodipine
–
690
–
24.4
Benzthiazide
9.32
–
415
–
Flecainide
–
419
355
5.2
–
Chlorothiazide
0.54
–
239
–
Hydralazine
–
193
132
1.9
–
Chlortalidone
1.28
367
308
–
Isoxsuprine
0.8
353
301
–
Clopamide
4.01
377
310
–
Labetalol
1.7
365
290
–
Clorexolone
7.26
–
391
–
Lidoflazine
0.6
530
–
–
Cyclopenthiazide
16.45
–
453
–
Lisinopril
–
271
250
1.5
–
Cyclothiazide
–
433
–
–
10.78, 11.91, and 12.81
Etacrynic acid
–
521
497
–
Furosemide
–
435
380
15.2
– 3 –
Lorcainide
1.8
425
–
6.6
Methyldopa
–
69
–
1.4
Mexiletine
1.2
329
278
–
3 11.5
Hydrochlorothiazide
0.7
294
255
–
Mefruside
8.67
–
417
–
Methyclothiazide
3.82
–
364
15.4
19.5
Metolazone
4.89
–
371
–
Minoxidil
–
297
–
Naftidrofuryl oxalate
–
–
409
Nifedipine
0.2
527
464
Pargyline
0.2
–
203
–
–
Spironolactone
–
592
539
20.7
Pentaerithrityl tetranitrate
–
663
–
–
–
Triamterene
–
298
263
8.7
–
–
–
–
–
341
14.9
23.1
355
274
–
18.8
Pentoxifylline
–
488
Perindopril
–
–
–
13.7
Drugs of abuse
–
314
–
–
–
0.1
396
–
–
–
A comprehensive HPLC method for the screening of common drugs of abuse is described in Chapter 1, Table 1.24. Furthermore, an additional eight systems (HBC, HBD, HBE, HBF, HBG, HBI and HBJ) are provided in Chapter 11, Table 11.4.
(pentaerithrityl)
(perindoprilat) Phenoxybenzamine
2.4 –
9.8 15.8
7.2
Trichlormethiazide 2.1 1.6
3.1 –
Xipamide
11.5
Phentolamine
1.7
368
–
Prajmalium bitartrate
2.2
–
340
Prazosin
0.8
352
–
2.5
10.6
Procainamide
1.3
208
160
1.9
–
HA
HC
HX
HY
HZ
HAA
3
–
–
1.8
–
k
k
RI
RI
RI
k
Quinapril
–
–
–
5.4
16.8
–
–
–
–
–
–
Quinidine
2.1
322
245
2.6
11
Amfetamine
0.9
0.98
244
–
–
–
–
–
Ramipril
4.2
15.7
Benzfetamine
1.2
0.15
–
0.6
496
407
–
–
Rescinnamine
–
–
Benzoylecgonine
0.9
1.7
467
351
–
–
236
–
–
Reserpine
16.4
Bufotenine
3.1
–
181
–
–
Sotalol
1.2
226
–
–
2
3.8
Cannabidiol
–
–
990
902
–
–
Tocainide
1.2
247
208
2.1
Tolazoline
2.1
225
179
Trandolapril
–
–
–
–
–
–
Trimetazidine
3
–
–
Verapamil
2.6
447
386
7
15.4
M (nor-)
1.7
–
–
6.6
–
N-Acetylprocainamide
3 –
– –
Drugs of abuse
5-Methyltryptamine
3.7 – 9.7
–
Cannabinol
–
–
1080
1028
–
–
–
Cocaine
2.8
–
348
289
3.3
11.9
6.1
17 –
D9-THC
–
–
–
–
–
–
2.1
Diamorphine
3
0.66
340
282
–
–
Diethyltryptamine
–
–
–
–
–
–
Dimethyltryptamine
–
–
–
228
–
–
Diuretics
DOM
–
1.13
340
–
–
–
System HN
Ketamine
–
–
311
262
2.4
R Gill et al., unpublished data, after Tisdall PA et al. (1980). Clin Chem 26: 702–706.
Lysergic acid
0.8
–
–
236
–
–
Lysergide
0.7
–
362
–
–
12
Trandolaprilat
–
–
6.1
9.6
table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:48
750
High Performance Liquid Chromatography Local anaesthetics
Drugs of abuse, continued HA
HC
HX
HY
HZ
HAA
k
k
RI
RI
RI
k
The general screening systems, previously described may be used, as well as system HQ or HR, below.
Mescaline
1.3
2.17
272
243
–
–
System HQ
Metamfetamine
2
2.07
262
216
2.4
8.4
Methadone
2.2
1.03
440
343
8.5
15.8
Methylenedioxyamfetamine
–
0.98
266
248
2.1
8.1
Methylenedioxymethamfetamine
–
–
278
252
2.2
9.1
Monoacetylmorphine
3.6
0.8
–
–
–
7.3
Morphine
3.8
1.3
200
182
1.8
N-Methyltryptamine
–
–
–
–
–
–
p-Methoxyamfetamine
–
–
–
–
–
–
Psilocin
3.1
–
240
226
–
–
Psilocybine
–
–
–
185
–
–
System HA, previously described, may be used or System HP, below. System HP
R Gill et al., unpublished data, after Twitchett PJ et al. (1978). J Chromatogr 150: 73–84. Column: ODS Hypersil (100 5 mm i.d., 5 mm). Mobile phase: Methanol–phosphate buffer (60 : 40). To prepare the phosphate buffer, dissolve 3.43 g (0.022 mol/L) sodium dihydrogenphosphate and 10.03 g (0.028 mol/L) disodium hydrogenphosphate in sufficient water to produce 1 L.
Ergot alkaloids HA
HP
k
k
Bromocriptine
–
44.3
Dihydroergocristine
–
18.3
Dihydroergocryptine
–
15.9
Dihydroergotamine
0.6
11.4
Ergocornine
0.4
10.2
Ergocristine
0.3
17.3
Ergocryptine
0.4
15.2
Ergometrine
0.4
Ergosine
0.3
Ergosinine
0.3
Ergotamine
0.4
n n
0.50 7.08 17.7 9.58
Column: ODS Hypersil (160 5 mm i.d., 5 mm). Mobile phase: Methanol–water–1% v/v solution of phosphoric acid– hexylamine (30 : 70 : 100 : 1.4).
System HR
Gill R et al. (1984). J Chromatogr 301: 155–163. n n
3.3
Ergot alkaloids
n n
Gill R et al. (1984). J Chromatogr 301: 155–163.
Column: As for System HQ above. Mobile phase: Methanol–1% v/v solution of phosphoric acid–hexylamine (100 : 100 : 1.4).
Local anaesthetics HA
HQ
HR
HX
HY
HZ
k
k
k
RI
RI
RT
Benzocaine
0.1
20.06
1.61
404
358
4.3
Bupivacaine
0.9
7.19
0.86
366
310
4.1
Butacaine
1.2
8.97
–
392
331
–
Butanilicaine
–
4.42
–
–
280
–
Chloroprocaine
–
0.24
–
–
250
–
Cinchocaine
1.9
–
371
–
Cocaine
2.8
2.68
–
348
289
3.3
Benzoylecgonine
0.9
5.68
–
–
–
–
Ecgonine
1.1
–
–
–
–
–
Cyclomethycaine
–
–
10.31
–
413
–
Dyclonine
–
–
2.78
–
347
–
Etomidate
–
–
475
417
–
Ketamine
–
–
–
311
262
Lidocaine
0.6
–
288
258
2.6
–
–
–
–
–
296
260
2.6
–
503
484
–
405
–
–
357
312
–
415
–
6.5
–
–
–
2.7 –
M (monoethylglycinexylidide)
1.2
–
5.51
0.79 – 1.09
2.4
Mepivacaine
0.9
Methohexital
–
–
Oxybuprocaine
–
16.25
Piperocaine
–
4.59
Pramocaine
0.6
Prilocaine
1
Procaine
1.9
–
–
264
225
Propofol
–
–
–
–
–
35
Proxymetacaine
2.1
–
–
269
–
Quinisocaine
2.2
–
–
–
–
Tetracaine
2
16.25
389
321
4.4
0.86 –
–
2.48 1.38
1.38
11.24 1.33
Iso-lysergic acid
–
0.83
Iso-lysergide
2.6
0.0
Lysergamide
0.5
0.33
Narcotic analgesics
Lysergic acid
0.8
0.0
Lysergic acid methylpropylamide
–
1.98
Systems HA or HC, previously described, may be used or System HS, below.
Lysergide
0.7
1.83
System HS
Lysergol
1.1
0.83
Baker PB, Gough TA (1981). J Chromatogr Sci 19: 483–489. n
Methylergometrine
0.4
0.83
Methysergide
0.4
2.33
2-Oxylysergide
–
0.92
n
Column: Aminopropyl-bonded silica Spherisorb S5NH2 (250 4 mm i.d., 5 mm). Mobile phase: Acetonitrile–tetrabutylammonium phosphate, 0.005 mol/L, pH 7.5 (85 : 15).
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:52
Recommended HPLC systems
Narcotic analgesics and narcotic antagonists HA
HC
HS
HX
HY
HZ
HAA
HAX
HAY
k
k
k
RI
RI
RT
RT
RT
RT
Alphaprodine
2.8
–
–
363
317
–
–
–
–
Bezitramide
0.2
–
–
564
–
22.5
–
–
–
Buprenorphine
0.4
0.05
–
397
339
5
14
–
Codeine
4.8
1.21
1.9
266
237
1.9
Morphine
3.8
1.3
5.16
–
–
–
–
–
–
M (nor-)
3.1
3.51
–
–
–
–
–
–
–
Cyclazocine
2.1
–
–
–
289
–
–
–
–
Dextromoramide
0.7
0.09
–
440
390
–
15.8
–
–
Dextropropoxyphene
1.9
0.19
–
–
374
15.8
–
–
Norpropoxyphene
1.3
–
–
–
–
–
–
–
7.6 –
5
– 6.1
3.4
3
0.66
0.35
340
282
–
–
6-Monoacetylmorphine
3.6
0.8
1
–
–
–
–
–
–
Morphine
3.8
1.3
5.16
–
–
–
–
–
–
Dihydrocodeine
7.2
2.5
–
261
208
–
–
Dihydromorphine
5.7
2.75
–
237
156
–
–
–
–
Dipipanone
2.2
1.61
–
500
363
–
–
–
–
Ethoheptazine
3.3
1.55
–
359
–
–
–
–
Ethylmorphine
3.7
1.06
1.45
291
244
–
–
Fentanyl
0.8
1.11
–
373
299
–
14.2
Hydromorphone
7.9
–
–
240
187
–
–
Ketobemidone
2.8
–
–
294
245
–
–
–
–
Levallorphan
1.9
1.46
–
356
291
–
–
–
–
Levorphanol
4.4
3.2
–
–
265
–
–
–
–
Meptazinol
3.1
–
–
–
269
–
–
–
–
Methadone
2.2
1.03
–
440
343
15.8
16.5
8.4
M (EDDP)
2.8
–
–
–
–
–
–
–
–
M (EMDP)
0.2
–
–
–
–
–
–
–
3.8
1.30
5.16
200
182
Morphine3-glucuronide
–
1.56
–
–
–
–
–
N-oxide
–
Diamorphine
Morphine
2
8.5
7.9
4.7
1.8
– 6.7
11.4 5.8
3.3
4.1
3.6 6 3.4
– 5.6
3.2
–
–
–
–
–
–
14
–
–
3.2
–
–
–
–
–
Nalorphine
1
0.29
–
260
237
–
Naloxone
1.4
0.17
–
–
238
Norcodeine
3.1
3.51
–
–
235
–
–
–
–
Normethadone
–
0.53
–
–
366
–
–
–
–
Normorphine
2.9
3.92
–
–
133
–
–
–
–
Norpipanone
–
0.35
–
466
–
–
–
–
Oxycodone
6.9
0.85
–
277
246
–
–
6.7
–
–
–
–
–
–
–
Oxymorphone
6.7
–
–
217
184
–
–
–
Pentazocine
1.8
0.67
–
372
288
3.8
12.5
9.9
5.5
Pethidine
2.8
0.55
–
345
281
3.2
11.8
9.2
4.8
M (nor-)
1.7
2.04
–
–
–
–
–
–
–
Pethidinic acid
2.8
–
–
–
–
–
–
–
–
Phenazocine
1.3
0.3
–
409
299
–
–
–
–
Phenoperidine
Oxymorphone
4.8 2
– 6.5
5.8 – –
0.8
0.1
–
434
–
–
–
–
–
Norpethidine
1.7
2.04
–
–
–
–
–
–
–
Pethidine
2.8
0.55
–
–
–
–
–
–
–
Piritramide
0.6
0.1
–
377
343
–
–
–
–
Thebacon
3.7
0.85
–
333
–
–
–
–
–
Tramadol
–
–
–
328
267
–
–
–
2.9
751
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:10:55
752
High Performance Liquid Chromatography
Oral hypoglyacemics and antidiabetics
Pesticides
The general screening systems, previously described, may be used.
System HAO
Osselton MD, Snelling RD (1986). J Chromatogr 368: 265–271.
HX
HY
HZ
HAA
Column: ODS Hypersil (160 5 mm i.d., 5 mm), stainless steel. Mobile phase: Acetonitrile–water (60 : 40). Flow rate: 2 mL/min. Detection: DAD (200–450 nm).
n n n n
Oral hypoglyacemics and antidiabetics
RI
RI
RT
RT
Carbutamide
–
321
–
14.5
Osselton MD, Snelling RD (1986). J Chromatogr 368: 265–271.
Chlorpropamide
450
411 and 413
17.7
Glibenclamide
637
571
14.4
Gliclazide
536
483
8.8
20.5
n n n n
Glipizide
478
423
4.5
17.6
Metformin
60
–
1.7
2.8
Tolazamide
452
445
6.8
–
Tolbutamide
477
424
5.9
–
5
System HAP
22
Column: Silica Spherisorb S5W (250 5 mm i.d.). Mobile phase: Dichloromethane–isoctane (60 : 40). Flow rate: 2 mL/min. Detection: DAD (200–450 nm).
For more information on screening pesticides, see Chapter 16, Table 16.1. Phenothiazines and other tranquillisers The general screening systems, previously described, may be used.
Phenothiazines and other tranquillisers HA
Acepromazine
HX
HY
HZ
HAA
HAX
HAY
HAZ
k
RI
RI
RT
RT
RT
RT
k
4.1
–
350
–
10.8
–
–
–
Azacyclonol
1.2
–
–
–
–
4.5
–
Benzoctamine
1.7
380
322
–
–
–
–
–
8.7
Butaperazine
3.4
464
406
–
–
–
–
–
Captodiame
–
561
–
–
20.2
–
–
–
Chlordiazepoxide
–
363
285
Chlormezanone
–
–
334
Chlorpromazine
3.2 –
6.9
5.3
1.68
6
5.3
–
4.1
456
350
16
17
BASE
2.64
M (nor-)
2.2
–
–
–
–
–
–
–
M (sulfoxide)
–
–
–
–
–
8.4
4.3
0.62
3
459
353
10.1
–
17.6
8.3
–
–
Chlorprothixene Clopenthixol
–
448
411
Clorazepic acid
–
475
388
9.1
15.2 15.5
5.6
–
–
–
–
–
–
–
–
–
–
–
–
18.4
–
–
–
–
423
349
–
–
–
–
–
Flupentixol
1.2
475
435
10.7
17.4
13.7
7.5
–
Sulfoxide
1.3
–
–
–
–
–
–
–
Fluphenazine
1.2
462
471
10.1
17.4
13.6
7.2
–
–
Clorazepate Fluanisone
Fluspirilene
–
538
–
Haloperidol
1.2
421
316
Levomepromazine
3.2
435
381
Loxapine
1.1
407
336
5.8 7.5 –
–
18.3
9.8
–
14.4
11.1
6.2
0.72
–
15.2
7.2
1.82
14.6
–
–
–
Mesoridazine
5
–
337
Oxypertine
0.7
402
–
Pecazine
3.9
443
382
–
–
15.3
7
–
Penfluridol
–
659
656
43.4
20.2
–
–
–
3.4 –
–
10.1
5
–
–
–
–
–
Perazine
–
403
371
6.3
–
–
–
–
Pericyazine
1.3
410
356
4.4
–
10.2
5.1
–
Perphenazine
1.9
428
395
3.28
Pimozide
0.7
504
–
Pipamperone
–
299
241
16
13.1
6.3
11.9
7.2
17.2
–
–
–
2.7
10.9
–
–
–
Pipotiazine
–
431
–
–
14.7
–
–
–
Prochlorperazine
3.9
450
323
10.4
–
–
–
–
Promazine
5.9
407
326
5.9
–
–
–
–
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:11:0
Recommended HPLC systems
753
Phenothiazines and other tranquillisers, continued HA
Prothipendyl
HX
HY
HZ
HAA
HAX
HAY
HAZ
k
RI
RI
RT
RT
RT
RT
k
4.4
388
–
–
–
–
–
–
Sulforidazine
–
421
–
4.8
Sulpiride
–
259
235
2
–
–
–
–
3.9
–
–
0.02
Thiopropazate
1
483
–
–
–
–
–
–
Thioproperazine
4.1
427
305
15.4
15.2
–
–
– 3.88
5.2
490
427
13.5
17.2
–
9.8
5
–
–
–
–
–
–
–
Tiotixene
3.8
442
374
6.8
–
–
–
–
Triflupromazine
2.7
484
454
12.3
–
17.3
8.9
–
Thioridazine Mesoridazine
n n
Steroids System HATa
System HAR
Walters MJ et al. (1990). J Assoc Off Anal Chem 73: 904–926. n n n n n
Column: ODS Zorbax (250 4.6 mm i.d., 5 mm), stainless steel. Eluent: (A) Methanol. Isocratic elution: (100). Flow rate: 1.5 mL/min. Detection: UV (l ¼ 240, 210 and 280 nm).
System HATb
Walters MJ et al. (1990). J Assoc Off Anal Chem 73: 904–926. n n n
Flow rate: 1.5 mL/min. Detection: UV (l ¼ 240, 210 and 280 nm).
Lurie I et al. (1994). J Forensic Sci 39: 74–85. n n n n n
Column: ODS Zorbax (250 4.6 mm i.d., 5 mm). Mobile phase: (A : B) Water–methanol. Gradient elution: (30 : 70) to (0 : 100) over 15 min with 15 min hold. Flow rate: 1.0 mL/min. Detection: DAD.
System HT
Column: ODS Zorbax (250 4.6 mm i.d., 5 mm), stainless steel. Eluent: (A : B) Methanol–water. Isocratic elution: (75 : 25).
Rose JQ, Jusko WJ (1979). J Chromatogr Biomed Appl 162: 273–280. n n
Column: Silica Zorbax SIL (250 4.6 mm i.d., 5 mm). Mobile phase: Methylene chloride–methanol (97 : 3).
Steroids
Beclometasone Dipropionate Betamethasone Betamethasone valerate Boldenone Undecylenate Cortisone
HT
HX
HY
HZ
HAA
HAR
HATa
HATb
k
RI
RI
RT
RT
RRT
RRT
RRT
4.2
444
–
–
–
–
–
–
–
–
711
–
–
–
–
– –
–
–
–
14.2
13.3
–
–
–
–
584
–
–
–
–
–
–
–
–
–
–
0.74
–
0.76
–
–
–
–
–
–
1.94
–
2.4
–
372
–
–
–
–
–
Dexamethasone
4.8
–
381
Fluoxymesterone
–
–
427
3.4 –
13.1
–
–
–
–
0.78
–
0.7
Hydrocortisone
5.8
403
349
–
17.7
–
–
–
Hydroxyprogesterone
–
1054
–
–
–
–
–
–
Metenolone
–
–
–
–
–
–
–
–
Acetate
–
–
–
–
–
–
1.26
3.54
Enantate
–
–
–
–
–
–
1.87
–
Methandienone
–
–
–
–
–
0.86
–
0.87
Methandriol
–
–
–
–
–
1.25
–
1.29
–
–
–
–
–
–
–
2.75
7.5
426
390
–
18.9
–
–
–
Dipropionate Methylprednisolone Methyltestosterone
–
–
587
–
–
1.17
–
1.27
Nandrolone
–
–
–
–
–
0.84
–
0.92
–
Norethisterone
–
536
676
Prednisolone
8.4
401
361
Prednisone
3.4
250
340
Progesterone
–
672
698
2.5 2.6 –
24
–
–
–
14.1
–
–
–
14.2
–
–
–
23.8
–
–
– table continued
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:11:2
754
High Performance Liquid Chromatography
Steroids, continued HT
HX
HY
HZ
HAA
HAR
HATa
HATb
k
RI
RI
RT
RT
RRT
RRT
RRT
Testosterone
–
534
508
–
–
–
–
–
Acetate
–
–
894
–
–
1.76
–
2.59
Propionate
–
–
1003
–
–
2.01
1.31
4.06
Methyltestosterone
–
–
–
–
–
1.17
–
1.27
Isobutyrate
–
–
–
–
–
2.17
–
–
Cipionate
–
–
–
–
–
2.63
–
–
Enantate
–
–
–
–
–
2.6
1.8
–
Undecanoate
–
–
–
–
–
3.18
–
–
Phenylpropionate
–
–
–
–
–
–
1.48
–
Isocaproate
–
–
–
–
–
–
1.62
–
Cipionate
–
–
–
–
–
–
2.05
–
Undecenoate
–
–
–
–
–
–
2.53
–
Decanoate
–
–
–
–
–
–
2.78
–
Undecylate
–
–
–
–
–
–
3.27
– –
–
438
312
–
–
–
–
2.5
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
Hexahydrobenzylcarbonate
–
–
–
–
–
–
1.65
–
Acetate
–
–
–
–
–
–
–
1.71
Triamcinolone Acetonide Trenbelone
Sulfonamides
Xanthine stimulants
System HU
Cobb PH, Hill GT (1976). J Chromatogr 123: 444–447. n n
Column: Silica Spherisorb (250 4 mm i.d., 5 mm). Mobile phase: Cyclohexane–ethanol–acetic acid (85.7 : 11.4 : 2.9).
Sulfonamides HU k
HA
HX
HY
HZ
HAA
k
RI
RI
RT
RT
Caffeine
0.2
305
259
1.9
6.7
Diprophylline
–
275
227
–
3.6
Fenetylline
–
336
277
–
–
Proxyphylline
0.1
293
–
–
–
Theobromine
0.1
262
201
1.6
3.8
Theophylline
0.1
276
249
1.7
4.9
Phthalylsulfathiazole
14.0
Additional systems
Succinylsulfathiazole
16.8
System HAD
Sulfadoxine
4.4
Aymard et al. (2000). J Chromatogr Biomed Sci Appl 744: 227–240.
Sulfamerazine
8.1
n
Sulfaquinoxaline
4.8
Sulfacetamide
7.7
Sulfachlorpyridazine
3.3
Sulfadiazine
8.7
Sulfadimidine
7.1
Sulfafurazole
6.0
Sulfamethoxazole
4.8
Sulfamethoxydiazine
8.2
Sulfamethoxypyridazine
7.5
Sulfamoxole
12.6
Sulfanilamide
8.9
Sulfapyridine
3.8
Sulfathiazole
13.4
Xanthine stimulants The general screening systems, previously described, may be used.
Column: C18 Symmetry Shield (250 4.6 mm i.d., 5 mm) protected by 2 mm Upchurch filter. Column temperature: 30 C. Mobile phase: (A : B) M/15 potassium dihydrogenphosphate with 1% (v/v) octane sulfonic acid : acetonitrile. Mobile phase (MP) 1: (95 : 5) at flow rate 1 mL/min; MP 2: (80 : 20) at flow rate 1 mL/min; MP 3: (30 : 70) at flow rate 1.2 mL/min. Eluent switching programme: At injection, MP1 to the column. From time 12 min to 30 min, MP2 to the column. From time 30 min, MP3 to the column to rinse it. From time 35 min to 40 min, equilibration with MP1. Detection: DAD (l ¼ 260 nm).
n n
n
n
k
Compound
2.7
Lamivudine
3.2
Didanosine
3.8
Stavudine
6.6
Zidovudine
8.1
Abacavir
11.1
Nevirapine
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:11:3
Recommended HPLC systems
755
System HAF
System HBB
Tanaka E et al. (1996). J Chromatogr B Biomed Sci Appl 682: 173–178.
Taninaka C et al. (2000). J Chromatogr B Biomed Sci Appl 738: 405–411.
n n n n n
Column: ODS TSK-gel Super (100 4.6 mm i.d., 2 mm). Mobile phase: (A : B) Acetonitrile–5 mmol/L sodium dihydrogenphosphate (pH 6). Isocratic elution: (45 : 55). Flow rate: 0.65 mL/min. Detection: UV (l ¼ 254 nm).
Retention time (min)
Compound
n n n n n
Retention time (min)
Clonazepam
6.8
Clarithromycin
6.6
Bromazepam
6.8
Erythromycin
9.1
Nitrazepam
9.6
Azithromycin
16.3
Roxithromycin
Triazolam
15.0
Lorazepam
18.4
Etizolam
21.0
Chlordiazepoxide
29.8
Diazepam
32.2
Flutazolam
System HAE
Proust V et al. (2000). J Chromatogr B Biomed Sci Appl 742: 453–458. n n
System HAV
Rutledge DR et al. (1994). J Pharm Biomed Anal 12: 135–140.
n n n n
Column: RP-short alkyl chain, silanol-deactivated (SCD 100; 250 4.6 mm i.d.), stainless steel. Mobile phase: (A : B) Methanol–0.04 mol/L dibasic potassium phosphate (pH 5.5). Isocratic elution: (50 : 50). Flow rate: 1 mL/min. Detection: UV (l ¼ 237 nm).
k
n n n
Celiprolol
2.3
Propranolol
3.6
Diltiazem deacetyldiltiazem
5.1
Diltiazem desmethyldiltiazem
6.1
Diltiazem
6.4
Imipramine
8.2
Verapamil
System HBA
n n n n
Column: C18 base-deactivated silica (125 4.6 mm i.d., 5 mm) with base-deactivated C18 precolumn (20 4.6 mm i.d., 5 mm). Eluent: (A : B) Acetonitrile–50 mmol/L potassium dihydrogenphosphate (pH 7.5, containing 500 mL triethylamine). Isocratic elution: (60 : 40). Flow rate: 2 mL/min. Detection: Fluorescence (lex ¼ 255 nm, lem ¼ 315 nm).
Retention time (min) 8.8
Compound
Compound
6.3
Delavirdine
7.0
Saquinavir
8.0
Nelfinavir
9.4
Amprenavir
22.2
Ritonavir
28.6
Efavirenz
System HAK
Le Guellec C et al. (1988). J Chromatogr Sci Appl 719: 227–233. n n
Sastre-Toran˜o J, Guchelaar H-J (1998). J Chromatogr B Biomed Sci Appl 720: 89–97.
Column: C18 (Lichrospher, 100 RP-18, 5 mm) with C18 precolumn (Lichrospher RP-18, 5 mm). Mobile phase: (A : B) Acetonitrile–25 mmol/L sodium phosphate modified with diethylamine (0.9%) and tetrahydrofuran (1%), pH 3.0. Isocratic elution: (44.8 : 55.2). Flow rate: 0.5 mL/min. Detection: UV (l ¼ 260 nm).
Retention time (min)
Compound
2.2
n
Compound
5.3
13.7
n
Column: C18 (250 6.0 mm i.d., 5 mm). Eluent: (A : B) Acetonitrile–50 mmol/L phosphate buffer (pH 7.2). Isocratic elution: (43 : 57). Flow rate: 1.7 mL/min. Detection: Electrochemical (working electrode: glassy carbon; reference electrode: Ag/AgCl).
n n n
Column: C18 Symmetry (250 4.6 mm i.d., 5 mm) with C18 precolumn Symmetry sentry. Mobile phase: (A : B) Acetonitrile–20 mmol/L potassium dihydrogenphosphate. Elution programme: (50 : 50) to (70 : 30) in 15 min. Flow rate: 1 mL/min. Detection: UV (l ¼ 313 nm).
Retention time (min)
Compound
4.7
Carbamazepine
6.2
Clonazepam
7.6
Nordazepam
9.3
Clobazamm
Not detected
Phenobarbital
Not detected
Phenytoin
Erythromycin
15.7
Clarithromycin
17.1
Roxithromycin
20.7
Azithromycin
System HAL
Boukhabza A et al. (1990). J Chromatogr 529: 210–216. n
Column: C18 Novapak (150 4.6 mm i.d., 5 mm).
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:11:4
756 n n n n n
High Performance Liquid Chromatography
Mobile phase: (A : B : C) Acetonitrile–methanol–6 mmol/L phosphate buffer (pH 5.7). Isocratic elution: (30 : 10 : 60). Flow rate: 1.3 mL/min. Detection: DAD (l ¼ 242 nm). Note: The phosphate buffer stock solution is prepared using 94 mL 0.2 mol/L sodium dihydrogenphosphate added to 6 mL 0.2 mol/L disodium phosphate heptahydrate.
9.1
Compound Desipramine Diazepam
Not detected
Alprazolam
Not detected
Bromazepam
Not detected
Clobazam
Not detected
Codeine
Compound
Not detected
Ephedrine
1.4
Barbital
Not detected
Levomepromazine
1.45
Clonazepam 7acetamidoclonazepam
Not detected
Lidocaine
Not detected
Medazepam
1.55
Clonazepam 7-aminoclonazepam
Not detected
Nortriptyline
2.0
Aprobarbital
Not detected
Propranolol
2.4
Hexobarbital
Not detected
Thioridazine
3.7
Flunitrazepam M (nor)
Not detected
Triazolam
4.4
Nordazepam oxazepam
4.4
Oxazepam
4.6
Nitrazepam
4.33
Clonazepam
5.1
Lorazepam
6.2
Flunitrazepam
6.3
Alprazolam
6.6
Triazolam
7.7
Chlordiazepoxide
7.8
Clobazam
7.9
Nordazepam
8.1
Bromazepam
8.2
Medazepam
13.2
Diazepam
System HAM
de Carvalho D, Lanchote VL (1991). Ther Drug Monit 13: 55–63.
n n n n
Retention time (min) 10.3
Retention time (min)
n
, continued
Column: C18 (150 4.0 mm i.d., 3 mm) with C18 precolumn (40 4.0 mm i.d., 3 mm). Mobile phase: (A : B ) Water–acetonitrile. Isocratic elution: (50 : 50). Flow rate: 0.7 mL/min. Detection: UV (l ¼ 313 nm).
Retention time (min)
Compound
1.8
Theophylline
1.98
Caffeine
2.0
Paracetamol
2.2
Primidone
2.7
Sulfamethoxazole
2.8
Phenobarbital
3.1
Chlordiazepoxide
3.4
Diazepam
3.4, 4.4
Oxazepam
3.5
Phenytoin
4.2
Lorazepam
4.3
Clonazepam
4.5
Nitrazepam
9.0
Imipramine
References Aquilante CL et al. (2006). Common laboratory methods in pharmacogenomic studies. Am J Health Syst Pharm 63: 2101–2110. Atanassova A et al. (2004). A high-performance liquid chromatography method for determining transition metal content in proteins. Anal Biochem 335: 103–111. Badoud F et al. (2010). Fast analysis of doping agents in urine by ultra-high pressure liquid chromatography quadrupole time of flight mass spectrometry II: Confirmatory analysis. J Chromatogr A 1217: 4109–4119. Bijlsma L et al. (2009). Simultaneous ultra-high pressure liquid chromatographytandem mass spectrometry determination of amphetamine and amphetaminelike stimulants, cocaine and its metabolites, and a cannabis metabolite in surface water and urban wastewater. J Chromatogr A 1216: 3078–3089. Bogusz M et al. (1993). An overview on the standardisation of chromatographic methods for screening analysis in toxicology by means of retention indices and secondary standards. Part II. High performance liquid chromatography. Fresenius Z Anal Chem 347: 73–81. Budowle B, van Daal A (2009). Extracting evidence from forensic DNA analyses: future molecular biology directions. Biotechniques 46: 339–340,342–350. Cheng X, Kaplan LA (2003). Simultaneous analysis of neutral carbohydrates and amino sugars in freshwaters with HPLC-PAD. J Chromatgr Sci 41: 434–438. Costabile M (2006). Molecular approaches in the diagnosis of primary immunodeficiency diseases. Hum Mutat 27: 1163–1173. Danielson PB (2005). Separating human DNA mixtures using denaturing highperformance liquid chromatography. Expert Rev Mol Diagn 5: 53–63. Engelhardt H (1977). The role of moderators in liquid-solid chromatography. J Chromatogr Sci 15: 380–384. Ettre LS (1980). Relative retention expressions in chromatography. J Chromatogr 198: 229–234. Hansen SH (1981). Column liquid chromatography on dynamically modified silica. J Chromatogr 209: 203–210. Halasz I et al. (1975). Ultimate limits in high-pressure liquid chromatography. J Chromatogr A 112: 37–60. Herzler M et al. (2003). Selectivity of substance identification by HPLC–DAD in toxicological analysis using a UV spectra library of 2682 compounds. J Anal Toxicol 27: 233–242. Huber S, George A, eds (1993). Applications of diode-array detection in HPLC. In: Chromatographic Science Series 62. New York: Marcel Dekker. Ibanez M et al. (2009). Screening of antibiotics in surface and wastewater samples by ultra-high pressure liquid chromatography coupled to hybrid quadrupole timeof-flight mass spectrometry. J Chromatogr A 1216: 2529–2539. Kosaki K et al. (2005). DHPLC in clinical molecular diagnostic services. Mol Genet Metab 86: 117–123. Lin SY et al. (2008). Mutation spectrum of 122 hemophilia A families from Taiwanese population by LD-PCR, DHPLC, multiplex PCR and evaluating the clinical application of HRM. BMC Med Genet 20(9): 53. Liu W et al. (1998). Denaturing high performance liquid chromatography (DHPLC) used in the detection of germline and somatic mutations. Nucleic Acids Res 26: 1396–1400. Luquin N et al. (2010). DHPLC can be used to detect low level mutations in amyotrophic lateral sclerosis. Amyotroph Lateral Scler 11(12): 76–82. Macnair JE et al. (1997). Ultra high-pressure reversed-phase liquid chromatography in packed capillary columns. Anal Chem 69: 983–989.
Clarke's Analysis of Drugs and Poisons Chapter No. 41 Dated: 10/3/2011 At Time: 16:11:4
Further reading Miura Y, Hamada H (1999). Ion chromatography of nitrite at the ppb level with photon measurement of iodine formed by post-column reaction of nitrite with iodide. J Chromatogr A 850: 153–160. Pragst F et al. (2001). UV Spectra of Toxic Compounds. Data Base of Photodiode Array UV Spectra of Illegal and Therapeutic Drugs, Pesticides, Ecotoxic Substances and Other Poisons. Heppenheim: Verlag Dieter Helm. Pragst F et al. (2002). Suchverfahren (General unknown). In: K€ ulpmann WR, ed. Klinisch-Toxikologische Analyse. Weinheim: Wiley-VCH Verlag GmbH, 49–124. Pragst F et al. (2004). Systematic toxicological analysis by high-performance liquid chromatography with diode array detection (HPLC-DAD). Clin Chem Lab Med 42: 1325–1340. Martin AJP, Synge RLM (1941). A new form of chromatogram employing two liquid phases. Biochem J 35: 1358–1368. Rozing G et al. (2001). A system and columns for capillary HPLC. Am Lab 33: 26–38. Small H et al. (1975). Novel ion exchange chromatographic method using conductrimetric analysis. Anal Chem 47: 1801–1809. Snyder LR (1968). Principles of Adsorption Chromatography. New York: Marcel Dekker, 194–195. Snyder LR et al. (1997). Practical HPLC Method Development. New York: Wiley. van Deemter JJ et al. (1956). Longtitudinal diffusion and resistance to mass transfer as cuases of non ideality in chromatography. Chem Eng Sci 5: 271–289. Ventura R et al. (2008). High throughput and sensitive screening by ultra-performance liquid chromatography tandem mass spectrometry if diuretics and other doping agents. Eur J Mass Spectrom (Chichester) 14: 191–200. Wells MJM, Clark CR (1981). Liquid chromatographic elution characteristics of some solutes used to measure column void volume on C18 bonded phases. Anal Chem 53: 1341–1345.
Further reading Aldridge AA et al. (2009). Ultra high performance liquid chromatography in the contract manufacturing environment. http://pharmtech.findpharma.com/ pharmtech/AnalyticsþArticle/Ultra-High-Performance-LiquidChromatography-in-th/ArticleStandard/Article/detail/584973 (accessed 14 December 2010). Aquilante CL et al. (2006). Common laboratory methods in pharmacogenomic studies. Am J Health Syst Pharm 63: 2101–2110. Armstrong D, Zhang B (2001). Chiral stationary phases for high performance liquid chromatography. Anal Chem 73: 557A–561A. Ayrton J et al. (1998). Use of generic fast gradient liquid chromatography– tandem mass spectroscopy in quantitative bioanalysis. J Chromatogr B 709: 243–254. Bobzin SC et al. (2000). LC–NMR: a new tool to expedite the dereplication and identification of natural products. J Ind Microbiol Biotechnol 25: 342–345. Budowle B, van Daal A (2009). Extracting evidence from forensic DNA analyses: future molecular biology directions. Biotechniques 46: 339–340, 342–350. Dai J (2009). Fast liquid chromatography for method development. Pharm Rev 12: 12–17. Fornstedt T, Guiochon G (2001). Nonlinear effects in LC and chiral LC. Anal Chem 73: 609A–617A. Fritz JS (2004). Early milestones in the development of ion-exchange chromatography: a personal account. J Chromatogr A 1039: 3–12. Gao VCX et al. (1998). Column switching in high performance liquid chromatography with tandem mass spectrometric detection for high-throughput preclinical pharmacokinetic studies. J Chromatogr A 828: 141–148. Guillarme D et al. (2010). New trends in fast and high-resolution liquid chromatography: a critical comparison of existing approaches. Anal Bioanal Chem 397: 1069–1082.
757
Haddad PR et al. (2008). Recent developments and emerging directions in ion chromatography. J Chromatogr A 1184: 456–473. Hamilton RJ, Sewell P (1977). Introduction to High Performance Liquid Chromatography, 2nd edn. London: Chapman & Hall. Heinig K, Bucheli F (2002). Application of column-switching liquid chromatography–tandem mass spectrometry for the determination of pharmaceutical compounds in tissue samples. J Chromatogr B 769: 9–26. Henion J et al. (1998). Sample preparation for LC–MS–MS: analyzing biological and environmental samples. Anal Chem 70: 650A–656A. Hicks RP (2001). Recent advances in NMR: expanding its role in rational drug design. Curr Med Chem 8: 627–650. Jerkovich AD et al. (2003). The use of micrometer-sized particles in ultrahigh pressure liquid chromatography. LC/GC North Am 21: 60–61. Johns D (1987). Resolving isomers on HPLC columns with chiral stationary phases. Am Lab Jan.: 72–76. Karnes HT, Sarkar MA (1987). Enantiomeric resolution of drug compounds by liquid chromatography. Pharm Res 4: 285–292. Lo´pez-Ruiz B (2000). Advances in the determination of inorganic anions by ion chromatography. J Chromatogr A 881: 607–627. Lunn G, Schmitt NR (1997, 2000). HPLC Methods for Pharmaceutical Analysis. Vols 1, 2–4. New York: Wiley. Majors RE (1997). New chromatography columns and accessories at the 1997 Pittsburgh Conference Part 1. LC–GC 15: 220–237. Majors RE (1998). New chromatography columns and accessories at the 1998 Pittsburgh Conference Part 1. LC–GC 16: 228–244. Majors RE (1999). New chromatography columns and accessories at the 1999 Pittsburgh Conference Part 1. LC–GC 17: 212–220. Majors RE (2000). New chromatography columns and accessories at the 2000 Pittsburgh Conference Part 1. LC–GC 18: 262–285. Meyer VR (1979). Practical High Performance Liquid Chromatography, 2nd edn. New York: Wiley. Peng SX et al. (1999). Direct determination of stability of protease inhibitors in plasma by HPLC with automated column-switching. J Pharm Biomed Anal 25: 343–349. Plumb RS et al. (1999). The application of fast gradient capillary liquid chromatography–mass spectrometry to the analysis of pharmaceuticals in biofluids. Rapid Commun Mass Spectrom 13: 865–872. Sch€ ufer C et al. (2001). HPLC columns: the next great leap forward – Part 1. Am Lab Feb.: 40–41. Sch€ ufer C et al. (2001). HPLC columns: the next great leap forward – Part 2. Am Lab Apr.: 25–26. Simpson CF (1976). Practical High Performance Liquid Chromatography. London: Heyden. Snyder LR (2000). HPLC past and present. Anal Chem 72: 412A–420A. Tanaka N et al. (2001). Monolithic LC columns. Anal Chem 72: 420A–429A. The MHE Research Foundation (2008). DHPLC Genetic Testing. Available at: www. mheresearchfoundation.org/DHPLC_Genetic_Testing.html (accessed 30 October 2008). Transgenomic Inc. (2008). Wave Systems for mutation detection. Available at: www.transgenomic.com/lib/br/602077.pdf (accessed 30 October 2008). Wehr T (2000). Configuring HPLC systems for LC–MS. LC–GC 18: 406–416. Weiss J (2005). Handbook of Ion Chromatography, 3rd revised updated edn. Weinheim: Wiley-VCH. Wilson I et al. (2000). Analytical chemistry: advancing hyphenated chromatographic systems. Anal Chem 71: 534A–542A. Wolfender JL et al. (2001). The potential of LC–NMR in phytochemical analysis. Phytochem Anal 12: 2–22. Yang LY et al. (2001). Applications of new liquid chromatography–tandem mass spectrometry technologies for drug development support. J Chromatogr A 926: 43–55.
Clarke's Analysis of Drugs and Poisons Chapter No. 42 Dated: 10/3/2011 At Time: 16:24:6
CHAPTER
42
Capillary Electrophoresis F Tagliaro, A Fanigliulo, J Pascali and F Bortolotti
Introduction
Theoretical aspects
Since its introduction in the early 1980s, and particularly in the first decade of this century, capillary electrophoresis (CE) has established a prominent role in many areas of applied analytical chemistry. This is demonstrated by an increasing number of applications and scientific papers in biomedical sciences, toxicology, biopharmaceutics, biotechnology, and environmental and forensic science. The spread of this application relies on its peculiar features:
Capillary electrophoresis can be defined as high-voltage electrophoresis (10–30 kV) carried out in a capillary-shaped separation compartment (typical dimensions: internal diameter 20–100 mm, length 20–100 cm). The geometry of this set-up, with respect to traditional slab gel electrophoresis, offers the neat advantages of minimal band diffusion and improved joule heating control, thus permitting the application of high voltages. The reduced zone broadening results in excellent separation efficiency, reaching up to 106 theoretical plates. The CE separation mechanism, as in any form of electrophoresis, is based on the principle that charged species subjected to an electric field tend to migrate, driven by electrostatic force, towards the electrode with the opposite charge. Their velocity depends on their electrophoretic mobility (m), which is specific for each individual ionic species on the basis of the mass-to-charge ratio, as described by Equation (42.1):
n n n n
n
Wide analytical applications (from inorganic ions to large DNA fragments and even viruses and cells) Variety of separation modes (electrophoretic, electrokinetic, chromatographic and more) Variety of detection systems (from ultraviolet (UV) spectroscopy to mass spectrometry (MS)) High separation efficiency (up to millions of theoretical plates) and mass sensitivity (from femtomoles (1015 moles) down to yoctomoles (1021 moles)); minimal consumption of samples (in the order of nanolitres) and solvents (a few millilitres per day) Simple and inexpensive operation coupled with instrumental ruggedness.
Moreover, being based on specific separation mechanisms, CE has established itself as an independent analytical technique complementary to chromatography. Capillary electrophoresis originated from the optimisation of basic electrophoretic principles traditionally applied using slab gels. The range of applications of CE soon extended to include hybrid separation mechanisms, partially borrowed from chromatography, and it has become a highly versatile and flexible tool in the hands of separation scientists. CE has been recognised as potentially admissible in the US courts as a form of evidence in accordance with the Daubert Standard (Kuffner et al. 1996). Although CE has received major attention in molecular biology, clinical chemistry, analytical toxicology and other areas of forensic analysis, it is still ‘in its infancy’. Forensic science laboratories have to deal with a range of diverse analytical problems involving, for example, gunshot residues, explosives, inks, dusts, soils, illicit drugs, poisons, DNA fragments and proteins and others, and CE has particular characteristics that make it applicable to all these areas. Forensic samples are often limited in quantity and heavily contaminated and must be conserved as far as possible in order to allow for further investigations. CE has great potential as a practical and productive investigation tool for analytical toxicologists and other forensic scientists dealing with such samples. In addition, scientists working in the fields of clinical and biochemical analysis, where analytical versatility, minimum use of sample volume and low operative costs are extremely important, derive considerable benefits from the technique. This chapter builds on that contributed to the third edition of this publication by Professor David Perrett in that it aims to illustrate the basic principles of CE by giving a description of its instrumentation and of the different modes of separation and detection. A major addition is the presentation of recent review papers that cover important topics of CE applications in analytical toxicology and to which the readers are referred for more detailed information. Further, a selected number of applications of particular interest are discussed.
758
mi ¼
qi 6phri
ð42:1Þ
where mi ¼ ion mobility, qi ¼ ion charge, h ¼ electrolyte solution viscosity, and ri ¼ ion effective radius. When an electric field of strength E is applied, the ion migration velocity (vi) will equal the product mi E, causing the physical separation of the components of a mixture of molecules with different mi (Equation 42.2): vi ¼ mi E
ð42:2Þ
where the electric field strength E is given by Equation (42.3): Electric field ðEÞ ¼
Applied voltage V ¼ Distance between electrodes d
ð42:3Þ
An additional phenomenon known as electroosmosis takes place inside the separation capillary and results from the double electrical layer that builds up at the solid–liquid interface whenever a solid surface is in contact with a solution of ions. This ionic double layer is described by the so-called zeta potential z. The inner wall of capillaries, silica being the most common material of which the CE capillaries are made, exhibits an excess of surface charge, since it contains a great number of silanol groups (SiOH), which at pH values higher than 2 are ionised as SiO. The resulting negative charge of the wall surface tends to be compensated for by cations attracted from the solution, thus building up a double electric layer. When an electric potential difference is established between the ends of the capillary (i.e. when electrophoresis starts), all the cations in the solution migrate towards the cathode (the negative electrode) and anions move in the opposite direction. The migration towards the cathode of excess cations close to the capillary wall (being not compensated by migration of the corresponding anions stationary in the wall) drags water in the same direction, thus producing a measurable flow of liquid inside the capillary (typically tens of nanolitres per minute), termed ‘electroosmotic flow’ (EOF). Capillaries made of materials other than silica (e.g. Teflon), or with the wall coated with neutral or charged coatings (negative or positive), will display the same phenomenon, depending on the degree of ionisation of the wall surface. It is
Clarke's Analysis of Drugs and Poisons Chapter No. 42 Dated: 10/3/2011 At Time: 16:24:6
Instrumental hardware
v ¼ ðmi þ mEOF Þ E
ð42:4Þ
The set-up of a capillary electropherograph is usually with the detector close to the end of the capillary towards which the EOF is directed, and liquid flow will drag towards the detector all the solutes contained in the injected sample, excluding those with an electrophoretic counter-migration velocity higher than the EOF itself (small ions with high charge-tomass ratio). Inside this flow of solvent, the ionic species migrate according to the respective mi, whereas neutral solutes (with mi ¼ 0) migrate all together at the velocity of the EOF. In short, with the usual instrumental arrangement, having the injector at the anode end and the detector close to the cathode end of an uncoated fused-silica capillary (negatively charged at the inner wall), the cations with the highest mobility will arrive at the detector first, followed by the cations with progressively lower mobility (lower charge-to-mass ratio); subsequently, the bulk of the neutrals will appear at the detector, followed by the anions in reversed order of mobility (slow anions first, fast anions last). Only the fastest anions with electrophoretic mobility higher than that of the EOF will not be detected, because they will escape from the opposite end of the separation capillary. The EOF, being driven by a force generated close to the capillary wall, has a peculiar ‘piston-like’ flow profile, which is particularly beneficial for molecular separations. In fact, it minimises band broadening, which is a typical drawback of capillary liquid chromatography, where the pressure-driven flow of the mobile phase, hindered by shear forces at the wall, yields a parabolic flow profile. The coexistence of both electrophoretic and electrokinetic phenomena, which can be tuned separately and in combination to achieve molecular separation, is a unique feature of CE that can be exploited to perform a great variety of separations modes. In addition, interactions taking place between analytes and other molecules (e.g. complexforming molecules, organic solvents, micelles, polymer gels) present in the medium in which separation occurs further contribute to differentiation of the migration velocities of different chemical species according to non-electrophoretic principles. For a detailed explanation of the theoretical principles of CE, readers are referred to publications listed in the ‘Further reading’ section of this chapter, and particularly to the book by Ahuja and Jimidar (2008). Finally, although quantification methods used in CE are similar to those applied in other forms of chromatography, in that they mainly use internal standardisation, two important differences must be emphasised. First, in CE most separations have an ‘on-column’ detection step. In this process, the velocity of the peaks crossing the detection window differs according to the respective ion mobilities of the analytes. As a result, the ‘apparent peak width’ of zones moving at different velocities will differ, even if the ‘real band width’ is the same. Consequently, two equal bands differing only in mobility will show different ‘peak areas’, because of the difference in their residence times inside the detector cell. To overcome this problem, for quantitative computations the ratio of peak area to migration time can be used instead of peak areas. An alternative is to use peak heights. Second, since only minute amounts of sample (of the order of a few tens of nanolitres) are injected onto the separation capillary, it is difficult to handle these volumes with precision using current technology. This seriously restricts the use of external standardisation for quantitative methods. Thus, as for example with gas chromatography (GC), internal standardisation is much preferred.
Instrumental hardware The outstanding feature of CE instrumentation is its basic simplicity. A schematic representation of a capillary electropherograph is given in Figure 42.1. In brief, as in other chromatographic techniques, a CE analysis is based on three major steps: sample injection, separation and detection. As a consequence, a capillary electropherograph can be described as an assembly of the following components: n n n n
An injection system A high-voltage power supply A separation capillary (generally located in a thermostatted compartment) A detector.
In CE, only a minute amount of sample can be introduced into the capillary, in order to maintain the separation efficiency. Overloading of samples causes peak broadening and peak distortion. Sample loading is limited by capillary dimensions. Typically, the length of the sample plug should not exceed 1–2% of the total capillary length and this corresponds to volumes of between 10 and 100 nL, for typical CE capillaries, which have an inner volume ranging between 1 and 4 mL. Sample injection is achieved by replacing the inlet buffer reservoir with the sample vial for a few seconds. The forces driving sample introduction into the separation capillary may be brought about by applying pressure, voltage or both. The most common modes are hydrodynamic and electrokinetic injection. Hydrodynamic injection is the most widely used and is carried out by the application of a positive pressure at the inlet end of the capillary, by the application of a vacuum at the terminal end or by exploiting siphoning between the two capillary ends. In this way, the amount of sample loaded is almost independent of the sample composition and is representative of the whole sample solution. Hydrodynamic injection is described by Equation (42.5): V¼
DPd4 pt 128hL
ðHagenPoiseuille equationÞ
ð42:5Þ
where V ¼ sample volume, DP ¼ applied pressure, d ¼ capillary internal diameter, t ¼ injection time, L ¼ capillary length and h ¼ buffer viscosity. The typical applied pressure range is between 5 and 30 psi (1 psi ¼ 69 mbar ¼ 6.9 kPa) and injection times are between 5 and 20 seconds, although these parameters depend strongly on method optimisation. The advantages of this injection mode are reproducibility and suitability for quantitative analysis. However, it is subject to limited sensitivity owing to the tiny volume of sample introduced into the capillary and hence the limited mass of analytes that can be introduced into the capillary.
PC control
Power supply (0–30 kV)
AU
important to point out that even ‘neutral’ capillaries may display a charged surface because of the adsorption on the wall of molecules present in the solution. Electrophoretic migration and electroosmotic flow may have either the same or the opposite direction. Thus, they sum as vectors and induce ionised species to migrate with an apparent velocity (v) resulting from the sum of their intrinsic mobility (mi) and the mobility of the EOF (mEOF) (Equation 42.4):
759
Time (min)
Capillary
Detector Electrodes Inlet reservoir
Outlet reservoir
Figure 42.1 Schematic representation of a capillary electropherograph.
Clarke's Analysis of Drugs and Poisons Chapter No. 42 Dated: 10/3/2011 At Time: 16:24:6
760
Capillary Electrophoresis
Electrokinetic injection is obtained by applying a voltage for a few seconds while the injection end of the capillary is dipped into the sample vial. In this way, analytes enter the separation capillary by both electrophoretic migration and the pumping effect of the EOF. The amount of sample introduced is given by Equation (42.6): Q¼
ðmi þmEOF ÞVpr 2 Ct L
ð42:6Þ
where Q ¼ grams or moles of injected amount, mi ¼ analyte electrophoretic mobility, mEOF ¼ EOF mobility, V ¼applied voltage, r ¼ capillary radius, t ¼ injection time, L ¼ capillary length and C ¼ analyte concentration. It can be seen that, in electrokinetic injection, discrimination occurs on the basis of the electrophoretic mobility of the sample components, so that ionic species with mobilities towards the detector will be preferentially introduced with respect to neutral species and ions with the opposite charge. This results in selectivity and sensitivity enhancements for the analytes that are preferentially loaded, but also in delicate reproducibility, strongly affected by the sample composition. Electrokinetic injection is the injection mode exploited to realise ‘sample stacking’ procedures that allow large increases in analytical sensitivity, as discussed below with regard to sample pretreatment and sample enrichment. Separation capillary Fused silica is by far the most widely used material for CE capillaries, since it is chemically and electrically inert, physically resistant, UV transparent (for the needs of in-capillary UV detection) and displays good thermal conductivity (in order to ensure high dissipation of the joule heat). To achieve the necessary mechanical resistance, capillaries are externally coated with a protective polyimide layer, in a similar way to GC capillaries. This external layer is optically opaque and must be removed in the region where an optical detector is to be placed. This can easily be achieved by burning or scraping off the material in a capillary segment corresponding to the detection window. In-capillary optical detection has the advantage of avoiding any post-separation added volumes and consequent band broadening, but has disadvantages in terms of optical path length and cell geometry. Typical ranges for inner and outer diameters of CE capillaries are 25–100 mm and 350–400 mm, respectively. Capillary length typically rages between 20 and 100 cm. Fused-silica capillaries can be internally ‘uncoated’ (naked) or ‘coated’ with thin layers of polymers. The most common internal coatings include amines, polyacrylamide, cellulose, poly(vinyl alcohol)s, amino acids, surfactants, aryl pentafluoro compounds, poly (vinylpyrrolidinone) and poly(ethyleneamine). Also, liquid chromatography (C2, C8, C18) or GC (poly(ethylene glycol) and phenyl methyl silicone) stationary phases can be used as capillary wall coatings. An inherent problem in CE separations is reproducibility, since the EOF and consequently migration times of analytes are affected by the inner surface conditions. A common practice adopted to refresh the inner surface of uncoated silica capillaries and remove adsorbed materials is that of flushing with strong bases, which dissolve a thin layer of the silica surface. Usually, 1 mol/L sodium hydroxide solution is flushed through the capillary, followed by 0.1 mol/L sodium hydroxide solution and then by the separation buffer for final conditioning. The frequency of base conditioning during the experimental work depends on the nature of the samples, the buffer employed, the working pH, the background electrolyte (BGE) concentration and the nature of the capillary surface. Acid washing and flushing with solvents are less popular but effective practices, particularly when coated capillaries are used that would suffer from exposure to basic solutions. Thermostatting is another key point to ensure reproducibility of capillary separations. Temperature variations due to joule heating should be minimised in CE analyses so as to minimise viscosity changes in both separation and injection. Thermostatting is better achieved by using a refrigerant fluid, but air streams are also effective when separations generate low currents.
Power supply A high-voltage power supply is a fundamental part of a CE instrument. It should be capable of delivering up to 30 kV, with currents up to 200–300 mA. Because of the typical direction of the EOF (towards the cathode, in naked fused-silica capillaries), the standard polarity configuration has the anode (positive electrode) placed at the injection end of the capillary and the cathode (negative electrode) at the opposite end, close to the detector (a configuration known as ‘normal polarity’). However, in many cases (e.g. when the EOF is directed towards the anode, because the capillary wall is coated with a positively charged wall modifier) the application of a ‘reversed polarity’ configuration is needed in order to allow the analytes to migrate to the detector. For this reason, dual-polarity power supplies that are capable of rapidly switching the electrode polarity are used in modern CE instrumentation. CE separations are usually carried out at a constant potential, but separations at constant current can sometimes be needed. An advantage of constant-current mode is the possibility of compensating by automatic adjustments of the voltage for viscosity changes caused by inadequate temperature control. Voltage programming, i.e. applying gradients of voltage or current during the analysis, has also been reported to improve separation efficiency in the case of complex samples, but the advantages of gradient application are not as important as in GC and high performance liquid chromatography (HPLC), where temperature or buffer composition gradients are the most popular mode for tuning separation. Detectors Although it might be expected that the detection of analytes directly inside the capillary through its transparent silica wall would be relatively simple, this is not the case. Inherently, the main problem related to detection is sensitivity, not in terms of detectable mass (picogram amounts of analytes are easily detectable inside the capillary with simple UV detectors), but in terms of concentration of analyte. Since only minute volumes (nanolitres) of samples can be introduced into the tiny separation compartment (i.e. the capillary, with a typical volume 1 mL), only very small amounts of analytes will reach the detector. On this basis, taking into account the high efficiency of CE (i.e. the ability to produce separations without generating dilution of the electrophoretic zones), it is mandatory to avoid zone broadening in order to achieve an acceptable sensitivity, either ‘in-capillary’ or ‘off-capillary’. The way to achieve this is by limiting any ‘dead volumes’ in the system, particularly at the detection side, and increasing the response time of the detectors. In the next subsections the most common CE detectors are described and their relative advantages and drawbacks discussed. Optical methods
UV is the most commonly employed means of detection in CE instruments. Absorbance detection is usually performed ‘in-capillary’, with the optical beam focused directly into the capillary, crossing a transparent window made in it by removing the protective polyimide external coating. The limited optical path inside the capillary and the poor optical shape of the capillary section are the major factors negatively affecting sensitivity. There are also other features peculiar to CE separation that have to be taken into account in quantitative analysis. As mentioned previously, detection in CE occurs when separation is still taking place, in contrast to chromatographic techniques where all analytes move at the same velocity in front of the detector, which is located after the separation column. In CE, the migration velocity differs from one analyte to another during detection such that their residence time in the detector is affected by their different velocities, and this affects the respective peak areas. To correct this artefact in quantitative analysis, peak areas in CE should be divided by their migration times. Moreover, in CE the dynamic linear range of optical detection is less extended than in HPLC, particularly with UV detection, since deviations from the Beer–Lambert Law occur owing to the small size and curvature of the capillary cell.
Clarke's Analysis of Drugs and Poisons Chapter No. 42 Dated: 10/3/2011 At Time: 16:24:6
Modes of separation Despite these limitations, UV detection is generally applicable, easy to perform and acceptably sensitive for most applications. Capillary design and optical cell design may be optimised to improve sensitivity. Two examples are Z-shaped cells and bubble cells. In Z-cells, a double right angle, in the capillary region where detection takes place, results in an increased optical path length. The light beam runs axially for the length included between the two right angles. In bubble cells, solutes pass through an expanded region (‘bubble’) inside the capillary, where the analyte zone expands radially and, consequently, contracts longitudinally, increasing the path length without sacrificing separation. The availability of multichannel, dispersive optical detectors (diode array detectors, DADs) for commercial CE instruments has greatly impacted CE instrumentation, allowing for enhanced information content at each analytical run and easier method development. Fluorescence detection, mainly based on laser-induced fluorescence (LIF), has been proficiently applied both to fluorescent analytes and to molecules that can be made fluorescent after chemical derivatisation. In this case, sensitivity can be as high as 1012 mol/L. In the case of non-UV-absorbing ionised molecules, ‘indirect detection’ can be performed by introducing a detectable (UV-absorbing) additive, with the same charge as the analyte of interest and similar mobility to the running buffer. Displacement of the additive by the nonabsorbing analyte will occur in the capillary in the zone of residence of the analyte and consequently a negative peak will be recorded by the detector (Beckers, Bocek, 2003; Johns et al. 2003). Indirect detection, for instance, makes possible the determination of small organic and inorganic ions, most of which do not absorb UV light, using the UV detectors present in all commercial CE instruments. Electrochemical detectors
Electrochemical (EC) detectors, particularly amperometric and conductimetric detectors, can be successfully coupled with CE. Electrochemical detection can be performed either ‘in-capillary’ or ‘off-capillary’. Amperometric detectors record the electronic current generated by an electrochemical reaction involving the analyte molecules, thus displaying, for oxidisable (or reducible) analytes, high sensitivity and good selectivity. This technique is mass sensitive and therefore independent of the cross-sectional pathlength of the capillary. Conductimetric detectors are simpler, although less selective, than amperometric detectors. Their application to CE allows direct detection of small ions, making possible ion analysis in a configuration resembling ion chromatography. A general problem with EC detection in CE is the isolation of the separation circuit from the detection circuit to avoid interferences. A further problem of the EC detection mode is the necessity of using miniaturised electrodes, compatible with capillary dimensions. These devices are not commercially available so far, making EC detection (particularly amperometric) still reliant on home-made components. Mass spectrometry
(See also Chapters 37 and 41.) Hyphenation with mass spectrometry (CE-MS) is the latest major innovation in CE. In recent years the application of CE-MS has grown substantially, and now accounts for a large number of publications in the pharmaceutical, biotechnological, environmental and toxicological science literature (Schmitt-Kopplin, Frommberger 2003; SchmittKopplin, Engelmann 2005). The key issue when coupling CE with MS is the efficient transfer of ions from the CE capillary to the mass analyser without sacrifice of separation performance and sensitivity (Smith et al. 1988). The electrospray ionisation (ESI) source has proved to be the most suitable for CE-MS coupling. This produces gas-phase ions from ions in solution, which are introduced into the mass spectrometer after being sprayed in a plume of droplets, as a result of a voltage applied on a holed metal tip. The ESI source is compatible with the CE-limited flows, since it is concentration sensitive and not mass sensitive. Other ion sources can be coupled with CE, for example atmospheric pressure chemical ionisation (APCI; Tanaka et al. 2003) and atmospheric pressure photoionisation (APPI; Mol et al. 2005), but the recent successful results of CE-MS interfacing are due to the development of the CE-ESI hyphenation.
761
Coupling CE with ESI involves joining a high-voltage-driven separation technique with a high-voltage-based molecule ionisation technique. Thus, it is necessary to ensure electrical continuity between the two systems while at the same time keeping the respective circuits independent. Second, a stable flow of ionised molecules must be constantly produced during analysis and therefore a BGE compatible with the ion source must be chosen. CE can be interfaced with ESI with essentially two possible alternatives, as well described by Cai and Henion (1995): the ‘sheath liquid’ interface and the ‘sheathless’ interface. In the former case the electrical continuity is brought about by an additional aqueous/organic flow (the sheath liquid) of the order of magnitude of mL/min, which dilutes the liquid stream coming out of the separation capillary (nL/min). This accessory flow also ensures the onset of a stable ion spray. The diluting effect on the sample may be seen as a drawback in terms of sensitivity, but, on the other hand, it allows for the use of non-volatile BGEs or small amounts of additives or coating agents in the separation buffer (van Wijk et al. 2007). In a sheathless interface, the electrical contact between the CE buffer and ion source is established by making the terminal end of the separation capillary conductive via several devices, such as stainless-steel connections, microelectrodes, conductive polymer coatings, and metal coatings of gold, silver, nickel or chromium. Unfortunately, the lack of reproducibility and robustness, due to delicate fabrication and manipulation of miniaturised components, limits its use on a large scale despite its advantages in terms of sensitivity (Smith 1990). Novel sheathless configurations have recently been designed, exploiting the results of the developments in nanospray and other miniaturised technology, with promising results (Janini et al. 2003; Kele et al. 2005). Fast mass analysers with a high sampling rate along the electropherogram are required to follow the tiny, fast and closely moving CE peaks, which reflect the intrinsic high efficiency of this separation technique. CE has been hyphenated with almost all types of MS detector: magnetic sectors (Perkins, Tomer 1994), single and triple quadrupoles (Baidoo et al. 2003), ion traps (IT; McClean et al. 2000; Wey et al. 2000; Wey, Thormann 2001a; Iio et al. 2003), Fourier transform ion cyclotron resonance (FTICR; Hofstadler et al. 1994, 1996) and time-offlight (TOF) mass spectrometers (Lazar et al. 1998a, 1998b; Ullsten 2004). At present, as in HPLC, ion traps remain the most widely employed detectors for CE because of their relative speed of scanning, relatively low cost and the possibility of performing multistep fragmentation experiments (MSn). The hyphenation of CE with TOF MS is seen as particularly promising, because of the high scan rate (10–200 ms per spectrum) and high sampling frequency of this detector (Lazar et al. 1998a, 1998b; 1999). TOF shows a particular advantage for toxicological analyses owing to the possibility of identifying molecules by determining their accurate mass and not requiring fragmentation databases. Microchip CE A specific advantage of CE is that instrumentation can easily be miniaturised in a microchip format, since a precise control of fluidics and other separation conditions can be obtained simply by changing the voltage and without the need of mechanical pumps, valves, etc. as in HPLC. CE microchips can also provide custom design, versatility, reduced consumption of reagents and sample, low waste generation, and increased analysis speed and portability. Because of the minimal amounts of sample injected, the most commonly used detection method is LIF, which offers the highest sensitivity. Microchip CE is mostly suitable for portable devices and fully automated analysers. Instrumentation based on this technique is now commercially available. Table 42.1, taken from the third edition of this work, gives an indication of the relative sensitivities of CE detectors towards various analytes in terms of mass and concentration limit of detection (LOD).
Modes of separation A particular advantage of CE is the possibility of using the same hardware to perform different separation modes by simply adjusting a few
Clarke's Analysis of Drugs and Poisons Chapter No. 42 Dated: 10/3/2011 At Time: 16:24:7
762
Capillary Electrophoresis
Table 42.1 Relative sensitivity of CE detection systems towards appropriate compounds Detection mode
LOD (moles injected) 13
–10
16
LOD (mol/L) 10
3
8
–10
Commercial availability
Comments
Yes; DAD supplied with the major instruments
At 3) of about 16 ng/mL in spiked urine using the sweeping-MEKC method with fluorescence detection (lex ¼ 320 nm; lem ¼ 390 nm), and a LOD of about 1 ng/mL with CSEI-sweep-MEKC. The sweeping-MEKC was also used for the online concentration and analysis of LSD in clandestine tablets (Fang et al. 2003). LIF detection, using a He–Cd laser at 325 nm wavelength, has also been applied to the determination of LSD in specimens of human blood (Frost, Koehler 1998). The CE method used a citrate–acetate system as running buffer. The use of DAD as a CE detector in routine methods has attracted particular attention in drug analysis. As expected from a multichannel detector, recording an entire signal spectrum enhances the capability of correct identification and determination of analytes and the possibility of studying the peak purity. An example of a CE-DAD application can be found in Nieddu et al. (2005). This rapid method realises the screening of a class of amfetamine designer drugs, comprising 10 methylenedioxy derivatives of amfetamines and phenethylamine in human blood. The detection limits and recoveries from blood samples were between 10 and 30 ng/mL and 81 and 90%, respectively. CSEI was used by Meng et al. (2006) as an on-line concentration method for the high-sensitivity analysis of illicit amfetamines. Using this approach (CSEI with micellar sweeping), a LOD lower than 50 pg/mL was achieved, whereas using normal MEKC it was about 10 ng/mL. The quantitative reproducibility of CSEI-micellar sweeping for the analysis of amfetamine, metamfetamine and MDMA using a benzylamine internal standard was satisfactory (standard deviation around 10%). The method was also tested on hair samples. Among multichannel detectors, mass-selective detectors have rapidly become the gold standard in toxicological analysis for both generic and specific determinations. Recently CE-MS coupling has begun to find applications in the field of drug analysis (Smyth 2006). Benzodiazepines are a challenging class of drugs to assay in biological fluids and represent a typical example of the proficient use of MS detection based on its outstanding identification power and selectivity. A double-dynamic coating CZE method with IT-MS detection was developed by Vanhoenacker et al. (2004a) for the separation of six benzodiazepines in spiked urine, and MS2 experiments were performed for confirmation. McClean et al. (2000) optimised a CZE-ESI-IT-MS method for the determination of selected 1,4-benzodiazepines, identified by sequential product ion fragmentation. MS3 fragmentation was also exploited by Wey et al. (2000) to unambiguously identify codeine, morphine, dihydrocodeine and their glucuronides in urine samples previously screened using opiate immunoassay. The simultaneous detection and quantification of a number of abused drugs in human hair (namely 6-monoacetylmorphine, morphine, amfetamine, metamfetamine (MA), methylenedioxyamfetamine (MDA), methylenedioxymetamfetamine (MDMA), benzoylecgonine, ephedrine and cocaine) was obtained by Gottardo et al. (2007a) with a rapid CZE-ESI-IT-MS method (Figure 42.3). Ammonium formate (25 mmol/L), pH 9.5 and 15 kV separation were employed for the separation using a bare fused-silica capillary. Under field-amplified sample stacking conditions, LODs were below 0.1 ng/mg for all drugs in hair matrix and good linearity was achieved in the concentration range 0.025–5 ng for each analyte per mg of sample. The same group investigated the application of these separation conditions when coupling CEESI with a TOF detector (Gottardo et al. 2007b), in order to exploit the advantages of high mass accuracy and fast scanning capability of this technique. Drugs and metabolites were identified in hair samples, after a single extraction procedure, by exact mass and isotopic pattern matching (Figure 42.4). Analytical precision in real matrices proved acceptable in both within-day and day-to-day tests.
Intensity × 106
766
MA + EPH A MDMA + MDA
3
COC
2 MOR IS + 6-MAM
1 BE
0 13
14
15
2
16 17 Time (min)
18
19
1
0 2
4
6
8
10
12
14
16
18
Time (min)
Figure 42.3 Toxicological analysis of drugs of abuse in hair using CZE-ion trap MS. Peak identification: A, amfetamine; MA, metamfetamine; MDA, methylenedioxyamfetamine; MDMA, methylenedioxymetamfetamine; EPH, ephedrine; COC, cocaine; BE, benzoylecgonine; MOR, morphine; 6-MAM, 6-acethylmorphine; IS, internal standard (folcodine). For analytical details, see text. (From Gottardo et al. 2007a, with permission.)
Clarke's Analysis of Drugs and Poisons Chapter No. 42 Dated: 10/3/2011 At Time: 16:24:8
Applications of CE to forensic and clinical drug analyses
767
2.5
Intensity × 106
2.0
1.5
1.0
0.5
(a)
0.0
8
10
12
14
16
18
20
Time (min) 2.0 6
Intensity × 106
1.5
1.0
2 5
0.5
IS
1 10 9
4 8
3
(b) 0.0
8
10
12
14
16
18
20
Time (min)
Figure 42.4 Toxicological analysis of drugs of abuse in hair using CZE-TOF MS. Peak identification: 1, MA, metamfetamine; 2, MDMA, methylenedioxymetamfetamine; 3, amfetamine; 4, MDA, methylenedioxyamfetamine; 5, ephedrine; 6, cocaine; 7, codeine; 8, benzoylecgonine; 9,6-acethylmorphine; 10, morphine, at a concentration of 0.2 ng/mg each drug; IS, internal standard (folcodine). For analytical details, see text. (From Gottardo et al. 2007b, with permission.)
the capillary, with resulting enantiomeric separation. When MS detection is used, the anionic chiral selector does not enter the MS ion source, because it is driven in the opposite direction by the applied voltage. Iio et al. (2003) used the complete filling technique in which the chiral selector, a diluted mixture of 3 mmol/L b-cyclodextrin and 10 mmol/L heptakis(2,6-di-O-methyl)-b-cyclodextrin, was instead added directly to the BGE. This method achieved the enantioseparation of metamfetamine and its metabolites in human urine samples with detection limits (with MS) in the range 0.03–0.05 mg/mL. Amfetamine, metamfetamine, MDA, MDMA and MDEA were also used as test compounds in a study by Souverain et al. (2006) on the different strategies for rapid chiral analysis using CE. The adoption of
hydroxypropyl-b-cyclodextrin as the chiral selector was investigated using different approaches (short-end injection, high electric fields, external pressure application, dynamic coating of the capillary) with a view to decreasing analysis time and increasing sensitivity. A general paper on the optimisation of chiral separations using dual neutral cyclodextrins (b-CD and dimethyl-b-CD) was published by Nhujak et al. (2005), showing the potential advantages of adding two chiral selectors to the running buffer. As previously discussed, non-aqueous capillary electrophoresis (NACE) is particularly eligible for CE-MS coupling, because of the high volatility of most organic solvents, which is beneficial for ionisation. Moreover, the observed change in selectivity in comparison with
Clarke's Analysis of Drugs and Poisons Chapter No. 42 Dated: 10/3/2011 At Time: 16:24:9
768
Capillary Electrophoresis
separations in aqueous buffer may offer additional practical advantages in resolution. A complete review on NACE-MS has been published by Scriba (2007), collecting all the published NACE-MS applications up to November 2006. The low number of papers reviewed is probably due to the difficulties in adopting a separation technique whose physical–chemical mechanisms are still scarcely understood. Peri-Okonny et al. (2003) exploited NACE-MS for the separation of nine basic drugs, including tricyclic antidepressant and bronchodilator drugs, with 80 mmol/L acetate buffer dissolved in methanol–acetonitrile (80 : 20 v/v). The results showed improved efficiency compared with HPLC separation. In a paper by Steiner and Hassel (2005), a few organic solvents (namely methanol, acetonitrile, DMSO, formamide, N-methylformamide and N,N-dimethylformamide) were compared against water for the preparation of ammonium acetate separation buffer. Selectivity, peak efficiency and average plate counts were evaluated in the separation of basic drugs. In the seven different solvents (including water), the shortest run time was obtained with acetonitrile, the best peak resolution with the amphiprotic solvents (especially methanol), best peak efficiency with methanol and formamide, and the most sensitive ESIMS detection with acetonitrile and methanol, but with only a slight advantage compared with water. Anderson et al. (2004) developed a NACE-MS method for the determination of lidocaine and two of its metabolites in human plasma. The effects of sheath liquid composition, drying gas temperature and nebulising gas pressure on separation efficiency were evaluated. Fluoxetine and related compounds were analysed with NACE-MS by Cherkaoui and Veuthey (2002), using 25 mmol/L ammonium acetate–1 mol/L acetic acid in acetonitrile as the running buffer and a 30 kV separation voltage. A significant increase in sensitivity was obtained compared with UV detection. NACE-MS was also reported by Geiser et al. (2000) for separation of amfetamine derivatives in spiked urine, after LLE. Although proposed as an attractive alternative to chromatography and supported by positive results, CE traditionally lacks sensitivity compared with chromatographic methods. However, on-line and offline sample enrichment and stacking injection techniques have been employed successfully to enhance sensitivity. As previously mentioned, CSEI was used by Meng et al. (2006) for the high-sensitivity analysis of illicit amfetamines and achieved a LOD of 15 mg/L, limit of detection, 1460 . Insoluble in water; very slightly
soluble in concentrated hydrochloric acid and nitric acid. Used in OTC stomach antacids. Aluminium Phosphide AlP = 58.0 CAS—20859-73-8 Synonym Aluminium monophosphide Proprietary Names Celphos; Delicia; Delicia Gastoxin; Detia GAS EX-B/EX-T;
Detia phospine pellets; Phostoxin; Quick-Phos; Quick-Fume.
Chemical Properties Dark grey or dark yellow crystals with a garlic odour. Does
not melt or decompose thermally at temperatures up to 1000 . Decomposes in water to give phosphine gas. Used as insecticidal grain fumigant. Aluminium Sulfate
Al2(SO4)3 = 342.1 CAS—10043-01-3 Synonyms Alum; aluminium sulphate (2 : 3); cake alum; dialuminium sulphate;
filter alum; papermakers’ alum; patent alum; peral alum; pickle alum. Proprietary Names Cake Alum; Patent Alum. Chemical Properties Odourless white lustrous crystals (also pieces, granules or
powder). Decomposes at 770 . Soluble in water and dilute acids; practically insoluble in alcohol. Used primarily for water purification systems and sewage treatment systems as a flocculent; in the paper and pulp industry; in fireproofing and waterproofing cloth; in clarifying oils and fats; in waterproofing concrete; in antiperspirants; in tanning leather; as a mordant in dyeing; in agricultural pesticides; as a soil conditioner to increase acidity for plants; and in cosmetics and soaps. Solutions containing 5–10% have been used as local applications to ulcers and to stop discharges from mucous surfaces. Colour Test Addition of sodium hydroxide to aluminium ions—forms a white precipitate that dissolves in excess sodium hydroxide. Quantification Specimen Collection Blood/serum/plasma—5 mL, plain or lithium heparin tube; urine—20 mL plastic universal container; dialysis fluid—20 mL plastic universal container. Note Aluminium is the most abundant element on earth and contamination from needles, syringes, collection tubes, glassware, analytical reagents, and equipments is a major problem. It is always advisable to analyse a sample of aluminium-free water alongside the test samples. Blood DPV Buffer: 0.5 mol/L ammonium acetate plus 0.5 mol/L ammonia with 13.3% 2 mol/L sodium chloride and dopamine. Scan rate: 20 mV/s. Pulse
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:28:2
Aluminium amplitude: 50 mV. Pulse width: 60 ms. Limit of detection, 19 nmol/L [Zhang et al. 2002]. Buffer: 0.08 mol/L ammonium acetate : ammonia (pH 8.5). Limit of detection, 76 nmol/L [Zhang et al. 2000]. ETAAS Dry cycle: 110 at 10 s for 5 s to 150 in 60 s for 15 s (to 400 at 1 s for 15 s in blood with a haematocrit of >0.5 L/L). Char cycle: 1400 at 40 s for 20 s (ramp time extended to 100 s in blood with a haematocrit of >0.5 L/L. Atomisation cycle: 260 for 8 s. Gas: N2, 100 mL/min. Limit of detection, 2.3 mg/L [van der Voet et al. 1985]. Dry cycle: 120 at 15 s for 15 s, 300 mL/min. Char cycle: 300 in 5 s for 5 s to 1530 in 25 s for 10 s, 300 mL/min. Atomisation cycle: 2700 in 1 s for 13 s, 20 mL/ min for first 6 s to 300 mL/min for last 7 s. Limit of detection, 20 pg [D’Haese et al. 1985]. ICP-AES Limit of detection, 0.1 mg/kg [Dinya et al. 2005]. Plasma gas: Ar, 12 L/ min. Nebuliser gas: Ar, 0.5 L/min. Nebuliser pressure: 2.3 bar (l ¼ 396.2 nm). Limit of detection, 0.12 mmol/L [Chappuis et al. 1992]. ICP-MS Plasma gas: 15 L/min. Auxiliary gas: 1.13 L/min. Nebuliser gas: 1.0 mL/ min. Limit of detection not reported [Rainska et al. 2007]. Plasma gas: Ar, 14.8 L/ min. Auxiliary gas: Ar, 0.9 L/min. Carrier gas: 1.1 L/min. Babington nebuliser (m/z 27). Limit of detection not reported [Botta et al. 2006]. Limit of detection, 0.1 mg/L [Nagaoka, Maitani 2005]. Plasma gas: 16 L/min. Auxiliary gas: 1 L/min. Nebuliser gas: 1.15 or 1.33 mL/min. Limit of detection, 40 mg/L [De Boer et al. 2004]. Limit of detection, 171 ng/L [Liao et al. 2004]. Outer gas: Ar, 15 L/min. Intermediate gas: 0.85 L/min. Carrier gas: 0.91 L/min. Limit of detection not reported [Nagaoka, Maitani 2001]. Plasma ETAAS Dry cycle: 120 at 1 s for 20 s to 160 in 10 s for 10 s. Char cycle: 1000 in 10 s for 20 s. Atomisation cycle: 2600 for 5 s to 2650 in 1 s for 7 s. Gas: Ar (l ¼ 309 nm). Limit of detection not reported [Progar et al. 1996]. Dry cycle: 110 at 10 s for 5 s to 150 in 60 s for 15 s. Char cycle: 1400 in 40 s for 20 s. Atomisation cycle: 260 for 8 s. Gas: N2, 100 mL/min. Limit of detection, 1.8 mg/L [van der Voet et al. 1985]. See Blood. Dry cycle: 100 at 15 s for 15 s to 120 in 10 s for 10 s, 300 mL/ min. Char cycle: 700 in 2 s for 1 s to 1580 in 120 s for 10 s, 300 mL/min [D’Haese et al. 1985]. ICP-AES Plasma gas: Ar, 0.01 L/min. Auxiliary gas: Ar, 10 L/min. Nebuliser gas: Ar, 0.8 L/min. McPherson 216 or Minuteman 310-SMP spectrometer (l ¼ 308 nm). Limit of detection not reported [Progar et al. 1996]. Serum ETAAS See Blood. Limit of detection, 1.9 mg/L [van der Voet et al. 1985]. Dry cycle: 350 for 60 s. Char cycle: 1500 for 60 s. Atomisation cycle: 2600 for 12 s. Aluminium hollow cathode lamp (l ¼ 309 nm). Limit of detection not reported [Gorsky, Dietz 1978]. ICP-AES Plasma gas: Ar, 12 L/min. Meinhard nebuliser (l ¼ 167 nm). Limit of quantification, 0.97 mg/L; limit of detection, 0.5 mg/L [Bianchi et al. 2007]. External gas: 14.0 L/min. Indirect gas: 0.5 L/min. Bearing gas: 1.0 L/min. Meinhard nebuliser (l ¼ 165–460 nm). Limit of detection not reported [Olszewski et al. 2006]. See Blood [Chappuis et al. 1992]. ICP-MS Outer gas: 15.0 L/min. Carrier gas: 0.8 L/min. Make-up gas: 0.17 L/min. Limit of detection, 5 mg/L [Murko et al. 2007]. FPLC-ICP-MS Column: Mono-Q HR 5/5 FPLC (50 5 mm i.d., 10 mm). Mobile phase: 0.05 mol/L TRIS hydrochloride : 0.05 mol/L TRIS hydrochloride with 0.25 mol/L ammonium acetate (100 : 0 to 0 : 100 at 15 min), flow rate 1 mL/min. UV-vis detection (proteins, l ¼ 295 nm). Carrier gas: 1.1 or 1.15 L/min. Intermediate gas: 1.0 or 0.9 L/min. Outer gas: 15.0 or 14.5 L/min for quadrupole and double-focusing ICP-MS, respectively. Limit of detection, 0.5–320 mg/L (mean, 76) were reported in 11 fatalities caused by amfetamine [Holmgren, Lindquist 1975]. In a 22-year-old man who died of cardiorespiratory arrest following amfetamine ingestion, the following postmortem tissue concentrations were reported for racemic, (R)- and (S)-amfetamine, respectively: blood 2.44, 1.26 and 1.18 mg/L; urine 33.4, 16.7 and 16.7 mg/L; liver 11.7, 6.07 and 5.64 mg/g; kidney 3.85, 2.00 and 1.85 mg/g; and brain 5.50, 2.95 and 2.55 mg/g [Meyer et al. 1997]. Half-life Plasma half-life, 4 to 8 h when the urine is acidic and approx. 12 h in subjects whose urinary pH values are uncontrolled. Volume of Distribution 3–4 L/kg. Distribution in Blood Plasma : whole blood, 1.0 Protein Binding 15–40%. Saliva Plasma : saliva ratio, ~0.35 Dose 20 to 100 mg of amfetamine sulfate daily has been used in the treatment of narcolepsy. Note For a review of the toxicokinetics of amfetamines, see Kraemer, Maurer [2002]; for a study of the pharmacokinetic and pharmacodynamic drug interactions in the treatment of attention-deficit hyperactivity disorder, see Markowitz, Patrick [2001]. Adjutantis G et al. (1975). Fatal intoxication with amphetamines (a case report). Med Sci Law 15: 62–63. Al-Dirbashi O et al. (1997). High-performance liquid chromatography of methamphetamine and its related compounds in human urine following derivatization with fluorescein isothiocyanate. J Chromatogr B Biomed Sci Appl 695: 251–258. Al-Dirbashi O et al. (1998). Enantioselective high-performance liquid chromatography with fluorescence detection of methamphetamine and its metabolites in human urine. Analyst 123: 2333–2337. Al-Dirbashi OY et al. (2000). Achiral and chiral quantification of methamphetamine and amphetamine in human urine by semi-micro column high-performance liquid chromatography and fluorescence detection. J Forensic Sci 45: 708–714. Al-Dirbashi OY et al. (2000). Quantification of methamphetamine, amphetamine and enantiomers by semi-micro column HPLC with fluorescence detection; applications on abusers’ single hair analyses. Biomed Chromatogr 14: 293–300. Anggard E et al. (1970). Relationships between pharmacokinetic and clinical parameters in chronic amphetamine abuse. Acta Pharmacol Toxicol (Copenh) 28: 92. Apollonio LG et al. (2006). Product ion mass spectra of amphetamine-type substances, designer analogues, and ketamine using ultra-performance liquid chromatography/tandem mass spectrometry. Rapid Commun Mass Spectrom 20: 2259–2264. Asghar SJ et al. (2001). A rapid method of determining amphetamine in plasma samples using pentafluorobenzenesulfonyl chloride and electron-capture gas chromatography. J Pharmacol Toxicol Methods 46: 111–115. Bartu A et al. (2009). Transfer of methylamphetamine and amphetamine into breast milk following recreational use of methylamphetamine. Br J Clin Pharmacol 67: 455–459. Baselt RC (2008). Disposition of Toxic Drugs and Chemicals in Man, 8th edn, Foster City, CA: Biomedical Publications. Bjork MK et al. (2010). Determination of 19 drugs of abuse and metabolites in whole blood by highperformance liquid chromatography–tandem mass spectrometry. Anal Bioanal Chem 396: 2393–2401. Bogusz MJ et al. (1997). Determination of phenylisothiocyanate derivatives of amphetamine and its analogues in biological fluids by HPLC-APCI-MS or DAD. J Anal Toxicol 21: >59–69. Bogusz MJ et al. (2000). Analysis of underivatized amphetamines and related phenethylamines with high-performance liquid chromatography–atmospheric pressure chemical ionization mass spectrometry. J Anal Toxicol 24: 77–84. Cairns T et al. (2004). Amphetamines in washed hair of demonstrated users and workplace subjects. Forensic Sci Int 145: 137–142. Campins Falco´ P et al. (1995). Improved amphetamine and methamphetamine determination in urine by normal-phase high-performance liquid chromatography with sodium 1,2-naphthoquinone 4-sulphonate as derivatizing agent and solid-phase extraction for sample clean-up. J Chromatogr B Biomed.Appl. 663: 235–245. Campins-Falco´ P et al. (1996). Amphetamine and methamphetamine determination in urine by reversed-phase high-performance liquid chromatography with simultaneous sample clean-up and derivatization with 1,2-naphthoquinone 4-sulphonate on solid-phase cartridges. J Chromatogr B Biomed Appl 687: 239–246. Casari C, Andrews AR (2001). Application of solvent microextraction to the analysis of amphetamines and phencyclidine in urine. Forensic Sci Int 120: 165–171. Centini F et al. (1996). Quantitative and qualitative analysis of MDMA, MDEA, MA and amphetamine in urine by headspace/solid phase micro-extraction (SPME) and GC/MS. Forensic Sci Int 83: 161–166. Cheung S et al. (1997). Simultaneous gas chromatographic determination of methamphetamine, amphetamine and their p-hydroxylated metabolites in plasma and urine. J Chromatogr B Biomed Sci Appl 690: 77–87. Cheze M et al. (2007). Simultaneous analysis of six amphetamines and analogues in hair, blood and urine by LC-ESI-MS/MS. Application to the determination of MDMA after low ecstasy intake. Forensic Sci Int 170: 100–104. Cirimele V et al. (1995). Detection of amphetamines in fingernails: an alternative to hair analysis. Arch Toxicol 70: 68–69. Clauwaert KM et al. (2000). Determination of the designer drugs 3,4-methylenedioxymethamphetamine, 3,4-methylenedioxyethylamphetamine, and 3,4-methylenedioxyamphetamine with HPLC and fluorescence detection in whole blood, serum, vitreous humor, and urine. Clin Chem 46: 1968–1977. Concheiro M et al. (2006). Determination of drugs of abuse and their metabolites in human plasma by liquid chromatography–mass spectrometry. An application to 156 road fatalities. J Chromatogr B Analyt Technol Biomed Life Sci 832: 81–89. Concheiro M et al. (2007). Confirmation by LC-MS of drugs in oral fluid obtained from roadside testing. Forensic Sci Int 170: 156–162. Cone EJ et al. (2007). Prevalence and disposition of drugs of abuse and opioid treatment drugs in oral fluid. J Anal Toxicol 31: 424–433. Dallakian P et al. (1996). Detection and quantitation of amphetamine and methamphetamine: electron impact and chemical ionization with ammonia–comparative investigation on Shimadzu QP 5000 GC-MS system. J Anal Toxicol 20: 255–261.
Dasgupta A, Spies J (1998). A rapid novel derivatization of amphetamine and methamphetamine using 2,2,2-trichloroethyl chloroformate for gas chromatography electron ionization and chemical ionization mass spectrometric analysis. Am J Clin Pathol 109: 527–532. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Elsohly MA et al. (1999). Immunoassay and GC-MS procedures for the analysis of drugs of abuse in meconium. J Anal Toxicol 23: 436–445. Farrell BM, Jefferies TM (1983). An investigation of high-performance liquid chromatographic methods for the analysis of amphetamines. J Chromatogr 272: 111–128. Fernandez MM et al. (2009). High-throughput analysis of amphetamines in blood and urine with online solid-phase extraction-liquid chromatography–tandem mass spectrometry. J Anal Toxicol 33: 578–587. Fisher DH, Bourque AJ (1993). Quantification of amphetamine in urine: solid-phase extraction, polymeric-reagent derivatization and reversed-phase high-performance liquid chromatography. J Chromatogr 614: 142–147. Franssen RM et al. (1994). Analysis of morphine and amphetamine in meconium with immunoassay and HPLC-diode-array detection. J Anal Toxicol 18: 294–295. Fritch D et al. (2009). Identification and quantitation of amphetamines, cocaine, opiates, and phencyclidine in oral fluid by liquid chromatography–tandem mass spectrometry. J Anal Toxicol 33: 569–577. Fuh MR et al. (2006). Determination of amphetamine and methamphetamine in urine by solid phase extraction and ion-pair liquid chromatography–electrospray-tandem mass spectrometry. Talanta 68: 987–991. Gray TR et al. (2009). A liquid chromatography tandem mass spectrometry method for the simultaneous quantification of 20 drugs of abuse and metabolites in human meconium. Anal Bioanal Chem 393: 1977–1990. Gunn JA et al. (2008). Simultaneous quantification of amphetamine and methamphetamine in meconium using ISOLUTE HM-N-supported liquid extraction columns and GC-MS. J Anal Toxicol 32: 485–490. Gunn JA et al. (2010). Identification and quantitation of amphetamine, methamphetamine, MDMA, pseudoephedrine, and ephedrine in blood, plasma, and serum using gas chromatography–mass spectrometry (GC/MS). Methods Mol Biol 603: 37–43. Hara K et al. (1997). Simple extractive derivatization of methamphetamine and its metabolites in biological materials with Extrelut columns for their GC-MS determination. J Anal Toxicol 21: 54–58. Hensley D, Cody JT (1999). Simultaneous determination of amphetamine, methamphetamine, methylenedioxyamphetamine (MDA), methylenedioxymethamphetamine (MDMA), and methylenedioxyethylamphetamine (MDEA) enantiomers by GC-MS. J Anal Toxicol 23: 518–523. Holmgren P, Lindquist O (1975). Lethal intoxications with centrally stimulating amines in Sweden 1966-1973. Z Rechtsmed 75: 265–273. Huang MK et al. (2002). One step and highly sensitive headspace solid-phase microextraction sample preparation approach for the analysis of methamphetamine and amphetamine in human urine. Analyst 127: 1203–1206. Jones J et al. (2009). Determination of amphetamine and methamphetamine in umbilical cord using liquid chromatography–tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 877: 3701–3706. Jonsson J et al. (1996). A convenient derivatization method for the determination of amphetamine and related drugs in urine. J Forensic Sci 41: 148–151. Joya X et al. (2010). Gas chromatography–mass spectrometry assay for the simultaneous quantification of drugs of abuse in human placenta at 12th week of gestation. Forensic Sci Int 196: 38–42. Jurado C et al. (2000). Rapid analysis of amphetamine, methamphetamine, MDA, and MDMA in urine using solid-phase microextraction, direct on-fiber derivatization, and analysis by GC-MS. J Anal Toxicol 24: 11–16. Kaddoumi A et al. (2004). High-performance liquid chromatography with fluorescence detection for the simultaneous determination of 3,4-methylenedioxymethamphetamine, methamphetamine and their metabolites in human hair using DIB-Cl as a label. Biomed Chromatogr 18: 202–204. Kala SV et al. (2008). Validation of analysis of amphetamines, opiates, phencyclidine, cocaine, and benzoylecgonine in oral fluids by liquid chromatography–tandem mass spectrometry. J Anal Toxicol 32: 605–611. Kalasinsky KS et al. (1993). Detection of amphetamine and methamphetamine in urine by gas chromatography/Fourier transform infrared (GC/FTIR) spectroscopy. J Anal Toxicol 17: 359–364. Kankaanp€a€a A et al. (2004). Single-step procedure for gas chromatography–mass spectrometry screening and quantitative determination of amphetamine-type stimulants and related drugs in blood, serum, oral fluid and urine samples. J Chromatogr B Analyt Technol Biomed Life Sci 810: 57–68. Karacic V, Skender L (2000). Analysis of drugs of abuse in urine by gas chromatography/mass spectrometry: experience and application. ArhHigRada Toksikol 51: 389–400. Katagi M et al. (1996). Direct high-performance liquid chromatographic and high-performance liquid chromatographic-thermospray-mass spectrometric determination of enantiomers of methamphetamine and its main metabolites amphetamine and p-hydroxymethamphetamine in human urine. J Chromatogr B Biomed Appl 676: 35–43. Kelly T et al. (2008). Development and validation of a liquid chromatography–atmospheric pressure chemical ionization-tandem mass spectrometry method for simultaneous analysis of 10 amphetamine-, methamphetamine- and 3,4-methylenedioxymethamphetamine-related (MDMA) analytes in human meconium. J Chromatogr B Analyt Technol Biomed Life Sci 867: 194–204. Kiely E et al. (2009). A fatality from an oral ingestion of methamphetamine. J Anal Toxicol 33: 557–560. Kim JY et al. (2008). Simultaneous determination of amphetamine-type stimulants and cannabinoids in fingernails by gas chromatography–mass spectrometry. Arch Pharm Res 31: 805–813. Kim JY et al. (2010). Rapid and simple determination of psychotropic phenylalkylamine derivatives in human hair by gas chromatography–mass spectrometry using micro-pulverized extraction. Forensic Sci Int 196: 43–50. Kim JY et al. (2010). Determination of amphetamine-type stimulants, ketamine and metabolites in fingernails by gas chromatography–mass spectrometry. Forensic Sci Int 194: 108–114. Kintz P et al. (1995). Simultaneous determination of amphetamine, methamphetamine, 3,4-methylenedioxyamphetamine and 3,4-methylenedioxymethamphetamine in human hair by gas chromatography–mass spectrometry. J Chromatogr B Biomed Appl 670: 162–166. Koide I et al. (1998). Determination of amphetamine and methamphetamine in human hair by headspace solid-phase microextraction and gas chromatography with nitrogen–phosphorus detection. J Chromatogr B Biomed Sci Appl 707: 99–104. Kraemer T, Maurer HH (2002). Toxicokinetics of amphetamines: metabolism and toxicokinetic data of designer drugs, amphetamine, methamphetamine, and their N-alkyl derivatives. Ther Drug Monit 24: 277–289.
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:28:39
Amicarbalide Kronstrand R et al. (2003). Quantitative analysis of desmethylselegiline, methamphetamine, and amphetamine in hair and plasma from Parkinson patients on long-term selegiline medication. J Anal Toxicol 27: 135–141. Kuwayama K et al. (2008). Analysis of amphetamine-type stimulants and their metabolites in plasma, urine and bile by liquid chromatography with a strong cation-exchange column-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 867: 78–83. Lee MR et al. (2000). Determination of amphetamine and methamphetamine in serum via headspace derivatization solid-phase microextraction-gas chromatography–mass spectrometry. J Chromatogr A 896: 265–273. Leis HJ et al. (2003). Enantioselective trace analysis of amphetamine in human plasma by gas chromatography/negative ion chemical ionization mass spectrometry. Rapid Commun Mass Spectrom 17: 569–575. Lin DL et al. (2004). Deposition characteristics of methamphetamine and amphetamine in fingernail clippings and hair sections. J Anal Toxicol 28: 411–417. Liu J et al. (2001). New method of derivatization and headspace solid-phase microextraction for gas chromatographic-mass spectrometric analysis of amphetamines in hair. J Chromatogr B Biomed Sci Appl 758: 95–101. Lowe RH et al. (2006). A validated positive chemical ionization GC/MS method for the identification and quantification of amphetamine, opiates, cocaine, and metabolites in human postmortem brain. J Mass Spectrom 41: 175–184. Marais AA, Laurens JB (2009). Rapid GC-MS confirmation of amphetamines in urine by extractive acylation. Forensic Sci Int 183: 78–86. Markowitz JS, Patrick KS (2001). Pharmacokinetic and pharmacodynamic drug interactions in the treatment of attention-deficit hyperactivity disorder. Clin Pharmacokinet 40: 753–772. Marquet P et al. (1997). Simultaneous determination of amphetamine and its analogs in human whole blood by gas chromatography–mass spectrometry. J Chromatogr B Biomed Sci Appl 700: 77–82. McCambly K et al. (1997). Robotic solid-phase extraction of amphetamines from urine for analysis by gas chromatography–mass spectrometry. J Anal Toxicol 21: 438–444. Meng P, Wang Y (2010). Small volume liquid extraction of amphetamines in saliva. Forensic Sci Int 197: 80–84. Meyer E et al. (1997). Tissue distribution of amphetamine isomers in a fatal overdose. J Anal Toxicol 21: 236–239. Miki A et al. (2003). Determination of methamphetamine and its metabolites incorporated in hair by column-switching liquid chromatography–mass spectrometry. J Anal Toxicol 27: 95–102. Molins Legua C et al. (1995). Amphetamine and methamphetamine determination in urine by reversed-phase high-performance liquid chromatography with sodium 1,2-naphthoquinone 4sulfonate as derivatizing agent and solid-phase extraction for sample clean-up. J Chromatogr B Biomed Appl 672: 81–88. Mortier KA et al. (2002). Determination of paramethoxyamphetamine and other amphetaminerelated designer drugs by liquid chromatography/sonic spray ionization mass spectrometry. Rapid Commun Mass Spectrom 16: 865–870. Musshoff F et al. (2002). Fully automated determination of amphetamines and synthetic designer drugs in hair samples using headspace solid-phase microextraction and gas chromatography– mass spectrometry. J Chromatogr Sci 40: 359–364. Myung SW et al. (1998). Determination of amphetamine, methamphetamine and dimethamphetamine in human urine by solid-phase microextraction (SPME)–gas chromatography/mass spectrometry. J Chromatogr B Biomed Sci Appl 716: 359–365. Nagasawa N et al. (1996). Rapid analysis of amphetamines in blood using head space-solid phase microextraction and selected ion monitoring. Forensic Sci Int 78: 95–102. Nakashima K et al. (2003). Determination of methamphetamine and amphetamine in abusers’ plasma and hair samples with HPLC-FL. Biomed Chromatogr 17: 471–476. Namera A et al. (2000). Simple and simultaneous analysis of fenfluramine, amphetamine and methamphetamine in whole blood by gas chromatography–mass spectrometry after headspace-solid phase microextraction and derivatization. Forensic Sci Int 109: 215–223. Namera A et al. (2002). Automated headspace solid-phase microextraction and in-matrix derivatization for the determination of amphetamine-related drugs in human urine by gas chromatography–mass spectrometry. J Chromatogr Sci 40: 19–25. Narasimhachari N et al. (1979). Quantitation of amphetamine in plasma and cerebrospinal fluid by gas chromatography–mass spectrometry-selected ion monitoring, using beta-methylphenethylamine as in internal standard. J Chromatogr 164: 386–393. Nishida M et al. (2003). Routine analysis of amphetamine and methamphetamine in biological materials by gas chromatography–mass spectrometry and on-column derivatization. J Chromatogr B Analyt Technol Biomed Life Sci 789: 65–71. Nishida M et al. (2006). Single hair analysis of methamphetamine and amphetamine by solid phase microextraction coupled with in matrix derivatization. J Chromatogr B Analyt Technol Biomed Life Sci 842: 106–110. Nishida K et al. (2006). High-performance liquid chromatographic-mass spectrometric determination of methamphetamine and amphetamine enantiomers, desmethylselegiline and selegiline, in hair samples of long-term methamphetamine abusers or selegiline users. J Anal Toxicol 30: 232–237. Okajima K et al. (2001). Highly sensitive analysis of methamphetamine and amphetamine in human whole blood using headspace solid-phase microextraction and gas chromatography–mass spectrometry. Forensic Sci Int 116: 15–22. Pellegrini M et al. (2002). Rapid screening method for determination of Ecstasy and amphetamines in urine samples using gas chromatography–chemical ionisation mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 769: 243–251. Peters FT et al. (2003). Screening for and validated quantification of amphetamines and of amphetamine- and piperazine-derived designer drugs in human blood plasma by gas chromatography/ mass spectrometry. J Mass Spectrom 38: 659–676. Peters FT et al. (2003). Concentrations and ratios of amphetamine, methamphetamine, MDA, MDMA, and MDEA enantiomers determined in plasma samples from clinical toxicology and driving under the influence of drugs cases by GC-NICI-MS. J Anal Toxicol 27: 552–559. Peters FT et al. (2007). Negative-ion chemical ionization gas chromatography–mass spectrometry assay for enantioselective measurement of amphetamines in oral fluid: application to a controlled study with MDMA and driving under the influence cases. Clin Chem 53: 702–710. Pichini S et al. (2004). Development and validation of a high-performance liquid chromatography– mass spectrometry assay for determination of amphetamine, methamphetamine, and methylenedioxy derivatives in meconium. Anal Chem 76: 2124–2132. Rohrich J, Kauert G (1997). Determination of amphetamine and methylenedioxyamphetaminederivatives in hair. Forensic Sci Int 84: 179–188. Sato Y et al. (2000). [Detection of methamphetamine in a severely burned cadaver–a case report]. Nihon Hoigaku Zasshi 54: 420–424. Scheidweiler KB, Huestis MA (2006). A validated gas chromatographic-electron impact ionization mass spectrometric method for methylenedioxymethamphetamine (MDMA), methamphetamine and metabolites in oral fluid. J Chromatogr B Analyt Technol Biomed Life Sci 835: 90–99.
875
Sergi M et al. (2009). Multiclass analysis of illicit drugs in plasma and oral fluids by LC-MS/MS. Anal Bioanal Chem 393: 709–718. Skender L et al. (2002). Quantitative determination of amphetamines, cocaine, and opiates in human hair by gas chromatography/mass spectrometry. Forensic Sci Int 125: 120–126. Slawson MH et al. (2002). Quantitative analysis of selegiline and three metabolites (N-desmethylselegiline, methamphetamine, and amphetamine) in human plasma by high-performance liquid chromatography–atmospheric pressure chemical ionization-tandem mass spectrometry. J Anal Toxicol 26: 430–437. Stanaszek R, Piekoszewski W (2004). Simultaneous determination of eight underivatized amphetamines in hair by high-performance liquid chromatography–atmospheric pressure chemical ionization mass spectrometry (HPLC-APCI-MS). J Anal Toxicol 28: 77–85. Stout PR et al. (2002). Rapid simultaneous determination of amphetamine, methamphetamine, 3,4methylenedioxyamphetamine, 3,4-methylenedioxymethamphetamine, and 3,4-methylenedioxyethylamphetamine in urine by solid-phase extraction and GC-MS: a method optimized for high-volume laboratories. J Anal Toxicol 26: 253–261. Strano-Rossi S et al. (2009). A rapid method for the extraction, enantiomeric separation and quantification of amphetamines in hair. Forensic Sci Int 193: 95–100. Takekawa K et al. (2007). Methamphetamine body packer: acute poisoning death due to massive leaking of methamphetamine. J Forensic Sci 52: 1219–1222. Terada M et al. (1982). Rapid and highly sensitive method for determination of methamphetamine and amphetamine in urine by electron-capture gas chromatography. J Chromatogr 237: 285–292. Ugland HG et al. (1997). Aqueous alkylchloroformate derivatisation and solid-phase microextraction: determination of amphetamines in urine by capillary gas chromatography. J Chromatogr B Biomed Sci Appl 701: 29–38. Ugland HG et al. (1999). Automated determination of ‘Ecstasy’ and amphetamines in urine by SPME and capillary gas chromatography after propylchloroformate derivatisation. J Pharm Biomed Anal 19: 463–475. Valentine JL et al. (1995). GC-MS determination of amphetamine and methamphetamine in human urine for 12 hours following oral administration of dextro-methamphetamine: lack of evidence supporting the established forensic guidelines for methamphetamine confirmation. J Anal Toxicol 19: 581–590. VanBocxlaer JF et al. (1997). Quantitative determination of amphetamine and alpha-phenylethylamine enantiomers in judicial samples using capillary gas chromatography. J Anal Toxicol 21: 5–11. Veress T (2000). Determination of amphetamine by HPLC after acetylation. J Forensic Sci 45: 161–166. Wan SH et al. (1978). Kinetics, salivary excretion of amphetamine isomers, and effect of urinary pH. Clin Pharmacol Ther 23: 585–590. Wang SM et al. (2001). Simultaneous supercritical fluid extraction and chemical derivatization for the gas chromatographic-isotope dilution mass spectrometric determination of amphetamine and methamphetamine in urine. J Chromatogr B Biomed Sci Appl 759: 17–26. Weinmann W et al. (2000). Automated solid-phase extraction and two-step derivatisation for simultaneous analysis of basic illicit drugs in serum by GC/MS. Int J Legal Med 113: 229–235. Wood M et al. (2003). Development of a rapid and sensitive method for the quantitation of amphetamines in human plasma and oral fluid by LC-MS-MS. J Anal Toxicol 27: 78–87. Wu J et al. (2001). Determination of stimulants in human urine and hair samples by polypyrrole coated capillary in-tube solid phase microextraction coupled with liquid chromatography– electrospray mass spectrometry. Talanta 54: 655–672. Wu TY, Fuh MR (2005). Determination of amphetamine, methamphetamine, 3,4-methylenedioxyamphetamine, 3,4-methylenedioxyethylamphetamine, and 3,4-methylenedioxymethamphetamine in urine by online solid-phase extraction and ion-pairing liquid chromatography with detection by electrospray tandem mass spectrometry. Rapid Commun Mass Spectrom 19: 775–780. Yashiki M et al. (1995). Detection of amphetamines in urine using head space-solid phase microextraction and chemical ionization selected ion monitoring. Forensic Sci Int 76: 169–177.
Amicarbalide Antiprotozoal (Veterinary) C15H16N6O = 296.3 CAS—3459-96-9 IUPAC Name 1,3-Bis(3-carbamimidoylphenyl)urea 3,30 -(Carbonyldiimino)bisbenzenecarboximidamide
Amicarbalide Isetionate C19H28N6O9S2 = 548.6 CAS—3671-72-5 Synonym M&B 5062A Proprietary Name Diampron Chemical Properties A white or slightly cream-coloured powder. Mp 200 to
204 . Soluble 1 in less than 1 of water and 1 in 250 of ethanol; practically insoluble in chloroform and ether.
Colour Tests Aromaticity (method 2)—yellow/orange; Liebermann’s test (100 )—yellow. Thin-layer Chromatography System TA—Rf 0.05, streaking (acidified iodoplatinate solution, positive). Ultraviolet Spectrum Aqueous acid—231 nm (A11¼1236a), inflexion at 256 nm. Infrared Spectrum Principal peaks at wavenumbers 1195, 1590, 1672, 1250, 1562, 1052 cm1 (KBr disk).
A
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:28:42
876
A
Amidefrine
Amidefrine Sympathomimetic C10H16N2O3S = 244.3 CAS—3354-67-4 IUPAC Name N-[3-[1-Hydroxy-2-(methylamino)ethyl]phenyl]methanesulfonamide Synonyms Amidephrine; MJ-1996.
Chemical Properties Crystals. Mp 159 to 161 . pKa 9.1. Amidefrine Mesilate C10H16N2O3S,CH3SO3H = 340.4 CAS—1421-68-7 Chemical Properties A white crystalline solid. Mp 207 to 209 . Soluble 1 in
about 5 of water; very slightly soluble in chloroform. Colour Test Mandelin’s test—violet. Thin-layer Chromatography System TA—Rf 0.15; system TB—Rf 0.00; system TC—Rf 0.01; system TE—Rf 0.00; system TL—Rf 0.02; system TAE—Rf 0.01 (acidified potassium permanganate solution, positive). Gas Chromatography System GA—not eluted. Ultraviolet Spectrum Aqueous acid—272 nm (A11¼22a).
0.50 mol/L sodium acetate buffer (pH 5.5): methanol (50 to 90%). Rf 0.70 (50% methanol), Rf 0.45 (60%) and Rf 0.03 (70 to 90%). Mobile phase 2: 0.1 mol/L phosphate buffer (pH 5.5): methanol (50 to 90%). Rf 0.03 [Mank et al. 1995]. Quantification Blood HPLC Column: LC-ABZ Supelcosil (15 cm 4.6 mm i.d., 5 mm). Mobile phase: methanol: 10 mmol/L ethylamine in 0.1 mol/L monochloroacetic acid (pH 2.8) (10:90), flow rate 1.0 mL/min. Fluorescence detection (lex¼385 nm, lem¼515 nm). Retention time: 4.0 min for amifostine metabolite, WR-1065. Limit of detection, 0.25 mg/L [Bonner, Shaw 2000]. Disposition in the Body Amifostine is rapidly cleared from plasma with 12 h after ingestion. The patient died 48 h later of potanoxic coma [Legras et al. 1996]. Legras A et al. (1996). Herbicide: fatal ammonium thiocyanate and aminotriazole poisoning. J Toxicol Clin Toxicol 34: 441–446.
Amiodarone Antiarrhythmic C25H29I2NO3 = 645.3 CAS—1951-25-3 IUPAC Name (2-Butyl-1-benzofuran-3-yl)[4-(2-diethylaminoethoxy)-3,5-diiodophenyl]methanone
Chemical Properties pKa 5.6. Extraction yield (chlorobutane), 0.95 [Demme et al. 2005]. Amiodarone Hydrochloride C25H29I2NO3,HCl = 681.8 CAS—19774-82-4 Proprietary Names Amiodar; Ancaron; Cordarex; Cordarone; Cordarone X;
Ortacrone; Tachydaron; Trangorex.
Chemical Properties A white crystalline powder. Mp about 161 . Very slightly
soluble in water; soluble in ethanol; freely soluble in chloroform. Colour Tests Iodine test (omitting MnO2)—positive; Liebermann’s reagent— brown-yellow; sulfuric acid—yellow. Thin-layer Chromatography System TA—Rf 0.72; system TB—Rf 0.62; system TC—Rf 0.68; system TE—Rf 0.82; system TL—Rf 0.55; system TAE—Rf 0.54; system TAF—Rf 0.64 (dragendorff spray, positive; acidified iodoplatinate solution, positive; Marquis reagent, yellow). Gas Chromatography System GA—RI 3335amiodarone; RI 2800O-desalkylamiodarone (art). High Performance Liquid Chromatography System HA—amiodarone k 2.4, monodesethylamiodarone k 1.8; system HX—RI 683; system HY—RI 476; system HZ—retention time 90.4 min.
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:29:25
Amiphenazole Ultraviolet Spectrum Aqueous acid—241 nm; aqueous alkali—251 nm.
Infrared Spectrum Principal peaks at wavenumbers 1630, 748, 1245, 1558, 1170, 998 cm1 (amiodarone hydrochloride, KBr disk).
885
Following oral administration of 400 mg to 7 subjects, peak plasma concentrations of about 0.5 to 1 mg/L were attained in about 7 h. After administration of 200 mg 8-hourly to 6 subjects for 1 month, plasma concentrations determined immediately before the morning dose ranged from 0.75 to 2.8 mg/L (mean 1.5 mg/L) [Andreasen et al. 1981]. During chronic treatment with 400 mg daily to 33 subjects, mean steadystate plasma concentrations of 2.2 mg/L of amiodarone and 2.0 mg/L of monodesethylamiodarone were reported. [Flanagan et al. 1982]. Half-life Plasma half-life during chronic dosing, 14 to 107 days (mean 50). Distribution in Blood Plasma:whole blood ratio, 1.3. Protein Binding Extensively bound. Note For a review of the pharmacokinetics of amiodarone, see Latini et al. [1984]. Dose Initially 600 mg of amiodarone hydrochloride daily; maintenance, 200 to 400 mg daily. Andreasen F et al. (1981). Pharmacokinetics of amiodarone after intravenous and oral administration. Eur J Clin Pharmacol 19: 293–299. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Flanagan RJ et al. (1982). Identification and measurement of desethylamiodarone in blood plasma specimens from amiodarone-treated patients. J Pharm Pharmacol 34: 638–643. Kollroser M, Schober C (2002). Determination of amiodarone and desethylamiodarone in human plasma by high-performance liquid chromatography-electrospray ionization tandem mass spectrometry with ion trap detector. J Chromatogr B Biomed Sci Appl 766: 219–226. Latini R et al. (1984). Clinical pharmacokinetics of amiodarone. Clin Pharmacokinet 9: 136–156. Plomp TA et al. (1983). Simultaneous determination of amiodarone and its major metabolite desethylamiodarone in plasma, urine and tissues by high-performance liquid chromatography. J Chromatogr 273: 379–392. Storey GCA et al. (1982). High-performance liquid chromatographic measurement of amiodarone and its desethyl metabolite: methodology and preliminary observations. Ther Drug Monit 4: 385–388.
Amiphenazole Narcotic Antagonist, Respiratory Stimulant C9H9N3S = 191.3 CAS—490-55-1 IUPAC Name 5-Phenyl-1,3-thiazole-2,4-diamine Synonyms DAPT; phenamizole; 5-phenyl-2,4-thiazolediamine. Proprietary Names Daptazile; Daptazole; Dizol; Fenamizol.
Chemical Properties Flakes which turn brown on exposure to light and air. Mp 163 to 164 , with decomposition. Amiphenazole Hydrochloride C9H9N3S,HCl = 227.7 CAS—942-31-4 Synonym Amiphenazole chloride Proprietary Names Daptazile; Daptazole. Chemical Properties A white, fine crystalline or granular, mobile powder.
Mass Spectrum Principal ions at m/z 86, 36, 87, 84, 58, 56, 44, 38.
Aqueous solutions hydrolyse slowly. Mp 236 . Soluble 1 in 16 of water and 1 in 50 of ethanol; slightly soluble in acetone, chloroform and ether.
Colour Test Liebermann’s reagent (100 )—green-blue. Thin-layer Chromatography System TA—Rf 0.61; system TB—Rf 0.02; system TC—Rf 0.33; system TE—Rf 0.62; system TL—Rf 0.57 (acidified iodoplatinate solution, positive). Gas Chromatography System GB—RI 1150; system GC—RI 2563. Ultraviolet Spectrum Aqueous acid—230 (A11¼998a), 261 nm. Quantification Plasma HPLC Limit of quantification, 50 mg/L for drug and metabolite. UV detection. Limit of detection, 1 mg/L for amiodarone and 0.5 mg/L for desethylamiodarone [Kollroser, Schober 2002]. UV detection. Limit of detection, 25 mg/L for amiodarone and monodesethylamiodarone [Plomp et al. 1983]. UV detection. Limit of detection, 100 mg/L for amiodarone and monodesethylamiodarone [Storey et al. 1982]. Urine HPLC See Plasma [Plomp et al. 1983]. Tissues HPLC See Plasma [Plomp et al. 1983]. Disposition in the Body Amiodarone is slowly and incompletely absorbed after oral administration; it is distributed to the tissues where it is strongly bound. It is metabolised by N-dealkylation to monodesethylamiodarone which is the major plasma metabolite during chronic dosing. High concentrations of amiodarone and monodesethylamiodarone are found in the liver, lungs and adipose tissue. Enterohepatic circulation may occur. Only a small amount is excreted in the urine as unchanged drug. Therapeutic Concentration Accumulates on chronic administration; steady state plasma concentrations are attained in about 1 month. Fifteen patients were administered 400 mg amiodarone 3 times daily and peak plasma concentrations of the drug were 1.478 to 1.983 mg/L. Peak plasma concentrations of the metabolite, desethylamiodarone, were 0.522 to 1.008 mg/L [Kollroser, Schober 2002].
Infrared Spectrum Principal peaks at wavenumbers 1495, 1637, 688, 750, 1665, 1050 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 191, 121, 77, 104, 122, 43, 51, 192. Dose Amiphenazole hydrochloride has been given parenterally in doses of 100 to 150 mg.
A
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:29:29
886
A
Amisometradine
Amisometradine Pyrimidine, Diuretic C9H13N3O2 = 195.2 CAS—550-28-7 IUPAC Name 6-Amino-1,2,3,4-tetrahydro-3-methyl-1-methyl-allyl-2,4-dioxopyrimidine Synonym Aminoisometradine Proprietary Name Rolicton
Infrared Spectrum Chemical Properties White crystalline powder. Mp 175 . Slightly soluble in cold water, the solubility increasing rapidly on heating; freely soluble in ethanol and acetone; insoluble in ether. It is extracted by organic solvents from aqueous acid or alkaline solutions. Amisometradine is an isomer of aminometradine. Log P (octanol/water) 0.13 [Meylan, Howard 1995]. Thin-Layer Chromatography System T1—Rf 0.68 (location reagent potassium permanganate spray, positive reaction). Ultraviolet Spectrum Aqueous acid (0.1 N sulfuric acid)—268.5 nm; methanol—267 nm. Infrared Spectrum Principal peaks at wavenumbers 1639, 1492 or 1604, 1680 cm1 (KBr disk). Disposition in the Body Toxicity LD50 (oral) in mice: 610 mg/kg. Dose Up to 1.6 g daily. Meylan WM, Howard PH (1995). Atom/fragment contribution method for estimating octanolwater partition coefficients. J Pharm Sci 84: 83–92.
Amisulpride Antipsychotic C17H27N3O4S = 369.5 CAS—71675-85-9 IUPAC Name 4-Amino-N-[(1-ethylpyrrolidin-2-yl)methyl]-5-ethylsulfonyl-2methoxybenzamide Synonyms Aminosultopride; 4-amino-N-[(1-ethyl-2-pyrrolidinyl)methyl]-5(ethylsulfonyl)-2-methoxybenzamide; DAN-2163. Proprietary Names Deniban; Solian; Sulamid.
Chemical Properties Crystals from acetone. Mp 126 to 127 . pKa 9.37. Log P (octanol/water), 1.10. Extraction yield (chlorobutane), 0.6 [Demme et al. 2005]. Stock solutions were stable for at least 1 month stored at 0–5 . Amisulpride is stable in human plasma for at least 24 h either at room temperature or at 37 [Malavasi et al. 1996]. Stability of amisulpride in methanol and human plasma was satisfactory at room temperature and at 37 . It was also satisfactory in human plasma following 2 freeze-thaw cycles and in human plasma diluted with borate buffer (pH 9.0) [Ascalone et al. 1996]. High Performance Liquid Chromatography System HAA—amisulpride RT 8.9 min, M1 RT 7.1 min, M2 RT 10.9 min. Column: Hypersil C18 BDS (150 4.6 mm i.d., 5 mm). Mobile phase: water-TEA1 mol/L potassium dihydrogen phosphate (pH 3.0, 974 : 1 : 25) : acetonitrile (850 : 150), flow rate 1 mL/min. Fluorescence detection (lex¼ 280 nm, lem¼ 370 nm). Retention time: 4 min [Malavasi et al. 1996]. Ultraviolet Spectrum Aqueous acid—230, 285 nm.
Mass Spectrum Principal ions at m/z 98, 44, 242, 70, 28, 111, 149, 369.
Quantification Blood HPLC Column: Nova-Pak C18 (300 3.9 mm i.d., 4 mm). Mobile phase: methanol : tetrahydrofuran : potassium dihydrogen phosphate (pH 2.6, 65 : 5 : 30). DAD. Retention time: 5.67 min. Limit of detection, 49 mg/L [Tracqui et al. 1995]. Plasma HPLC Column: Chiralpak AS (250 4.6 mm i.d.). Mobile phase: n-hexane: ethanol (67 : 33) containing 0.2% diethylamine, flow rate 0.5 mL/min. UV (l¼ 280 nm) or fluorescence (lex¼ 280 nm, lem¼ 370 nm) detection. Limit of quantification, 2.5 mg/L for both enantiomers [Ascalone et al. 1996]. Column: Hypersil C18 (150 4.6 mm i.d., 5 mm). Mobile phase: 850 mL 1 mol/L potassium dihydrogen phosphate (25 mL in 950 mL water with 1 mL TEA, pH 3.0 to 1 L with
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:29:35
Amitriptyline water) to 1 L with acetonitrile, flow rate 1.0 mL/min. Fluorescence detection (lex¼ 280 nm, lem¼ 370 nm). Limit of quantification, 0.5 mg/L [Malavasi et al. 1996]. Column: RP-18 (250 4.6 mm i.d., 5 mm). Mobile phase: methanol : water : diethylamine (532 : 468 : 0.8), flow rate 1 mL/min. UV detection (l¼ 226 nm). Retention time: 10.3 min. Limit of detection, 5 mg/L [Bohbot et al. 1987]. Serum LC-MS Column: C18 (50 4.6 mm i.d., 5 mm). Mobile phase: methanol : 5 mmol/L acetic acid (pH 3.9, 20 : 80 to 70 : 30 over 4 min for 1 min), flow rate 1.0 mL/min. ESI, positive ion mode, MRM acquisition mode. Retention time: 1.5 min. Limit of quantification, 28.3 mg/L [Kirchherr, K€ uhn-Velten 2006]. Urine HPLC See Plasma. Limit of quantification, 50 mg/L for both enantiomers [Ascalone et al. 1996]. See Plasma. Limit of quantification, 100 mg/L [Malavasi et al. 1996]. Disposition in the Body Following oral administration, amisulpride is readily absorbed, with 2 absorption peaks observed, the first after ~1 h and the second between 3 and 4 h after administration. It is only weakly metabolised; metabolites account for only ~4% of the dose and are inactive. It is excreted largely unchanged in urine (70%) and a small amount in faeces as the unchanged drug. Amisulpride is widely distributed throughout the body and is only very weakly dialysed. Therapeutic Concentration Following administration of a single oral dose of 50 mg amisulpride to 18 healthy subjects the mean plasma concentrations were 18.8, 17.9 and 17.2 mg/L at 2, 4 and 6.5 h, respectively. Following administration of a single 200 mg dose, the plasma concentrations were 76.2, 83.0, and 92.1 mg/L at 2, 4 and 6.5 h, respectively [Mattila et al. 1996]. Toxicity In a non-fatal overdose, a 30-year-old Caucasian woman who had taken ~3 g amisulpride and an unknown amount of dosulepin had a blood amisulpride concentration of 9.63 mg/L. Her gastric fluid concentration was 14.3 mg/L. The patient experienced generalised convulsions, which resolved spontaneously, followed by coma, motor restlessness, tachycardia and slight prolongation of the QT interval. The subject was treated with gastric lavage and recovered within 48 h. The drug blood concentration was 10–50 times higher than that observed at therapeutic levels. A second subject who had been found dead had a blood amisulpride concentration of 41.7 mg/L. This is 40– 200 times the observed therapeutic concentration [Tracqui et al. 1995]. Half-life Plasma, 12 h. Volume of Distribution 5.8 L/kg. Clearance 20 L/h Protein Binding 16%. Dose Up to 1200 mg daily orally; up to 400 mg daily has been given IM. Ascalone Vet al. (1996). Stereospecific determination of amisulpride, a new benzamide derivative, in human plasma and urine by automated solid-phase extraction and liquid chromatography on a chiral column. Application to pharmacokinetics. J Chromatogr B Biomed Appl 676: 95–105. Bohbot M et al. (1987). Determination of a new benzamide, amisulpride, in human plasma by reversed-phase ion-pair high-performance liquid chromatography. J Chromatogr 416: 414–419. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid–liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Kirchherr H, K€ uhn-Velten WN (2006). Quantitative determination of forty-eight antidepressants and antipsychotics in human serum by HPLC tandem mass spectrometry: a multi-level, singlesample approach. J Chromatogr B Analyt Technol Biomed Life Sci 843: 100–113. Malavasi B et al. (1996). Determination of amisulpride, a new benzamide derivative, in human plasma and urine by liquid–liquid extraction or solid-phase extraction in combination with high-performance liquid chromatography and fluorescence detection. application to pharmacokinetics. J Chromatogr B Biomed Appl 676: 107–115. Mattila MJ et al. (1996). Single oral doses of amisulpride do not enhance the effects of alcohol on the performance and memory of healthy subjects. Eur J Clin Pharmacol 51: 161–166. Tracqui A et al. (1995). Amisulpride poisoning: a report on two cases. Hum Exp Toxicol 14: 294–298.
887
Proprietary Name Tryptizol (syrup). Chemical Properties A pale yellow to brownish-yellow powder. Mp ~140 .
Practically insoluble in water; soluble 1 in 120 of ethanol, 1 in 6 of acetone, and 1 in 8 of chloroform. Amitriptyline Hydrochloride C20H23N,HCl = 313.9 CAS—549-18-8 Proprietary Names Adepril; Amavil; Amiline; Amineurin; Amioxid; Amitid;
Amitril; Amitrip; Amitrol; Domical; Elatrol; Elavil; Endep; Equilibrin; Klotriptyl; Laroxyl; Lentizol; Levate; Meravil; Novoprotect; Novotriptyn; Redomex; Saroten; Sarotex; Syneudon; Triptyl; Tryptanol; Tryptil; Tryptizol. It is an ingredient of Etrafon; Limbatril; Limbitrol; Triavil; Triptafen. Chemical Properties Colourless crystals or white powder. Mp 196 to 197 . Soluble 1 in 1 of water, 1 in 1.5 of ethanol, 1 in 56 of acetone, 1 in 1.2 of chloroform, and 1 in 1 of methanol; practically insoluble in ether. pKa 9.4 (25 ). Log P (octanol/ water), 2.18 [Meylan, Howard 1995]. Colour Tests Mandelin’s test—brown!green; Marquis test—brown-orange; sulfuric acid—orange. Thin-layer Chromatography System TA—Rf 0.51; system TB—Rf 0.50; system TC—Rf 0.32; system TE—Rf 0.69; system TL—Rf 0.15; system TAE—Rf 0.27; system TAF—Rf 0.51; system TAJ—Rf 0.13; system TAK—Rf 0.05; system TAL—Rf 0.56 (Dragendorff spray, positive; acidified iodoplatinate solution, positive; Marquis test, brown). Gas Chromatography System GA—amitriptyline RI 2194, M (cis-10-OH-) RI 2348, M (trans-10-OH-) RI 2348, M (N-oxide) RI 1975, M (nortriptyline) RI 2215, M (cyclobenzaprine) RI 2235, M (OH-) RI 2380, M (nor-OH-) RI 2390; system GB—amitriptyline RI 2284, M (cis-10-OH-) RI 2454, M (cis-10-OH-Noxide) RI 2215, M (trans-10-OH-) RI 2466, M (trans-10-OH-N-oxide) RI 2239, M (cyclobenzaprine) RI 2330, M (N-oxide) RI 2051; system GF—amitriptyline RI 2510, M (10-OH-) RI 2830 or 2880 (stereoisomers); system GM— amitriptyline RRT 0.723, M (cis-10-OH-) RRT 1.149, M (trans-10-OH-) RRT 1.168, M (cyclobenzaprine) RRT 0.850 (all relative to iprindole); system GS—RT 16.1 min. High Performance Liquid Chromatography System HA—amitriptyline k 3.3, M (10-OH-) k 2.9, M (nor-10-OH-) k 1.8, M (nortriptyline) k 2.0; system HF—amitriptyline k 5.42, M (nortriptyline) k 4.58; system HX—RI 440; system HY—RI 375; system HZ—RT 7.5 min; system HAA—RT 15.9 min; system HAX— RT 15.8 min; system HAY—RT 7.3 min; system HAZ—amitriptyline k 1.76, M (nortriptyline) k 1.71 [Aymard et al. 1997]. Ultraviolet Spectrum Aqueous acid—239 nm (A11¼ 504a).
Amitriptyline Tricyclic Antidepressant C20H23N = 277.4 CAS—50-48-6 IUPAC Name 3-(10,11-Dihydro-5H-dibenzo[a,d]cyclohepten-5-ylidene)-N,Ndimethyl-1-propanamine Synonym 10,11-Dihydro-N,N-dimethyl-5H-dibenzo[a,d]cycloheptene-D5,gpropylamine
Chemical Properties A colourless oil, which becomes yellow on standing through oxidation to a ketonic product. pKa 9.4 [Sangster 1997]. Log P (octanol/ water), 4.92 [Hansch et al. 1995], 4.94. Extraction yield (chlorobutane), 1 [Demme et al. 2005]. Amitriptyline Embonate (C20H23N)2,C23H16O6 = 943.2 CAS—17086-03-2
Infrared Spectrum Principal peaks at wavenumbers 756, 770, 746, 969, 1014, 1258 cm1 (amitriptyline hydrochloride, KBr disk).
A
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:29:38
888
A
Amitriptyline
Mass Spectrum Principal ions at m/z 58, 59, 202, 42, 203, 214, 217; 10-OHamitriptyline 58, 42, 69, 41, 30, 215, 202, 59; 10-OH-nortriptyline 44, 45, 26, 218, 215, 203, 202, 42; nortriptyline 44, 202, 45, 220, 218, 215, 91.
Quantification Blood GC Carrier gas: He, 4 mL/min. Temperature programme: 100 for 1 min to 300 at 15 /min. FID. Limit of detection, 32 mg/L [Lee et al. 1997]. GC-MS Column: AT-5MS (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 20 cm/s. Temperature programme: 70 for 5 min to 310 at 20 /min for 10 min. EI ionisation at 70 eV, SIM acquisition mode. Retention time: 18.4 min. Limit of detection, 0.12 mg/L [Stiakakis et al. 2009]. Ultra 2 capillary 5% phenylmethylsiloxane (12 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1 mL/min. Temperature programme: 150 for 2 min to 200 at 15 /min for 2 min to 270 at 30 /min for 7 min. EI ionisation, SIM acquisition mode. Limit of quantification, 100 mg/L, limit of detection, 9 mg/L [Margalho et al. 2007]. Column: DB-5 cross-linked 5% phenylmethylsiloxane (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1 mL/min. Temperature programme: 50 for 2 min to 180 at 30 /min to 280 at 5 /min for 19 min. Full scan mode. Retention time: 17.8 min. Limit of quantification, 0.05 mg/ L [Paterson et al. 2004]. HPLC Column: Spheri-5 RP-18 (100 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : 0.1 mol/L sodium dihydrogen phosphate : diethylamine (40 : 57.5 : 2.5), flow rate 2.0 mL/min. UV detection (l¼ 220 and 254 nm). Limit of detection, 0.05 mg/L [McIntyre et al. 1993]. LC-MS Column: Xterra RP-18. Mobile phase: acetonitrile : 4 mmol/L ammonium formate buffer (pH 3.2). ESI, MRM acquisition mode. Limit of quantification, 2 mg/L [Titier et al. 2007]. Plasma GC Column: 5% phenylmethylsilicone (12 m 0.2 mm i.d., 0.33 mm). Carrier gas: He, 0.8 mL/min. Temperature programme: 150 to 300 at 10 /min. NPD. Limit of quantification, 19.3 mg/L, limit of detection, 5.8 mg/L [de la Torre et al. 1998]. NPD. Limit of quantification, 125 mg/L [Ulrich, Martens 1997; Vandel et al. 1992]. Column: 3% SP-2250 on Supelcoport 80/100 mesh (2 m 2 mm i.d.). Carrier gas: Ar, 3 105 N/m2. Temperature: 250 . AFID. Retention time: 3.5 min. Limit of detection, 10 mg/L [Dawling, Braithwaite 1978]. GC-MS Column: DB-5MS (30 m 0.32 mm i.d., 0.25 mm). Carrier gas: He, 2.0 mL/min. Temperature programme: 100 for 1 min to 300 at 20 /min. EI ionisation, positive ion mode, SIM acquisition mode. Limit of quantification, 2 mg/L, limit of detection, 0.5 mg/L [Lee et al. 2008]. PBMS. Limit of detection, 2 ng/g for amitriptyline and 5 ng/g for nortriptyline [Kudo et al. 1997]. HPLC Column: mBondapak C18 (250 3.9 mm i.d.). Mobile phase: phosphate buffer : acetonitrile : TEA (65 : 35 : 0.1, pH 5.1). UV detection (l¼ 239 nm). Limit of detection, 5 mg/L [Zarghi et al. 2001]. Column: Supelcosil LC-PCN cyanopropyl (150 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : methanol : 0.005 mol/L ammonium phosphate buffer (pH 7.0, 70 : 15 : 15), flow rate 1.5 mL/min. UV detection (l¼ 254 nm). Limit of detection, 29 nmol/L [Johansen, Rasmussen 1998]. Column: Nova-Pak RP C18 (150 4.6 mm i.d., 4 mm). Mobile phase: 5 mmol/L potassium dihydrogen phosphate : acetonitrile : diethylamine (pH 8, 500 : 500 : 2), flow rate, 0.9 mL/min. UV detection (l¼ 242 nm). Retention time: 17 min. Limit of detection, 60 nmol/L [Theurillat, Thormann 1998]. Column: Symmetry RP C18 (250 4.6 mm i.d., 5 mm). Mobile phase: 0.067 mol/L potassium dihydrogen phosphate (pH 3, 65 : 35); flow rate, 1.2 mL/min. DAD (l¼ 200 nm or 450 nm). Retention time: 11.3 min. Limit of detection, 5 mg/L [Aymard et al. 1997]. Column: silica. Mobile phase: acetonitrile : 0.1 mol/L ammonium acetate (94 : 6). UV detection. Limit of detection, 5 ng/g for amitriptyline and 10 ng/g for nortriptyline [Kudo et al. 1997]. Column: Nova-Pak C18 (5 mm). Mobile phase: water : acetonitrile (70 : 30) containing 1% TEA (pH 3.0), flow rate 2.0 mL/min. UV detection (l¼ 240 nm). Retention time: 10.5 min. Limit of detection, 2 mg/L [Ghahramani, Lennard 1996]. See also H€artter, Hiemke [1992] and Queiroz et al. [1995]. LC-MS Column: Symmetry C18 (150 3.0 mm i.d., 5 mm). Mobile phase : acetonitrile: 0.1% formic acid (28 : 72 for 4 min to 70 : 30 in 1 min for 3 min to 28 : 72 in 0.7 min). APCI, positive ion mode, full scan mode. Limit of quantification, 10 mg/L, limit of detection, 5 mg/L [Kollroser, Schober 2002]. Column: C18 (15 2.1 mm i. d.). Mobile phase: 3 mmol/L ammonium acetate (pH 3.3) : acetonitrile (66 : 34), flow rate 1.4 mL/min. API, TIS. Limit of quantification, 1–2 mg/L, limit of detection, 5 mg/L [Zhang et al. 2000]. Note For a fluorescence polarisation immunoassay for amitriptyline and other tricyclic antidepressants and its comparison with HPLC see Hackett et al. [1998]. Serum GC Column: HP-5 (25 m 0.2 mm i.d., 0.33 mm). Carrier gas: N2, 0.7 mL/ min. Temperature programme: 120 to 240 at 30 /min for 20 min. NPD. Limit of detection, 1.5 ng/mL [Ulrich et al. 1996].
GC-MS Column: HP-5 MS (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1 mL/min. Temperature programme: 60 for 1 min to 280 at 10 /min for 20 min. EI ionisation at 70 eV. Retention time: 22.5 min. Limit of quantification, 0.025 mg/ L, limit of detection, 1 g (moderate intoxication) and >2 g (severe or fatal). In 3 deaths attributed to amitriptyline overdose, postmortem tissue concentrations were:
Blood (mg/L) Liver (mg/g) Urine (mg/L) [Munksgaard 1969]
Amitriptyline
Nortriptyline
6, 18, 3 72, 66, 58 6, 28, 7
5, –, 2 98, 24, 60 10, 12, 7
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:29:39
Amlodipine In 7 deaths attributed to amitriptyline overdose, postmortem concentrations (mean), were:
Blood (mg/L) Liver (mg/g)
Amitriptyline
Nortriptyline
0.43–8.30 (3.4) 10.4–243 (92)
0.29–6.50 (1.6) 4.2–456 (94)
[Bailey, Shaw 1980]
In a fatality involving the suicidal ingestion of amitriptyline, 10 blood samples taken 21 h after discovery of the body and ~28.5 h after the ingestion revealed concentrations of 2.5–12 mg/L amitriptyline, 0.7–3.1 mg/ L nortriptyline, and 81–244 salicylate; a further 10 h later, the concentrations were 1–39 mg/L, 0.6–7.0 mg/L, and 86–310 mg/L, respectively. Of tissue samples, drug concentrations were highest in the liver (amitriptyline 301 mg/g, salicylates 670 mg/g) [Pounder et al. 1994]. A 44-year-old female was found dead in bed. Postmortem blood amitriptyline concentration was 85.9 mg/L [Margalho et al. 2007]. A 37-year-old female was found dead in her apartment. Amitriptyline and nortriptyline were quantified in her blood at concentrations of 7.0 and 7.4 mg/ L, respectively [Stiakakis et al. 2009]. Bioavailability 30–60%. Half-life Plasma half-life, 9–36 h, increased in overdosage. Volume of Distribution 15 L/kg. Clearance Plasma clearance, 11.5 mL/min/kg. Distribution in Blood Plasma : whole blood ratio, 1.2. Saliva Saliva : plasma ratio, 3. Protein Binding 91–97%. Note For a review of the pharmacokinetics of tricyclic antidepressants see Molnar, Gupta [1980]. Dose For depression, 50 to 150 mg amitriptyline hydrochloride daily; up to 300 mg daily has been given. Aymard G et al. (1997). Sensitive and rapid method for the simultaneous quantification of five antidepressants with their respective metabolites in plasma using high-performance liquid chromatography with diode-array detection. J Chromatogr B Biomed Sci Appl 700: 183–189. Bailey DN, Shaw RF (1980). Interpretation of blood and tissue concentrations in fatal self-ingested overdose involving amitriptyline: an update (1978-1979). J Anal Toxicol 4: 232–236. Couper FJ et al. (1995). Detection of antidepressant and antipsychotic drugs in postmortem human scalp hair. J Forensic Sci 40: 87–90. Dawling S, Braithwaite RA (1978). Simplified method for monitoring tricyclic antidepressant therapy using gas–liquid chromatography with nitrogen detection. J Chromatogr 146: 449–456. de la Torre R et al. (1998). Quantitative determination of tricyclic antidepressants and their metabolites in plasma by solid-phase extraction (Bond-Elut TCA) and separation by capillary gas chromatography with nitrogen–phosphorous detection. Ther Drug Monit 20: 340–346. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid–liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Dolezalova M (1992). On-line solid-phase extraction and high-performance liquid chromatographic determination of nortriptyline and amitriptyline in serum. J Chromatogr 579: 291–297. Edelbroek PM et al. (1984). Amitriptyline metabolism in relation to antidepressive effect. Clin Pharmacol Ther 35: 467–473. Fischer D, Breyer-Pfaff U (1995). Comparison of procedures for measuring the quaternary Nglucuronides of amitriptyline and diphenhydramine in human urine with and without hydrolysis. J Pharm Pharmacol 47: 534–538. Garland WA (1977). Quantitative determination of amitriptyline and its principal metabolite, nortriptyline, by GLC–chemical ionization mass spectrometry. J Pharm Sci 1977 66: 77–81. Ghahramani P, Lennard MS (1996). Quantitative analysis of amitriptyline and nortriptyline in human plasma and liver microsomal preparations by high-performance liquid chromatography. J Chromatogr B Biomed Appl 685: 307–313. Hackett LP et al. (1998). A comparison of high-performance liquid chromatography and fluorescence polarization immunoassay for therapeutic drug monitoring of tricyclic antidepressants. Ther Drug Monit 20: 30–34. Hansch C et al. (1995). Exploring QSAR: Hydrophobic, Electronic, and Steric Constants. Washington DC: American Chemical Society. H€artter S, Hiemke C (1992). Column switching and high-performance liquid chromatography in the analysis of amitriptyline, nortriptyline and hydroxylated metabolites in human plasma or serum. J Chromatogr 578: 273–282. Johansen K, Rasmussen KE (1998). Automated on-line dialysis for sample preparation and HPLC analysis of antidepressant drugs in human plasma: inhibition of interaction with the dialysis membrane. J Pharm Biomed Anal 16: 1159–1169. Kollroser M, Schober C (2002). Simultaneous determination of seven tricyclic antidepressant drugs in human plasma by direct-injection HPLC-APCI-MS-MS with an ion trap detector. Ther Drug Monit 24: 537–544. Kudo K et al. (1997). Selective determination of amitriptyline and nortriptyline in human plasma by HPLC with ultraviolet and particle beam mass spectrometry. J Anal Toxicol 21: 185–189. Lee XP et al. (1997). Detection of tricyclic antidepressants in whole blood by headspace solid-phase microextraction and capillary gas chromatography. JChromatogr Sci 35: 302–308. Lee XP et al. (2008). Determination of tricyclic antidepressants in human plasma using pipette tip solid-phase extraction and gas chromatography–mass spectrometry. J SepSci 31: 2265–2271. Li S et al. (1994). Identification and quantitation of drugs of abuse in urine using the generalized rank annihilation method of curve resolution. J Chromatogr B Biomed Appl 655: 213–223. Maresova Vet al. (2008). Screening and semiquantitative analysis of drugs and drugs of abuse in human serum samples using gas chromatography–mass spectrometry. Neuroendocrinol Lett 29: 749–754. Margalho C et al. (2007). Massive intoxication involving unusual high concentration of amitriptyline. Hum Exp Toxicol 26: 667–670. McIntyre IM et al. (1993). Dual ultraviolet wavelength high-performance liquid chromatographic method for the forensic or clinical analysis of seventeen antidepressants and some selected metabolites. J Chromatogr 621: 215–223.
889
Meylan WM, Howard PH (1995). Atom/fragment contribution method for estimating octanol– water partition coefficients. J Pharm Sci 84: 83–92. Molnar G, Gupta RN (1980). Plasma levels and tricyclic antidepressant therapy. Part 2. Pharmacokinetic, clinical and toxicologic aspects. Biopharm Drug Dispos 1: 283–305. Munksgaard EC (1969). Concentrations of amitriptyline and its metabolites in urine, blood and tissue in fatal amitriptyline poisoning. Acta Pharmacol Toxicol 27: 129–134. Paterson S et al. (2004). Screening and semi-quantitative analysis of post mortem blood for basic drugs using gas chromatography/ion trap mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 813: 323–330. Pounder DJ et al. (1994). Postmortem changes in blood amitriptyline concentration. Am J Forensic Med Pathol 15: 224–230. Queiroz RH et al. (1995). Simultaneous HPLC analysis of tricyclic antidepressants and metabolites in plasma samples. Pharm Acta Helv 70: 181–186. Rao ML et al. (1994). Monitoring tricyclic antidepressant concentrations in serum by fluorescence polarization immunoassay compared with gas chromatography and HPLC. Clin Chem 40: 929–933. Sangster J (1997). Octanol–Water Partition Coefficients: Fundamentals and Physical Chemistry. Chichester, UK: Wiley. Sauvage FL et al. (2006). A fully automated turbulent-flow liquid chromatography–tandem mass spectrometry technique for monitoring antidepressants in human serum. Ther Drug Monit 28: 123–130. Scoggins BA et al. (1980). Measurement of tricyclic antidepressants. Part I. A review of methodology. Clin Chem 26: 5–17. Segatti MP et al. (1991). Rapid and simple high-performance liquid chromatographic determination of tricyclic antidepressants for routine and emergency serum analysis. J Chromatogr 536: 319–325. Shu Y et al. (1998). Determination of amitriptyline and nortriptyline in human liver microsomes with reversed-phase HPLC in vitro. Zhongguo Yao Li Xue Bao 19: 343–346. Stiakakis I et al. (2009). Disputed case of homicide by smothering due to severe amitriptyline intoxication of the victim. J Forensic Leg Med 16: 280–283. Tanaka E et al. (1997). Forensic analysis of eleven cyclic antidepressants in human biological samples using a new reversed-phase chromatographic column of 2 micron porous microspherical silica gel. J Chromatogr B Biomed Sci Appl 692: 405–412. Theurillat R, Thormann W (1998). Monitoring of tricyclic antidepressants in human serum and plasma by HPLC: characterization of a simple, laboratory developed method via external quality assessment. J Pharm Biomed Anal 18: 751–760. Titier K et al. (2007). Quantification of tricyclic antidepressants and monoamine oxidase inhibitors by high-performance liquid chromatography–tandem mass spectrometry in whole blood. J Anal Toxicol 31: 200–207. Tracqui A et al. (1992). Determination of amitriptyline in the hair of psychiatric patients. Hum Exp Toxicol 11: 363–367. Ulrich S, Martens J (1997). Solid-phase microextraction with capillary gas–liquid chromatography and nitrogen–phosphorus selective detection for the assay of antidepressant drugs in human plasma. J Chromatogr B Biomed Sci Appl 696: 217–234. Ulrich S et al. (1996). Simultaneous determination of amitriptyline, nortriptyline and four hydroxylated metabolites in serum by capillary gas–liquid chromatography with nitrogen–phosphorusselective detection. J Chromatogr B Biomed Appl 685: 81–89. Vandel S et al. (1992). [Comparative study of two techniques for the determination of amitriptyline and nortriptyline: EMIT and gas chromatography.]. Therapie 47: 41–45. Zarghi A et al. (2001). Determination of amitriptyline in plasma samples by high-performance liquid chromatography. BollChim Farm 140: 458–461. Zhang H et al. (2000). Atmospheric pressure ionization time-of-flight mass spectrometry coupled with fast liquid chromatography for quantitation and accurate mass measurement of five pharmaceutical drugs in human plasma. J Mass Spectrom 35: 423–431.
Amlodipine Calcium Channel Blocker C20H25ClN2O5 = 408.9 CAS—88150-42-9 IUPAC Name 3-O-Ethyl 5-O-methyl 2-(2-aminoethoxymethyl)-4-(2-chlorophenyl)-6-methyl-1, 4-dihydropyridine-3,5-dicarboxylate Synonyms 2-[(2-Aminoethoxy)methyl]-4-(2-chlorophenyl)-1,4-dihydro-6methyl-3,5-pyridinedicarboxylic acid 3-ethyl 5-methyl ester; UK-48340.
Chemical Properties pKa 8.6. Log P (octanol/water), 3.00. Extraction yield (chlorobutane), 1 [Demme et al. 2005]. Amlodipine Maleate C20H25ClN2O5,C4H4O4 = 525.0 CAS—88150-47-4 Synonym UK-48340-11 Chemical Properties A white crystalline powder. Mp 178 to 179 .
Amlodipine Besilate C20H25ClN2O5,C6H5SO3H = 567.1 CAS—111470-99-6 Synonyms Amlodipine besylate; amlodipine monobenzenesulfonate. Proprietary Names Amlodin; Amlor; Antacal; Astudal; Istin; Monopina; Norvas;
Norvasc.
Chemical Properties A white crystalline powder. It is slightly soluble in water; sparingly soluble in ethanol.
A
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:29:43
890
A
Amobarbital
Thin-layer Chromatography System TE—Rf 0.36; system TB—Rf 0.02; system TAE—Rf 0.11. Gas Chromatography System GB—RI 2982; system GP—M (dehydro-2HOOC)-ME-, RI 2430, M (dehydro-desamino-HOOC)-ME-, RI 2635. High Performance Liquid Chromatography System HX—RI 428; system HZ—retention time 4.9 min; system HAA—retention time 15.1 min. Column: ODS Cosmosil 5 C18-P (150 4.6 mm, 5 mm). Mobile phase: 0.05 mol/ L phosphate buffer solution (pH 3.1) : acetonitrile (65:35) containing 0.005 mol/L sodium octane sulfonate and 5 mg/L EDTA, flow rate 1 mL/min. Electrochemical detection. Retention time: 10.1 min [Shimooka et al. 1989]. Ultraviolet Spectrum Aqueous acid (0.2 mol/L NH2SO4)—239 nm; basic— 238 nm (besilate).
Infrared Spectrum Principal peaks at wavenumber 1208, 1182, 1094 cm1 (KBr disk).
320 at 20 /min, for 9 min. Carrier gas: N2, flow rate 1 mL/min. ECD. Retention time: 4.4 min [Faulkner et al. 1986]. Limit of detection, 0.2 mg/L [Faulkner et al. 1986]. HPLC Electrochemical detection. Limit of detection, about 0.2 mg/L for each enantiomer [Josefsson, Norlander 1996]. Serum HPLC Electrochemical detection. Limit of detection, 0.1 mg/L [Shimooka et al. 1989]. Gingival Crevicular Fluid GC-MS ECD. Limit of detection, 0.5 mg/L [Monkman et al. 1996]. Tissue GC-MS ECD. Limit of detection, 0.5 ng/g for each enantiomer [Scharpf et al. 1994]. Note For GC-MS method see Beresford et al. [1988]. Disposition in the Body Amlodipine is slowly and almost completely absorbed after oral administration; peak plasma concentrations occur after 6 to 12 h. Absorption is not affected by food. It undergoes minimal presystemic metabolism; bioavailability is about 60 to 65% and it undergoes extensive but slow metabolism in the liver. Metabolites lack significant pharmacological activity and are excreted predominantly in urine (about 60% of a dose); 355 . Very soluble in water, slightly
soluble in alcohol. Used as a herbicide. Methanearsonic Acid CH3H2AsO3 = 134.0 CAS—124-58-3 Synonyms Arsonic acid, methyl-; MA; MMA; monomethylarsonic acid. Chemical Properties White solid. Pleasant acid taste. Mp 161 . Soluble in water
and alcohol. pKa1 3.6, pKa2 8.2 [Sur, Dunemann 2004]. Used as a herbicide. Roxarsone C6H6AsNO6 = 263.1 CAS—121-19-7 Synonyms 4-Hydroxy-3-nitrophenylarsonic acid; NSC-2101; nitrophenolarsonic
acid. Proprietary Name Ren-o-sal Chemical Properties Pale yellow solid. Puffs up and deflagrates on heating.
Slightly soluble in cold water; soluble in approx. 30 parts boiling water; freely soluble in methanol, ethanol, acetic acid, acetone and alkalis; sparingly soluble in dilute mineral acids. Insoluble in ether, ethyl acetate. Used as an antibacterial. Sodium Arsanilate C6H7AsNNaO3 = 239.1 CAS—127-85-5 Synonyms Arsanilic acid
sodium
salt;
sodium
aminarsonate;
sodium
anilarsonate. Proprietary Names (4-Aminophenyl)-arsonic acid sodium salt; Arsamin; Atoxyl;
Nuarsol; Protoxyl; Soamin; Sonate; Piglet Pro-Gen V; Trypoxyl.
Chemical Properties The tetrahydrate is a white odourless crystalline powder.
Soluble in water and alcohol. Formerly used as an antisyphilitic. Sodium Cacodylate C2H6AsNaO2 = 160.0 CAS—124-65-2
Quantification Specimen collection: Blood—10 mL, K-EDTA tube; urine— 20 mL plastic universal container. Blood AAS Perkin-Elmer 3300. Arsenic hollow cathode lamp (l ¼ 193.7 nm). Limit of detection, 1.0–2.0 mg/L [Alauddin et al. 2003]. Limit of detection, 1.95 ng [Concha et al. 1998]. GBC hollow cathode lamp (l ¼ 193.7 nm). Carrier gas: N2, 200 mL/min. Limit of detection, 0.02 mg/L [Tripathi et al. 1997]. PerkinElmer 5000 (l ¼ 193.7). Arsenic hollow cathode lamp. Limit of detection, 0.5 mg/L [Foa et al. 1984]. FAAS Flame: air-acetylene. Carrier gas: N2, 90 mL/min. Hollow cathode lamp (l ¼ 193.7 nm). Limit of detection not reported [Le et al. 1992]. ETAAS Carrier gas: Ar. Dry cycle: 200 at 35 s for 30 s. Char cycle: 1600 at 20 s for 15 s. Atomisation cycle: 2500 for 4 s, gas stop. Limit of detection, 2 mg/L [Campillo et al. 2000]. HPLC-AAS Column: Phenomenex Bondclone (300 3.9 mm i.d., 10 mm). Arsenic hollow cathode lamp (l ¼ 193.7 nm). Limit of detection, 10 mg/L for arsenobetaine, DMA, and arsenite; 15 mg/L for MMA; and 20 mg/L for arsenate [Le et al. 1994]. HPLC-ICP-MS Column: Inertsil AS (150 2.1 mm i.d., 3.0 mm). Mobile phase: 10 mmol/L sodium butanesulfate : 4 mmol/L tetramethyl ammonium hydroxide : 4 mmol/L malonic acid : 0.05% methanol (pH 3.0), flow rate 0.2 mL/min. Limit of quantification, 0.1 mg/L for As3, As5 and the methylated metabolites [Fujisawa et al. 2007]. Column: Phenomenex Bondclone (300 3.9 mm i.d., 10 mm). Mobile phase: 50 mmol/L phosphate or carbonate buffer (pH 7.5, 9.0, or 10.3), flow rate 1.0 mL/ min. Outer gas: 13.8 L/min. Auxiliary gas: 0.70 L/min. Nebuliser gas: 0.96 L/min. Meinhard nebuliser. Limit of detection not reported [Le et al. 1994]. ICP-MS Limit of detection, 0.97 mg/L [Liao et al. 2004]. Coolant gas: 16 L/min. Auxiliary gas: 1.4 L/min. Nebuliser gas: 0.7 L/min. Limit of quantification, 5 mg/L for DMA, arsenocholine, and As5; 4 mg/L for arsenobetaine; 0.8 mg/L for As3; and 0.6 mg/L for MMA. Limit of detection, 2 mg/L for DMA and As5, 1 mg/L for arsenobetaine and arsenocholine, and 0.2 mg/L for As3 and MMA [Milstein et al. 2003]. Plasma gas: 15 L/min. Auxiliary gas: 1.0 L/min. Method of detection, 30, 45, 27, and 61 ng/L for AsB, DMA, MMA, and As5, respectively [Wei et al. 2000]. Plasma gas: Ar, 16 L/min. Auxiliary gas: 1.4 L/min. Carrier gas: 0.5 L/min. Limit of detection not reported [Tanaka et al. 1996]. Note For spectrophotometric methods for the detection of arsenic in blood, see Pillai et al. [2000] and Lakso et al. [1979]. Serum ETAAS See Blood [Campillo et al. 2000]. ICP-MS Column: Inertsil AS (150 2.1 mm i.d., 3 mm). Mobile phase: 10 mmol/ L butane sulfonic sodium : 4 mmol/L malonic acid: 4 mmol/L tetramethylammonium hydroxide (pH 3.0), flow rate 0.2 mL/min. Plasma gas: Ar, 18 L/min. Limit of detection,not reported [Fukai et al. 2006]. Urine LC-MS Column: weak anion exchange. Mobile phase: methanol ammonium dihydrogen phosphate : ammonium acetate : glacial acetic acid. Limit of
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:34:25
Arsenic detection, 0.5 mg/L for As3 and As5, 2.0 mg/L for MMA, 1.5 mg/L for DMA, and 1.0 mg/L for arsenobetaine [Apostoli et al. 1999]. AAS Perkin-Elmer MHS-20. Varian AA 1275. Limit of detection, 1 mg/L (approx. 1 ng) [Heilier et al. 2005]. Phillips AAS-PU-9100 (l ¼ 193.7 nm). Arsenic hollow cathode lamp. Limit of detection, 0.5 mg/L [Dang et al. 1999]. Limit of detection, 1.95 ng [Concha et al. 1998]. Carrier gas: Ar, 200 mL/min. Dry cycle: 50 to 80 for 10 s to 120 for 10 s. Char cycle: 120 to 400 for 20 s. Atomisation cycle: 2600 for 5 s, gas stop. Limit of detection not reported [Das et al. 1995]. See Blood [Foa et al. 1984]. HPLC-AFS Column: Hamilton PRP-X100 anion exchange (250 4.1 mm i.d., 10 mm). Mobile phase: 10 mmol/L ammonium dihydrogen phosphate (pH 5.8) : 60 mmol/L ammonium dihydrogen phosphate (pH 5.8, 100 : 0 for 2 min to 0 : 100 at 2 min for 4 min to 100 : 0 for 6 min), flow rate 1.0 mL/min. Retention time: 2.6, 3.6, 6.4 and 8.8 min for As3, DMA, MA, and As5, respectively. Arsenic-boosted discharge hollow cathode super lamp (l ¼ 197.3, 193.7 and 189.0 nm). Carrier gas: Ar, 0.25 L/min. Drying gas: N2, 2.5 L/min. Limit of quantification, 10, 11, 6, and 6 mg/L for As3, DMA, MMA, and As5, respectively. Limit of detection, 3, 4, 2, and 2 mg/L for As3, DMA, MMA, and As5, respectively [Lindberg et al. 2007]. FAAS See Blood [Le et al. 1992]. ETAAS Dry cycle: 130 at 20 s for 10 s. Char cycle: 1400 in 5 s for 30 s. Atomisation cycle: 2500 for 5 s. Limit of detection, 0.03 mg/L [Horng et al. 2002]. See Blood [Campillo et al. 2000]. Limit of detection, 0.5 mg/L [Gebel et al. 1998]. ICP-MS See Serum [Fukai et al. 2006]. Limit of quantification, 0.18 mg/L; limit of detection, 0.05 mg/L [Brima et al. 2006], 9.98 mg/L [Liao et al. 2004]. Coolant gas: 16 L/min. Auxiliary gas: 1.4 L/min. Nebuliser gas: 0.7 L/min. Limit of quantification, 5 mg/L for DMA, arsenocholine and As5; 4 mg/L for arsenobetaine; 0.8 mg/L for As3; 0.6 mg/L for MMA. Limit of detection, 2 mg/L for DMA and As5; 1 mg/L for arsenobetaine and arsenocholine; 0.2 mg/L for As3 and MMA [Milstein et al. 2003]. Plasma gas: 15 L/min. Auxiliary gas: 1.0 L/min. Limit of detection, 30, 45, 27 and 61 ppt for AsB, DMA, MMA, and As5, respectively [Wei et al. 2000]. See also Apostoli et al. [1999], Amarasiriwardena et al. [1998] and Alves et al. [1993]. GC-ICP-MS Limit of detection, 2–12 pg/L [Kresimon et al. 2001]. HPLC-ICP-MS Column: Inertsil AS (150 2.1 mm i.d., 3.0 mm). Mobile phase: 10 mmol/L sodium butanesulfate : 4 mmol/L tetramethyl ammonium hydroxide : 4 mmol/L malonic acid : 0.05% methanol (pH 3.0), flow rate 0.2 mL/min. Limit of quantification, 0.1 ppb for As3, As5, and the methylated metabolites [Fujisawa et al. 2007]. Column: Hamilton PRP-X100 (250 4.1 mm i.d., 10 mm). Mobile phase: 10 mmol/L ammonium carbonate in 2% methanol (pH 9.0) : 50 mmol/L ammonium carbonate in 2% methanol (pH 9.0; 100 : 0 for 0.5 min to 0 : 100 at 0.5 min for 12.5 min), flow rate 1.0 mL/min. Plasma gas: 15 L/min. Auxiliary gas: 1.1 L/min. Nebuliser gas: 1.0 L/min. Limit of detection, 0.003, 0.01, 0.004, 0.003 and 0.002 mg/L for arsenobetaine, As3, DMA, MMA and As5, respectively [Wang et al. 2007]. Column: Shodex RSpak NN-614 cation exchange (150 4.6 mm i.d.) Mobile phase: 5 mmol/L nitric acid : 6 mmol/L ammonium nitrate : 1.5 mmol/L 2, 6-pyridinedicarboxylic acid, flow rate 1.0 mL/min. Limit of detection, 0.1 mg/L [Hata et al. 2007]. Column: Hamilton PRPX100 spherical poly(styrene-divinyl-benzene) trimethylammonium exchanger (250 4.1 mm i.d.). Mobile phase: 10 mmol/L ammonium carbonate : 20 mmol/L ammonium carbonate (100 : 0 for 5 min to 0 : 100 at 6 min for 16 min to 100 : 0 at 23 min for 4 min), flow rate 1 mL/min. Nebuliser gas: 0.85 L/ min. Limit of detection, 0.8 mg/L [Morton, Mason 2006]. Limit of detection, 0.3 mg/ L for As3, MMA, and DMA; 0.4 mg/L for As5 [Christian et al. 2006]. See also Chowdhury et al. [2003]; Kavanagh et al. [1998]; Lai et al. [2004]; Le et al. [1994]; Mandal et al. [2003]; Meza et al. [2004]; Sur, Dunemann [2004]; Zheng et al. [1999]. Note For spectrophotometric methods for the detection of arsenic in urine, see Lakso et al. [1979] and Kneip et al. [1977]. Hair AAS See Urine [Das et al. 1995]. Perkin-Elmer 403 (l ¼ 193.7 nm). Carrier gas: Ar. Fuel: H2. Oxidant: Air. Limit of detection, 0.02 mg [Curatola et al. 1978]. ETAAS Dry cycle: 80 to 120 in 10 s. Char cycle: 300 to 400 at 10 s. Atomisation cycle: 2700 to 2800 in 5 s. Carrier gas: 200 mL/min. Hitachi model 180-50, S.N.5721-2 (l ¼ 193.8 nm). Limit of detection not reported [Kazi et al. 2006]. See Urine. Limit of detection, 5 mg/kg [Gebel et al. 1998]. Dry cycle: 140 at 5 s for 15 s. Char cycle: 1300 in 15 s for 30 s. Atomisation cycle: 2700 for 5 s. Perkin-Elmer 5000 (l ¼ 193.7 nm). Limit of detection, 10 pg [Koons and Peters 1994]. ICP-MS Plasma gas: Ar, 13 L/min. Auxiliary gas: Ar, 0.9 L/min. Nebuliser gas: Ar, 0.95 L/min. Limit of quantification, 0.036 mg/kg [Kintz et al. 2007]. Limit of detection, 0.13 mg/kg [Nadal et al. 2005]. Plasma gas: 15 L/min. Auxiliary gas: 0.8 L/min. Nebuliser gas: 0.8 L/min. Limit of detection not reported [Samanta et al. 2004]. HPLC-ICP-MS Column: RP-C18 (100 4.6 mm i.d.). Mobile phase: 0.35 mmol/ L tetrabutylammonium hydrogen sulfate (pH 5.75), flow rate 1 mL/min. Carrier gas: Ar, 0.75 L/min. Plasma gas: 16 L/min. Auxiliary gas: 1 L/min. Limit of detection not reported [Yan˜ez et al. 2005]. Column: Shodex Asahipak ES-502N 7C (100 7.6 mm i.d.). Mobile phase: 15 mmol/L citric acid monohydrate (pH 2.0), flow rate 1.0 mL/min. Plasma gas: 15 L/min. Auxiliary gas: 1.2 L/min. Limit of detection not reported [Mandal et al. 2003]. Liver AAS See Urine [Das et al. 1995]. Nail AAS See Urine [Das et al. 1995]. ICP-MS See Hair [Samanta et al. 2004]. Perkin-Elmer Sciex ELAN 5000. Plasma gas: Ar. Limit of detection, 0.07 mg/L [Chen et al. 1999].
917
HPLC-ICP-MS Column: Shodex Asahipak ES-502N 7C (100 7.6 mm i.d.). Mobile phase: 15 mmol/L citric acid monohydrate (pH 2.0), flow rate 1.0 mL/ min. Plasma gas: 15 L/min. Auxiliary gas: 1.2 L/min. Limit of detection not reported [Mandal et al. 2003]. Milk AAS See Urine [Concha et al. 1998]. ICP-MS Plasma gas: 12 to 13 L/min. Auxiliary gas: 0.9 to 1.0 L/min. Sample gas: 1.0 to 1.2 L/min. Limit of detection, 0.2 mg/L [Krachler et al. 2000]. Other GC-MS Water. Column: HP-5MS (30 m 0.25 mm i.d., 0.25 mm). Temperature programme: 75 for 0.5 min to 280 at 30 /min for 1 min. Carrier gas: He, 0.75 mL/min. EI ionisation at 70 eV. Limit of detection, 0.1 mg/L for DMA and MMA, 3 mg/L for total inorganic arsenic [Claussen 1997]. AAS Fresh and Freeze-dried Plant Ssamples. Carrier gas: Ar, 50 mL/min. Atomisation cycle: 770 . Hollow cathode lamp (l ¼ 193.7 nm). Limit of detection not reported [Krachler, Emons 2000]. Seafood Products. Perkin-Elmer 5000. Limit of detection, 0.7 mg/kg [Mun˜oz et al. 1999]. Water. See Urine [Das et al. 1995]. ETAAS Naphtha. Dry cycle: 50 for 10 s to 90 in 25 s to 120 in 10 s, Ar, 200 mL/ min. Atomisation cycle: 2500 for 8 s (gas stop; l ¼ 193.7 nm). Limit of detection, 2.7 mg/L [Cassella et al. 2004]. ICP-MS Seafood. Perkin-Elmer Elan 6000. Limit of detection, 0.05 mg/kg [Falco´ et al. 2006]. Infant Food Products. Coolant gas: Ar, 12 L/min. Auxiliary gas: Ar, 1.0 L/min. Nebuliser gas: 0.8 L/min. Limit of detection, 5 mg/kg for As3, MMA and DMA; 10 mg/kg for As5 [Vela, Heitkemper 2004]. Food. Varian-Vista with an ultrasonic nebuliser. Limit of detection, 0.06 mg/kg [Llobet et al. 2003]. Meals from a Catering Establishments. Plasma gas: 15 L/min. Auxiliary gas: 0.9 L/ min. Nebuliser gas: 0.73 L/min. Limit of quantification, 16 mg/kg [Noel et al. 2003]. Vegetables. Limit of detection, 0.04 mg/L [Alam et al. 2003]. Well Water. Limit of detection, 3 mg/L [Rahman et al. 2001]. Beverages. Limit of detection, 0.3 mg/kg [MacIntosh et al. 2000]. Method 200.8 of the EPA. Limit of detection, 1 mg/L [McCarty et al. 2004]. HPLC-ICP-MS Rice. Column: CAPCELL PAK C18MG (250 4.6 mm i.d.). Mobile phase: 10 mmol/L sodium 1-butanesulfonate : 4 mmol/L tetramethylammonium hydroxide : 4 mmol/L malonic acid : 0.05% methanol (pH 3.0), flow rate 0.75 mL/min. Plasma gas: 15 L/min. Carrier gas: 0.70 L/min. Auxiliary gas: 0.9 L/ min. Makeup gas: 0.43 L/min. Limit of quantification, 7.9 mg/kg; limit of detection, 2.4 mg/kg [Hamano-Nagaoka et al. 2008]. Drinking Water. Column: Agilent (250 4.6 mm i.d., 5 mm). Mobile phase: 2.0 mmol/L sodium dihydrogen phosphate : 0.2 mmol/L EDTA (pH 6.0), flow rate 1.0 mL/min. Plasma gas: Ar, 15 L/min. Auxiliary gas: 1.0 L/min. Carrier gas: 1.0 L/min. Limit of detection, 67, 74, 35 and 89 ng/L for As3, DMA, MMA and As5, respectively [Day et al. 2002]. Note For colourimetric assays for arsenic, see George et al. [1973] and Crawford and Tavares [1974]. For a study of arsenic in soils, plants, water and sediments in Korea, see [Jung et al. [2002]. Disposition in the Body Absorption Inhalation exposure Arsenic absorption via the lungs is a two-step process: deposition of the particles on to the lung surface, and absorption of arsenic from the deposited material. In patients with lung cancer exposed to arsenic in cigarette smoke, deposition was estimated to be approx. 40% and absorption 75 to 85%. Thus, overall absorption was approx. 30 to 34%. In workers exposed to arsenic trioxide dusts in smelters, the amount excreted in the urine was approx. 40 to 60% of the inhaled dose. Oral exposure Arsenates and arsenites are well absorbed from the gastrointestinal tract. In one study where faecal excretion was measured in humans given oral doses of arsenite, less than 5% was recovered. This indicates absorption was at least 95%. This is supported in studies where urinary excretion in humans was found to account for 55 to 80% of daily oral intakes of arsenate or arsenite. However, gastrointestinal absorption may be lower if highly insoluble forms or arsenic are ingested. Dermal exposure Studies investigating percutaneous absoption of 73As as arsenic acid alone and mixed with soil has been measured in skin from cadavers. Labelled arsenic was applied to skin in diffusion cells and transit through the skin into receptor fluid measured. After 24 h, 0.93% of the dose passed through the skin and 0.98% remained in the skin after washing. Absorption was lower with 73As mixed with soil. Arsenates and arsenites are well absorbed following both oral and inhalation exposure. Data on distribution are limited but it appears arsenic is transported to nearly all tissues with the highest concentrations found in muscle. Metabolism involves mainly reduction-oxidation reactions that interconvert Asþ5and Asþ3; and methylation of Asþ3 to form MA and DMA. Most arsenic is rapidly excreted in urine as a mixture of inorganic arsenics, MA and DMA, although some may remain bound in tissues (especially skin, hair and fingernails). The capacity of the body to methylate inorganic arsenic is very important in terms of detoxification. Limited data suggest that the methylation system might begin to become saturated at intakes of approx. 0.2 to 1 mg per day. Urinary excretion is complete within 6 days and accounts for approx. 90% of the dose. Trivalent arsenic salts are excreted as DMA (50%), MA (14%), As(þ5) (8%), and As(þ3) (8%). Organo-arsenic compounds (e.g. arsenobetaine, arsenochlorine), which are present in various types of seafood, are excreted unchanged. Normal Concentrations Blood—0.2 mg/L); poorly soluble in a variety of organic solvents. Ultraviolet Spectrum Basic—252, 276 nm.
Infrared Spectrum Principal peaks at wavenumbers 1659, 1278, 1370 cm1 (KBr pellet).
931
The mean peak plasma concentration occurring 6 h after administration of 750 mg three times a day for 5 days followed by 750 mg twice a day for 16 days, with a standard breakfast, was 51 mg/L in 4 male subjects, aged over 18 years, with CD4 counts 250 cells/mm3. Single doses of 100, 250, 750, 1500 and 3000 mg administered to the same group produced peak concentrations of 4.5, 14.3, 37.9, 33.4 and 39.0 mg/L, respectively, observed between 4 and 8 h [Hughes et al. 1991]. Toxicity It is safe to give doses of up to 3000 mg daily. Half-life Plasma, between 60 and 70 h. Volume of Distribution 0.6 L/kg. Clearance Plasma, 0.15 mL/min/kg; also, reported as 1.56 L/h. Protein Binding >99%. Note For reviews of atovaquone see Haile, Flaherty [1993] and Spencer, Goa [1995]. Dose 750 mg as an oral suspension twice a day or 750 mg as tablets three times a day. DeAngelis DV et al. (1994). High-performance liquid chromatographic assay for the measurement of atovaquone in plasma. J Chromatogr 652: 211–219. Dixon R et al. (1996). Single-dose and steady-state pharmacokinetics of a novel microfluidized suspension of atovaquone in human immunodeficiency virus-seropositive patients. Antimicrob Agents Chemother 40: 556–560. Haile LG, Flaherty JF (1993). Atovaquone: a review. Ann Pharmacother 27: 1488–1494. Hannan SL et al. (1996). Determination of the potent antiprotozoal compound atovaquone in plasma using liquid-liquid extraction followed by reversed-phase high-performance liquid chromatography with ultraviolet detection. J Chromatogr B Biomed Sci Appl 678: 297–302. Hansson AG et al. (1996). Rapid high-performance liquid chromatographic assay for atovaquone. J Chromatogr B Biomed Sci Appl 675: 180–182. Hughes WT et al. (1991). Safety and pharmacokinetics of 566C80, a hydroxynaphthoquinone with anti-Pneumocystis carinii activity: a phase I study in human immunodeficiency virus (HIV)infected men. J Infect Dis 163: 843–848. Rolan PE et al. (1994). Examination of some factors responsible for a food-induced increase in absorption of atovaquone. Br J Clin Pharmacol 37: 13–20. Spencer CM, Goa KL (1995). Atovaquone. A review of its pharmacological properties and therapeutic efficacy in opportunistic infections. Drugs 50: 176–196. Studenberg SD et al. (1995). A robotics-based liquid chromatographic assay for the measurement of atovaquone in plasma. J Pharm Biomed Anal 13: 1383–1393.
Atracurium Skeletal Muscle Relaxant C53H72N2O12 = 929.2 CAS—64228-79-1 IUPAC Name 2,20 -[1,5-Pentanediylbis[oxy(3-oxo-3,1-propanediyl)]]bis[1[(3,4-dimethoxyphenyl)-methyl]-1,2,3,4-tetrahydro-6,7-dimethoxy-2methylisoquinolinium]
Atracurium Besilate Mass Spectrum Principal ions at m/z 202, 366, 115, 77, 368, 125, 105, 213. Quantification Plasma GC ECD. Sensitivity, 0.01 mg/L [Rolan et al. 1994]. HPLC UV detection (l¼254 nm). Limit of quantification, 0.1 mg/L [Hannan et al. 1996]. Column: C6 Spherisorb (250 4.6 mm i.d., 5 mm). Mobile phase: methanol : 10 mmol/L triethylamine in 0.2% aqueous trifluoroacetic acid, pH 2 (76:24), flow rate 1 mL/min. Retention time: 8.8 min. UV detection. Limit of detection, 0.5 mg/L [Hansson et al. 1996]. UV detection. Limit of detection, 0.25 mg/L [Studenberg et al. 1995]. UV detection (l¼254 nm). Limit of quantification, 0.25 mg/L [DeAngelis et al. 1994]. UV detection. Limit of detection, 0.05 mg/L [Rolan et al. 1994]. Disposition in the Body Atovaquone is rapidly but poorly absorbed after oral administration; bioavailability shows considerable inter-individual variation and is increased by concomitant administration with food, especially food with high fat content. The drug is excreted almost entirely in faeces (94% of an administered dose over 21 days) as the unchanged drug, with little present in urine. There is no evidence that atovaquone is metabolised in the body. It may undergo enterohepatic recycling. Plasma concentrations do not increase proportionally with dose. Therapeutic Concentration Six male individuals, HIV-sero-positive, with CD4 counts >200 cells/mm3 were administered with single 500, 1000 and 1500 mg doses after an overnight fast. Peak plasma concentrations reached 2.7, 5.6 and 4.3 mg/L, respectively, between 1.5 and 2.5 h. Following administration of 1000 mg daily with a high-fat meal and 1000 mg twice daily with or without a highfat meal, for 14 days, the mean concentrations were 24.2, 41.3 and 34.4 mg/L, respectively. These levels were detected at 9.1, 3.7 and 2.1 h [Dixon et al. 1996].
C53H72N2O12,2C6H5O3S = 1243.5 CAS—64228-81-5 Synonyms Atracurium besylate; BW-33A. Proprietary Name Tracrium Chemical Properties An off-white powder. Mp 85 to 90 . Softens at 60 .
Ultraviolet Spectrum Ethanol—231, 280 nm; basic—281 nm.
Infrared Spectrum Principal peaks at wavenumber 1725, 1513, 1189 cm1 (KBr pellets).
A
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:35:10
932
Atrazine Beemer GH et al. (1990). Pharmacokinetics of atracurium during continuous infusion. Br J Anaesth 65: 668–674. Bjorksten AR et al. (1990). Simple high-performance liquid chromatographic method for the analysis of the non-depolarizing neuromuscular blocking drugs in clinical anaesthesia. J Chromatogr 533: 241–247. Charlton AJ et al. (1989). Atracurium overdose in a small infant. Anaesthesia 44: 485–486. Durcan J, Carter JA (1986). Overdose of atracurium. Anaesthesia 41: 767. Peat SJ et al. (1988). The prolonged use of atracurium in a patient with tetanus. Anaesthesia 43: 962–963. Simmonds RJ (1985). Determination of atracurium, laudanosine and related compounds in plasma by high-performance liquid chromatography. J Chromatogr 343: 431–436. Varin F et al. (1990). Determination of atracurium and laudanosine in human plasma by highperformance liquid chromatography. J Chromatogr 529: 319–327. Yate PM et al. (1987). Clinical experience and plasma laudanosine concentrations during the infusion of atracurium in the intensive therapy unit. Br J Anaesth 59: 211–217.
A
Atrazine Herbicide C8H14ClN5 = 215.7 CAS—1912-24-9 IUPAC Name 6-Chloro-N-ethyl-N0 -(1-methylethyl)-1,3,5-triazine-2,4-diamine
Quantification Plasma HPLC Column: mPorasil (150 3.9 mm i.d.). Mobile phase: acetonitrile : 0.002 mol/L sulfuric acid (50:50), flow rate 2 mL/min. UV detection (l¼210 nm). Retention time: 3.5 min. Limit of detection, 0.025 mg/L for atracurium and laudanosine [Bjorksten et al. 1990]. Column: Spherisorb C8 (100 4.9 mm, 5 mm). Mobile phase: (A) acetonitrile : methanol : 0.03 mol/L dibasic potassium phosphate buffer (37.5:5:57.5); (B) acetonitrile : methanol : 0.1 mol/L dibasic potassium phosphate buffer (37.5:15:47.5), final pH 5. Elution programme: 0 to 100% (B) over 8 min, flow rate 1.7 mL/min. Fluorescence detection (lex¼240 nm, lem¼320 nm). Retention time: 6.2 min. Limit of detection, 0.02 mg/L for atracurium, 0.01 mg/L for laudanosine [Varin et al. 1990]. Fluorescence detection. Limit of detection, about 0.01 mg/L for atracurium, 0.005 mg/L for laudanosine [Simmonds 1985]. Disposition in the Body After IVadministration, it is inactivated in plasma by Hofmann elimination, a non-enzymatic breakdown process occurring at physiological pH and temperature, to produce laudanosine and other metabolites. It also undergoes ester hydrolysis by non-specific plasma esterases. The metabolites have no neuromuscular blocking activity although laudanosine may have some stimulatory action on the CNS. Excreted in urine and bile, mostly as metabolites. It crosses the placenta. Therapeutic Concentration The serum therapeutic concentration range is 0.1 to 1.0 mg/L. A study in 20 male and female subjects undergoing major surgery, with a mean age of 59.4 years, found that 90% paralysis was maintained by a mean infusion rate of 4.25 mg/kg/min of atracurium (mean plasma concentration 1.13 mg/L) [Beemer et al. 1990]. A study in 20 subjects on mechanical ventilation receiving atracurium infusion found that stable neuromuscular blockade was associated with a plasma-atracurium concentration of 1.5 mg/L, but this had increased to 5.3 mg/L at 38 h. Plasma-laudanosine concentrations were measured in 6 of the subjects and the maximum concentrations measured ranged from 1.9 to 5.1 mg/L [Yate et al. 1987]. An atracurium infusion was given for 71 days to a 35-year-old IV drug abuser who contracted tetanus. The mean rate of infusion over the period was 1.3 mg/kg/h. There was no evidence of accumulation of laudanosine as plasma concentration at the end of the infusion was 0.985 mg/L. The plasmaatracurium concentration was 1.5 mg/L [Peat et al. 1988]. Toxicity A 59-day-old infant weighing 2.75 kg received 37 mg of atracurium over 75 min when the syringe driver was set up incorrectly. The infant made a full recovery by 135 min after the infusion was stopped. It was estimated that the plasma-laudanosine concentration at the time neuromuscular function recovered was about 19 mg/L. No atracurium was detectable in the plasma sample taken [Charlton et al. 1989]. A 3-week-old ventilated neonate weighing 3.9 kg received a bolus overdose of atracurium of 5.1 mg/kg in error. Intense bronchospasm occurred, systolic blood pressure fell to 28 mmHg and pulse rate increased from 140/min to 180/ min. The neonate recovered within 3 h following treatment with adrenaline and ventilatory support [Durcan, Carter 1986]. Half-life Plasma, atracurium 20 min; laudanosine 3 h. Volume of Distribution 0.16 L/kg. Clearance Plasma, 4.4 to 6.5 mL/min/kg. Protein Binding 80% Dose Initially 300 to 600 mg/kg of atracurium besilate intravenously with subsequent doses of 100 to 200 mg/kg as necessary; 5 to 10 mg/kg/min has been given as a continuous IV infusion.
Chemical Properties Colourless crystals. Mp 171 to 174 . Practically insoluble in water; soluble 1 in 20 of chloroform, 1 in 80 of ether and 1 in 55 of methanol. pKa 1.7. Log P (octanol/water), 2.6. Thin-layer Chromatography System TA—Rf 0.77; system TAB—Rf 0.04; system TAC—Rf 0.08. Gas Chromatography System GA—atrazine RI 1714, M (desethyl-) RI 1680, M (desethyl-deschloro-methoxy-) RI 1670; system GK—RRT 0.79(relative to caffeine). High Performance Liquid Chromatography System HY—RI 401; system HAA—retention time 18.2 min; system HAO—k 10.75; system HAP—k 1.24. Ultraviolet Spectrum Ethanol—263 nm (A11¼195b).
Infrared Spectrum Principal peaks at wavenumbers 1539, 1612, 1295, 804, 1161, 1121 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 43, 58, 44, 200, 68, 215, 41, 42. Quantification Urine HPLC Limits of detection, 0.2 to 0.5 mg/L for atrazine and other pesticide metabolites [Baker et al. 2000]. Limit of detection, 0.5 mg/L for atrazine metabolites [Beeson et al. 1999]. For atrazine metabolites, see Buchholz et al. [1999]. Note For an ELISA for the quantification of atrazine and its major metabolites, see Lucas et al. [1993]. Disposition in the Body Toxicity In a fatality involving the ingestion of 500 mL of a herbicide mix containing atrazine (100 g), aminotriazole (25 g), ethylene glycol (25 g), and formaldehyde (0.15 g), the postmortem tissue concentrations of atrazine were reported to be: liver 32.04 mg/g, pancreas 30.99 mg/g, small intestine mg/g, kidney 97.62 mg/g, lung 79.53 mg/g, heart 15.27 mg/g, muscle 19.93 mg/g. At the time of death, about 3 days after ingestion, the plasma atrazine concentration was 1.49 mg/L [Pommery et al. 1993]. Baker SE et al. (2000). Quantification of selected pesticide metabolites in human urine using isotope dilution high-performance liquid chromatography/tandem mass spectrometry. J Expo Anal Environ Epidemiol 10: 789–798.
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:35:13
Atropine
933
Beeson MD et al. (1999). Isotope dilution high-performance liquid chromatography/tandem mass spectrometry method for quantifying urinary metabolites of atrazine, malathion, and 2,4dichlorophenoxyacetic acid. Anal Chem 71: 3526–3530. Buchholz BA et al. (1999). HPLC-accelerator MS measurement of atrazine metabolites in human urine after dermal exposure. Anal Chem 71: 3519–3525. Lucas AD et al. (1993). Determination of atrazine metabolites in human urine: development of a biomarker of exposure. Chem Res Toxicol 6: 107–116. Pommery J et al. (1993). Atrazine in plasma and tissue following atrazine-aminotriazole-ethylene glycol-formaldehyde poisoning. J Toxicol Clin Toxicol 31: 323–331.
A
Atropine Anticholinergic C17H23NO3 = 289.4 CAS—51-55-8 IUPAC Name [(1R,5S)8-Methyl-8-azabicyclo[3.2.1]octan-3-yl] 3-hydroxy-2phenylpropanoate Synonyms ()-Hyoscyamine; (3-endo)-8-methyl-8-azabicyclo[3.2.1]oct-3-yla-(hydroxymethyl)benzene acetate; (1R,3R,5S,8R)-tropan-3-yl (RS)-tropate. Proprietary Name Atropinol. Atropine and its salts are ingredients in and also used as adjuncts in many proprietary preparations [Sweetman 2007].
Mass Spectrum Principal ions at m/z 124, 82, 94, 83, 42, 96, 103, 67.
Chemical Properties An alkaloid obtained from Duboisia spp. and other solanaceous plants, or prepared by synthesis. Colourless crystals or white crystalline powder. Mp 114 to 118 . Soluble 1 in 455 of water, 1 in 50 of boiling water, 1 in 2 of ethanol, 1 in 1 of chloroform, 1 in 25 of ether and 1 in 27 of glycerol. pKa 9.9 (20 ). Log P (octanol), 1.8. Extraction yield (chlorobutane), 0.6 [Demme et al. 2005]. Atropine Sulfate (C17H23NO3)2,H2SO4,H2O = 694.8 CAS—55-48-1 (anhydrous); CAS—5908-99-6 (monohydrate). Proprietary Names AtroPen; Atropisol; Atropocil; Atropt; Atrospan; Liotropina;
Ocu-Tropine; Sal-Tropine. Chemical Properties Colourless crystals or white crystalline powder. It effloresces in dry air. Mp 190 , with decomposition, after drying at 135 for 15 min. Soluble 1 in 0.4 of water and 1 in 5 of ethanol; practically insoluble in chloroform and ether. Colour Test Liebermann’s reagent—red-orange. Thin-layer Chromatography System TA—Rf 0.18; system TB—Rf 0.05; system TC—Rf 0.03; system TE—Rf 0.24; system TL—Rf 0.01; system TAE—Rf 0.05; system TAF—Rf 0.28 (acidified iodoplatinate solution—positive; Dragendorff spray—positive; Marquis test—pink). Gas Chromatography System GA—atropine RI 2190, M (-CH2O) RI 1980, M (-H2O) RI 2085, M (-AC) RI 2275; system GB—atropine RI 2293, M (-CH2O) RI 2051, M (-H2O) RI 2250; system GF—RI 2660. High Performance Liquid Chromatography System HA—k 3.9 (tailing peak); system HX—RI 306; system HY—RI 251; system HZ—RT 2.2 min; system HAA—RT 10.4 min. Ultraviolet Spectrum Aqueous acid—252, 258 (A11¼ 6.3 c), 264 nm. No alkaline shift.
Infrared Spectrum Principal peaks at wavenumbers 1720, 1035, 1153, 1163, 1063, 1204 cm1 (KBr disk).
Quantification Blood GC-MS Column: cross-linked methyl silicone capillary (25 m 0.2 mm i.d., 0.33 mm). Temperature programme: 100 to 240 at 20 /min. EI ionisation, SIM acquisition mode. Retention time: 1.7 min. Limit of quantification, 10 mg/L [Saady, Poklis 1989]. LC-MS Column: LiChroCART LiChrospher 60 RP-select B. Mobile phase: 0.1% formic acid in water and in acetonitrile. APCI. Atropine, fentanyl and scopolamine. Limit of quantification, 0.9 mg/L, limit of detection, 0.6–0.7 mg/L [Skulska et al. 2007]. CE Capillary: 50 cm. Buffer: borate-phosphate (pH 9.2) with 50 mol/L SDS. Limit of quantification, 0.2 mg/L, limit of detection, 0.06 mg/L for atropine (in presence of strychnine and tetracaine [Plaut, Staub 1998]. Plasma HPLC Column: ODS. Mobile phase: acetonitrile : 50 mmol/L potassium phosphate buffer (pH 3.0) containing 2 mmol/L sodium heptane sulfonate (16 : 84). UV detection (l¼ 220 nm). Limit of quantification, 1 mg/L [Rbeida et al. 2005]. LC-MS Column: Atlantis T3 C18 (150 4.6 mm i.d., 5 mm). Mobile phase: 0.1% formic acid : acetonitrile-water and 0.1% formic acid (80 : 20, 77 : 23 to 62 : 38 at 5 min to 20 : 80 at 6 min for 1 min to 77 : 23 at 7.5 min for 1 min), flow rate 1.0 mL/ min. ESI, positive ion mode, MRM acquisition mode. Limit of quantification, 0.05 mg/L, limit of detection, 0.01 mg/L [John et al. 2010]. Column: XTerraMS C8 (100 2.1 mm i.d., 3.5 mm). Mobile phase: 2 mmol/L formate buffer (pH 3) : acetonitrile (99 : 1 for 2 min to 98 : 2 at 3 min to 5 : 95 at 3.1 min for 7.5 min to 99 : 1 at 8 min until 11 min), flow rate 0.2 mL/min. ESI, positive ion mode, MRM acquisition mode. Limit of quantification, 0.25 mg/L, limit of detection, 0.075 mg/L [Abbara et al. 2008]. Column: C8 Superspher 60 RP select B (125 2 mm i.d.). Mobile phase: 50 mmol/L ammonium formate (pH 3.5) : acetonitrile (90 : 10 for 2 min to 20 : 80 at 5 min for 2 min to 90 : 10 at 7.01 min until 10 min), flow rate 0.4 mL/min for 2 min to 0.6 mL/min at 2.01 min to 0.4 mL/min at 7.01 min. APCI or ESI. Limit of quantification, 5 mg/L (APCI) and 0.1 mg/L (ESI) [Beyer et al. 2007]. Radioimmunoassay Limit of detection, 2.5 mg/L for atropine and hyoscyamine [Virtanen et al. 1980]. Serum GC-MS Column: HP-5MS (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 0.8 mL/min. Temperature programme: 50 for 1 min to 300 at 20 /min for 5 min. EI ionisation at 70 eV, SIM acquisition mode. Limit of detection, 5 mg/L for tropane alkaloids [Namera et al. 2002]. LC-MS Column: Hypurity C18 (50 2.1 mm i.d., 5 mm). Mobile phase: aqueous buffer (10 g/L ammonium acetate, 15 mL/L acetic acid and 2 mL/L trifluoroacetic anhydride) : water : acetonitrile (5 : 95 : 0 for 1 min to 5 : 0 : 95 at 5 min to 5 : 95 : 0 at 5.5 min), flow rate 0.2 mL/min. ESI, positive ion mode, SRM acquisition mode. Limit of quantification, 5 mg/L [Boermans et al. 2006]. Radioimmunoassay See Plasma [Virtanen et al. 1980]. Urine GC-MS See Serum [Namera et al. 2002]. LC-MS See Blood [Skulska et al. 2007]. See Serum [Boermans et al. 2006]. Stomach Contents CE See Blood [Plaut, Staub 1998].
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:35:18
934
A
Atropine Methobromide
Biological Specimens HPLC Column: TSK gel ODS-120A (250 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : water (2 : 8) containing 6 mmol/L phosphoric acid (pH 2.7), flow rate 1.0 mL/min. UV detection (l¼ 215 nm). Limit of detection, 8.5 mg/L [Okuda et al. 1991]. Disposition in the Body Atropine is readily absorbed from mucous membranes, skin and the gastrointestinal tract but not from the stomach. Approximately 80–90% of a dose is excreted in the urine in 24 h, 50% of the dose as unchanged drug, 40 h. Volume of Distribution 23 to 31 L/kg. Clearance Plasma clearance, 630 mL/min. Protein Binding Decreasing from 51% at 0.02 to 0.05 mg/L to 7% at 1 mg/L. Note For reviews of azithromycin, see Hopkins [1991], Peters et al. [1992] and Lalak, Morris [1993]. Dose Usually 500 mg daily. Bergan T et al. (1992). Azithromycin pharmacokinetics and penetration to lymph. Scand J Infect Dis Suppl 83: 15–21. Hopkins S (1991). Clinical toleration and safety of azithromycin. Am J Med 91: 40S–45S. Lalak NJ, Morris DL (1993). Azithromycin clinical pharmacokinetics. Clin Pharmacokinet 25: 370–374. Nahata MC et al. (1993). Pharmacokinetics of azithromycin in pediatric patients after oral administration of multiple doses of suspension. Antimicrob Agents Chemother 37: 314–316. Peters DH et al. (1992). Azithromycin. A review of its antimicrobial activity, pharmacokinetic properties and clinical efficacy. Drugs 44: 750–799. Riedel KD et al. (1992). Equivalence of a high-performance liquid chromatographic assay and a bioassay of azithromycin in human serum samples. J Chromatogr 576: 358–362. Sastre Toran˜o J, Guchelaar HJ (1998). Quantitative determination of the macrolide antibiotics erythromycin, roxithromycin, azithromycin and clarithromycin in human serum by high-performance liquid chromatography using pre-column derivatization with 9-fluorenylmethyloxycarbonyl chloride and fluorescence detection. J Chromatogr B Biomed Sci Appl 720: 89–97. Shepard RM et al. (1991). High-performance liquid chromatographic assay with electrochemical detection for azithromycin in serum and tissues. J Chromatogr 565: 321–337. Taninaka C et al. (2000). Determination of erythromycin, clarithromycin, roxithromycin, and azithromycin in plasma by high-performance liquid chromatography with amperometric detection. J Chromatogr B Biomed Sci Appl 738: 405–411.
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:35:51
Aztreonam
Azlocillin Antibacterial C20H23N5O6S = 461.5 CAS—37091-66-0 IUPAC Name (6R)-6-[D-2-(2-Oxoimidazolidine-1-carboxamido)-2-phenylacetamido]penicillin Synonym Bay e 6905
Azlocillin Sodium C20H22N5NaO6S = 483.5 CAS—37091-65-9 Proprietary Names Azlin; Securopen. Chemical Properties A white to pale yellow hygroscopic powder. It is freely
soluble in water; soluble in dimethyl formamide and methanol; slightly soluble in ethanol and isopropanol; practically insoluble in acetone, chloroform and ether. Ultraviolet Spectrum Water—257, 320 nm (sodium salt).
Infrared Spectrum Principal peaks at wavenumber 1722, 1606, 1532, 1772, 1272, 3295 cm1 (sodium salt) (KBr disk).
941
Quantification Plasma HPLC UV detection (l¼220 nm). Limit of detection, 0.1 mg/L [Kn€ oller et al. 1988]. UV detection (l¼220 nm). Limit of quantification, 5 mg/mL, limit of detection, 1.3 mg/mL [Hildebrandt, Gundert-Remy 1982]. UV detection. Limit of detection, 0.7 mg/L [Gundert-Remy and De Vries 1979]. Serum HPLC See Plasma [Kn€ oller et al. 1988]. UV detection (l¼214 nm). Limit of detection, 0.1 mg/L [Jehl et al. 1987]. UV detection. Limit of detection, 0.4 mg/L [Weber et al. 1983]. Urine HPLC See Plasma [Kn€ oller et al. 1988]. See Serum [Jehl et al. 1987]. UV detection. Limit of detection, 1 to 2 mg/L, azlocillin, 5 to 7 mg/L penicilloic acid metabolite [Gau, Horster 1979]. Bile HPLC See Serum [Jehl et al. 1987]. Tissues HPLC See Plasma [Kn€ oller et al. 1988]. Disposition in the Body Azlocillin is poorly absorbed from the gastro-intestinal tract but is widely distributed following IV administration in body tissues and fluids. It crosses the placenta and small amounts are distributed into breast milk. There is little diffusion into CSF unless meninges are inflamed. It is metabolised to a limited extent and about 50 to 70% of a dose is excreted unchanged in the urine by glomerular filtration and tubular secretion. This occurs within 24 h of administration, therefore, producing high urinary concentrations. It is partly excreted in bile. Concomitant administration of probenecid increases plasma concentrations. It can be removed by haemodialysis. Therapeutic Concentration Following administration of 4 g as an IV infusion over 20 min to 12 healthy male and female subjects, the mean peak plasma concentration was 451 mg/L. This decreased to 223 mg/L after 30 min and 33 mg/L 4 h from the beginning of infusion [Colaizzi et al. 1986]. Half-life Plasma half-life, 1 h; 2 to 6 h in renal impairment. Volume of Distribution 0.22 L/kg. Clearance Serum clearance, 153 mL/min/1.73 m2. Protein Binding Between 20 and 46%. Note For a review of the pharmacokinetics of azlocillin, see Bergan [1983]. Dose Up to 5 g (of the base) intravenously every 8 h; reduced in patients with renal dysfunction. Bergan T (1983). Review of the pharmacokinetics and dose dependency of azlocillin in normal subjects and patients with renal insufficiency. J Antimicrob Chemother 11: 101–114. Colaizzi PA et al. (1986). Comparative pharmacokinetics of azlocillin and piperacillin in normal adults. Antimicrob Agents Chemother 29: 938–940. Gau W, Horster FA (1979). [High pressure liquid chromatographic analysis of azlocillin and its penicilloate in urine]. Arzneimittelforschung 29: 1941–1943. Gundert-Remy U, De Vries JX (1979). Determination of the ureidopenicillins azlocillin, mezlocillin and bay K 4999 in plasma by high performance liquid chromatography. Br J Clin Pharmacol 8: 589–592. Hildebrandt R, Gundert-Remy U (1982). Improved procedure for the determination of the ureidopenicillins azlocillin and mezlocillin plasma by high-performance liquid chromatography. J Chromatogr 228: 409–412. Jehl F et al. (1987). Hospital routine analysis of penicillins, third-generation cephalosporins and aztreonam by conventional and high-speed high-performance liquid chromatography. J Chromatogr 413: 109–119. Kn€ oller J et al. (1988). Application of high-performance liquid chromatography of some antibiotics in clinical microbiology. J Chromatogr 427: 257–267. Weber A et al. (1983). High-pressure liquid chromatographic quantitation of azlocillin. Antimicrob Agents Chemother 24: 750–753.
Aztreonam Antibacterial C13H17N5O8S2 = 435.4 CAS—78110-38-0 IUPAC Name [2S-[2a,3b(Z)]]-2-[[[1-(2-Amino-4-thiazolyl)-2-[(2-methyl-4oxo-1-sulfo-3-azetidinyl)amino]-2-oxoethylidene]amino]oxy]-2-methylpropanoic acid Synonyms Azthreonam; SQ-26776. Proprietary Names Azactam; Azonam; Aztreon; Nebactam; Primbactam; Urobactam.
Chemical Properties A white crystalline odourless powder. Decomposes at 227 . It is very slightly soluble in dehydrated alcohol; slightly soluble in methanol; soluble in dimethylformamide and dimethylsulfoxide; practically insoluble in chloroform, ethyl acetate and toluene. pKa, less than 2.0. Aztreonam Disodium C13H15N5Na2O8S2 = 479.4
A
Clarke's Analysis of Drugs and Poisons Chapter No. A Dated: 15/3/2011 At Time: 21:36:1
942
A
Aztreonam
Ultraviolet Spectrum Aqueous acid (ethanol)—268, 281 nm; basic—289 nm.
Infrared Spectrum Principalpeaksatwavenumber1640,1041,633 cm1 (KBrdisk).
Quantification Serum HPLC UV detection. Limit of detection, 0.5 mg/L [Jehl et al. 1987]. UV detection. Limit of detection, 1 mg/L [Mihindu et al. 1983]. UV detection (l¼293 nm). Limit of detection, 1 mg/L [Pilkiewicz et al. 1983]. UV detection. Limit of detection, 1 mg/L [Swabb et al. 1981].
Urine HPLC See Serum [Jehl et al. 1987]. See Serum [Mihindu et al. 1983]. UV detection (l¼293 nm). Limit of detection, 5 mg/L [Pilkiewicz et al. 1983]. UV detection. Limit of detection, 5 mg/L [Swabb et al. 1981]. Bile HPLC See Serum [Jehl et al. 1987]. Faeces HPLC UV detection. Limit of detection, about 2 mg/g [Ehret et al. 1987]. Bioassay Limit of detection, 0.03 mg/L [Ehret et al. 1987]. Disposition in the Body Aztreonam is completely absorbed after IM administration, with peak concentrations reached within 1 h. It takes 1 to 3 h for equilibrium to be reached between tissues and plasma. It is widely distributed in body tissues and fluids, including bile, but diffusion into CSF is poor unless meninges are inflamed. It crosses the placenta and small amounts are distributed into breast milk. Only about 6 to 16% is metabolised to inactive metabolites by hydrolysis. The drug is excreted predominantly in urine by renal tubular secretion and glomerular filtration; about 60 to 75% of a dose appears within 8 h as the unchanged drug with only small quantities of metabolites. Only small amounts of the unchanged drug and metabolites are excreted in faeces. It is removed by haemodialysis and to a lesser extent by peritoneal dialysis. Therapeutic Concentration The trough serum therapeutic concentration range is 1 to 10 mg/L and the peak, 50 to 250 mg/L. Twenty-four male volunteers were divided into 4 groups according to the degree of creatinine clearance. Group 1 (mean age, 27 years): creatinine clearance, 80 mL/min, group 2 (mean age, 43): 30 to 79 mL/min, group 3 (mean age, 44): 10 to 29 mL/min, group 4 (mean, 40 years old): 350 .
Non-hygroscopic. Freely soluble in water and isotonic saline; sparingly soluble in methanol and ethanol; practically insoluble in organic solvents. Mass Spectrum Principal ions at m/z 30, 138, 195, 140, 103, 197, 77, 196.
Quantification Plasma GC ECD. Limit of detection, 50 mg/L [Degen, Riess 1976]. HPLC Electrochemical detection. Limit of quantification, 10 mg/L, limit of detection, 2.5 mg/L [Millerioux et al. 1996]. Serum GC–MS Limit of detection, 5 mg/L [Swahn et al. 1979]. Urine GC See Plasma [Degen, Riess 1976]. Cerebrospinal Fluid GC–MS See Serum [Swahn et al. 1979]. Disposition in the Body Baclofen is rapidly absorbed after oral administration. About 80% of a dose is excreted in the urine in 24 h, mostly as unchanged drug. About 15% of a dose is metabolised, mainly to the deaminated derivative; about 20% of a dose may be eliminated in the faeces as unchanged drug and metabolites. Therapeutic Concentration A 45-year-old, healthy, woman administered 20 mg baclofen orally produced a peak plasma concentration of 270 mg/L after 2.5 h [Tosunoglu, Ersoy 1995]. After an oral dose of 40 mg to 1 subject, a peak plasma concentration of unchanged drug of about 0.6 mg/L was attained in 2 h; a peak concentration of metabolites of about 0.1 mg/L was attained in about 4 h [Faigle, Keberle 1972]. Toxicity Recovery has occurred after the ingestion of 1.5 g. A fatality occurred after ingestion of baclofen. Baclofen was detected in serum at a concentration of 17 mg/L and in urine at 760 mg/L which was collected 12 h after ingestion of the drug [Fraser et al. 1991]. Coma, respiratory failure and severe seizures occurred in a 39-year-old female subject after ingestion of 450 mg of baclofen; following treatment, the patient regained consciousness within 36 h, at which time a plasma concentration of 0.2 mg/L was reported; the plasma half-life was found to be 35 h [Ghose et al. 1980]. Half-life Plasma half-life, about 2 to 4 h. Protein Binding About 30%. Dose 15 to 60 mg daily; maximum of 100 mg daily. Degen PH, Riess W (1976). The determination of gamma-amino-beta-(p-chlorophenyl)butyric acid (baclofen) in biological material by gas-liquid chromatography. J Chromatogr 117: 399–405. Faigle JW, Keberle H (1972). The chemistry and kinetics of Lioresal. Postgrad Med J 48: 9–13. Fraser AD et al. (1991). Toxicological analysis of a fatal baclofen (Lioresal) ingestion. J Forensic Sci 36 (5): 1596–1602. Ghose K et al. (1980). Complications of baclofen overdosage. Postgrad Med J 56: 865–867. Millerioux L et al. (1996). High-performance liquid chromatographic determination of baclofen in human plasma. J Chromatogr A 729: 309–314. Swahn CG et al. (1979). Mass fragmentographic determination of 4-amino-3-p-chlorophenylbutyric acid (baclofen) in cerebrospinal fluid and serum. J Chromatogr 162: 433–438. Tosunoglu S, Ersoy L (1995). Determination of baclofen in human plasma and urine by highperformance liquid chromatography with fluorescence detection. Analyst 120: 373–375.
Balsalazide Mesalamine, Antiinflammatory C17H15N3O6 = 357.3 CAS—80573-04-2 IUPAC Name (3Z)-3-[[4-(2-Carboxyethylcarbamoyl)phenyl]hydrazinylidene]6-oxocyclohexa-1,4-diene-1-carboxylic acid
Disposition in the Body Balsalazide is metabolised in the colon by bacterial azoreductases to 5-aminosalicyclic acid (5-ASA). 5-ASA is responsible for the antiinflammatory action, and 4-aminobenzoyl-b-alanine (4-ABA) is an inert carrier. Twenty-five percent of the released 5-ASA is inactivated in the colonic mucosa and liver to the N-acetylated metabolite NASA. 4-ABA is also converted to its Nacetylated metabolite NABA. Only NASA and NABA are detected in urine; the parent drug is excreted mainly in the faeces. Therapeutic Concentration Twenty healthy male and female volunteers were administered 2.25 g balsalazide (equivalent to 800 mg 5-ASA). A mean peak plasma 5-ASA concentration of 348 mg/L was reached at 9.1 h [Sandborn et al. 2004]. Fifty-four adult patients with ulcerative colitis (in remission) were administered with 3 to 6 g daily for at least 1 year. Peak plasma concentrations of 0.324 mg/L were reached within 2 h. [Green et al. 1998]. Peak plasma concentrations of the metabolite 4-ABA were 99%.
Infrared Spectrum Principal peaks at wavenumbers 1694, 757, 1587, 1162, 1204, 1219 cm1. Mass Spectrum Principal ions at m/z 230, 109, 82, 187, 243, 363, 42, 123.
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:29:54
Benzaldehyde Quantification Plasma GC Column: 2% OV3 or 3% Dexil 300 on HP Chrom W AW DMCS 80/ 100 mesh (1 m 2 mm i.d.). Carrier gas: N2, 35 mL/min. Temperature programme: 230 to 285 at 2 /min to 290 at 3 /min. FID. Limit of detection, 30 mg/L [Quaglio et al. 1982]. HPLC Column: CPS (3 mm). Mobile phase: 0.15 mol/L acetate buffer (pH 4.7) : acetonitrile (75 : 25). Electrochemical detection. Limit of quantification, 2.4 mg/L; limit of detection, 0.8 mg/L [Seiler et al. 1994]. Column: Hypersil-CPS (250 4.6 mm i.d., 5 mm). Mobile phase: 0.15 mol/L acetate buffer (pH 4.7) : acetonitrile (75 : 25), flow rate 1.5 mL/min. Electrochemical detection. Limit of detection 0.5 mg/L [Suss et al. 1991]. Column: C18 Nucleosil (250 4 mm i.d., 5 mm). Mobile phase: acetonitrile : 0.05 mol/L potassium dihydrogen phosphate (pH 2.8; 32 : 68), flow rate 1.0 mL/min. UV detection (l ¼ 254 nm). Retention time: 5.9 min. Limit of detection, 0.5–1 mg/L [Kruger et al. 1984]. Serum HPLC Column: C8 (250 cm 2.6 mm i.d., 10 mm). Mobile phase: 0.12 mol/L acetate buffer (pH 4.2) : acetonitrile (60 : 40), flow rate 1 mL/min. Electrochemical detection. Limit of detection, 0.2 mg/L [Furlanut et al. 1987]. LC-MS Column: Chromolith Speed ROD C18 (50 4.6 mm i.d., 5 mm). Mobile phase: methanol : 5 mmol/L acetic acid (pH 3.9) in ammonia (20 : 80 to 70 : 30 at 4 min for 1 min to 20 : 80), flow rate 1.0 mL/min. ESI, positive ion mode, MRM acquisition mode. Limit of quantification, 0.17 mg/L [Kirchherr, K€ uhn-Velten 2006]. Disposition in the Body Therapeutic Concentration Thirteen schizophrenic patients were orally administered 6 mg benperidol as liquid and 6 mg as tablets. The mean peak plasma concentration was 10.2 mg/L at 1.0 h for the liquid and 7.3 mg/L at 2.7 h for the tablet formulation [Seiler et al. 1994]. Dose 0.25 to 1.5 mg daily.
957
B
Infrared Spectrum Principal peaks at wavenumbers 1500, 1665, 1593, 1195, 1570, 1265 cm1 (KBr disk).
Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Furlanut M et al. (1987). Electrochemical detection of benperidol in serum for drug monitoring in humans. Ther Drug Monit 9: 343–346. Hansch C et al. (1995). Exploring QSAR: Hydrophobic, Electronic, and Steric Constants. Washington DC: American Chemical Society. Kirchherr H, K€ uhn-Velten WN (2006). Quantitative determination of forty-eight antidepressants and antipsychotics in human serum by HPLC tandem mass spectrometry: a multi-level, singlesample approach. J Chromatogr B Analyt Technol Biomed Life Sci 843: 100–113. Kruger R (1984). Determination of benperidol in human plasma by high-performance liquid chromatography. J Chromatogr 311: 109–116. Quaglio MP et al. (1982). Determination of benperidol, droperidol and pimozide in human plasma by GLC. Boll Chim Farm 121: 276–284. Seiler W et al. (1994). Pharmacokinetics and bioavailability of benperidol in schizophrenic patients after intravenous and two different kinds of oral application. Psychopharmacology (Berl) 116: 457–463. Suss S et al. (1991). Determination of benperidol and its reduced metabolite in human plasma by high-performance liquid chromatography and electrochemical detection. J Chromatogr 565: 363–373.
Benserazide Dopa-Decarboxylase Inhibitor, Antiparkinsonian C10H15N3O5 = 257.2 CAS—322-35-0 IUPAC Name DL-Serine 2-[(2,3,4-trihydroxyphenyl)methyl] hydrazide
Mass Spectrum Principal ions at m/z 60, 42, 88, 30, 31, 43, 29, 70.
Synonym Serazide
Chemical Properties Extraction yield (chlorobutane), 0 [Demme et al. 2005]. Benserazide Hydrochloride C10H15N3O5,HCl = 293.7 CAS—14919-77-8; 14046-64-1 Proprietary Names It is an ingredient of Madopar and Prolopa. Chemical Properties An off-white crystalline powder. Mp 146 to 148 . Soluble
1 in 3 of water, 1 in 118 of ethanol, 1 in 66 of acetone, 1 in 180 of chloroform and 1 in 455 of ether. Colour Tests Ammoniacal silver nitrate—black; p-dimethyl-aminobenzaldehyde—red/-; ferric chloride—green-brown; Folin-Ciocalteu reagent—blue; methanolic potassium hydroxide—red; Millon’s reagent—red-orange; Nessler’s reagent—black; palladium chloride—orange!brown; potassium dichromate— brown. Thin-layer Chromatography System TA—Rf 0.01; system TB—Rf 0.00; system TC—Rf 0.01; system TL—Rf 0.03; system TAE—Rf 0.07. High Performance Liquid Chromatography System HX—RI 35. Ultraviolet Spectrum Aqueous acid—203, 269 nm.
Disposition in the Body Rapidly absorbed after oral administration and appears to be rapidly metabolised. About 50 to 60% of an oral dose and 80% of an IV dose is excreted in the urine in 24 h, and about 30% and 10% of the respective doses are eliminated in the faeces in 7 days. Therapeutic Concentration After an oral dose of 50 mg of 14C-benserazide hydrochloride, peak plasma concentrations of about 1 mg/L were attained in 1 h [Schwartz et al. 1974]. Dose Usually the equivalent of 50 mg of benserazide daily, increasing to 100 to 250 mg daily (given in combination with levodopa in the treatment of parkinsonism). Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Schwartz DE et al. (1974). Pharmacokinetics of the decarboxylase inhibitor benserazide in man; its tissue distribution in the rat. Eur J Clin Pharmacol 7: 39–45.
Benzaldehyde Flavouring Agent C6H5CHO = 106.1 CAS—100-52-7
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:30:1
958
Benzalkonium Bromide
Synonym Artificial essential oil of almond
Proprietary Names Benzalchlor-50; Cetal Conc. A and B; Empigen BAC; Hyamine 3500; Laudamonium; Morpan BC; Ovules Pharmatex; Quartamon; Roccal; Sabol; Silquat B10; Silquat B50; Vantoc CL; Zephiran. It is an ingredient of Polycide, Stomosol and Timodine.
B Chemical Properties A clear, colourless, strongly refractive liquid, with a characteristic odour of bitter almonds. It becomes yellowish on storage and oxidises in air to benzoic acid. Bp about 179 . Refractive index, at 20 , 1.544 to 1.5465. Soluble 1 in 350 of water; miscible with ethanol and ether. Log P (octanol/water), 1.5. Gas Chromatography System GA—RI 947; system GI—retention time 34.2 min. Ultraviolet Spectrum Ethanol—245 (A11¼1294b), 279 (A11¼126b), 289 nm (A11¼105b).
Chemical Properties A white or yellowish-white amorphous powder, thick gel or gelatinous pieces. A solution in water gives a clear, colourless or pale yellow, syrupy liquid, which foams strongly when shaken. Very soluble in water, ethanol and acetone; practically insoluble in ether. Thin-layer Chromatography System TL—Rf 0.00; system TAE—Rf 0.04; system TAF—Rf 0.66. Ultraviolet Spectrum Water—256 (A11¼4.9b), 262 (A11¼5.8b), 268 nm (A11¼4.6b). Infrared Spectrum Principal peaks at wavenumbers 702, 726, 780, 875, 1618, 1211 cm1 (thin film).
Benzamine Piperidine, Anaesthetic (Local) C15H21NO2 = 247.3 CAS—500-34-5 IUPAC Name 4-Benzoyloxy-2,2,6-trimethylpiperidine Synonyms Betacaine; racemic benzamine. Infrared Spectrum Principal peaks at wavenumbers 1700, 1210, 1601, 1175, 1318, 753 cm1 (thin film). Mass Spectrum Principal ions at m/z 77, 106, 105, 51, 50, 78, 52, 39.
Quantification Blood GC For detection and identification of volatile compounds using headspace analysis and FID, see Ramsey and Flanagan [1982]. Serum GC–MS For head-space analysis, see Zlatkis et al. [1974]. Ramsey JD, Flanagan RJ (1982). Detection and identification of volatile organic compounds in blood by headspace gas chromatography as an aid to the diagnosis of solvent abuse. J Chromatogr 240: 423–444. Zlatkis A et al. (1974). Analysis of trace volatile metabolites in serum and plasma. J Chromatogr 91: 379–383.
Benzalkonium Bromide Quaternary Ammonium, Antiseptic CAS—8001-54-5 Proprietary Name Morpan BB Chemical Properties A mixture of alkylbenzyldimethylammonium bromides of the general formula [C6H5CH2N(CH3)2R]Br in which R represents a mixture of the alkyls from C8H17 to C18H37. An aqueous solution containing the equivalent of 50% of [C6H5CH2N(CH3)2C13H27]Br is a colourless or pale-yellow syrupy liquid, which is miscible with water, ethanol, and acetone. Note See Benzalkonium Chloride.
Chemical Properties Extracted by organic solvents from aqueous alkaline solutions. Log P (octanol/water) 2.95 [Meylan, Howard 1995]. Benzamine Hydrochloride Synonyms Beta-eucaine hydrochloride; eucaine hydrochloride. Chemical Properties White crystalline powder. Mp 274 , with decomposition. Soluble 1 in 30 of water, 1 in 35 of ethanol and 1 in 30 of chloroform. Benzamine Lactate
Chemical Properties White crystalline powder. Mp 152 . Soluble 1 in 5 of water and 1 in 8 of ethanol.
Thin-layer Chromatography System T1—Rf 0.57 (location reagent acidified iodoplatinate spray, positive reaction). Ultraviolet Spectrum Aqueous acid (0.1 N sulfuric acid)—233, 274 nm. Infrared Spectrum Principal peaks at wavenumbers 1714, 1276, 711 or 1108 cm1 (KBr disk). Toxicity The minimum IV lethal dose in rats is 15 to 25 mg/kg. Dose Up to 30 mg. Meylan WM, Howard PH (1995). Atom/fragment contribution method for estimating octanolwater partition coefficients. J Pharm Sci 84: 83–92.
Benzathine Benzylpenicillin Benzalkonium Chloride Cationic Disinfectant, Antiseptic CAS—8001-54-5 Note A mixture of alkylbenzyldimethylammonium chlorides of the general formula [C6H5CH2N(CH3)2R]Cl, in which R represents a mixture of the alkyls from C8H17 to C18H37.
Natural Penicillin, Antibiotic C16H20N2(C16H18N2O4S)2 = 909.1 CAS—1538-09-6 (anhydrous benzathine benzylpenicillin); 5928-83-6 (benzathine benzylpenicillin monohydrate); 41372-02-5 (benzathine benzylpenicillin tetrahydrate) IUPAC Name N, N0 -Dibenzylethylenediammonium bis[(6R)-6-(2-phenylacetamido)penicillanate]
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:30:9
Benzatropine Synonyms Benzathine benzylpenicillin; benzathine penicillin G; benzethacil; dibenzylamine penicillin G; penzaethinum G. Proprietary Names Benzatron; Benzetacil; Bepeben; Bicillin; Dibencil; Extencilline; Galtamicina; Longacilin; Neolin; Penadur; Pen di Ben; Penditan; Pendysin; Penadur; Penidural; Permapen; Retarpen. It is also an ingredient in Bicillin C-R; Pecivax; Tardocillin.
959
han L et al. (1993). Penicillin levels following the administration of benzathine penicillin G in pregnancy. Obstet Gynecol 82: 338–342.
Benzatropine Dopamine Uptake Inhibitor, Anticholinergic C21H25NO = 307.4 CAS—86-13-5 IUPAC Name (1R,5R)-3-[Di(phenyl)methoxy]-8-methyl-8-azabicyclo[3.2.1] octane Synonyms Benztropine; 3-(diphenylmethoxy)-8-methyl-8-azabicyclo[3.2.1] octane.
Chemical Properties White powder. Mp 129 to 133 with decomposition. Soluble 1 in 6000 of water and 1 in 1000 of ethanol; freely soluble in dimethylformamide and in formamide; almost insoluble in ether and chloroform. The tetrahydrate is a white, odourless, crystalline powder. Soluble 1 in 5000 of water and 1 in 65 of alcohol. Dissolve 50 mg of dehydrate alcohol in 50 mL of water, the pH will be between 4.0 and 6.5. High Performance Liquid Chromatography Column: Hypersil C18 (100 4.6 mm i.d., 5 mm). Mobile phase: phosphate buffer (4.32 g/L potassium hydrogen phosphate; 6.30 g/L sodium heptane-1-sulfonate) : acetonitrile (pH 3.17; 71.5 : 28.5), flow rate 1.0 mL/min. UV detection (l ¼ 258 nm). Retention times: penicillin G 4 min, benzathine 8 min. Limit of quantification not reported [Irwin et al. 1984]. Ultraviolet Spectrum Methanol—251, 257, 263, 266, 320 nm. Disposition in the Body Relatively stable in the presence of gastric juice or serum, and penicillin is slowly released after PO or IM administration. When benzathine benzylpenicillin is given by mouth, the maximum concentration of penicillin in the blood is obtained less rapidly than after a comparable dose of a soluble salt of penicillin by injection, but the level is maintained for a longer period; doses of 225 mg usually give effective blood levels for 6 h. When given by IM injection, benzathine benzylpenicillin forms a depot from which penicillin is released over several days. After a single dose of 450 mg, an effective concentration in the blood may be maintained for 1 week. Distribution into the CSF is reported to be poor. Due to the slow absorption from the injection site, benzylpenicillin has been detected in the urine for up to 12 weeks after a single dose. Therapeutic Concentration In a group of 193 patients receiving a 4-weekly dose of IM benzathine benzylpenicillin (1 200 000 units) for the prevention of secondary rheumatic fever, mean serum concentrations at days 1, 3, 10, 21 and 28 were reported as being 0.17, 0.11, 0.04, 0.03 and 0.02 mg/L, respectively [Kaplan et al. 1989]. Twenty-five healthy pregnant volunteers at 38–39 weeks’ gestation scheduled for elective caesarean delivery were administered benzathine benzylpenicillin (2400000 units) IM preoperatively. Ten women delivered 1 day after injection, 5 delivered 2–3 days after and 10 delivered 7 days after. Penicillin concentrations (mg/L) in compartments of the maternal-placentalfetal unit were reported as follows: Sample
Day 1
Days 2–3
Day 7
Maternal serum Maternal CSF Cord serum Amniotic fluid Maternal/cord serum
0.14 0.005 0.12 0.16 4.03
0.14 0.005 0.04 0.04 19.2
0.08 0.005 0.04 0.03 5.8
[Nathan et al. 1993]
Toxicity Allergic reactions may occur. It should not be injected IV because ischaemic reactions may occur. Dose Benzathine benzylpenicillin has the same antimicrobial action as benzylpenicillin, to which it is hydrolysed gradually after deep IM injection. This results in a prolonged effect but, because of the relatively low blood concentrations of benzylpenicillin produced, its use should be restricted to micro-organisms that are highly susceptible to benzylpenicillin. In acute infections (and if bacteraemia is present), the initial treatment should be with benzylpenicillin by injection. Usually up to 900 mg daily by mouth or up to 1.8 g IM as a single dose. Irwin WJ et al. (1984). Controlled-release penicillin complexes. High-performance liquid chromatography and assay. J Chromatogr 287: 85–96. Kaplan EL et al. (1989). Pharmacokinetics of benzathine penicillin G: serum levels during the 28 days after intramuscular injection of 1,200,000 units. J Pediatr 115: 146–150. Nat-
Chemical Properties Soluble in chloroform. Log P (octanol/water) 4.28 [Meylan, Howard 1995]. Extraction yield (chlorobutane), 1 [Demme et al. 2005]. Benzatropine Mesilate C21H25NO,CH4O3S = 403.5 CAS—132-17-2 IUPAC Name (1R,5R)-3-Benzhydryloxy-8-methyl-8-azabicyclo[3.2.1]octane
methanesulfonic acid Synonyms Benzatropine methanesulfonate; tropine diphenylmethyl ether. Proprietary Names Bensylate; Cogentin. Chemical Properties A white, slightly hygroscopic, crystalline powder. Mp 141 to 145 . Soluble 1 in 0.7 of water, 1 in 1.5 of ethanol, and 1 in 2 of chloroform; practically insoluble in ether. pKa 10.0 (20 ). Log P (octanol/water) 3.91 [Meylan, Howard 1995], (heptane) 0.4. Colour Tests Mandelin’s test—yellow; Marquis test—yellow. Thin-layer Chromatography System TA—Rf 0.13; system TAE—Rf 0.06; system TAG—Rf 0.02; system TB—Rf 0.26; system TC—Rf 0.06 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 2302; system GB—RI 2423. High Performance Liquid Chromatography System HA—k 3.7 (tailing peak). Ultraviolet Spectrum Aqueous acid—253, 259 nm (A11 ¼ 14.5a).
B
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:30:15
960
Benzbromarone
Infrared Spectrum Principal peaks at wavenumbers 1054, 700, 742, 1191, 1204, 1075 cm1 (KBr disk).
B Chemical Properties Yellowish crystals. Mp 151 . Log P (octanol/water), 6.0. Thin-layer Chromatography System TF—Rf 0.49; system TAE—Rf 0.94. High Performance Liquid Chromatography System HX—RI 860; system HAA—retention time 26.1 min. Ultraviolet Spectrum Methanolic acid—237 (A11¼666b), 274 (inflexion), 281 (A11¼314b); aqueous alkali—240 nm (A11¼440b), 281 (inflexion), 355 nm (A11¼513b).
Mass Spectrum Principal ions at m/z 83, 140, 82, 124, 96, 97, 42, 125.
Quantification Blood GC-MS Column: DB-5MS 5% phenyl-methylpolysiloxane capillary (15 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1 mL/min. Temperature programme: 125 to 290 at 20 /min for 5 min. EI ionisation. Limit of detection not reported [Rosano et al. 1994]. Plasma GC-MS Column: Silanised glass with 5% dimethyldichlorosilane in toluene packed with 1.5% OV-17 on 100/200 mesh gas chrom Q (2 mm i.d.). Carrier gas: He, 20.0 mL/min. Temperature: 220 . EI ionisation at 70 eV, full scan mode. Limit of detection, 2 mg/L [Jindal et al. 1981]. HPLC Column: C8 Spherisorb (150 3.9 mm i.d., 5 mm). Mobile phase: 0.15% triethylamine (pH 3) : acetonitrile (40 : 60), flow rate 1.5 mL/min. UV detection (l ¼ 199 nm). Retention time: 7.0 min. Limit of quantification, 0.25 mg/L [Selinger et al. 1989]. Serum GC-MS See Blood [Rosano et al. 1994]. Urine GC-MS See Blood [Rosano et al. 1994]. See Plasma [Jindal et al. 1981]. Disposition in the Body Absorbed after oral administration. Toxicity In a fatality attributed to the ingestion of an unknown quantity of benzatropine tablets, the following postmortem concentrations were reported: blood 0.7 mg/L, liver 1.6 mg/g, and urine 0.8 mg/L. In a second case, a liver concentration of 2.3 mg/g was found [del Villar, Liddy 1976]. A 41-year-old male with a history of schizophrenia and drug abuse was found dead. Benzatropine at a concentration of 0.183 mg/L in his blood and 7.12 mg/L in his urine was found [Rosano et al. 1994]. Dose 0.5 to 6 mg of benzatropine mesilate daily. del Villar G, Liddy M (1976). TIAFT Bull 12: 11–12. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Jindal SP et al. (1981). A stable isotope dilution assay for the antiparkinsonian drug benztropine in biological fluids. Clin Chim Acta 112: 267–273. Meylan WM, Howard PH (1995). Atom/fragment contribution method for estimating octanolwater partition coefficients. J Pharm Sci 84: 83–92. Rosano TG et al. (1994). Benztropine identification and quantitation in a suicidal overdose. J Anal Toxicol 18: 348–353. Selinger K et al. (1989). High-performance liquid chromatographic method for the analysis of benztropine in human plasma. J Chromatogr 491: 248–252.
Benzbromarone Uricosuric C17H12Br2O3 = 424.1 CAS—3562-84-3 IUPAC Name (3,5-Dibromo-4-hydroxyphenyl)(2-ethyl-3-benzofuranyl) methanone Proprietary Names Desuric; Narcaricin; Normurat; Uricovac M.
Quantification Plasma HPLC UV detection. Limit of detection, 0.01 mg/L for benzarone [de Vries et al. 1986]. Serum HPLC UV detection. Limit of detection, 140 mg/L for benzbromarone, 90 mg/L for benzarone [Vergin, Bishop 1980]. Urine HPLC See Plasma [de Vries et al. 1986]. Disposition in the Body Benzbromarone is incompletely absorbed after oral administration. It is metabolised by debromination to bromobenzarone and benzarone which are both active metabolites. About 50% of a dose is excreted in the bile as free and conjugated (glucuronide) metabolites and less than 10% of a dose is excreted in the urine. Therapeutic Concentration Following a single oral dose of 100 mg to 7 subjects, peak plasma concentrations of 1.4 to 2.9 (mean 2.2) mg/L of benzbromarone and 0.6 to 1.2 (mean 0.8) mg/L of benzarone were attained in about 3 and 6 h, respectively [Ferber et al. 1981]. Half-life Plasma half-life, benzbromarone about 3 h, benzarone about 13 h. Note For a review of benzbromarone, see Heel et al. [1977]. Dose Usually 100 mg daily; up to 300 mg daily has been given. de Vries JX et al. (1986). Determination of benzarone in human plasma and urine by high-performance liquid chromatography and gas chromatography-mass spectrometry. Identification of the conjugates. J Chromatogr 382: 167–174. Ferber H et al. (1981). Pharmacokinetics and biotransformation of benzbromarone in man. Eur J Clin Pharmacol 19: 431–435. Heel RC et al. (1977). Benzbromarone: a review of its pharmacological properties and therapeutic use in gout and hyperuricaemia. Drugs 14: 349–366. Vergin H, Bishop G (1980). High-performance liquid chromatographic determination of benzbromarone and the main metabolite benzarone in serum. J Chromatogr 183: 383–386.
Benzene Solvent C6H6 = 78.11 CAS—71-43-2 Synonyms Cyclohexatriene; phenyl hydride.
Chemical Properties A clear, colourless, mobile, inflammable liquid, with a characteristic aromatic odour, which burns with a smoky flame. Bp 80.1 . It solidifies when cooled to 0 . Practically immiscible with water; miscible with dehydrated alcohol, acetone, chloroform, ether and glacial acetic acid. Log P (octanol/ water), 2.1. Gas Chromatography System GA—RI 1000; system GI—retention time 14.8 min.
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:30:25
Benzethonium Chloride High Performance Liquid Chromatography System HAA—retention time 19.5 min. Ultraviolet Spectrum Ethanol—255 nm (A11¼28b).
Mass Spectrum Principal ions at m/z 78, 77, 52, 51, 50, 39, 79, 74. Quantification Blood GC FID. For head-space analysis, see Collom, Winek [1970]. Urine GC FID. Limit of quantification, 23 mg/L, limit of detection, 7 mg/L [Ljungkvist et al. 2001]. FID. Limit of detection, 10 mg/L for phenol [Rick et al. 1982]. HPLC Fluorescence detection (lex¼395 nm; lem¼470 nm). Limit of detection, 1 mg/L [Einig, Dehnen 1995]. SIM acquisition mode. Limit of detection, 0.01 mg/L for trans, trans-muconic acid (metabolite) [Ruppert et al. 1995]. UV detection (l ¼ 265 nm). Limit of detection, 125 ng [Lee et al. 1993]. Tissue GC See Snyder et al. [1977]. See Blood Collom, Winek [1970]. Disposition in the Body Benzene is absorbed from the gastrointestinal tract, through the skin and from the lungs. About 50% of inhaled benzene is retained in the body; eventually up to 50% of the retained benzene may be eliminated via the lungs, only very small amounts (~0.1%) appearing unchanged in the urine. The remainder of the retained benzene is excreted in the urine, mainly as phenol together with small amounts of catechol and quinol. The excretion of phenol is highest in the first 24 h and is complete within 48 h of exposure. The phenolic metabolites are excreted mainly in the conjugated form as ethereal sulfates and glucuronides. Toxicity The maximum permissible atmospheric concentration is 10 ppm or 30 mg/m3. The maximum exposure limit is 1 ppm. Chronic poisoning may result from exposure to low concentrations over a period of time; 1500 ppm for an hour causes marked depression of the CNS and 7500 ppm for half an hour or 20 000 ppm for a few minutes may cause death; chronic inhalation of as little as 100 ppm is likely to cause severe bone marrow depression. The ingestion of 10 mL has caused death. In a death attributed to inhalation of benzene vapours (glue sniffing), the postmortem concentration of benzene in the blood was 0.94 mg/L and in the kidney 5.5 mg/g [Collom, Winek 1970]. The following postmortem tissue concentrations were reported in a fatality due to the ingestion of benzene: blood 38 mg/L, brain 253 mg/g, kidney 21 mg/g, liver 105 mg/g, urine 20 mg/L [Alha et al. 1975]. A 3-year-old child drank some benzene and died 2 h later; postmortem concentrations were: brain 250 mg/g, kidney 20 mg/g, liver 800 mg/g [Heyndrickx et al. 1966]. Half-life Plasma half-life, 9 to 24 h. Note For a review of the effects on health of benzene inhalation, see Haley [1977]. Alha A et al. (1975). Bull Int Assoc Forensic Toxicol 11(3): 9. Collom WD, Winek CL (1970). Detection of glue constitutents in fatalities due to “glue sniffing”. Clin Toxicol 3: 125–130. Einig T, Dehnen W (1995). Sensitive determination of the benzene metabolite S-phenylmercapturic acid in urine by high-performance liquid chromatography with fluorescence detection. J Chromatogr A 697(1–2): 371–375. Haley TJ (1977). Evaluation of the health effects of benzene inhalation. Clin Toxicol 11: 531–548. Heyndrickx A et al. (1966). Distribution of benzene in a fatal child poisoning. J Pharm Belg 21: 406–408. Lee BL et al. (1993). Urinary trans,trans-muconic acid determined by liquid chromatography: application in biological monitoring of benzene exposure. Clin Chem 39(9): 1788–1792. Ljungkvist G et al. (2001). Determination of low concentrations of benzene in urine using multidimensional gas chromatography. Analyst 126(1): 41–45. Rick DL et al. (1982). Determination of phenol and pentachlorophenol in plasma and urine samples by gas liquid chromatography. J Anal Toxicol 6: 297–300. Ruppert T et al. (1995). Determination of urinary trans,trans-muconic acid by gas chromatographymass spectrometry. J Chromatogr B Biomed Sci Appl 666(1): 71–76. Snyder CA et al. (1977). An extraction method for determination of benzene in tissue by gas chromatography. Am Ind Hyg Assoc J 38: 272–276.
Benzene Hexachloride Insecticide C6H6Cl6 = 290.8
961
CAS—319-84-6 (a-isomer); 319-85-7 (b-isomer); 58-89-9 (g-isomer); 31986-8 (d-isomer); 6108-10-7 (e-isomer) Synonyms BHC; HCH; hexachlorocyclohexane; technical benzene hexachloride.
Chemical Properties A mixture of the several isomers of 1,2,3,4,5,6-hexachlorocyclohexane; it contains not less than 12% of the gamma isomer. Benzene hexachloride has 5 known isomers designated alpha, beta, gamma, delta and epsilon; of these only the gamma isomer (see Lindane) is outstandingly active as an insecticide. White to light brown granules, flakes or powder, with a characteristic musty odour. Mp 159 . Practically insoluble in water; its solubility in organic solvents depends on the proportions of the various isomers present. Gas Chromatography System GA—a-isomer RI 1690, b-isomer RI 1710, d-isomer RI 1755, g-isomer RI 1745. Infrared Spectrum Principal peaks at wavenumbers 688, 855, 704, 781, 917, 957 cm1 (Nujol mull). Quantification See under Lindane. Disposition in the Body Absorbed after ingestion, inhalation or through the skin. It is stored in the body fat and adrenal glands. The b-isomer accumulates on chronic exposure (see under Lindane). Toxicity Benzene hexachloride has a greater chronic toxicity than lindane. Ingestion of 20 to 30 g may produce serious toxic effects but death is unlikely unless it is dissolved in an organic solvent.
Benzethidine Narcotic Analgesic C23H29NO3 = 367.5 CAS—3691-78-9 IUPAC Name Ethyl-1-(2-benzyloxyethyl)-4-phenylpiperidine-4-carboxylate Synonyms Benzyloxyethylnorpethidine; TA-28.
Chemical Properties Extracted by organic solvents from aqueous alkaline solutions. Log P (octanol/water) 4.46 [Meylan, Howard 1995]. Colour Tests Ammonium molybdate test—orange-brown (limit of detection 0.5 mg); sulfuric acid-formaldehyde test—dull orange (limit of detection 0.5 mg). Thin-layer Chromatography System T1—Rf 0.74 (location reagent acidified iodoplatinate spray, positive reaction). Gas Chromatography System G2/225—retention time 2.53 relative to codeine; system G4—retention time 1.70 relative to codeine. Ultraviolet Spectrum Aqueous acid (0.1 N sulfuric acid)—252, 258, 264 nm and an inflexion at 267 nm.
Meylan WM, Howard PH (1995). Atom/fragment contribution method for estimating octanolwater partition coefficients. J Pharm Sci 84: 83–92.
Benzethonium Chloride Cationic Disinfectant, Antiseptic C27H42ClNO2 = 448.1 CAS—121-54-0 IUPAC Name N,N-Dimethyl-N-[2-[2-[4-(1,1,3,3-tetramethylbutyl)phenoxy] ethoxy]-ethyl]-benzenemethanaminium chloride Proprietary Names Hyamine 1622; Phemerol Chloride.
B
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:30:31
962
Benzfetamine Ultraviolet Spectrum Aqueous acid—252, 258 nm (A11¼19c), 262, 268 nm.
B Chemical Properties White crystals. A solution in water foams strongly when shaken. Mp 160 to 165 . Soluble 1 in 0.6 of water, 1 in 0.6 of ethanol and 1 in 1 of chloroform; slightly soluble in ether; practically insoluble in light petroleum. Log P (octanol/water), 4.0. Colour Test Marquis test—orange. Thin-layer Chromatography System TA—Rf 0.03 (acidified iodoplatinate solution, positive). Ultraviolet Spectrum Aqueous acid—263, 269 (A11¼29c), 274 nm (A11¼28c). No alkaline shift.
Infrared Spectrum Principal peaks at wavenumbers 1497, 740, 698, 1028, 1125, 1220 cm1 (KBr disk).
Infrared Spectrum Principal peaks at wavenumbers 1240, 1505, 1120, 826, 1063, 769 cm1 (KBr disk). Use Used as 0.1 to 0.2% solutions.
Benzfetamine Anorectic C17H21N = 239.4 CAS—156-08-1 IUPAC Name (aS)-N,a-Dimethyl-N-(phenylmethyl)benzeneethanamine Synonym Benzphetamine Proprietary Name Didrex
Chemical Properties A liquid. Bp 127 . Practically insoluble in water; soluble in methanol, ethanol, chloroform, acetone, benzene and ether. pKa 6.6. Log P (octanol/water), 4.1. Benzfetamine Hydrochloride C17H21N,HCl = 275.8 CAS—5411-22-3 Proprietary Names Didrex; Inapetyl. Chemical Properties A white crystalline powder. Mp about 131 . Freely soluble
in water, ethanol and chloroform; slightly soluble in ether. Colour Test Marquis test—orange Thin-layer Chromatography System TA—Rf 0.73; system TB—Rf 0.67; system TC—Rf 0.70; system TE—Rf 0.87; system TL—Rf 0.70; system TAE—Rf 0.60 (dragendorff spray, positive; acidified iodoplatinate solution, positive; Marquis reagent, brown). Gas Chromatography System GA—benzfetamine RI 1855, amfetamine RI 1125, metamfetamine RI 1175; system GB—benzfetamine RI 1899, amfetamine RI 1150, metamfetamine RI 1200; system GC—benzfetamine RI 2172, amfetamine RI 1536, metamfetamine RI 1722; system GF—benzfetamine RI 2050, amfetamine RI 1315, metamfetamine RI 1335. Column: TC-1 cross-linked methylsilicone (20 m 0.25 mm i.d., 0.25 mm). Temperature programme: 60 for 0.5 min, to 280 at 20 /min. Carrier gas: He, flow rate 1 mL/min. SIM acquisition mode. Retention time: amfetamine, 4.6 min; metamfetamine, 5.4 min; benzfetamine, 8.2 min [Kikura, Nakahara 1995]. High Performance Liquid Chromatography System HA—benzfetamine k 1.2, amfetamine k 0.9, metamfetamine k 2.0; system HC—benzfetamine k 0.15, amfetamine k 0.98, metamfetamine k 2.07.
Mass Spectrum Principal ions at m/z 91, 148, 149, 65, 92, 42, 56, 39.
Quantification Urine GC-MS Column: HP-1 (12 m 0.2 mm i.d., 0.33 mm). Temperature programme: 80 for 1 min, to 210 at 20 /min for 2 min. Carrier gas: He, flow rate 1 mL/min. Retention time: amfetamine, 4.2 min; metamfetamine, 5.1 min; benzfetamine, 8.2 min. Limit of detection, 5 mg/L for amfetamine and metamfetamine, 2 mg/L for benzfetamine [Cody, Valtier 1998]. HPLC Benzfetamine metabolites Fujinami et al. [1998]. Hair GC-MS. Limit of detection, 0.5 ng/mg [Nakahara 1995]. Disposition in the Body Readily absorbed after oral administration and mainly excreted in the urine as amfetamine and metamfetamine; the metabolism of benzfetamine to desmethylbenzfetamine may be a major pathway in its metabolism. It is also excreted in the urine as conjugated metabolites, p-hydroxy-Nbenzylamfetamine glucuronide and p-hydroxybenzfetamine glucuronide; very little is excreted as unchanged drug. Therapeutic Concentration Urinary concentrations of p-hydroxy-N-benzylamfetamine glucuronide (pHBAG) and p-hydroxybenzfetamine glucuronide (pHBZG) were determined in 2 healthy volunteers after the administration of 10 mg benzfetamine hydrochloride. After 6 h, pHBAG concentrations were 450 nM (subject 1) and 800 nM (subject 2), and pHBZG 800 nM (subject 1) and 1900 nM (subject 2). After 24 h, pHBAG levels dropped to 50 nM for both subjects and pHBZG concentrations were negligible [Fujinami et al. 1998]. Toxicity A 16-year-old male was found dead with several medicine bottles in the room. Postmortem examination and toxicological analysis revealed the following benzfetamine concentrations: blood 13.9 mg/L, urine 8.0 mg/L, liver 106.3 mg/kg, brain 30.5 mg/kg, and gastric contents (total) 53.1 mg [Brooks et al. 1982].
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:30:44
Benzocaine Dose 25 to 150 mg of benzfetamine hydrochloride daily. Brooks JP et al. (1982). A case of benzphetamine poisoning. Am J Forensic Med Pathol 3: 245–247. Cody JT, Valtier S (1998). Detection of amphetamine and methamphetamine following administration of benzphetamine. J Anal Toxicol 22: 299–309. Fujinami A et al. (1998). Development of a method for the quantitation of benzphetamine metabolites in human urine by high-performance liquid chromatography. Ann Clin Biochem 35: 775–779. Kikura R, Nakahara Y (1995). Hair analysis for drugs of abuse. XI. Disposition of benzphetamine and its metabolites into hair and comparison of benzphetamine use and methamphetamine use by hair analysis. Biol Pharm Bull 18: 1694–1699. Nakahara Y (1995). Detection and diagnostic interpretation of amphetamines in hair. Forensic Sci Int 70: 135–153. Cody JT, Valtier S (1998). Detection of amphetamine and methamphetamine following administration of benzphetamine. J Anal Toxicol 22: 299–309.
Benzilonium Bromide Anticholinergic C22H28BrNO3 = 434.4 CAS—1050-48-2 IUPAC Name 1,1-Diethyl-3-[(hydroxydiphenylacetyl)oxy]pyrrolidinium bromide Proprietary Names Portyn; Ulcoban.
Chemical Properties A white crystalline powder. Mp 203 to 204 . Soluble in water. Log P (octanol/water), 0.3. Colour Tests The following tests are performed on benzilonium nitrate: Liebermann’s reagent—brown; Marquis test—orange!green!blue; sulfuric acid—orange. Thin-layer Chromatography System TA—Rf 0.03; system TAJ—Rf 0.00; system TAK—Rf 0.00; system TAL—Rf 0.05 (acidified iodoplatinate solution, positive). Ultraviolet Spectrum Aqueous acid—252, 258 (A11¼11a), 264 nm.
963
Proprietary Name Amplivix
B Chemical Properties A yellowish powder. Mp 167 . Soluble 1 in about 500 of water at 25 and 1 in about 100 of water at 45 ; soluble in acetone and chloroform. Log P (octanol/water) 6.6. Thin-layer Chromatography System TD—Rf 0.62; system TE—Rf 0.23; system TF—Rf 0.58; system TAD—Rf 0.65. Ultraviolet Spectrum Methanol—240 nm (A11¼1280b), 275, 357 nm.
Infrared Spectrum Principal peaks at wavenumbers 1618, 756, 1577, 1135, 1293, 1176 cm1 (Nujol mull). Mass Spectrum Principal ions at m/z 518, 173, 264, 519, 373, 376, 520, 249.
Dose Benziodarone has been given in initial doses of 600 mg daily and maintenance doses of 300 to 400 mg daily.
Benzocaine Anaesthetic (Local) C9H11NO2 = 165.2 CAS—94-09-7 IUPAC Name 4-Aminobenzoic acid ethyl ester Synonyms Anaesthesinum; anesthamine; ethoforme; ethyl aminobenzoate. Proprietary Names Americaine; Anaesthesin; Flavamed; Subcutin. It is an ingredient of AAA, Audicort, Auralgan, Auralgicin, Auraltone, Merocaine, Transvasin, Tyrosolven and Tyrozets. Infrared Spectrum Principal peaks at wavenumbers 1728, 1220, 1190, 1162, 690, 1052 cm1 (KBr disk). Quantification Plasma GC–MS Limit of detection, 5 mg/L [Dahlstr€ om et al. 1980]. Dose 30 to 70 mg daily. Dahlstr€ om H et al. (1980). Quantitative determination of benzilonium bromide in plasma by gas chromatography-mass spectrometry after oxidation to benzophenone. J Chromatogr 183: 511–513.
Benziodarone Uricosuric, Vasodilator C17H12I2O3 = 518.1 CAS—68-90-6 IUPAC Name 2-Ethyl-3-benzofuranyl 4-hydroxy-3,5-diiodophenyl methanone
Chemical Properties Colourless crystals or white crystalline powder. Mp 88 to 92 . Soluble 1 in 2500 of water, 1 in 8 of ethanol, 1 in 2 of chloroform and 1 in 4 of ether; soluble in dilute acids. pKa 2.5 (25 ). Log P (octanol/water), 1.9. Colour Test Diazotisation—red. Thin-layer Chromatography System TA—Rf 0.67; system TB—Rf 0.06; system TC—Rf 0.57; system TD—Rf 0.56; system TE—Rf 0.77; system TF—Rf 0.62; system TL—Rf 0.66; system TAD—Rf 0.63; system TAE—Rf 0.84; system TAF—Rf 0.87 (ninhydrin spray, positive; acidified potassium permanganate solution, positive; Van Urk reagent, bright yellow). Gas Chromatography System GA—benzocaine RI 1555, 4-aminobenzoic acid RI 1547; system GF—RI 2100.
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:30:58
964
B
Benzoctamine
High Performance Liquid Chromatography System HA—k 0.1; system HQ—k 20.06; system HR—k 1.61; system HX—RI 404; system HY—RI 358; system HZ—retention time 4.3 min. Ultraviolet Spectrum Aqueous acid—272 (A11¼90c), 278 nm; aqueous alkali—285 nm (A11¼930a); ethanol—293 nm (A11¼1238a). Chemical Properties pKa 7.6. Log P (octanol/water), 3.5. Benzoctamine Hydrochloride C18H19N,HCl = 285.8 CAS—10085-81-1 Synonym Ba-30803 Proprietary Name Tacitin(e) Chemical Properties A white crystalline powder. Mp 320 to 322 . Soluble in
water, ethanol and chloroform; sparingly soluble in acetone and ether.
Infrared Spectrum Principal peaks at wavenumbers 1280, 1680, 1598, 1170, 1315, 1634 cm1 (KBr disk).
Colour Tests Mandelin’s test—blue-green; Marquis test—red-violet. Thin-layer Chromatography System TA—Rf 0.59; system TB—Rf 0.57; system TC—Rf 0.52; system TL—Rf 0.43; system TAE—Rf 0.38; system TAJ—Rf 0.31; system TAK—Rf 0.14; system TAL—Rf 0.65 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 2078; system GB—RI 2172; system GF—RI 2445. High Performance Liquid Chromatography System HA—k 1.7; system HX—RI 380; system HY—RI 322. Ultraviolet Spectrum Aqueous acid—264, 271 nm (A11¼57a).
Infrared Spectrum Principal peaks at wavenumbers 757, 743, 732, 1134, 1060, 1020 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 218, 44, 191, 221, 219, 178, 42, 180.
Mass Spectrum Principal ions at m/z 120, 165, 92, 65, 137, 39, 121, 93; 4aminobenzoic acid 137, 120, 92, 65, 39, 138, 121, 63. Dose 30 to 60 mg of benzoctamine hydrochloride daily.
Benzoic Acid
Disposition in the Body Benzocaine is metabolised by hydrolysis to 4aminobenzoic acid. At the concentrations normally used (2 to 10%) it is comparatively non-irritant and non-toxic, having only about one-tenth the toxicity of cocaine. The maximum safe amount for topical use is 5000 mg (25 mL of a 20% solution).
Benzoctamine Tranquilliser C18H19N = 249.4 CAS—17243-39-9 IUPAC Name N-Methyl-9,10-ethanoanthracene-9(10H)-methenamine
Preservative C7H6O2 = 122.1 CAS—65-85-0 Synonyms Benzenecarboxylic acid; phenylformic acid. Proprietary Names It is an ingredient of Aserbine and Malatex.
Chemical Properties Colourless, light feathery crystals or white scales or powder. Mp 122.4 . It sublimes on heating. Soluble 1 in about 350 of water, 1 in 20 of boiling water, 1 in 3 of ethanol, 1 in 5 of chloroform and 1 in 3 of ether; freely soluble in acetone. pKa 4.2 (25 ). Log P (octanol/water), 1.9. Sodium Benzoate C7H5O2Na = 144.1 CAS—532-32-1
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:31:5
Benzoylecgonine Chemical Properties A white, amorphous, granular, flaky, or crystalline powder. Soluble 1 in 2 of water and 1 in 90 of ethanol.
Thin-layer Chromatography System TD—Rf 0.26; system TE—Rf 0.07; system TF—Rf 0.28; system TAD—Rf 0.35; system TAJ—Rf 0.55; system TAK—Rf 0.81; system TAL—Rf 0.92. Gas Chromatography System GA—RI 1180. High Performance Liquid Chromatography System HX—RI 360; system HY—RI 327; system HZ—retention time 3.0 min (benzoate). Ultraviolet Spectrum Aqueous acid—230 (A11¼923a), 273 nm (A11¼85a); methanol—227 (A11¼895a), 272 (A11¼73a), 280 nm (A11¼61b).
965
Proprietary Name Tessalon
B Chemical Properties A clear, pale yellow, viscous liquid. Miscible with water, ethanol, chloroform and ether. Log P (octanol/water), 2.4. Colour Tests Aromaticity (method 2)—yellow/red; Liebermann’s test (at 100 )—blue. Thin-layer Chromatography System TA—Rf 0.61; system TAJ—Rf 0.56; system TAK—Rf 0.23; system TAL—Rf 0.90 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—not eluted. Ultraviolet Spectrum Ethanol—308 nm (A11¼473c).
Infrared Spectrum Principal peaks at wavenumbers 709, 1689, 1296, 667, 935, 685 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 105, 77, 51, 122, 50, 39, 74, 76; hippuric acid 105, 135, 51, 134, 77, 106, 50, 78.
Infrared Spectrum Principal peaks at wavenumbers 1095, 1605, 1266, 1172, 1698, 1527 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 176, 59, 132, 150, 45, 193, 105, 29.
Quantification Plasma GC FID. Limit of detection, 0.1 mg/L [Li, Zhao 2001]. ECD. Limit of detection, 10 mg/L [Sioufi, Pommier 1980]. HPLC UV detection (l¼283 nm). Limit of quantification, 0.05 mg/L [Nomeir et al. 1994]. Urine GC FID. Limit of detection, 0.2 mg/L [Li, Zhao 2001]. ECD. Limit of detection, 0.5 mg/L [Aprea et al. 1997]. See Plasma [Sioufi, Pommier 1980]. Disposition in the Body Benzoic acid is metabolised in the liver by conjugation with glycine and is rapidly and completely excreted in the urine as hippuric acid. Normal urinary excretion of hippuric acid is 1 to 2.5 g daily, equivalent to 0.7 to 1.7 g of benzoic acid. When taken in large doses, a proportion may be excreted as benzoylglucuronic acid. Benzoic acid may be found in the urine as a metabolite of benzaldehyde, and is also a metabolite of numerous other compounds. Uses Benzoic acid is used as a preservative in a concentration of 0.1%. Sodium benzoate is given by mouth in a dose of 6 g to test liver function.
Dose 300 to 600 mg daily.
Benzoylecgonine Alkaloid C16H19NO4 = 289.3 CAS—519-09-5 IUPAC Name 3-(Benzoyloxy)-8-methyl-8-azabicyclo[3.2.1]octane-2-carboxylic acid Synonym Ecgonine benzoate
Aprea C et al. (1997). Analytical method for the determination of urinary 3-phenoxybenzoic acid in subjects occupationally exposed to pyrethroid insecticides. J Chromatogr B Biomed Sci Appl 695 (2): 227–236. Li AQ, Zhao XL (2001). Determination and pharmacokinetic study of 1-p-(3.3-dimethyl-1-triazeno) benzoic acid in cancer patients by capillary gas chromatography. Biomed Chromatogr 15 (2): 75–78. Nomeir AA et al. (1994). Liquid chromatographic analysis, stability and protein binding studies of the anti-HIV agent benzoic acid, 2-chloro-5[[(1-methylethoxy)thioxomethyl]amino]-,1-methylethyl ester. J Pharm Biomed Anal 12(5): 693–698. Sioufi A, Pommier F (1980). Gas chromatographic determination of low concentrations of benzoic acid in human plasma and urine. J Chromatogr 181(7); B Biomed. Appl.: 161–168.
Benzonatate Cough Suppressant C30H53NO17 = 603.7 CAS—104-31-4 IUPAC Name 4-(Butylamino)benzoic acid 3,6,9,12,15,18,21,24,27-nonaoxaoctacos-1-yl ester Synonyms Benzonatine; benzononatine.
Chemical Properties An alkaloid obtained from coca leaves, Erythroxylum coca (Erythroxylaceae) and its varieties. The hydrated form occurs as crystals. Mp 86 to 92 (anhydrous) 195 (with decomposition). Very soluble in hot water; soluble in ethanol; practically insoluble in ether; soluble in dilute acids and alkalis. Log P (octanol/water) 1.3. Extraction yield (chlorobutane), 0 [Demme et al. 2005]. Thin-layer Chromatography System TA—Rf 0.21; system TB—Rf 0.00; system TC—Rf 0.01 (acidified iodoplatinate solution, positive). Plate: silica 60 F254 (10 10 cm). Mobile phase: hexane : toluene : diethylamine (65 : 20 : 5). UV detection. Rf 0.02 [Antonilli et al. 2001]. Gas Chromatography System GA—benzoylecgonine RI 2570; cocaine RI 2187; system GB—benzoylecgonine RI 2663, cocaine RI 2289.
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:31:12
966
Benzquinamide
High Performance Liquid Chromatography System HA—k 0.9 (tailing peak); system HQ—k 5.68; system HY—RI 236; system HZ—RT 1.7 min; system HAA—RT 9.7 min. Ultraviolet Spectrum Aqueous acid—234 (A11 ¼ 376a), 274 nm.
B
Antonilli L et al. (2001). Analysis of cocaethylene, benzoylecgonine and cocaine in human urine by high-performance thin-layer chromatography with ultraviolet detection: a comparison with high-performance liquid chromatography. J Chromatogr B Biomed Sci Appl 751: 19–27. de la Torre R et al. (1995). Determination of cocaine and its metabolites in human urine by gas chromatography/mass spectrometry after simultaneous use of cocaine and ethanol. J Pharm Biomed Anal 13: 305–312. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Jeanville PM et al. (2001). Rapid confirmation/quantitation of ecgonine methyl ester, benzoylecgonine, and cocaine in urine using on-line extraction coupled with fast HPLC and tandem mass spectrometry. J Anal Toxicol 25: 69–75. Kintz P, Mangin P (1995). Simultaneous determination of opiates, cocaine and major metabolites of cocaine in human hair by gas chromotography/mass spectrometry (GC/MS). Forensic Sci Int 73: 93–100. Sosnoff CS et al. (1996). Analysis of benzoylecgonine in dried blood spots by liquid chromatography–atmospheric pressure chemical ionization tandem mass spectrometry. J Anal Toxicol 20: 179–184. Wang WL et al. (1994). Simultaneous assay of cocaine, heroin and metabolites in hair, plasma, saliva and urine by gas chromatography-mass spectrometry. J Chromatogr B Biomed Appl 660: 279–290.
Benzquinamide Infrared Spectrum Principal peaks at wavenumbers 1275, 1720, 1618, 717, 1116, 1316 cm1.
Antiemetic C22H32N2O5 = 404.5 CAS—63-12-7 IUPAC Name 2-(Acetyloxy)-N,N-diethyl-1,3,4,6,7,11b-hexahydro-9,10-dimethoxy2H-benzo[a]-quinolizine-3-carboxamide
Chemical Properties A yellowish crystalline powder. Mp 130 to 131.5 . Soluble in dilute acetic acid. pKa 5.9. Log P (octanol/water), 1.9. Benzquinamide Hydrochloride Mass Spectrum Principal ions at m/z 124, 82, 168, 77, 105, 42, 94, 83.
C22H32N2O5,HCl = 441.0 CAS—113-69-9 Proprietary Name Emete-con
Colour Tests Liebermann’s reagent—black; Marquis test—yellow; sulfuric acid—orange. Thin-layer Chromatography System TA—Rf 0.65; system TB—Rf 0.07; system TC—Rf 0.69; system TL—Rf 0.36; system TAJ—Rf 0.53; system TAK—Rf 0.06; system TAL—Rf 0.91 (acidified iodoplatinate solution, positive). High Performance Liquid Chromatography System HA—k 0.3. Ultraviolet Spectrum Aqueous acid—282 nm (A 11¼95b). No alkaline shift.
Quantification See also under Cocaine. Blood LC-MS CI. Limit of detection, 2 mg/L [Sosnoff et al. 1996]. Plasma GC-MS SIM acquisition mode (m/z 404, 140 and 298). Limit of detection, 5 mg/L [Wang et al. 1994]. Urine GC-MS SIM acquisition mode (m/z 318, 439). Limit of detection, 1 mg/L [de la Torre et al. 1995]. See Plasma [Wang et al. 1994]. HPLC Column: LiChrocart-LiCrospher 100 RP-18 (250 4 mm i.d., 5 mm). Mobile phase: 0.045 mol/L ammonium acetate-methanol-acetonitrile (80 : 10 : 10) : 0.045 mol/L ammonium acetate-methanol-acetonitrile (20 : 40 : 40; 100 : 0 to 47.2 : 52.8 in 20 min), flow rate 1 mL/min. UV detection (l ¼ 235 nm). Retention time: 8.48 min. Limit of quantification, 200 mg/L, limit of detection, 25 mg/L [Antonilli et al. 2001 LC-MS Column: Allure Basix (30 2.1 mm, 5 mm). Mobile phase: 50 mmol/L formic acid-100 mmol/L ammonium formate-acetonitrile : acetone (60 : 40). Tandem MS detection (m/z 290 to 168 transition). Retention time: 1.5 min. Limit of detection, 0.5 mg/L [Jeanville et al. 2001]. Oral Fluid See Plasma [Wang et al. 1994]. Hair GC-MS SIM acquisition mode (m/z 240 and 361). Limit of detection, 0.2 mg/ g using a 30 mg sample [Kintz, Mangin 1995]. See Plasma. Limit of detection, 0.5 mg/g [Wang et al. 1994]. Disposition in the Body Benzoylecgonine is the first hydrolysis product formed in the metabolism of cocaine; it is then further hydrolysed to ecgonine.
Infrared Spectrum Principal peaks at wavenumbers 1244, 1733, 1633, 1204, 1515, 1123 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 205, 244, 191, 345, 206, 72, 272, 246.
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:31:20
Benzydamine
967
B Quantification Plasma GC–MS Limit of detection, 10 mg/L [Hobbs, Connolly 1978]. Disposition in the Body Readily absorbed after oral administration; bioavailability about 35%. Metabolised by O-demethylation, deacetylation and N-dealkylation. After an oral dose, up to about 7% is excreted unchanged in the urine in 24 h. Therapeutic Concentration Following oral administration of 200 mg to 20 subjects, a mean peak plasma concentration of 0.6 mg/L was attained in 1.2 h. Following intramuscular injection of 50 mg to 20 subjects, a mean peak plasma concentration of 0.7 mg/L was attained in 0.26 h [Hobbs, Connolly 1978]. Half-life Plasma half-life, about 1 h. Volume of Distribution About 1 L/kg. Dose The equivalent of 50 mg of benzquinamide by intramuscular injection, repeated as necessary. Hobbs DC, Connolly AG (1978). Pharmacokinetics of benzquinamide in man. J Pharmacokinet Biopharm 6: 477–485.
Mass Spectrum Principal ions at m/z 91, 121, 65, 122, 309, 64, 230, 123.
Benzthiazide Diuretic, Antihypertensive C15H14ClN3O4S3 = 431.9 CAS—91-33-8 IUPAC Name 6-Chloro-3-[[(phenylmethyl)thio]methyl]-2H-1,2,4-benzothiadiazine-7-sulfonamide 1,1-dioxide Proprietary Names Aquatag; Exna; Hydrex. It is an ingredient of Decaserpyl Plus and Dytide.
Chemical Properties A white crystalline powder. Mp 231 to 232 . Practically insoluble in water, chloroform and ether; slightly soluble in ethanol; soluble 1 in 100 of acetone; freely soluble in dimethylformamide and in solutions of alkalis. pKa 6.0. Log P (octanol/water), 1.7. Colour Tests Koppanyi-Zwikker test—violet; Liebermann’s reagent—redbrown; sulfuric acid—yellow. Thin-layer Chromatography System TD—Rf 0.14; system TE—Rf 0.09; system TF—Rf 0.51; system TAD—Rf 0.30; system TAJ—Rf 0.31; system TAK—Rf 0.06; system TAL—Rf 0.71. Gas Chromatography System GA—RI 2680. High Performance Liquid Chromatography System HN—k 9.32; system HY—RI 415. Ultraviolet Spectrum Aqueous acid—283 nm (A11¼292a); aqueous alkali— 297 nm (A11¼310b).
Quantification Plasma HPLC Column: reversed phase. UV detection (l¼280 nm). Limit of detection, 10 mg/L [Meyer et al. 1982]. Urine HPLC See Plasma [Meyer et al. 1982]. Faeces HPLC See Plasma [Meyer et al. 1982]. Disposition in the Body Poorly absorbed after oral administration. Less than 10% of a dose is excreted in the urine as unchanged drug in 48 h, and about 80% of a dose is eliminated in the faeces. Therapeutic Concentration Following an oral dose of 50 mg to 4 subjects, peak plasma concentrations of about 0.008 and 0.02 mg/L were attained in 3 h in 2 subjects; in the remaining 2 subjects benzthiazide was not detectable in the plasma [Meyer et al. 1982]. Half-life Derived from urinary excretion data, about 10 to 15 h. Dose 50 to 200 mg daily. Meyer MC et al. (1982). HPLC determination of benzthiazide in biologic material. Biopharm Drug Dispos 3: 1–9.
Benzydamine Analgesic C19H23N3O = 309.4 CAS—642-72-8 IUPAC Name N,N-Dimethyl-3-[[1-(phenylmethyl)-1H-indazol-3-yl]oxy]-1propanamine Synonym Benzindamine
Chemical Properties Bp 160 . Log P (octanol/water), 4.2. Benzydamine Hydrochloride Infrared Spectrum Principal peaks at wavenumbers 1180, 1160, 1310, 1502, 1620, 1590 cm1 (KBr disk).
C19H23N3O,HCl = 345.9 CAS—132-69-4 Proprietary Names Afloben;
Tantum; Verax.
A-Termadol;
Difflam;
Imotryl;
Multum;
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:32:15
968
Benzyl Alcohol
Chemical Properties A white crystalline powder. Mp 160 . Soluble 1 in 1 of
water, 1 in 8 of ethanol and 1 in 4 of chloroform; practically insoluble in ether.
B
Colour Test Mandelin’s test—brown-green. Thin-layer Chromatography System TA—Rf 0.44; system TB—Rf 0.36; system TC—Rf 0.22; system TL—Rf 0.09; system TAE—Rf 0.16 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 2380. High Performance Liquid Chromatography System HAA—retention time 15.0 min. Ultraviolet Spectrum Aqueous acid—307 nm (A11¼144c). No alkaline shift.
Mass Spectrum Principal ions at m/z 79, 77, 108, 101, 51, 50, 39, 40. Quantification Blood GC–MS Limit of detection, 1 mg/L [Dasgupta, Steinagel 1997]. Plasma HPLC Column: C18. Mobile phase: water : acetonitrile : glacial acetic acid. UV detection (l¼254 nm). Limit of detection, 10 ng [Tan et al. 1991]. Serum GC–MS SIM (m/z 69, 77, 91, 105, 121, 504 and 518). Limit of detection, 0.1 mg/L [Dasgupta, Humphrey 1998]. See Blood [Dasgupta, Steinagel 1997]. Use Topically in concentrations of up to 10%.
Infrared Spectrum Principal peaks at wavenumbers 1529, 740, 1491, 1613, 1182, 1141 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 85, 58, 86, 91, 84, 70, 42, 225.
Dasgupta A, Steinagel G (1997). Gas chromatographic-mass spectrometric identification and quantitation of benzyl alcohol from human serum and postmortem blood after derivatization with 4carbethoxy hexafluorobutyryl chloride: a novel derivative. J Forensic Sci 42(4): 697–700. Dasgupta A, Humphrey PE (1998). Gas chromatographic-mass spectrometric identification and quantitation of benzyl alcohol in serum after derivatization with perfluorooctanoyl chloride: a new derivative. J Chromatogr B Biomed Sci Appl 708(1–2): 299–303. Tan HS et al. (1991). Determination of benzyl alcohol and its metabolite in plasma by reversed-phase high-performance liquid chromatography. J Chromatogr 568(1): 145–155.
Benzyl Benzoate Acaricide C14H12O2 = 212.2 CAS—120-51-4 Proprietary Names Antiscabiosum Mago; Ascabiol; Benzemul; Scabanca. Quantification Plasma HPLC Limit of quantification, 0.5 mg/L [Baldock et al. 1990]. Urine HPLC Limit of quantification, 1.0 mg/L [Baldock et al. 1990]. Disposition in the Body Absorbed after oral administration. About 50% of a dose is excreted unchanged in the urine. Therapeutic Concentration An oral dose of 1 mg/kg produced a blood concentration of about 0.8 mg/L within 2 h and a significant concentration was maintained for several hours [Catanese et al. 1966]. Protein Binding A fraction of benzydamine is bound to proteins in the blood. Dose 150 to 200 mg of benzydamine hydrochloride daily.
Chemical Properties Colourless crystals or a clear, colourless, oily liquid. Bp 323 to 324 . Practically insoluble in water; miscible with ethanol, acetone, carbon disulfide, chloroform and ether. Log P (octanol/water), 4.0. Thin-layer Chromatography System TF—Rf 0.73; system TAD—Rf 0.09. Gas Chromatography System GA—RI 1738. Ultraviolet Spectrum Ethanol—230 nm (A11¼843b).
Baldock GA et al. (1990). Determination of benzydamine and its N-oxide in biological fluids by high-performance liquid chromatography. J Chromatogr 529(1): 113–123. Catanese B et al. (1966). Studies on the absorption and elimination of benzydamine in the mouse, rat, dog, and man. Arzneimittelforschung 16: 1354–1357.
Benzyl Alcohol Anaesthetic (Local), Disinfectant C7H8O = 108.1 CAS—100-51-6 Synonyms Phenylcarbinol; phenylmethanol.
Chemical Properties A colourless liquid with a faint aromatic odour. Mass per mL 1.043 to 1.046 g. Bp 203 to 208 . Soluble 1 in 25 of water; miscible with ethanol, chloroform and ether. pKa 15.4. Log P (octanol/water), 1.1. Thin-layer Chromatography System TAE—Rf 0.86. Gas Chromatography System GA—RI 1040. Ultraviolet Spectrum Methanol—252 (A11¼14b), 258 (A11¼17b), 264 nm (A11¼13b).
Infrared Spectrum Principal peaks at wavenumbers 1263, 1706, 1106, 713, 698, 1067 cm1 (thin film). Mass Spectrum Principal ions at m/z 77, 105, 91, 51, 65, 212, 50, 78. Disposition in the Body Rapidly hydrolysed to benzoic acid and benzyl alcohol; benzyl alcohol then undergoes further oxidation to benzoic acid followed by conjugation with glycine to form hippuric acid. Excreted in the urine mainly as hippuric acid. Uses Topically as a 25% application; a 5% solution is used as an insect repellent.
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:32:55
Benzylpenicillin
969
Benzyl Nicotinate Rubefacient (Topical) C13H11NO2 = 213.2 CAS—94-44-0 Proprietary Names Rubriment. It is an ingredient of Bayolin.
Chemical Properties A liquid. Log P (octanol/water), 2.4. Colour Tests Cyanogen bromide—red-orange; Liebermann’s reagent—brown; sulfuric acid—orange. Thin-layer Chromatography System TA—Rf 0.63; system TB—Rf 0.42; system TC—Rf 0.71; system TL—Rf 0.60 (acidified potassium permanganate solution, positive). Ultraviolet Spectrum Methanol—262 nm (A11¼152b).
B
Infrared Spectrum Principal peaks at wavenumbers 1274, 1040, 1502, 1017, 763, 1056 cm1. Mass Spectrum Principal ions at m/z 284, 91, 375, 81, 42, 36, 285, 175. Dose Benzylmorphine hydrochloride was formerly given in doses of 8 to 30 mg.
Benzylpenicillin Antibiotic C16H18N2O4S = 334.4 CAS—61-33-6 IUPAC Name (2S,5R,6R)-3,3-Dimethyl-7-oxo-6-[(phenylacetyl)amino]-4-thia1-azabicyclo[3.2.0]heptane-2-carboxylic acid Synonyms Crystalline penicillin G; penicillin; penicillin G; penicillin II.
Infrared Spectrum Principal peaks at wavenumbers 1278, 1724, 1108, 740, 699, 1587 cm1 (KCl disk). Use Topically in a concentration of 2.5%.
Note The name ‘benzylpenicillin’ and its synonyms are commonly used to describe either benzylpenicillin potassium or benzylpenicillin sodium. Chemical Properties An antimicrobial acid produced by the growth of certain strains of Penicillium notatum. pKa 2.8 (25 ). Log P (octanol/water), 1.8. Benzylpenicillin Potassium
Benzylmorphine Narcotic Analgesic C24H25NO3 = 375.5 CAS—14297-87-1 IUPAC Name 7,8-Didehydro-4,5-epoxy-17-methyl-3-(phenylmethoxy)morphinan-6-ol
C16H17KN2O4S = 372.5 CAS—113-98-4 Proprietary Names Abbocillin-G; Crystapen G; Falapen; Hyasorb; M-Cillin B;
Megacillin; Novopen; P-50; Paclin G; Pentids; Pfizerpen; Sugracillin. Note Megacillin is also used as a proprietary name for clemizole penicillin, phenoxymethylpenicillin and procaine penicillin (procaine benzylpenicillin). Chemical Properties A white, finely crystalline powder. Mp 214 to 217 , with decomposition. Very soluble in water; soluble in ethanol; practically insoluble in chloroform and ether. Benzylpenicillin Sodium C16H17N2NaO4S = 356.4 CAS—69-57-8 ecilline G. It is an ingredient of Bicillin Proprietary Names Crystapen; Gonopen; Sp
and Triplopen.
Note Bicillin is also used as a proprietary name for benzathine benzylpenicillin. Chemical Properties A white to slightly yellow, finely crystalline powder. Very
soluble in water; soluble in ethanol; practically insoluble in chloroform and ether. High Performance Liquid Chromatography System HY—RI 376. Ultraviolet Spectrum Benzylpenicillin sodium: water—257 (A11¼7b), 264, 325 nm. Chemical Properties A colourless crystalline powder. Mp 132 . Soluble 1 in 2500 of cold water; 1.6 parts dissolves in 100 parts ether; readily soluble in 50% ethanol and in benzene. pKa 8.1 (20 ). Log P (octanol/water), 3.0. Benzylmorphine Hydrochloride C24H25NO3,HCl = 411.9 CAS—630-86-4 Proprietary Name Peronine Chemical Properties A colourless microcrystalline powder. Soluble 1 in 200 of
water and 1 in 160 of ethanol; practically insoluble in chloroform and ether. Colour Test Marquis test—red!violet. Thin-layer Chromatography System TA—Rf 0.41; system TB—Rf 0.06; system TC—Rf 0.23; system TL—Rf 0.08; system TAE—Rf 0.20; system TAF—Rf 0.23 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 3015. High Performance Liquid Chromatography System HA—k 4.4 (tailing peak); system HC—k 1.03. Ultraviolet Spectrum Aqueous acid—284 nm (A11¼48b).
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:33:6
970
N-Benzylpiperazine
Infrared Spectrum Principal peaks at wavenumbers 1620, 1777, 1500, 1310, 1700, 703 cm1 (benzylpenicillin sodium, KBr disk).
B
Quantification Serum HPLC UV detection (l¼208 nm). Limit of quantification, 0.5 to 50 mg/L [Mendez-Alvarez et al. 1991]. Disposition in the Body About 30% of an oral dose is absorbed, the remainder being inactivated by gastric acid; maximum concentrations are attained about 1 h after oral administration. After IM injection it is rapidly absorbed, peak concentrations being attained in 15 to 30 min. About 60 to 90% of an IM dose is excreted in the urine, mainly in the first few hours; the urinary material consists of unchanged drug and penicilloic acid (about 20% of the dose). Biliary excretion also occurs. Therapeutic Concentration In plasma, minimum inhibitory concentration 0.006 to 2 mg/L. Half-life Plasma half-life, about 0.5 to 1 h; increased in infants and elderly subjects, and in renal impairment. Distribution in Blood Plasma : whole blood ratio, 1.6. Protein Binding 45 to 65%. Note For a review of the pharmacokinetics of penicillin antibiotics, see Barza, Weinstein [1976]. For a review of clinical pharmacology and therapeutic use of penicillins, see Nathwani, Wood [1993]. Dose 0.6 to 2.4 g daily, given parenterally; up to 24 g daily in severe infections. Barza M, Weinstein L (1976). Pharmacokinetics of the penicillins in man. Clin Pharmacokinet 1: 297–308. Mendez-Alvarez E et al. (1991). A reversed phase liquid chromatographic method for the simultaneous determination of several common penicillins in human serum. Biomed Chromatogr 5(2): 78–82. Nathwani D, Wood MJ (1993). Penicillins. A current review of their clinical pharmacology and therapeutic use. Drugs 45: 866–894.
N-Benzylpiperazine Stimulant C11H16N2 = 176.3 CAS—2759-28-6 IUPAC Name 1-(Phenylmethyl)piperazine Synonyms 1-Benzyl-1,4-diazacyclohexane; 1-benzylpiperazine; BZP. Street Names A2; Bliss; Charge; Frenzy; Herbal ecstasy; Legal E; Legal X. Chemical Properties Pale yellow viscous liquid. Mp 17 to 20 . pKa 9.59 [Bishop et al. 2005]. Log P (octanol/water), 1.36 [Meylan, Howard 1995]. Human urine samples were stable for up to 3 months at –20 [Nordgren, Beck 2004]. There was no indication of instability of processed samples over a time period of 30 h. Freeze–thaw stability was established [Peters et al. 2003]. N-Benzylpiperazine Hydrochloride C11H16N2,HCl = 212.8 CAS—72878-35-4 Chemical Properties White solid.
N-Benzylpiperazine Dihydrochloride
C11H16N2,2HCl = 249.3 CAS—5321-63-1 Chemical Properties White solid. Mp 287 to 292 .
Ultraviolet Spectrum Neutral—211 nm. Mass Spectrum Principal ions at m/z 91, 134, 56, 176, 120, 118, 119, 77, 91, 146, 134, 132, 85, 56, 218, 175 (AC derivative), Staack et al. [2002]; 91, 272, 181, 56, 69, 175, 146, 196 (TFAA derivative), Wikstrom et al. [2004]; 91, 281, 372, 175, 295 (HFBA derivative).
Quantification Blood GC-MS Column: HP-5MS 5% phenylmethylsilicone (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 0.9 mL/min. EI ionisation, SIM acquisition mode. Limit of quantification, 0.02 mg/g [Wikstrom et al. 2004]. Plasma GC-MS Column: HP-5MS capillary 5% phenylmethylsiloxane (30 m 0.25 mm i.d., 250 nm). Carrier gas: He, 0.6 mL/min. Temperature programme: 100 to 250 at 10 /min to 310 at 30 /min for 1 min. EI ionisation at 70 eV, SIM acquisition mode. Limit of quantification, 5 mg/L; limit of detection, approx. 1 mg/L [Peters et al. 2003]. Column: HP-5MS (19 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1 mL/ min. Temperature programme: 90 for 1 min to 300 at 15 /min for 5 min. EI ionisation at 70 eV. Limit of quantification not reported [de Boer et al. 2001]. Urine GC-MS Column: Fused silica capillary (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1 mL/min. Temperature programme: 80 for 1 min to 250 at 10 /min. EI ionisation at 70 eV. Limit of detection, 20 mg/L for BZP and 500 mg/L for 3-hydroxy-BZP and 4-hydroxy-BZP [Tsutsumi et al. 2005]. LC-MS Direct injection, positive ion mode, MRM acquisition mode. Limit of detection, 8.8 mg/L (screening), 0.9 mg/L (confirmation) [Nordgren et al. 2005]. Column: SCX (150 0.2 mm i.d.). Mobile phase: acetonitrile : 40 mmol/L ammonium acetate (pH 4; 75 : 25), flow rate 0.15 mL/min. ESI, positive ion mode, SIM acquisition mode. Limit of detection, 0.2 mg/L for BZP and 0.4 mg/L for 3-hydroxyBZP and 4-hydroxy-BZP [Tsutsumi et al. 2005]. Column: HyPURITY Advance (30 2.1 mm i.d., 3 mm). Mobile phase: 10 mmol/L ammonium acetate : methanol (95 : 5 at 0.1 min to 20 : 80 at 1 min to 95 : 5 for 5 min), flow rate 400 mL/min. APCI, positive ion mode. Retention time: 1.44 min. Limit of detection, 0.9 mg/L [Nordgren, Beck 2004]. Other GC-MS Rat Urine. Column: Fused silica capillary DB-5MS (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1 mL/min. Temperature programme: 80 for 1 min to 250 at 10 /min. EI ionisation at 70 eV. Limit of detection not reported [Tsutsumi et al. 2006]. LC-MS Rat Urine Column: SCX (150 0.2 mm i.d.). Mobile phase: acetonitrile : 40 mmol/L ammonium acetate (pH 4; 75 : 25), flow rate 0.15 mL/min. ESI, positive ion mode. Limit of detection not reported [Tsutsumi et al. 2006]. Disposition in the Body Metabolised by CYP2D6 to form 4-hydroxy-BZP, 3-hydroxy-BZP, 4-hydroxy-3-methoxy-BZP, piperazine, benzylamine and N-benzylethylenediamine. The 4-hydroxy-BZP and 4-hydroxy-methoxy-BZP metabolites are also excreted as glucuronic and/or sulfuric acid conjugates in urine. BZP has been measured in 13 non-fatal (0.02 to 1.2 mg/g blood) and 1 fatal instance of ingestion (1.7 mg/g blood) [Wikstrom et al. 2004]. Note For further reading on the metabolism of BZP, see Staack et al. [2002]. BZP is a metabolite of the antidepressant drug piberaline [Olajos, Sztaniszlav 1986]. Toxicity There have been few published cases of fatal BZP toxicity. It stimulates the release of dopamine and noradrenaline and also inhibits dopamine, noradrenaline and serotonin uptake. It acts as a CNS stimulant, producing effects comparable to amfetamine [Maurer et al. 2004; Wikstrom et al. 2004]. Bishop S et al. (2005). Simultaneous separation of different types of amphetamine and piperazine designer drugs by capillary electrophoresis with a chiral selector. J Forensic Sci 50: 326–335. deBoer D et al. (2001). Piperazine-like compounds: a new group of designer drugs-of-abuse on the European market. Forensic Sci Int 121: 47–56. Maurer HH et al. (2004). Chemistry, pharmacology, toxicology, and hepatic metabolism of designer drugs of the amphetamine (ecstasy), piperazine, and pyrrolidinophenone types: a synopsis. Ther Drug Monit 26: 127–131. Meylan WM, Howard PH (1995). Atom/fragment contribution method for estimating octanol– water partition coefficients. J Pharm Sci 84: 83–92. Nordgren HK, Beck O (2004). Multicomponent screening for drugs of abuse: direct analysis of urine by LC-MS-MS. Ther Drug Monit 26: 90–97. Nordgren H K et al. (2005). Application of direct urine LC-MS-MS analysis for screening of novel substances in drug abusers. J Anal Toxicol 29: 234–239. Olajos S, Sztaniszlav D (1986). Gas chromatographic method for determination of a piperazine derivative (Trelibet) and its metabolites in human plasma and urine. J Chromatogr 378: 155–162. Peters FT et al. (2003). Screening for and validated quantification of amphetamines and of amphetamine- and piperazine-derived designer drugs in human blood plasma by gas chromatography/ mass spectrometry. J Mass Spectrom 38: 659–676. Staack RF et al. (2002). Studies on the metabolism and toxicological detection of the new designer drug N-benzylpiperazine in urine using gas chromatography-mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 773: 35–46. Tsutsumi H et al. (2005). Development of simultaneous gas chromatography-mass spectrometric and liquid chromatography–electrospray ionization mass spectrometric determination method for the new designer drugs, N-benzylpiperazine (BZP), 1-(3-trifluoromethylphenyl)piperazine (TFMPP) and their main metabolites in urine. J Chromatogr B Analyt Technol Biomed Life Sci 819: 315–322. Tsutsumi H et al. (2006). Metabolism and the urinary excretion profile of the recently scheduled designer drug N-benzylpiperazine (BZP) in the rat. J Anal Toxicol 30: 38–43. Wikstrom M et al. (2004). A2 (N-benzylpiperazine) a new drug of abuse in Sweden. J Anal Toxicol 28: 67–70.
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:33:8
Beryllium
Bephenium Hydroxynaphthoate Anthelmintic C28H29NO4 = 443.5 CAS—7181-73-9 (bephenium); 3818-50-6 (hydroxynaphthoate) IUPAC Name N,N-Dimethyl-N-(2-phenoxyethyl)benzenemethanaminium 3hydroxy-2-naphthoate Synonym Naphthammonum Proprietary Name Alcopar(a)
971
Berberine Sulfate C20H18NO4,HSO4 = 433.4 CAS—633-66-9 Synonyms Berberine acid sulfate; berberine bisulfate. Chemical Properties Bright yellow acicular crystals or dark yellow powder.
Soluble 1 in about 100 of water; slightly soluble in ethanol. Colour Tests Mandelin’s test—blue-green!brown; Marquis test—green. Thin-layer Chromatography System TA—Rf 0.07; system TL—Rf 0.00; system TAE—Rf 0.04; system TAF—Rf 0.56; system TAJ—Rf 0.00; system TAK—Rf 0.00; system TAL—Rf 0.20 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 2070. High Performance Liquid Chromatography System HY—RI 327. Ultraviolet Spectrum Aqueous acid—264 (A11¼554b), 345 nm.
Chemical Properties A yellow to greenish-yellow crystalline powder, which gives a green fluorescence when examined under ultraviolet light. Mp 170 to 171 . Practically insoluble in water; soluble 1 in 50 of ethanol. Log P (octanol/water), 0.5. Colour Tests Mandelin’s test—green; Marquis test—red-violet. Thin-layer Chromatography System TA—Rf 0.77 (acidified potassium permanganate solution, positive). Ultraviolet Spectrum Methanol—234 (A11¼1300b), 263, 270, 283, 295, 352 nm.
Infrared Spectrum Principal peaks at wavenumbers 1233, 768, 1653, 1590, 858, 726 cm1 (KBr disk). Quantification Urine Colorimetry See Rogers [1958]. Disposition in the Body Bephenium hydroxynaphthoate is poorly absorbed after oral administration; less than 1% of a dose is excreted in the urine in 24 h. Dose The equivalent of 2.5 g of bephenium, as a single dose. Rogers EW (1958). Excretion of bephenium salts in urine of human volunteers. BMJ 2: 1576–1577.
Berberine Alkaloid [C20H18NO4]þ = 336.4 CAS—2086-83-1 (C20H18NOþ4); 117-74-8 (C20H18NO4OH) IUPAC Name 5,6-Dihydro-9,10-dimethoxybenzo[g]-1,3-benzodioxolo[5,6-a] quinolizinium
Chemical Properties A quaternary alkaloid present in Hydrastis, in various species of Berberis, and in many other plants. Yellow crystals. Mp 145 . Soluble 1 in 4.5 of water and 1 in 100 of ethanol; very slightly soluble in ether. Log P (octanol/ water), 2.1. Berberine Hydrochloride C20H18ClNO4,2H2O = 407.8 CAS—633-65-8 (anhydrous); 5956-60-5 (dihydrate) Synonym Berberine chloride Chemical Properties Bright yellow acicular crystals or powder. Soluble 1 in 400
of water; freely soluble in boiling water; soluble in ethanol; practically insoluble in chloroform and ether.
Infrared Spectrum Principal peaks at wavenumbers 1505, 1271, 1234, 1030, 1587, 1098 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 321, 278, 320, 292, 306, 191, 322, 304. Quantification Plasma HPLC UV detection (l¼346 nm). Limit of detection, 0.4 ng [Zeng 1999]. Limit of detection, 18.1 mg/L [Chen, Chang 1995]. Urine GC–MS Limit of detection, 1 mg/L [Miyazaki et al. 1978]. HPLC Limit of detection, 2.3 mg/L [Chen, Chang 1995]. Bile HPLC Limit of detection, 90.4 mg/L [Chen, Chang 1995]. Disposition in the Body After oral administration, 85%) and undergoes moderate first pass metabolism. It is extensively metabolised to 4 metabolites; the principal metabolite makes up 16% of the dose and 3 minor metabolites in trace amounts. It is excreted in urine (72%) mostly as metabolites and small amounts of the unchanged drug (400 units. Half-life Plasma, 2 to 4 h. Volume of Distribution 0.35 to 0.45 L/kg. Clearance Plasma, 119 to 128 mL/min/m2. Protein Binding 1%. Note For review of bleomycin, see Dorr [1992]. Dose Usually 15 to 60 units weekly intravenously. The maximum total cumulative dose is 400 or 500 units. Bitran JD (1985). Intraperitoneal bleomycin. Pharmacokinetics and results of a phase II trial. Cancer 56: 2420–2423. Dorr RT (1992). Bleomycin pharmacology: mechanism of action and resistance, and clinical pharmacokinetics. Semin Oncol 19: 3–8. Shiu GK et al. (1979). Rapid high-performance liquid chromatographic determination of bleomycin A2 in plasma. J Pharm Sci 68: 232–234.
very hygroscopic powder. It is very soluble in water; slightly soluble in ethanol; practically insoluble in acetone and in ether.
Boldenone
Ultraviolet Spectrum Aqueous acid (0.2 mol/L H2SO4)—290 nm: basic— 290 nm.
Anabolic Steroid C19H26O2 = 286.4 CAS—846-48-0 IUPAC Name (17b)-17-Hydroxyandrosta-1,4-dien-3-one Synonym Dehydrotestosterone
Chemical Properties Crystals. Mp 164 to 166 . Log P (octanol/water), 3.05. Boldenone Acetate C21H28O3 = 328.5 Chemical Properties Crystals. Mp 151 to 153 .
Boldenone Benzoate C26H30O5 = 422.5
Infrared Spectrum Principal peaks at wavenumbers 1653, 1548, 1118, 618 cm1 (sulfate salt).
Boldenone Undecylenate C30H44O3 = 452.7 CAS—13103-34-9 Synonyms Ba-29038; 10-undecenoate. Proprietary Names Parenabol; Vebonol.
Thin-layer Chromatography System TA—Rf 0.98; system TE—Rf 0.98; system TAJ—Rf 0.90; system TAK—Rf 0.88; system TAL—Rf 0.98; system TAM—Rf 0.99. Gas Chromatography System GAG—boldenone RRT 1.05, boldenone undecylenate RRT 2.62 (both relative to testosterone); system GAI—boldenone RRT 0.961, 5b-androst-1-en-17b-ol-3-one metabolite RRT 0.96 (both relative to 17amethyl-5a-androstan-3b, 17b-diol); system GAR—boldenone retention time 12.8 min, boldenone acetate retention time 13.6 min, boldenone benzoate retention time 18.7 min, boldenone undecylenate retention time 22.4 min. Column: silica SE54, cross-linked 5% phenyl, 1% vinyl methyl silicone (17 m 0.2 mm i.d., 0.33 mm). Carrier gas: He, flow rate 1 mL/min. Split 1:10. MS detection (EI), SIM. Internal standard: 5a-androstane-3,17-dione,bis-TMS. RI: boldenone (TMS derivative) 2685. Column: silica OV1, cross-linked methyl silicone (20 m 0.25 mm i.d., 0.33 mm). Carrier gas: He, flow rate 1 mL/min. MS detection (EI), SIM. Internal standard: 5a-androstane-3,17-dione,bis-TMS. RI: boldenone (TMS derivative) 2672 [Schanzer, Donike 1992]. High Performance Liquid Chromatography System HAR—RRT 0.74 (relative to testosterone); system HATa—boldenone undecylenate RRT 1.94(relative to testosterone); system HATb—boldenone RRT 0.76(relative to testosterone). Column: C18 Nucleosil (250 10 mm i.d., 7 mm). Mobile phase: (A) water, (B) acetonitrile : water (90:10). Elution programme: (A:B) (70:30) to (20:80) over 25 min, flow rate 6 mL/min. Retention time: 13.1 min [Schanzer, Donike 1992]. Ultraviolet Spectrum Aqueous acid—248 nm; aqueous base—250 nm; ethanol—243 nm.
B
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:35:33
986
Boldione
Boldione Anabolic Steroid C19H24O2 = 284.4 CAS—897-06-3 IUPAC Name (8R,9S,10R,13S,14S)-10,13-Dimethyl-7,8,9,11,12,14,15,16-octahydro-6H-cyclopenta[a]phenanthrene-3,17-dione Synonyms Androstadienedione; 1,4-androstadiene-3,17-dione; 1-dehydroandrostenedione.
B
Infrared Spectrum Principal peaks at wavenumbers 1654, 1400, 1055 cm1; (acetate)1725, 1231, 1034 cm1; (benzoate)1658, 1276, 716 cm1; (undecylenate) 1725, 1654, 1175 cm1 (KBr disks). Chemical Properties Prohormone of the anabolic steroid boldenone. Log P (octanol/water), 2.92 [Meylan, Howard 1995]. Stability of boldione in bovine urine was verified for 4 weeks in the dark at –20 [Draisci et al. 2003]. Ultraviolet Spectrum
Mass Spectrum Principal ions at m/z 122, 121, 91, 123, 147, 77, 107, 93.
Quantification Urine GC–MS Column: 5% phenylmethylpolysiloxane Ultra-2 (30 m 0.2 mm i. d., 0.33 mm), cross-linked capillary. Temperature programme: 150 held for 2 min, step rate of 20 /min to 300 , held for 2 min. MS detection negative chemical ionisation (NCI): Internal standard: 1,2-d2-testosterone. Retention time: boldenone metabolite, 9.17 min. Limit of detection, 2 ppb [Choi et al. 1998]. Disposition in the Body Boldenone is metabolised to 5b-androst-1-en-17bol-3-one, 5b-androst-1-ene-3a,17b-diol, 5b-androst-1-en-3a-ol-17-one, 5bandrost-1-ene-3,17-dione, androsta-1,4-diene-6b,17b-diol-3-one, androsta-1,4dien-6b-ol-3,17b-dione and 5b-androstan-3a-ol-17-one, by 5b-reduction. Excreted, via the kidneys, to a large extent as a conjugate. Choi MH et al. (1998). Determination of four anabolic steroid metabolites by gas chromatography/ mass spectrometry with negative ion chemical ionization and tandem mass spectrometry. Rapid Commun Mass Spectrom 12: 1749–1755. Schanzer W, Donike M (1992). Metabolism of boldenone in man: gas chromatographic/mass spectrometric identification of urinary excreted metabolites and determination of excretion rates. Biol Mass Spectrom 21: 3–16.
Quantification Urine GC-MS Column: Ultra-2 cross linked phenylmethylsiloxane (17 m 0.2 mm i.d., 0.33 mm). Carrier gas: He, 1.0 mL/min. Temperature programme: 180 for 2 min to 260 at 5 /min to 310 at 6 /min for 2 min. EI ionisation, scan mode. Limit of detection, 4.6 and 4.2 mg/L for boldione and boldenone, respectively [Kim et al. 2006]. LC-MS Column: Agilent Hypersil ODS C18 (150 2.0 mm i.d., 5.0 mm). Mobile phase: acetonitrile : water (20 : 80 to 45 : 55 at 5 min). ESI, positive ion mode. Limit of detection not reported [Kim et al. 2006]. Other GC-MS Bovine Urine. Column: 30 m 0.25 mm i.d., 0.25 mm. Carrier gas: He, 1.0 mL/min. Temperature programme: 120 for 2 min to 250 at 15 /min to 300 at 5 /min for 10 min. EI ionisation, negative ion mode. Limit of detection, 0.05 mg/L [Le Bizec et al. 2006]. HPLC Bovine Liver and Kidney. Column: SYNERGY fusion RP 80 (150 4.6 mm i.d., 4.0 mm). Mobile phase: acetonitrile : water (20 : 80 for 9 min to 40 : 60 at 42 min to 20 : 80 at 47 min). Retention time: approx. 41.9 min. Limit of detection, 7.11 mg/L [Merlanti et al. 2007]. LC-MS Bovine Urine. Column: Uptisphere C18 TF (50 2.1 mm i.d., 3.0 mm). Mobile phase: methanol : 0.5% acetic acid (10 : 90 to 0 : 100 at 9 min for 6 min). ESI, positive ion mode, SRM acquisition mode. Limit of detection not reported [Le Bizec et al. 2006]. Equine Urine. Column: Supelcosil LC-8-DB reversed phase (100 2.1 mm i.d., 3.0 mm). Mobile phase: 0.1% acetic acid : methanol (60 : 40 to 0 : 100 at 5 min for 5 min), flow rate 0.2 mL/min. API, ESI, positive ion mode, MRM acquisition mode. Retention time: 4.88 min. Limit of detection, 5 mg/L [Yu et al. 2005]. Bovine Urine. Column: Allure C18 reversed phase. Mobile phase: acetonitrile : 5 mol/L ammonium acetate (60 : 40), flow rate 130 mL/min. APCI, positive ion mode. Limit of quantification, 0.2 mg/L [Draisci et al. 2003]. Note Neoformation of 17a-boldenone in bovine faeces requires that only uncontaminated urine be analysed, see Pompa et al. [2006]. There is a natural occurrence of various boldenone metabolites in the urine of non-treated animals; for more information, see Le Bizec et al. [2006]. Disposition in the Body Rapidly metabolised to boldenone and 2 minor metabolites, M2 and M3. M2 appears to be a hydroxylated version of boldione and M3 the reduced form of M2. A healthy male volunteer (age 26 years, weight 71 kg) was administered one 100 mg boldione tablet orally. Urine samples were collected over 48 h and analysed by GC-MS. The maximum amount of boldione excreted occurred after 1.8 h and was 16.1 mg (in 750 mL urine), whereas figures for boldenone (the main metabolite) were 6.8 mg (in
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:35:37
Borates 700 mL urine) after 3.6 h. The total amount excreted after the 48 h period was 34.5 mg for boldione and 16.0 mg for boldenone (in 4.16 L urine) [Kim et al. 2006]. Draisci R et al. (2003). Confirmatory analysis of 17beta-boldenone, 17alpha-boldenone and androsta-1,4-diene-3,17-dione in bovine urine by liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 789: 219–226. Kim Y et al. (2006). Characterization of boldione and its metabolites in human urine by liquid chromatography/electrospray ionization mass spectrometry and gas chromatography/mass spectrometry. Rapid Commun Mass Spectrom 20: 9–20. Le Bizec B et al. (2006). Criteria to distinguish between natural situations and illegal use of boldenone, boldenone esters and boldione in cattle 1. Metabolite profiles of boldenone, boldenone esters and boldione in cattle urine. Steroids 71: 1078–1087. Merlanti R et al. (2007). An in vitro study on metabolism of 17beta-boldenone and boldione using cattle liver and kidney subcellular fractions. Anal Chim Acta 586: 177–183. Meylan WM, Howard PH (1995). Atom/fragment contribution method for estimating octanol– water partition coefficients. J Pharm Sci 84: 83–92. Pompa G et al. (2006). Neoformation of boldenone and related steroids in faeces of veal calves. Food Addit Contam 23: 126–132. Yu NH et al. (2005). Screening of anabolic steroids in horse urine by liquid chromatography–tandem mass spectrometry. J Pharm Biomed Anal 37: 1031–1038.
Bopindolol Antihypertensive C23H28N2O3 = 380.5 CAS—62658-63-3 IUPAC Name 1-[(1,1-Dimethylethyl)amino]-3-[(2-methyl-1H-indol-4-yl)oxy]2-propanol benzoate ester
987
1.2-g dose antipyrine on day 1 and 2-mg bopindolol on day 3. The patients with cirrhosis reached a mean peak plasma concentration of 5.8 mg/L at 1.6 h and the healthy volunteers 6.9 mg/L within 1.5 h. No significant difference was observed [Wensing et al. 1990]. Half-life Mean, 4.4 h (range 3.1 to 6.0 h). Volume of Distribution Mean, 202 L (range, 102 to 504 L) after oral administration; 148 L intravenously. Clearance After single dose administration, plasma clearance is 30.9 L/h and for multiple dosing, 38.0 L/h. Dose Between 0.5 and 4 mg. Holmes D et al. (1991). Steady state pharmacokinetics of hydrolysed bopindolol in young and elderly men. Eur J Clin Pharmacol 41: 175–178. Humbert H et al. (1987). Column liquid chromatographic determination of hydrolysed bopindolol, in the picogram per millilitre range in plasma, using cartridge extraction and dual electrochemical detection. J Chromatogr 422: 205–215. Wensing G et al. (1990). Pharmacokinetics after a single oral dose of bopindolol in patients with cirrhosis. Eur J Clin Pharmacol 39: 569–572.
Borates Anion Boric Acid H3BO3 = 61.8 CAS—10043-35-3 Synonyms Boracic acid; orthoboric acid; occurs in nature as the mineral sassolite. Proprietary Name Borofax Chemical Properties Colourless, odourless, transparent crystals, or white gran-
ules or powder. Mp 171 . Very soluble in water, alcohol, glycerol. Used for weatherproofing wood and fireproofing fabrics; as a preservative; in manufacture of cements, crockery, porcelain, enamels, leather, soaps, artificial gems; in cosmetics; photography; hardening steel; as an insecticide. Sodium Borate Decahydrate
Na2B4O7,10H2O = 381.2 CAS—1330-43-4 Synonyms Sodium biborate; sodium pyroborate; sodium tetraborate; fused
sodium borate, borax glass, or fused borax (anhydrous form). Chemical Properties It is soluble in ether and methylene chloride. Bopindolol Maleate C27H32N2O7 = 496.6 Proprietary Name Sandonorm
Bopindolol Malonate C26H32N2O7 = 484.5 CAS—82857-38-3 Synonyms Bopindolol hydrogen malonate; LT-31-200. Proprietary Name Wandonorm
Ultraviolet Spectrum Principal peaks at 221, 267 nm.
Quantification Plasma HPLC Electrochemical detection. Limit of detection, 0.025 mg/L [Humbert et al. 1987]. Disposition in the Body Bopindolol is rapidly hydrolysed in plasma and no measurable amounts of the parent drug can be detected after oral administration. The two main metabolites identified are benzoic acid and an active hydrolysed metabolite. Therapeutic Concentration Twenty young male volunteers, aged between 18 and 42 years, and 20 elderly males, 62 to 83 years old, were administered with a single 1-mg dose of bopindolol once daily for 7 days. Mean peak plasma concentrations of 4.5 and 5.2 mg/L were reached for the young and elderly volunteers, respectively. All peak concentrations were observed approximately 1.5 h after administration. No significant differences were observed between the young and elderly [Holmes et al. 1991]. Fourteen patients with cirrhosis (male and female, aged between 40 and 66 years) and 15 healthy volunteers (21 to 28 years) were administered with a
Proprietary Names Borax; Jaikin (decahydrate). Chemical Properties Hard odourless crystals, granules or crystal powder. Mp
75 ; loses 5H2O at 100 and 9H2O at 150 ; becomes anhydrous at 320 . Soluble in water; very soluble in glycerol; insoluble in alcohol. Used for soldering metals; in manufacture of glazes and enamels; in tanning; in cleaning compounds; artificially ageing wood; as a preservative; fireproofing fabrics and wood; in cockroach control; pharmaceutical aid.
Colour Test Test applicable to gastric contents and scene residues. A portion of the sample is acidified with 1 mol/L hydrochloric acid and applied to turmeric paper (prepared by soaking strips of filter paper in turmeric spice dissolved in methanol (10 g/L) and allowing them to dry at room temperature)—A brown-red colour, which intensifies when the paper dries, indicates the presence of borate; moisten the filter paper with 4 mol/L ammonium hydroxide solution—A colour change to green-black suggests the presence of borates (oxidising agents such as bromates, chlorates, iodates and nitrates interfere by bleaching the turmeric). Limit of detection, 20 mg/L. Confirmation Test Stomach contents, 5 mL, is filtered into a 10 mL glass tube and 0.5 mL of the filtrate is added to a clean glass tube before slowly adding 0.5 mL of carminic acid solution (0.5 g/L in concentrated sulfuric acid) down the side of the tube so that it forms a layer underneath the sample—A blue-violet ring at the interface of the two layers suggests the presence of borate. Strong oxidising agents (e.g. bromates, chlorates, iodates and nitrates) also give a positive result with this test. Quantification Specimen collection: Blood—10 mL EDTA tube; urine—20 mL plastic universal container; gastric contents are useful in postmortem examinations. Blood ICP-AES Nebuliser gas: 0.70 L/min (l ¼ 249.7 nm). Limit of detection not reported [Svantesson et al. 2002]. ICP-MS Plasma gas: Ar, 15 L/min. Auxiliary gas: Ar, 0.8 L/min. Nebuliser gas: Ar, 0.9 L/min. Limit of detection, 5 mg/L [Moreton, Delves 1999]. Jobin-Yvon JY 48 spectrometer. Plasma-Therm source. Limit of detection, 0.06 mmol/L [Mauras et al. 1986]. Note For a DEAE cellulose method for the analysis of boron-10, see Schremmer and Noonan [1987]. For a colorimetric method for the detection of boron in blood, see Dill et al. [1977]. For a spectrophotometric method for the detection of boron in blood, see Edwall et al. [1979]. Plasma ICP-MS Plasma gas: Ar, 15 L/min. Auxiliary gas: Ar, 0.8 L/min. Nebuliser gas: Ar, 0.9 L/min. Limit of detection, 5 mg/L [Moreton, Delves 1999]. Serum FAAS Gas: nitrous oxide–acetylene. Perkin-Elmer (l ¼ 249.7 nm). Limit of detection, 15 mg/L [Bader, Brandenberger 1968]. Gas: air–acetylene. Limit of quantification, 0.2 mg [Siemer 1982]. Urine FAAS See Serum [Bader, Brandenberger 1968; Siemer 1982]. ICP-AES See Blood [Svantesson et al. 2002]. CSF FAAS See Serum [Bader, Brandenberger 1968]. Dialysis Fluid ICP-MS See Blood [Mauras et al. 1986]. Brain ICP-MS See Plasma [Moreton, Delves 1999]. Note For a review of the considerations for measuring boron at low concentrations, see Downing et al. [1998]. For a spectrometric method for the determination of boron, see Aznarez and Mir [1984]. Disposition in the Body Boric acid is well absorbed through mucous membranes and damaged skin, but not when the skin is intact. Intestinal absorption of
B
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:35:41
988
B
Botulinum Toxin
boron is rapid and almost complete following ingestion of boric acid or borates, with peak blood concentrations being reached within 2 h [Locatelli et al. 1987]. The main route of elimination of borates is via the kidneys and they are excreted substantially unchanged, regardless of the route of administration. Urinary excretion accounts for 85 to 100% of a dose within 5 to 7 days. Smaller amounts are excreted in the faeces and in sweat. Boric acid is not metabolised in humans or animals [Murray 1998]. Note For a possible role of dietary boron, see Hunt [1998]. Normal Concentrations Blood—10000 mg/kg [Iezhitsa et al. 2002]. Use Bromantane is used as a performance-enhancing agent for muscles in athletes and as an immunostimulant [Burnat et al. 1997]. Burnat P et al. (1997). Bromontan, a new doping agent. Lancet 350: 963–964. Iezhitsa IN et al. (2002). Toxic effect of single treatment with bromantane on neurological status of experimental animals. Bull Exp Biol Med 133: 380–383.
Bromazepam Anxiolytic, Benzodiazepine, Tranquilliser C14H10BrN3O = 316.2 CAS—1812-30-2 IUPAC Name 7-Bromo-5-pyridin-2-yl-1,3-dihydro-1,4-benzodiazepin-2-one Synonyms Bromazepamas; bromazepamum; 7-bromo-1,3-dihydro-5-(2-pyridinyl)-2H-1,4-benzodiazepin-2-one; Ro-5-3350. Proprietary Names Compendium; Creosedin; Durazanil; Lectopam; Lexomil; Lexotan; Lexotanil; Normoc.
Infrared Spectrum Principal peaks at wavenumbers 1685, 825, 750, 802, 1315, 1230 cm1.
Clarke's Analysis of Drugs and Poisons Chapter No. B Dated: 15/3/2011 At Time: 21:35:53
Bromazepam Mass Spectrum Principal ions at m/z 236, 317, 315, 288, 316, 286, 208, 78 (bromazepam); 79, 78, 52, 105, 304, 314, 316, 51 (3-hydroxybromazepam).
Quantification Blood GC Column: SE-54 fused silica 5% phenyl methyl silicone (25 m 0.31 mm i.d., 0.17 mm). Carrier gas: He, 2–3 mL/min. Temperature programme: 200 for 1 min to 290 at 10 /min. NPD. Retention time: 6.99 min. [Lillsunde, Sepp€al€a 1990]. GC-MS Column: DB5-MS (20 m 0.18 mm i.d., 0.18 mm). Temperature programme: 70 for 0.5 min to 320 at 40 /min for 1 min. TOF-MS. Limit of detection, 0.63 ng [Aebi et al. 2002]. Column: HP-1 (13 m 0.2 mm i.d., 0.33 mm). Carrier gas: He, 60 kPa. Temperature programme: 100 to 250 at 30 /min to 300 at 20 /min for 5 min. SIM acquisition mode (m/z 317). Limit of detection, 2–5 ng/g [Zhang et al. 1996]. LC-MS Column: Restek Allure C18 (150 3.2 mm i.d., 5 mm). Mobile phase: 5 mmol/L ammonium acetate (pH 4.75) : acetonitrile : methanol (90 : 5 : 5 to 50 : 25 : 25 at 7 min to 10 : 45 : 45 at 27 min for 3 min to 90 : 5 : 5 at 31 min), flow rate 0.45 mL/min. DAD-ESI detection. Limit of detection, 6 mg/L [Dussy et al. 2006]. Column: Uptisphere ODB 18. Mobile phase: 2 mmol/L formate buffer and acetonitrile. Limit of detection, 0.45 mg/ L can be fatal. Other toxic effects include tachycardia, hallucinations and loss of consciousness. A 35-year-old male was found dead next to an empty box of thirty 150 mg slow-release bupropion tablets. Blood and urine concentrations (mg/L) of bupropion and its metabolites were as shown below: Sample
Bupropion
Hydroxybupropion
Threohydrobupropion
Femoral blood Urine
N.D.
5.8
30.4
42.9
100
617
N.D. Not detected [Mercerolle et al. 2008].
In 3 fatal overdose cases involving bupropion, the victims had blood concentrations of 4.0, 0.16 and 4.2 mg/L bupropion. In the 2 cases where bupropion was highest, the lethal dose was estimated to be 95% of an administered dose is excreted in urine, with ~3% unchanged and 5% as FBAL, the major metabolite. 2.5% of the dose is recovered in faeces. Therapeutic Concentration A single oral dose of 892 to 2510 mg/m2 was administered and taken within 30 min of the end of a meal. Mean peak plasma concentrations of the drug ranged from 2.4 to 3.9 mg/L and were reached within ~1.5 h [Roche Laboratories Inc. 1998]. Toxicity In patients treated with doses of up to 3514 mg/m2 daily (i.e. cases of acute overdose), nausea, vomiting, diarrhoea and bleeding were observed. Half-life 0.5 to 1 h. Protein Binding 70%. Metabolic reactions, catalysed by P450 isoforms CYP3A4 and CYP2C8, include epoxidation to form the 10,11-epoxide, which is active, followed by hydroxylation to trans-10,11dihydro-10,11-dihydroxycarbamazepine; glucuronic acid conjugation also occurs. The rate of metabolism is higher in children than in adults and levels of the epoxide may be higher in children than in adults. Approximately 25% of a dose is excreted in the urine as the dihydroxy metabolite, together with 2% as the 10,11epoxide and 99%. Note For a review of carbenoxolone, see Pinder et al. [1976]. Dose 150 to 300 mg of carbenoxolone sodium daily. Baron JH et al. (1975). Factors affecting the Absorption of Carbenoxolone in Patients with Peptic Ulcer. In: Jones FA, Parke DV, eds. 4th Symposium on Carbenoxolone. London: Butterworths, 115–124 Downer HD et al. (1970). The absorption and excretion of carbenoxolone in man. J Pharm Pharmacol 22: 479–487. Peskar BM et al. (1976). Radioimmunoassay for carbenoxolone. J Pharm Pharmacol 28: 720–721. Pinder RM et al. (1976). Carbenoxolone: a review of its pharmacological properties and therapeutic efficacy in peptic ulcer disease. Drugs 11: 245–307. Rhodes C, Wright PA (1974). A gas chromatographic determination of carbenoxolone in human serum. J Pharm Pharmacol 26: 894–898.
Carbetapentane Cough Suppressant C20H31NO3 = 333.5 CAS—77-23-6 IUPAC Name 2-(Diethylaminoethoxy)ethyl 1-phenylcyclopentanecarboxylate Synonym Pentoxyverine
1045
Mass Spectrum Principal ions at m/z 86, 91, 87, 145, 58, 144, 30, 44.
C Dose 25 to 150 mg of carbetapentane citrate daily.
Carbidopa Dopa-Decarboxylase Inhibitor C10H14N2O4, H2O = 244.2 CAS—28860-95-9 (anhydrous); 38821-49-7 (monohydrate) IUPAC Name (2S)-3-(3,4-Dihydroxyphenyl)-2-hydrazinyl-2-methylpropanoic acid hydrate Synonyms HMD; (aS)-a-hydrazino-3,4-dihydroxy-a-methylbenzenepropanoic acid monohydrate; ()-L-a-Methyldopa hydrazine; Mk-486. Proprietary Name It is an ingredient of Sinemet.
Chemical Properties A white or creamy-white powder. Mp 203 to 205 , with decomposition. Soluble 1 in 500 of water; practically insoluble in ethanol, chloroform, and ether; freely soluble in 3 mol/L hydrochloric acid. Log P (octanol/water), 0.1. Extraction yield (chlorobutane), 0 [Demme et al. 2005]. Colour Tests Ammoniacal silver nitrate (on warming) —silver mirror; ferric chloride—brown; Folin-Ciocalteu reagent—blue; Liebermann’s reagent—black; Marquis test—brown; methanolic potassium hydroxide—orange; Millon’s reagent (cold)—orange; Nessler’s reagent—orange!black; palladium chloride—orange!brown; potassium dichromate (method 1)—red. Thin-layer Chromatography System TD—Rf 0.00; system TE—Rf 0.00; system TF—Rf 0.02; system TAD—Rf 0.04. Gas Chromatography System GA—carbidopa, not eluted; carbidopa-Me2 RI 1660. High Performance Liquid Chromatography System HY—RI 190. Ultraviolet Spectrum Methanolic acid—282 nm (A11¼130a); aqueous alkali—291 nm.
Chemical Properties Log P (octanol/water), 4.2. Carbetapentane Citrate C20H31NO3, C6H8O7 = 525.6 CAS—23142-01-0 Proprietary Names Atussil; Germapect; Sedotussin; Toclase; Tuclase; Tussa-
Tablinen.
Chemical Properties A white crystalline powder. Mp 93 . Freely soluble in water
and chloroform; soluble in ethanol, acetone and ethyl acetate; practically insoluble in ether and in benzene. Colour Tests Liebermann’s reagent—red-orange; Mandelin’s test—brown (slow); Marquis test—orange (slow). Thin-layer Chromatography System TA—Rf 0.48; system TB—Rf 0.48; system T—Rf 0.22 (Dragendorff spray, positive; acidified iodoplatinate solution, positive; Marquis reagent, brown). Gas Chromatography System GA—RI 2232; system GF—RI 2455. Ultraviolet Spectrum Aqueous acid—252, 258 (A11¼6.1a), 264 nm. No alkaline shift. Infrared Spectrum Principal peaks at wavenumbers 1625, 1121, 1260, 1525, 1290, 875 cm1 (KBr disk).
Infrared Spectrum Principal peaks at wavenumbers 1725, 1120, 1158, 1176, 694, 1234 cm1 (KBr disk).
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:44:21
1046
Carbimazole
Mass Spectrum Principal ions at m/z 123, 57, 42, 103, 44, 85, 124, 51.
Ultraviolet Spectrum Aqueous acid—291 nm (A11¼557a); aqueous alkali— 244 nm.
C Quantification Plasma HPLC Electrochemical detection. Limit of detection, 8 mg/L [Sagar, Smyth 2000]. Electrochemical detection. See Lucarelli et al. [1990]. Electrochemical detection. See Titus et al. [1990]. Electrochemical detection. Limit of detection, 15 mg/L [Nissinen, Taskinen 1982]. Urine HPLC See Plasma Titus et al. [1990]. Disposition in the Body Readily but incompletely absorbed after oral administration. About 50% of an oral dose is excreted in the urine in 48 h and 47% is eliminated in the faeces. Of the urinary material, about 30% is unchanged drug, 10 to 14% is 2-(4-hydroxy-3-methoxybenzyl)propionic acid, 10% is 2-(3,4-dihydroxybenzyl)propionic acid, 5% is 3,4-dihydroxyphenylacetone and 10% is 2-(3hydroxybenzyl)propionic acid. The metabolites are excreted mainly as glucuronide conjugates. Therapeutic Concentration Following an oral dose of 50 mg to 10 subjects, a mean peak plasma concentration of 0.2 mg/L was attained in 2 to 4 h [Vickers et al. 1974]. Half-life Plasma half-life, about 2 h. Protein Binding About 36%. Note For a review of the pharmacokinetics of carbidopa, see Pinder et al. [1976]. Dose Usually the equivalent of 75 to 150 mg of anhydrous carbidopa daily, given in combination with levodopa. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Lucarelli C et al. (1990). Simultaneous measurement of L-dopa, its metabolites and carbidopa in plasma of parkinsonian patients by improved sample pretreatment and high-performance liquid chromatographic determination. J Chromatogr 511: 167–176. Nissinen E, Taskinen J (1982). Simultaneous determination of carbidopa, levodopa and 3,4-dihydroxyphenyl-acetic acid using high-performance liquid chromatography with electrochemical detection. J Chromatogr B Biomed Appl 231: 459–462. Pinder RM et al. (1976). Levodopa and decarboxylase inhibitors: a review of their clinical pharmacology and use in the treatment of parkinsonism. Drugs 11: 329–377. Sagar KA, Smyth MR (2000). Simultaneous determination of levodopa, carbidopa and their metabolites in human plasma and urine samples using LC-EC. J Pharm Biomed Anal 22: 613–624. Titus DC et al. (1990). Simultaneous high-performance liquid chromatographic analysis of carbidopa, levodopa and 3-O-methyldopa in plasma and carbidopa, levodopa and dopamine in urine using electrochemical detection. J Chromatogr 534: 87–100. Vickers S et al. (1974). Metabolism of carbidopa (1-(-)-alpha-hydrazino-3,4-dihydroxy-alphamethylhydrocinnamic acid monohydrate), an aromatic amino acid decarboxylase inhibitor, in the rat, rhesus monkey, and man. Drug Met Disp 2: 9–22.
Infrared Spectrum Principal peaks at wavenumbers 1574, 1275, 740, 1246, 1150, 767 cm1 (KBr disk).
Mass Spectrum Principal ions at m/z 186, 114, 29, 72, 42, 113, 27, 109; thiamazole 114, 42, 72, 113, 69, 81, 54, 115.
Carbimazole Antithyroid Agent C7H10N2O2S = 186.2 CAS—22232-54-8 IUPAC Name Ethyl 3-methyl-2-sulfanylideneimidazole-1-carboxylate Synonym 2,3-Dihydro-3-methyl-2-thioxo-1H-imidazole-1-carboxylic acid ethyl ester Proprietary Names Carbazole; Carbotiroid; Neo-Mercazole; Neo-Morphazole; Neo-Thyreostat; Neo-Tireol.
Chemical Properties A white or creamy-white crystalline powder. Mp 122 to 125 . Soluble 1 in 500 of water, 1 in 50 of ethanol, 1 in 17 of acetone, 1 in 3 of chloroform and 1 in 330 of ether. Log P (octanol/water), 0.5. Colour Tests Palladium chloride—orange; to a small quantity add 1 drop of iodobismuthous acid solution—red. Thin-layer Chromatography System TA—Rf 0.72; system TB—Rf 0.01; system TD—Rf 0.63; system TE—Rf 0.42; system TF—Rf 0.47; system TAD—Rf 0.68; system TAE—Rf 0.75; system TAF—Rf 0.75 (acidified iodoplatinate solution, positive; acidified potassium permanganate solution, positive). Gas Chromatography System GA—carbimazole RI 1678, thiamazole RI 1550. High Performance Liquid Chromatography System HX—RI 318; system HAA—retention time 11.1 min.
Quantification Plasma GC AFID. Limit of detection, 30 mg/L for thiamazole [Bending, Stevenson 1978]. GC–MS Limit of detection, 2 mg/L for thiamazole [Floberg et al. 1980]. HPLC Absorbance detection at 405 nm. Limit of detection, 5 mg/L for thiamazole [Meulemans et al. 1980]. Disposition in the Body Carbimazole is rapidly and almost completely absorbed after oral administration and converted to the active metabolite thiamazole. It is almost completely excreted in the urine in 24 h as metabolites; 3-methyl-2thiohydantoin has been identified as a minor metabolite in urine and plasma. About 3% of a dose is eliminated in the faeces. Therapeutic Concentration After an oral dose of 60 mg, given to 11 subjects, peak serum concentrations of thiamazole of 0.5 to 3.4 (mean 0.9) mg/L were attained in about 0.7 to 3 h [Melander et al. 1980]. Half-life Plasma half-life, thiamazole about 3 to 5 h. Volume of Distribution Thiamazole, about 0.5 L/kg. Protein Binding Thiamazole, not significantly bound. Note For a review of the pharmacokinetics of antithyroid drugs, see Kampmann and Hansen [1981]. Dose Initially 30 to 60 mg daily; maintenance, 5 to 20 mg daily. Bending MR, Stevenson D (1978). Measurement of methimazole in human plasma using gas-liquid chromatography. J Chromatogr 154: 267–271. Floberg S et al. (1980). Determination of methimazole in plasma using gas chromatography–mass spectrometry after extractive alkylation. J Chromatogr 182: 63–70.
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:44:32
Carbocromen Kampmann JP, Hansen JM (1981). Clinical pharmacokinetics of antithyroid drugs. Clin Pharmacokinet 6: 401–428. Melander A et al. (1980). Comparative in vitro effects and in vivo kinetics of antithyroid drugs. Eur J Clin Pharmacol 17: 295–299. Meulemans A et al. (1980). Determination of methimazole in plasma by high performance liquid chromatography. J Liq Chromatogr 3(2): 287–298.
Carbinoxamine Antihistamine C16H19ClN2O = 290.8 CAS—486-16-8 IUPAC Name 2-[(4-Chlorophenyl)-2-pyridinylmethoxy]-N,N-dimethylethanamine Synonym Paracarbinoxamine
1047
HPLC Coulometric detection. Limit of quantification, 2 mg/L, limit of detection, 0.5 mg/L [Stockis et al. 1995]. Coulometric detection. For method see Stockis et al. [1992]. Disposition in the Body Therapeutic Concentration Twenty healthy males and females (20 to 42 years) were administered a single medication containing 20 mg phenylephrine and 4 mg carbinoxamine maleate. The peak plasma carbinoxamine concentration was 6.5 mg/ L, 2 to 6 h after dosing for a single dose and a steady state concentration of 13.5 mg/L, approximately 4.8 h after dosing was observed over a 4-day dosing period [Stockis et al. 1995]. Dose 12 to 32 mg of carbinoxamine maleate daily. Hoffman DJ et al. (1983). Capillary GLC assay for carbinoxamine and hydrocodone in human serum using nitrogen-sensitive detection. J Pharm Sci 72: 1342–1344. Stockis A et al. (1995). Relative bioavailability of carbinoxamine and phenylephrine from a retard capsule after single and repeated dose administration in healthy subjects. Arzneimittelforschung 45: 1009–1112. Stockis A et al. (1992). Relative bioavailability of carbinoxamine and phenylpropanolamine from a retard suspension after single dose administration in healthy subjects. Arzneimittelforschung 42 (12): 1478–1481.
Carbocromen
Chemical Properties A liquid. pKa 8.1 (25 ). Log P (octanol/water), 2.6. Carbinoxamine Maleate C16H19ClN2O, C4H4O4 = 406.9 CAS—3505-38-2 Proprietary Names Allergefon; Clistin; Histex; Ziriton. It is an ingredient of
Davenol, Extil, and Rondec. Chemical Properties A white crystalline powder. Mp 117 to 119 . Soluble 1 in less than 1 of water, 1 in 1.5 of ethanol, and 1 in 1.5 of chloroform; very slightly soluble in ether. Colour Test Cyanogen bromide—orange-pink. Thin-layer Chromatography System TA—Rf 0.48; system TB—Rf 0.26; system TC—Rf 0.19; system TE—Rf 0.50; system TL—Rf 0.04; system TAE—Rf 0.13; system TAF—Rf 0.16; system TAJ—Rf 0.04; system TAK—Rf 0.00; system TAL—Rf 0.27 (Dragendorff spray, positive; acidified iodoplatinate solution, positive; Marquis reagent, pink). Gas Chromatography System GA—carbinoxamine RI 2080, M (chlorobenzoylpyridine) RI 1645, M (carbinol) RI 1670, M (nor-) RI 2150; system GB—RI 2147; system GC—RI 2430. High Performance Liquid Chromatography System HA—k 4.7 (tailing peak); system HX—RI 359; system HAA—retention time 12.8 min. Ultraviolet Spectrum Aqueous acid—263 nm (A11¼323a); aqueous alkali— 261 nm (A11¼181a).
Antianginal Vasodilator C20H27NO5 = 361.4 CAS—804-10-4 IUPAC Name Ethyl 2-[3-(2-diethylaminoethyl)-4-methyl-2-oxochromen-7-yl] oxyacetate Synonyms Chromonar; [[3-[2-(diethylamino)ethyl]-4-methyl-2-oxo-2H-1benzopyran-7-yl]oxy]-acetic acid ethyl ester.
Chemical Properties Practically insoluble in water; soluble in chloroform and ether. Log P (octanol/water), 3.4. Carbocromen Hydrochloride C20H27NO5, HCl = 397.9 CAS—655-35-6 Synonym Cassella 4489 Proprietary Name Intensain Chemical Properties A white crystalline powder. Mp 159 to 160 . Freely soluble
in water, ethanol, methylene chloride, and chloroform; sparingly soluble in acetone, benzene, and ether. Colour Tests Aromaticity (method 2)—yellow/orange; Liebermann’s reagent (100 )—blue (3 min); sulfuric acid—violet (blue fluorescence at 350 nm). Thin-layer Chromatography System TA—Rf 0.48; system TB—Rf 0.17; system TC—Rf 0.24; system TE—Rf 0.62; system TL—Rf 0.12; system TAE—Rf 0.18 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 2835. Ultraviolet Spectrum Aqueous acid—202, 216, 321 nm (A11¼527a).
Infrared Spectrum Principal peaks at wavenumbers 1084, 1110, 1584, 1041, 763, 1010 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 58, 71, 26, 54, 167, 72, 42, 44.
Quantification Plasma GC AFID. Limit of detection, 200 ng/L [Hoffman et al. 1983].
Infrared Spectrum Principal peaks at wavenumbers 1708, 1608, 1210, 1755, 1178, 1085 cm1 (KBr disk).
C
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:44:53
1048
Carbon Tetrachloride
Mass Spectrum Principal ions at m/z 86, 87, 58, 30, 29, 84, 56, 42.
C Quantification Plasma Spectrofluorimetry Limit of detection, 40 mg/L [Martin, Wiegand 1970]. Urine Spectrofluorimetry See Plasma [Martin, Wiegand 1970]. Disposition in the Body Incompletely absorbed after oral administration (about 35%); rapidly hydrolysed to the corresponding carboxylic acid. About 70% of an IV dose and 20% of an oral dose are excreted in the 24-h urine as the carboxylic acid metabolite. Therapeutic Concentration Following oral administration of 150 mg three times a day to 6 subjects, a peak plasma concentration of 0.9 mg/L of the carboxylic acid metabolite was reported, declining to 0.06 mg/L, 10 h after the last dose [Martin, Wiegand 1970]. Half-life Plasma half-life, carboxylic acid metabolite about 0.8 h. Volume of Distribution Carboxylic acid metabolite, about 0.4 L/kg. Dose Carbocromen hydrochloride has been given in doses of 225 to 675 mg daily. Martin YC, Wiegand RG (1970). Metabolism and excretion of chromonar and its metabolite in dog and man. J Pharm Sci 59: 1313–1318.
Carbon Tetrachloride Anthelmintic (Veterinary), Solvent CCl4 = 153.8 CAS—56-23-5 IUPAC Name Tetrachloromethane Synonyms Carboneum tetrachloratum medicinale; methane tetrachloride; perchloromethane; tetrachlorocarbon; tetracloruro de carbono.
Chemical Properties A heavy, clear, colourless, volatile liquid, with a chloroform-like odour. It is non-inflammable, but in contact with a flame it decomposes and gives rise to toxic products (phosgene, carbon dioxide and hydrochloric acid), which have an acrid odour. Weight per millilitre, 1.592–1.595 g. Bp 76 to 78 . Refractive index 1.4607. Soluble 1 in 2000 water; miscible with dehydrated alcohol, benzene, chloroform and ether. Log P (octanol), 2.8. Colour Test Fujiwara test—red. Gas Chromatography System GA—RI 1000; system GI—RT 8.6 min; system GAA—RI 661. Mass Spectrum Principal ions at m/z 117, 119, 47, 35, 121, 82, 84, 49. Quantification Blood GC Column: Carbowax 20 M on Carbopak P 60/80 mesh (2 m 2 mm i. d.). Carrier gas: N2, 30 mL/min. Temperature: 90 . FID. Limit of detection, 50 mg/L [Tombolini, Cingolani 1996]. Column: 2.5% OV 17 silicone on Chromosorb G 80/ 100 mesh (6 ft 1/8 in i.d.). Carrier gas: N2, 30 mL/min. Temperature: 150 . FID. Limit of detectin, 2 mg/L [Goldermann et al. 1983]. Column: 20% Supelco SP-2100 plus 0.1% Carbowax 1500 on Supelcoport 80/100 mesh (3.0 m 2.0 mm i.d.). Carrier gas: N2, 20 mL/min. Temperature: 95 . FID. Limit of detection, 5 mg [Reddrop et al. 1980]. GC-MS See GC. Carrier gas: 45 mL/min. Temperature: 75 . ECD. Limit of detection, 15 ng [Reddrop et al. 1980]. Urine GC See Blood [Tombolini, Cingolani 1996]. Bile GC See Blood [Tombolini, Cingolani 1996]. Vitreous Humour GC See Blood [Tombolini, Cingolani 1996]. Brain GC See Blood [Tombolini, Cingolani 1996]. Kidney GC See Blood [Tombolini, Cingolani 1996]. Liver GC See Blood [Tombolini, Cingolani 1996]. See Blood [Reddrop et al. 1980]. GC-MS See Blood [Reddrop et al. 1980]. Lung GC See Blood [Tombolini, Cingolani 1996]. Expired Air GC See Blood [Reddrop et al. 1980]. GC-MS See Blood [Reddrop et al. 1980]. Muscle GC See Blood [Tombolini, Cingolani 1996]. Myocardium GC See Blood [Tombolini, Cingolani 1996]. Spleen GC See Blood [Tombolini, Cingolani 1996]. Disposition in the Body Carbon tetrachloride is readily absorbed after inhalation and also absorbed after ingestion or through the skin; the rate of absorption is increased by the concomitant ingestion of alcohol. It is excreted mainly from the lungs as carbon tetrachloride and carbon dioxide; it is excreted in urine as urea and an unidentified metabolite, and is eliminated in faeces.
Toxicity The minimum lethal dose is 3–5 mL, but recoveries have occurred following ingestion of 30–40 mL. Carbon tetrachloride injures almost all cells of the body including those of the blood, CNS, liver and kidney; the kidneys and liver of those who have died often show marked fatty degeneration. The maximum permissible atmospheric concentration is 10 ppm. Inhalation of concentrations of 1000 ppm, even for short periods, may cause acute toxic reactions. Continued exposure to concentrations of approx. 100 ppm may give rise to chronic poisoning. When carbon tetrachloride is ingested together or immediately after alcohol, its toxicity, particularly nephrotoxicity, is greatly increased. Toxic effects are associated with blood concentrations of 20–50 mg/L and a postmortem blood concentration of 260 mg/L has been reported in one fatality. A 75-year-old man died in hospital after ingesting an unknown amount of carbon tetrachloride. Concentrations were 328.5 mg/L in urine, 169.8 mg/L in bile, 143.4 mg/L in systemic venous blood, 57.5 mg/L in arterial blood and 170.5 mg/L in vitreous humour. In tissues, the concentrations were 657.9 mg/kg in pancreas, 243 mg/kg in brain, 237.3 mg/kg in testis, 127.3 mg/kg in lungs, 150.5 mg/kg in kidneys, 71.1 mg/kg in muscle, 78.5 mg/kg in myocardium, 68.3 mg/kg in spleen and 58.6 mg/kg in liver [Tombolini, Cingolani 1996]. The following postmortem tissue concentrations were reported in a fatality caused by inhalation of carbon tetrachloride: blood 18 mg/L, brain 175 mg/g, lung 12.5 mg/g [Franc 1983]. A 36-year-old man drank 50 mL of carbon tetrachloride. At the beginning of hyperventilation therapy blood concentrations were 120 mg/L. These levels fell to 30 mg/L within 6 days but the patient died from pneumonia 39 days after admission [Goldermann et al. 1983]. A 61-year-old man ingested a large amount of carbon tetrachloride, well in excess of the adult lethal dose. He was treated and survived with relatively mild clinical and biochemical evidence of toxicity [Mathieson et al. 1985]. A middle-aged alcoholic ingested ~30 mL carbon tetrachloride under the impression that it was alcohol. He was seriously ill but recovered. On his admission to hospital, his serum concentration of carbon tetrachloride was 20 mg/L. The first 24-h urine collection contained 8 mg/L and the first peritoneal dialysate contained 1 mg/L of carbon tetrachloride [SL Tompsett, personal communication 1967]. The following postmortem tissue concentrations were reported in a fatality caused by inhalation of carbon tetrachloride: kidney 32 mg/g, liver 142 mg/g, lung 39 mg/g, muscle 46 mg/g [Korenke, Pribilla 1969]. Franc A (1983). TIAFT Bull 17: 22–25. Goldermann L et al. (1983). Quantitative assessment of carbon tetrachloride levels in human blood by head-space gas chromatography: application in a case of suicidal carbon tetrachloride intoxication. Intensive Care Med 9: 131–135. Korenke HD, Pribilla O (1969). [Suicide by single inhalation of carbon tetrachloride (CCl4), with resulting leukoencephalopathy.]. Arch Toxikol 25: 109–126. Mathieson PW et al. (1985). Survival after massive ingestion of carbon tetrachloride treated by intravenous infusion of acetylcysteine. Hum Toxicol 4: 627–631. Reddrop CJ et al. (1980). Two rapid methods for the simultaneous gas–liquid chromatographic determination of carbon tetrachloride and chloroform in biological material and expired air. J Chromatogr 193: 71–82. Tombolini A, Cingolani M (1996). Fatal accidental ingestion of carbon tetrachloride: a postmortem distribution study. J Forensic Sci 41: 166–168.
Carbromal Hypnotic C7H13BrN2O2 = 237.1 CAS—77-65-6 IUPAC Name 2-Bromo-N-carbamoyl-2-ethylbutanamide Synonyms N-(Aminocarbonyl)-2-bromo-2-ethylbutanamide; bromadal; bromodiethylacetylurea; karbromal; uradal. Proprietary Names Adalin; Mirfudorm.
Chemical Properties A white crystalline powder. Mp 116 to 119 . Soluble 1 in 3000 of water, 1 in 18 of ethanol, 1 in 3 of chloroform and 1 in 14 of ether. Log P (octanol/water), 1.5. Colour Test Nessler’s reagent—brown-orange. Thin-layer Chromatography System TB—Rf 0.12; system TD—Rf 0.53; system TE—Rf 0.75; system TF—Rf 0.55; system TAD—Rf 0.64; system TAE—Rf 0.85; system TAF—Rf 0.87 (fluorescein solution, pink). Gas Chromatography System GA—carbromal RI 1513, M (carbromide) RI 1215, M (OH-carbromide) RI 1340, M (desbromo-) RI 1380, carbromal-Art RI 1450. High Performance Liquid Chromatography System HX—RI 410; system HY—RI 377; system HZ—retention time 3.9 min. Ultraviolet Spectrum No significant absorption, 230 to 360 nm.
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:44:59
Carbutamide
1049
In 11 fatalities attributed to carbromal overdose, the following post-mortem tissue concentrations, mg/L or mg/g (mean, N), were reported
Blood Brain Kidney Liver Urine
Infrared Spectrum Principal peaks at wavenumbers 1694, 660, 1600, 1094, 1212, 834 cm1 (KBr disk).
Carbromal
Bromoethylbutyramide
19.9–26.1 (23, 3) 0.6–120.9 (45, 8) 0.4–13 (6, 4) 1.1–2.8 (2.3, 4) 0.85–47.4 (24, 5)
0.2–27.2 (15, 10) 23.3–118.4 (66, 11) 16.1–79.6 (45, 9) 1.75–50.5 (14, 9) 2.5–36.9 (23, 8)
Total bromide concentrations in the blood ranged from 145 to 1898 (mean 536) mg/L [Kaferstein, Sticht 1978]. Post-mortem serum-carbromal concentrations of 158 and 71 mg/L were found in 2 fatalities resulting from acute intoxication [Gruska et al. 1970 and [Gruska et al. 1971]. Half-life Plasma half-life, carbromal 7 to 15 h, bromide about 15 days. Dose 0.3 to 1 g, as a hypnotic. Eichelbaum M et al. (1978). Determination of monoureides in biological fluids by high-pressureliquid-chromatography. Arch Toxicol 41(3): 187–193. Gruska H et al. (1970). Klinik, toxikologie und therapie einer schweren carbromalvergiftung mit letalem ausgang. Arch Toxicol 26(2): 149–160. Gruska H et al. (1971). Klinik, toxikologie und therapie einer schweren carbamazepin-vergiftung. Arch Toxicol 27: 193–203. Kaferstein H, Sticht G (1978). [Post mortem determination of bromureides (author’s transl)]. Z Rechtsmed 81: 269–283. Vohland HW et al. (1976). [On the toxicology of carbromal. I. Estimation of carbromal and its hypnotically active metabolites in rats and humans (author’s transl)]. Arch Toxicol 36: 31–42. Wells J, Cimbura G (1973). The determination of elevated bromide levels in blood by gas chromatography. J Forensic Sci 18(4): 437–440.
Carbutamide
Mass Spectrum Principal ions at m/z 44, 69, 41, 208, 210, 55, 71, 43; 2-bromo2-ethylbutyramide 69, 43, 41, 44, 71, 167, 165, 55; 2-bromo-2-ethyl-3-hydroxybutyramide 150, 152, 165, 41, 167, 43, 44, 130; 2-ethylbutyrylurea 45, 130, 44, 71, 42, 61, 115, 55.
Quantification Blood GC FID. For determination of inorganic bromide, see Wells, Cimbura [1973]. Plasma HPLC UV detection. Carbromal, 2-bromo-2-ethylbutyramide and 2-ethylbutyrylurea. Limit of detection, 200 mg/L for carbromal [Eichelbaum et al. 1978]. Disposition in the Body Carbromal is readily absorbed after oral administration. The major metabolite is free bromide ion; hydrolysis to an active metabolite, 2-bromo-2-ethylbutyramide, also occurs followed by oxidation to 2-bromo-2ethyl-3-hydroxybutyramide; other metabolites include 2-ethylbutyrylurea and 2ethyl-2-hydroxybutyric acid. Carbromal is excreted in the urine mainly as bromide ion and partly as 2-ethyl-2-hydroxybutyric acid, with very little as unchanged drug. Peak bromide excretion is attained after about 48 h. Therapeutic Concentration Following a single oral dose of 1 g given to 4 subjects, mean peak serum concentrations of about 6 mg/L of carbromal and 3 mg/L of 2-bromo-2ethylbutyramide were attained in 0.5 and 4 h respectively; the total bromide concentration reached about 12 mg/L in 9 h and was still increasing [Vohland et al. 1976]. Toxicity Fatalities have occurred in adults following the ingestion of 10 to 25 g but they are rare. Long-term use of carbromal may give rise to symptoms of chronic toxicity resembling bromism. Toxic effects are associated with serum-bromide concentrations of 300 to 1200 to 3200 mg/L.
Antidiabetic C11H17N3O3S = 271.3 CAS—339-43-5 IUPAC Name 1-(4-Aminophenyl)sulfonyl-3-butylurea Synonyms 4-Amino-N-[(butylamino)carbonyl]benzenesulfonamide; BZ-55; glybutamide; U-6987. Proprietary Names Diabetoplex; Dia-Tablinen; Glucidoral; Insoral; Invenol; Nadisan. Note Insoral is also used as a proprietary name for phenformin hydrochloride.
Chemical Properties A white finely crystalline powder. M.p. 144 to 145 . Soluble in water at pH 5–8. Practically insoluble in chloroform, and ether; soluble in ethanol and acetone. pKa 6.0 (20 ). Log P (octanol/water), 1.0. Thin-layer Chromatography System TA—Rf 0.78; system TT—Rf 0.90; system TU—Rf 0.27; system TV—Rf 0.07; system TAE—Rf 0.87. High Performance Liquid Chromatography System HY—RI 321; system HAA—retention time 14.5 min. Ultraviolet Spectrum Aqueous acid—266 nm (A11¼149a); aqueous alkali— 255 nm (A11¼616a).
Infrared Spectrum Principal peaks at wavenumbers 1661, 1147, 1599, 1089, 1635, 1310 cm1 (KBr disk).
C
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:45:9
1050
C
Carfenazine
Quantification Serum HPLC UV detection. See Saffar et al. [1982]. Disposition in the Body Absorbed after oral administration. It is excreted in the urine mainly as the acetyl derivative. Therapeutic Concentration Following an oral dose of 250 mg to 5 subjects, a mean peak serum concentration of 48.8 mg/L was reported at 2.6 h [Saffar et al. 1982]. Half-life Plasma half-life, about 24 h. Dose Carbutamide has been given in doses of 0.5 to 1 g daily. Saffar F et al. (1982). Biopharmaceutical studies on the clinical inequivalence of two carbutamide tablets. Chem Pharm Bull 30: 679–683.
Dose 75 to 400 mg of carfenazine maleate daily.
Carisoprodol Carfenazine Antipsychotic, Tranquilliser C24H31N3O2S = 425.6 CAS—2622-30-2 IUPAC Name 1-(10-[3-[4-(2-Hydroxyethyl)-1-piperazinyl]propyl]-10H-phenothiazin-2-yl)-1-propanone Synonym Carphenazine
Chemical Properties Log P (octanol/water), 3.3. Carfenazine Maleate C24H31N3O2S, 2C4H4O4 = 657.7 CAS—2975-34-0 Proprietary Name Proketazine Chemical Properties A fine yellow powder. Mp 176 to 185 , with decomposi-
tion. Soluble 1 in 600 of water and 1 in 400 of ethanol; practically insoluble in chloroform and ether. Colour Tests Formaldehyde–sulfuric acid—blue; Forrest reagent—red; FPN reagent—red-orange; Liebermann’s reagent—red-brown; Mandelin’s test—redviolet; Marquis test—orange!red-violet. Thin-layer Chromatography System TA—Rf 0.54; system TB—Rf 0.05; system TC—Rf 0.27; system TE—Rf 0.39; system TL—Rf 0.07; system TAE—Rf 0.39; system TAJ—Rf 0.08; system TAK—Rf 0.00; system TAL—Rf 0.51 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 3590. High Performance Liquid Chromatography System HA—k 1.7; system HX—RI 419. Ultraviolet Spectrum Aqueous acid—243 (A11¼606a), 277 nm. No alkaline shift.
Skeletal Muscle Relaxant C12H24N2O4 = 260.3 CAS—78-44-4 IUPAC Name [2-(Carbamoyloxymethyl)-2-methylpentyl] N-propan-2-ylcarbamate Synonyms 2-[[(Aminocarbonyl)oxy]methyl]-2-methylpentyl (1-methylethyl) carbamate; carisoprodate; carisoprodolum; isobamate; N-isopropylmeprobamate. Proprietary Names Artifar; Caridolin; Carisoma; Chinchen; Dolaren; Flexartal; Mio Relax; Mioxom; Muslax; Myolax; Neotica; Rela; Rotalin; Sanoma; Scutamil-C; Soma; Somadril; Somalgit. It is also an ingredient of Flectomas; Flexidone; Lagaflex; Relaxibys; Sodol Compound; Soma Compound; Teknadone. Note The name Soma has also been applied to a hallucinogenic fungus.
Chemical Properties White crystalline powder. Mp 92 to 93 . Soluble in water: 30 mg/100 mL at 25 , 140 mg/100 mL at 50 ; soluble in most common organic solvents; practically insoluble in vegetable oils. Log P (octanol/water) 2.36. Stable in dilute acids and alkalis. Carisoprodol in plasma was stable at 20 for up to 37 days [Kucharczyk et al. 1986]. Thin-layer Chromatography System TB—Rf 0.04; system TD—Rf 0.36; system TE—Rf 0.75; system TF—Rf 0.53; system TAD—Rf 0.59; system TAE—Rf 0.85; system TAF—Rf 0.79 (furfuraldehyde reagent, positive). Gas Chromatography System GA—RI 1830; RI 1785 meprobamate. Column: 2% GE-SE 30 on Aeropak 30* 80/100 mesh (1.8 m 2 mm i.d.). Carrier gas: N2, 50 mL/min. Temperature programme: 100 to 210 at 4.2 /min. FID. Retention time: 16 min [Cardini et al. 1968]. Ultraviolet Spectrum No significant absorption, 230 to 360 nm. Infrared Spectrum Principal peaks at wavenumbers 1695, 1527, 1075, 1246, 1101, 1319 cm1 (Nujol mull).
Mass Spectrum Principal ions at m/z 55, 57, 43, 97, 41, 56, 158, 44; meprobamate 83, 84, 55, 56, 43, 71, 41, 62 (no peaks above 160).
Infrared Spectrum Principal peaks at wavenumbers 1585, 1557, 1666, 1204, 751, 1298 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 268, 143, 425, 70, 269, 42, 394, 157.
Quantification Blood GC Column: 5% phenyl methyl silicone (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He. Temperature programme: 175 for 30 s to 220 at 10 /min to 260 at
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:45:23
Carisoprodol 20 /min for 6 min. FID. Limit of detection, 0.5 mg/L [Logan et al. 2000]. Column: 3% SE-30 on 80/100 mesh Chromosorb WAW-DMDS (40 0.25" o.d.). Carrier gas: He, 60 mL/min. Temperature: 190 . FID. Retention time: 2.25 min. Limit of detection not reported [Maes et al. 1969]. GC-MS Column: DB-35 ms cross-linked silica capillary (30 m 0.32 mm i.d., 0.25 mm). Carrier gas: He, 1.5 mL/min. Temperature programme: 120 for 1 min to 320 at 15/min for 4 min. EI ionisation at 70 eV, SIM acquisition mode. Limit of quantification, 1 mg/L; limit of detection, 0.1 mg/L [Gunnar et al. 2004]. Column: DB-5 capillary (30 m 0.25 mm i.d.). Temperature programme: 100 to 280 at 10/ min for 11 min. Retention time: 7.53 and 8.44 for carisoprodol and meprobamate, respectively. Limit of detection not reported [Backer et al. 1990]. Plasma GC Column: 3% OV-17 on 100/120 mesh Chromosorb WHP (60 2 mm i.d.). Carrier gas: N2, 14 psi. Temperature: 180 . FID. Retention time: 5.25 and 4.81 min for carisoprodol and meprobamate, respectively. Limit of detection, 0.5 and 1 mg/L for carisoprodol and meprobamate, respectively [Kintz et al. 1988]. Column: 3% GP SP2100 DB on 100/120 mesh Supelcoport (2 m 2 mm i.d.). Carrier gas: N2, 30 mL/min. Temperature: 180 . Retention time: 5.1 min. Limit of detection, 230 mg/L [Kucharczyk et al. 1986]. GC-MS Column: HP-5 MS (30 m 0.25 mm i.d., 0.25 mm). Temperature programme: 190 for 5 min to 245 at 30 /min to 255 at 2 /min. EI ionisation at 70 eV, SIM acquisition mode. Limit of quantification, 1.93 mg/L, limit of detection, 0.58 mg/L, both for meprobamate [Daval et al. 2006]. LC-MS Column: Symmetry C18 reversed phase ODS (150 2.1 mm i.d., 3.5 mm). Mobile phase: 10 mmol/L ammonium acetate : acetonitrile (90 : 10 for 5 min to 20 : 80 at 15 min for 10 min), flow rate 150 mL/min. ESI, full scan and SIM acquisition modes. Limit of quantification, 0.5 mg/L [Matsumoto et al. 2003]. Serum GC Column: GP 2% SP-2110/1% SP-2510 DA on 100/120 Supelcoport (1.2 m 2 mm i.d.). Carrier gas: N2, 30 mL/min. Temperature programme: 180 for 2 min to 240 at 8 /min for 2 min. FID. Limit of detection not reported [Olsen et al. 1994]. Column: 3% OV-1 or 3% OV-17 (60 0.25" o.d.). Carrier gas: N2. Temperature programme: 140 for 1 min to 275 at 24 /min for 12 min. FID. Limit of detection not reported [Adams et al. 1975]. GC-MS See Blood [Gunnar et al. 2004]. Oral Fluid LC-MS Column: Atlantis dC18(50 2.1 mm i.d., 3.5 mm). Mobile phase: acetonitrile : 5 mmol/L aqueous ammonium acetate (pH 5.0; 10 : 90 to 40 : 60 at 4.0 min to 90 : 10 at 4.1 min until 8.0 min to 10 : 90 at 8.1 min), flow rate 0.3 mL/min. ESI, positive ion mode, MRM acquisition mode. Limit of quantification, 97%), with approximately 57% of the dose excreted in the faeces and 27% in urine [Paulson et al. 2000]. CYP2C9 is the major isoform responsible for the metabolism of celecoxib [Davies et al. 2000], although CYP3A4 also plays a part [Tang et al. 2000]. Note For studies on the influence of CYP2C9 genetic polymorphisms, see Kirchheiner et al. [2003] or Brenner et al. [2003]. Therapeutic Concentration Twelve healthy volunteers were administered a single 200 mg oral dose of celecoxib. The mean peak plasma concentration was 700 mg/L at 18 h [Zarghi et al. 2006]. After an overnight fast, 12 healthy male volunteers received 200 mg celecoxib. After 2 days, a once-daily dose of 600 mg rifampicin was given for 5 consecutive days, and on day 9 another 200 mg dose of celecoxib was administered. Before rifampicin, the mean peak plasma celecoxib concentration was 545 274 mg/L reached at 4 0.9 h, and after rifampicin the mean peak plasma celecoxib concentration was 239 146 mg/L at 4 0.8 h. Rifampicin pretreatment decreased the AUC of celecoxib by 64% and increased clearance by 185% [Jayasagar et al. 2003]. Twelve healthy volunteers were administered a single oral dose of 200 mg celecoxib. The mean maximum plasma concentration was 840 280 mg/L reached at 2.9 1.2 h [Werner et al. 2002]. Four healthy young volunteers were administered a single 200 mg dose of celecoxib. Peak plasma concentrations of 0.797 mg/L were reached within 1.8 h for those who had celecoxib administered without food, and 0.877 mg/ L within 6.3 h for the volunteers who had the drug administered with a highfat breakfast [Davies et al. 2000]. Toxicity Acute overdose can result in coma, hypertension, acute renal failure and respiratory depression. Several cases of cholestatic hepatitis have been reported after celecoxib therapy [Alegria et al. 2002; Galan et al. 2001; Nachimuthu et al. 2001; O’Beirne, Cairns 2001], one case also involved pancreatitis [CarrilloJimenez, Nurnberger 2000]. Celecoxib has been associated with toxic epidermal necrolysis [Friedman et al. 2002; Giglio 2003] and allergic vasculitis [Drago et al. 2004; Jordan et al. 2002], in one case fatally [Schneider et al. 2002]. Celecoxib has also been associated with acute febrile neutrophilic dermatoses [Fye et al. 2001], acute generalised exanthematous pustulosis [Yang et al. 2004] as well as ‘drug rash with eosinophilia and systemic symptoms (DRESS)’ syndrome [Lee et al. 2008]. Half-life 11.2 to 15.6 h. Volume of Distribution 5.7 to 7.1 L/kg (455 166 L) [Davies et al. 2000]. Clearance 27.7 L/h. Distribution in Blood Red blood cells : plasma ratio is 0.89. Protein binding 97% to albumin. Dose 200 mg daily and increased (if necessary) to 400 mg. Alegria P et al. (2002). Celecoxib-induced cholestatic hepatotoxicity in a patient with cirrhosis. Ann Intern Med 137: 75. Brautigam L et al. (2001). Determination of celecoxib in human plasma and rat microdialysis samples by liquid chromatography tandem mass spectrometry. J Chromatogr B Biomed Sci Appl 761: 203–212. Brenner S et al. (2003). Influence of age and cytochrome P450 2C9 genotype on the steady-state disposition of diclofenac and celecoxib. Clin Pharmacokinet 42: 283–292. Carrillo-Jimenez R, Nurnberger M (2000). Celecoxib-induced acute pancreatitis and hepatitis: a case report. Arch Intern Med 160: 553–554. Chow HH et al. (2004). Determination of celecoxib in human plasma using solid-phase extraction and high-performance liquid chromatography. J Pharm Biomed Anal 34: 167–174. Davies NM et al. (2000). Clinical pharmacokinetics and pharmacodynamics of celecoxib: a selective cyclo-oxygenase-2 inhibitor. Clin Pharmacokinet 38: 225–242. Drago F et al. (2004). Cutaneous vasculitis induced by cyclo-oxygenase-2 selective inhibitors. J Am Acad Dermatol 51: 1029–1030. Friedman B et al. (2002). Toxic epidermal necrolysis due to administration of celecoxib (Celebrex). South Med J 95: 1213–1214. Fye KH et al. (2001). Celecoxib-induced Sweet’s syndrome. J Am Acad Dermatol 45: 300–302. Galan MV et al. (2001). Celecoxib-induced cholestatic hepatitis. Ann Intern Med 134: 254. Giglio P (2003). Toxic epidermal necrolysis due to administration of celecoxib (Celebrex). South Med J 96: 320–321. Hamama AK et al. (2005). Simultaneous determination of rofecoxib and celecoxib in human plasma by high-performance liquid chromatography. J Chromatogr Sci 43: 351–354. Jalalizadeh H et al. (2004). Determination of celecoxib in human plasma by high-performance liquid chromatography. J Pharm Biomed Anal 35: 665–670.
C
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:47:30
1064
C
Celiprolol
Jayasagar G et al. (2002). Validated HPLC method for the determination of celecoxib in human serum and its application in a clinical pharmacokinetic study. Pharmazie 57: 619–621. Jayasagar G et al. (2003). Influence of rifampicin pretreatment on the pharmacokinetics of celecoxib in healthy male volunteers. Drug Metabol Drug Interact 19: 287–295. Jordan KM et al. (2002). Allergic vasculitis associated with celecoxib. Rheumatology (Oxford) 41: 1453–1455. Kirchheiner J et al. (2003). Influence of CYP2C9 genetic polymorphisms on pharmacokinetics of celecoxib and its metabolites. Pharmacogenetics 13: 473–480. Lee JH et al. (2008). Drug Rash with Eosinophilia and Systemic Symptoms (DRESS) syndrome induced by celecoxib and anti-tuberculosis drugs. J Korean Med Sci 23: 521–525. Nachimuthu S et al. (2001). Acute hepatocellular and cholestatic injury in a patient taking celecoxib. Postgrad Med J 77: 548–550. O’Beirne JP, Cairns SR (2001). Drug points: cholestatic hepatitis in association with celecoxib. Brit Med J 323: 23. Paulson SK et al. (2000). Metabolism and excretion of [(14)C]celecoxib in healthy male volunteers. Drug Metab Dispos 28: 308–314. Pavan Kumar VV et al. (2006). Simultaneous quantitation of etoricoxib, salicylic acid, valdecoxib, ketoprofen, nimesulide and celecoxib in plasma by high-performance liquid chromatography with UV detection. Biomed Chromatogr 20: 125–132. Rao RN et al. (2005). Development and validation of a reversed-phase liquid chromatographic method for separation and simultaneous determination of COX-2 inhibitors in pharmaceuticals and its application to biological fluids. Biomed Chromatogr 19: 362–368. Rose MJ et al. (2000). Determination of celecoxib in human plasma by normal-phase high-performance liquid chromatography with column switching and ultraviolet absorbance detection. J Chromatogr B Biomed Sci Appl 738: 377–385. Schneider F et al. (2002). Fatal allergic vasculitis associated with celecoxib. Lancet 359: 852–853. Sch€ onberger F et al. (2002). Simple and sensitive method for the determination of celecoxib in human serum by high-performance liquid chromatography with fluorescence detection. J Chromatogr B Analyt Technol Biomed Life Sci 768: 255–260. St€ ormer E et al. (2003). Simultaneous determination of celecoxib, hydroxycelecoxib, and carboxycelecoxib in human plasma using gradient reversed-phase liquid chromatography with ultraviolet absorbance detection. J Chromatogr B Analyt Technol Biomed Life Sci 783: 207–212. Tang C et al. (2000). Major role of human liver microsomal cytochrome P450 2C9 (CYP2C9) in the oxidative metabolism of celecoxib, a novel cyclooxygenase-II inhibitor. J Pharmacol Exp Ther 293: 453–459. Ventura CA et al. (2006). Influence of modified cyclodextrins on solubility and percutaneous absorption of celecoxib through human skin. Int J Pharm 314: 37–45. Werner U et al. (2002). Investigation of the pharmacokinetics of celecoxib by liquid chromatography-mass spectrometry. Biomed Chromatogr 16: 56–60. Yang CC et al. (2004). Acute generalized exanthematous pustulosis caused by celecoxib. J Formos Med Assoc 103: 555–557. Zarghi A et al. (2006). Simple and rapid high-performance liquid chromatographic method for determination of celecoxib in plasma using UV detection: application in pharmacokinetic studies. J Chromatogr B Analyt Technol Biomed Life Sci 835: 100–104. Zhang M et al. (2006). Determination of celecoxib in human plasma and breast milk by highperformance liquid chromatographic assay. J Chromatogr B Analyt Technol Biomed Life Sci 830: 245–248.
Infrared Spectrum Principal peaks at wavenumbers 1674, 1636, 823, 591 cm1.
Celiprolol b-Blocker C20H33N3O4 = 379.5 CAS—56980-93-9 IUPAC Name 3-[3-Acetyl-4-[3-(tert-butylamino)-2-hydroxypropoxy]phenyl]1,1-diethylurea Synonyms N0 -[3-Acetyl-4-[3-[(1,1-dimethylethyl)-amino]-2-hydroxypropoxy] phenyl]-N,N-diethylurea; ST-1396.
Mass Spectrum Principal ions at m/z 86, 58, 250, 44, 57, 72, 291, 71. Chemical Properties Crystals. Mp 110 to 112 . pKa 9.68 (25 ). Log P (octanol/aqueous phosphate buffer, pH 7.4), 0.8 (37 ); log P (chloroform/water), 0.14; log P (chloroform/pH 7.1 aqueous phosphate buffer), 2.42; log P (octanol/water), 1.92. Extraction yield (chlorobutane), 0.1 [Demme et al. 2005]. Celiprolol Hydrochloride C20H33N3O4, HCl = 416.0 CAS—57470-78-7 Synonyms REV-5320A; RG-5320A; RHC-5320A; ST-1236. Proprietary Names Cardem; Celectol; Celipro; Corliprol; Dilanorm; Jofurol;
Moderator; Selectol.
Chemical Properties White odourless crystals. Mp 197 to 200 , with decom-
position. Solubility at 25 in water, 15.1 g/100 mL; methanol, 18.2 g/100 mL; ethanol, 1.61 g/100 mL; chloroform, 0.42 g/100 mL.
Thin-layer Chromatography System TAE—Rf 0.15; system TE—Rf 0.33. Gas Chromatography System GA— RI 2610. High Performance Liquid Chromatography System HAV—k 2.2; system HAA—retention time 11.5 min; system HZ—RI 2.5 min. Ultraviolet Spectrum Aqueous acid (0.01 mol/L hydrochloric acid)—231 (A11¼660) and 324 (A11¼60) nm; aqueous alkali (0.01 mol/L sodium hydroxide)— 231 (A11¼640) and 324 (A11¼60) nm; methanol—232 (A11¼775) and 329 (A11¼58) nm.
Quantification Blood HPLC For fluorescence detection and UV detection methods, see Buskin et al. [1982].
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:47:46
Cerivastatin Plasma HPLC UV detection. Limit of detection, about 16 mg/L [Braza et al. 1998]. Fluorescence detection. Limit of detection, 5 mg/L [Chiu, Raymond 1996]. UV and fluorescence detection. Limit of detection, 5 mg/L (total celiprolol), 2.5 mg/L (each enantiomer) [Verbesselt et al. 1996]. UV detection. Limit of detection, 4 mg/L [Rutledge et al. 1994a]. UV detection. Limit of detection, 25.7 mg/L [Rutledge et al. 1994b]. Fluorescence detection. Limit of detection, 1.5 mg/L (each enantiomer) [Hartmann et al. 1989]. Fluorescence detection. Limit of detection, 10 mg/L [Hippmann, Takacs 1983]. UV detection. Limit of detection, 10 mg/L. Fluorescence detection. Limit of detection, 5 mg/L [Buskin et al. 1982]. Urine HPLC Fluorescence detection. Limit of detection, 2.5 mg/L (each enantiomer) [Hartmann et al. 1989]. See Plasma [Hippmann, Takacs 1983]. See Blood Buskin et al. [1982]. Disposition in the Body Absorbed after oral administration in a non-linear fashion; the percentage of dose absorbed increases with increasing dose. The extent of absorption is reduced in the presence of food. Peak plasma concentrations are reached after 2 to 3 h. Crosses the placenta. Low lipid solubility. Undergoes only minimal metabolism and is excreted unchanged in the urine (~11%) and faeces (84% in 24 h after an oral dose). After an IV dose, ~50% is excreted in urine and 31% in faeces. A very low percentage of the drug is excreted as three metabolites. Bioavailability 30 to 70%, which is reduced in the presence of food. Therapeutic Concentration Considerable inter-individual variation in peak plasma concentrations. Mean peak plasma concentration measured after administration of a 200 mg single oral dose to 12 subjects was 687 mg/L after 3.71 h. Following administration of 200 mg and 400 mg daily for 7 days to 12 subjects, the mean peak plasma concentration was 597 mg/L and 1676 mg/L, respectively [Norris et al. 1986]. Half-life Plasma, 5 to 6 h. Protein Binding 25%. Note For reviews of celiprolol, see Riddell et al. [1987] and Milne, Buckley [1991]. Dose 200 mg of the hydrochloride daily, increased to 400 mg daily if necessary. Braza AJ et al. (1998). Determination of celiprolol and oxprenolol in human plasma by highperformance liquid chromatography and the analytical error function. J Chromatogr B Biomed Sci Appl 718: 267–272. Buskin JN et al. (1982). Specific and sensitive assay of celiprolol in blood, plasma and urine using high-performance liquid chromatography. J Chromatogr 230: 454–460. Chiu FC, Raymond K (1996). Validated assay for the determination of celiprolol in plasma using high-performance liquid chromatography and a silanol deactivated reversed-phase support. J Chromatogr B Biomed Appl 687(2): 462–465. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings at the 12th TIAFT, Seoul: 481–486. Hartmann C et al. (1989). Simultaneous determination of (R)- and (S)-celiprolol in human plasma and urine: high-performance liquid chromatographic assay on a chiral stationary phase with fluorimetric detection. J Chromatogr 496: 387–396. Hippmann D, Takacs F (1983). Arzneimittelforschung 33: 8–12. Milne RJ, Buckley MM (1991). Celiprolol. An updated review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy in cardiovascular disease. Drugs 41: 941–969. Norris RJ et al. (1986). A pharmacokinetic evaluation of celiprolol in healthy elderly volunteers. J Cardiovasc Pharmacol 8: S91–S92. Riddell JG et al. (1987). Celiprolol. A preliminary review of its pharmacodynamic and pharmacokinetic properties and its therapeutic use in hypertension and angina pectoris. Drugs 34: 438–458. Rutledge DR et al. (1994a). Simultaneous determination of verapamil and celiprolol in human plasma. J Chromatogr Sci 32: 153–156. Rutledge DR et al. (1994b). Liquid chromatographic determination of celiprolol, diltiazem, desmethyldiltiazem and deacetyldiltiazem in plasma using a short alkyl chain silanol deactivated column. J Pharm Biomed Anal 12: 135–140. Verbesselt R et al. (1996). Liquid chromatographic determination of total celiprolol or (S)-celiprolol and (R)-celiprolol simultaneously in human plasma. J Chromatogr B Biomed Sci Appl 683: 231–236.
€ line Cephae Emetic, Expectorant C28H38N2O4 = 466.6 CAS—483-17-0 IUPAC Name (1R)-1-[[(3R,11bS)-3-ethyl-9,10-dimethoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinolin-2-yl]methyl]-7-methoxy-1,2,3,4-tetrahydroisoquinolin-6-ol Synonyms Desmethylemetine; dihydropsychotrine; 70 ,10,11-trimethoxyemetan-60 -ol.
Chemical Properties An alkaloid present in ipecacuanha, the dried root, or rhizome and root, of Cepha€elis ipecacuanha (¼Uragoga ipecacuanha) (Rubiaceae) or
1065
C. acuminata. Needles. Mp 115 to 116 . Practically insoluble in water; freely soluble in acetic acid, acetone, ethanol, and chloroform; slightly soluble in ether. Log P (octanol/water), 4.9. € line Hydrochloride Cephae C28H38N2O4,2HCl = 539.5 CAS—5853-29-2 Chemical Properties White crystals or crystalline powder. Solutions turn yellow.
Soluble in water; less soluble in alcohol, acetone and chloroform. Practically insoluble in benzene. Colour Test Liebermann’s reagent—black. Thin-layer Chromatography System TA—Rf 53; system TB—Rf 01; system TC—Rf 19; system TL—Rf 08 (acidified iodoplatinate solution, positive). High Performance Liquid Chromatography System HA—k 7.7 (tailing peak). Ultraviolet Spectrum Aqueous acid—283 nm (A11¼127b); ethanol—235, 276 nm (A11¼144b).
Infrared Spectrum Principal peaks at wavenumbers 1264, 1515, 1116, 1229, 1213, 1616 cm1 (cepha€eline hydrochloride, KBr disk). Mass Spectrum Principal ions at m/z 178, 192, 272, 466, 244, 288, 191, 273. Quantification See under Emetine. Plasma HPLC Fluorescence detection. Limit of quantification, 1.0 mg/L [Asano et al. 2001]. Urine HPLC See Plasma. Limit of quantification, 5 mg/L [Asano et al. 2001]. Disposition in the Body Therapeutic Concentration Six healthy male volunteers were administered Ipecac syrup containing 0.843 g/L cepha€eline and 0.503 g/L emetine at doses of 5, 10, 15, 20, 25, and 30 mL. Cepha€eline was found in plasma at a concentration of 1.7, 3.0, 3.1, 3.6, 5.6, and 3.1 mg/L, respective, to the dosings at times of 2.1, 1.7, 0.8, 1.1, 2.4, and 1.5 h, respectively [Asano et al. 2001]. Dose Cepha€eline hydrochloride has been given in doses of 5 to 10 mg. Asano T et al. (2001). High-performance liquid chromatographic assay with fluorescence detection for the determination of cephaeline and emetine in human plasma and urine. J Chromatogr B Biomed Sci Appl 757: 197–206.
Cerivastatin Lipid-Regulating Agent C26H34FNO5 = 459.6 CAS—145599-86-6 IUPAC Name (E,3R,5S)-7-[4-(4-Fluorophenyl)-5-(methoxymethyl)-2,6-di(propan-2-yl)pyridin-3-yl]-3,5-dihydroxyhept-6-enoic acid Synonym (3R,5S,6E)-7-[4-(4-Fluorophenyl)-5-(methoxymethyl)-2,6-bis(1methylethyl)-3-pyridinyl]-3,5-dihydroxy-6-heptenoic acid
Cerivastatin Sodium C26H33FNNaO5 = 481.5 CAS—143201-11-0
C
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:48:11
1066
Cetalkonium Chloride
Synonym Bay-W-6228 Proprietary Names Baycol; Lipobay. Chemical Properties A white to off-white hygroscopic amorphous powder. It is
soluble in water, ethanol, and methanol; very slightly soluble in acetone. Ultraviolet Spectrum Methanol—228, 263 nm.
C
hydroxylated metabolite, M23, showed a concentration of 1.6 mg/L at approx. 4 to 6 h [Muck et al. 2000]. Bioavailability Absolute bioavailability, 60%. Half-life Plasma, 2 to 3 h. Volume of Distribution 0.3 L/kg. Clearance Plasma, 0.2 L/h/kg. Protein Binding 99%; 80% to albumin. Dose Initially 0.1 mg of cerivastatin sodium daily, increasing to 0.3 mg daily. Jemal M et al. (1999). Quantitation of cerivastatin and its seven acid and lactone biotransformation products in human serum by liquid chromatography-electrospray tandem mass spectrometry. J Chromatogr B, Biomed Sci Appl 736(1–2): 19–41. Krol GJ et al. (1993). LC separation and induced fluorometric detection of rivastatin in blood plasma. J Pharm Biomed Anal 11: 1269–1275. Muck W et al. (2000). Pharmacokinetics of cerivastatin when administered under fasted and fed conditions in the morning or evening. Int J Clin Pharmacol Ther 38(6): 298–303.
Cetalkonium Chloride Cationic Disinfectant C25H46ClN = 396.1 CAS—122-18-9 IUPAC Name Benzyl-hexadecyl-dimethylazanium chloride Synonym N-Hexadecyl-N,N-dimethylbenzenemethanaminium chloride Proprietary Names Baktonium. It is an ingredient of AAA, Bonjela, and Teejel.
Infrared Spectrum Principal peaks at wavenumbers 1603, 1098, 1381, 1222 cm1 (KBr disc).
Quantification Plasma HPLC Fluorescence detection. Limit of quantification, 0.025 mg/L [Krol et al. 1993]. Serum HPLC MS detection. Limit of quantification, 0.01 mg/L for cerivastatin and the lactone form, 0.05 to 0.5 mg/L for its biotransformation products [Jemal et al. 1999]. Disposition in the Body Cerivastatin is readily and almost completely absorbed after oral administration. It undergoes metabolism in the liver and is catalysed by at least two cytochrome P450 isoenzymes, CYP2C8 and CYP3A4. Three active metabolites are formed. The demethylated metabolite (about 50% as potent as the parent drug) and the hydroxylated metabolite (about 80% as potent) are the major metabolites formed; the product of both demethylation and hydroxylation also has activity, but it is only a minor metabolite. The metabolites are excreted in urine (about 30%) and in faeces (about 70%). There is no unchanged drug found in urine. There is no significant clearance by haemodialysis. Therapeutic Concentration Twenty-four healthy, Caucasian males, aged between 21 and 44 years, were administered with a single dose of 0.8 mg after an overnight fast (group 1), with breakfast (group 2), in the evening with dinner (group 3) or 4 h after dinner (group 4). The peak plasma concentrations were 7.7, 8.7, 7.1 and 7.1 mg/L for the four groups, respectively. These concentrations were reached within 1.5, 2.3 and 2.5 h, respectively. The mean concentration detected for the demethylated metabolite, M1, was 0.6 mg/L for the four groups and this was observed approx. 4 h after administration. The
Chemical Properties A white crystalline powder. Mp 58 to 60 . Sparingly soluble in cold water, soluble in hot water; soluble in acetone, ethanol, chloroform, ether, ethyl acetate, propylene glycol, and glycerol. Log P (octanol/water), 4.9. Thin-layer Chromatography System TA—Rf 0.12, streaking (acidified iodoplatinate solution, positive). Ultraviolet Spectrum Aqueous acid—258 (A11¼8b), 264 (A11¼10b), 270 (A11¼8b) nm; methanol—253, 258, 263 (A11¼11b), 270 nm.
Infrared Spectrum Principal peaks at wavenumbers 722, 698, 1612, 1204, 875, 790 cm1 (KBr disk). Use In a concentration of 0.01% in preparations applied to the mouth or throat.
Cetirizine Histamine H1-Antagonist C21H25ClN2O3 = 388.9 CAS—83881-51-0 IUPAC Name 2-[4-[(4-Chlorophenyl)phenylmethyl]-1-piperazinyl]ethoxy] acetic acid
Chemical Properties Crystals from ethanol. Mp 110 to 115 . Cetirizine Dihydrochloride C21H27Cl3N2O3 = 461.8 CAS—83881-52-1
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:48:23
Cetoxime Synonyms P-071; UCB-P071. Proprietary Names Alerlisin; Formistin; Reactine; Virlix; Voltric; Zirtek; Zyrlex;
Zyrtec.
Chemical Properties White-to-almost-white crystalline powder. Mp 225 .
Freely soluble in water; practically insoluble in acetone and dichloromethane. Log P octanol/water) 0.61. Thin-layer Chromatography Plate: silica 60 F254. Solvent system: chloroform followed by chloroform : methanol (85 : 15). Rf 0.31 [Pandya et al. 1996]. High Performance Liquid Chromatography. System HAA—RT 15.7 min; system HAX—RT 8.89 min; system HAY—RT 5.29 min; system HZ—RT 3.6 min. Ultraviolet Spectrum Aqueous acid (pH 2.38)—192, 258 nm.
Mass Spectrum Principal ions at m/z 201, 165, 203, 56, 187, 242, 299, 388.
Quantification Plasma GC Column: fused silica methylsilicone (25 m 0.31 mm i.d., 0.17 mm). Temperature: 260 . Carrier gas: He, 35 cm/s. NPD or FID. Retention time: 8.5 min. Limit of detection, 0.02 mg/L [Baltes et al. 1988]. HPLC Column: C18 Nucleosil 100-3 (150 3.2 mm, 3 mm). Mobile phase: 30 mmol/L potassium dihydrogen phosphate (pH 6.8). UV detection (l¼ 232 nm). Retention time: 8.25 min. Limit of quantification, 0.01 mg/L. [Macek et al. 1999]. Column: Beckman 5 gm octyl reversed phase (25 cm 2 mm). Mobile phase: acetonitrile : 0.01 mol/L potassium dihydrogen phosphate-0.02 mol/L sodium octane sulfonic acid (pH 3.0; 40 : 60), flow rate 0.25 mL/min. UV detection (l ¼ 230 nm). Limit of detection, 10 mg/L [Pariente-Khayat et al. 1995]. Column: reversed phase C18 RP-18 (250 4.6 mm i.d., 5 mm). Mobile phase: 0.01 mol/L potassium dihydrogen phosphate-0.02 mol/L sodium dodecyl sulfate (pH 2.9) : acetonitrile (55 : 45), flow rate 2.5 mL/min. UV detection (l¼ 230 nm). Retention time: 15 min. Limit of detection, 5 mg/L [Muscara, De Nucci 1995]. Serum HPLC Column: C18 (150 3.9 mm i.d., 4 mm). Mobile phase: acetonitrile : water (39 : 61) containing 13 mmol/L phosphate buffer (pH 2.8). UV detection (l¼ 230 nm). Retention time: 1.69 min. Limit of detection, 0.005 mg/L [Zaater et al. 2000]. Column: Spherisorb S5 ODS2 reversed phase (250 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : methanol: 0.05 mol/L ammonium phosphate (pH 2.5; 33 : 9 : 58), flow rate 2 mL/min. UV detection (l¼ 211 nm). Retention time: 6.4 min. Limit of detection, 0.02 mg/L [Moncrieff 1992]. Column: Spherisorb C8 (5 mm). Mobile phase: methanol : acetonitrile : 0.2 mol/L sodium hydrogen phosphate (30 : 20 : 50). UV detection. Limit of detection, 5 mg/L [Awni et al. 1990]. Urine HPLC See Plasma [Pariente-Khayat et al. 1995]. Column: Spherisorb 5ODS-2 (25 cm 4.6 mm i.d.). Mobile phase: Pic A: methanol : tetrahydrofuran (33 : 65 : 2), flow rate 1.0 mL/min. DAD (l ¼ 230 nm). Retention time: 6.17 min. Limit of detection, 20 mg/L [Rosseel, Lefebvre 1991]. Renal Dialysate HPLC See Serum. Limit of detection, 2.1 mg/L [Awni et al. 1990]. Disposition in the Body Cetirizine is rapidly absorbed after oral administration and undergoes metabolism in the liver, to a very limited extent by O-dealkylation. The metabolite produced has negligible antihistaminic activity. Cetirizine is primarily excreted in urine (70%) and 10% in the faeces, as the unchanged drug [Wood et al. 1987]. It is secreted in breast milk. Therapeutic Concentration Fifteen infants and toddlers, with a mean age of 12.3 months, were admitted to hospital for reoccurring respiratory infections or other hypersensitivity-
1067
related diseases and treated with a single dose of 0.25 mg/kg cetirizine solution. A mean peak plasma concentration of 0.39 mg/L was observed at 2.0 h [Spicak et al. 1997]. Eight children (2–6-years-old) were administered a single 5-mg oral dose of 10 mg/mL cetirizine 1.5 h before anaesthesia. The mean maximum plasma concentration of 607 231 mg/L was reached at 1.93 1.39 h [ParienteKhayat et al. 1995]. Fourteen healthy male volunteers (age: 21 to 46 years) received a single oral dose of 10 mg cetirizine hydrochloride as Zyrtek or Zetir. Mean peak plasma concentrations were 307 and 302 mg/L for Zetir and Zyrtek, respectively, both reached at 0.5 h. Ten elderly volunteers (aged between 60 and 90 years) and 10 healthy, young volunteers (21–29-years-old) were administered with a single oral dose of 10 mg. Mean plasma concentrations were 0.362 mg/L for the elderly individuals and 0.337 mg/L for the young. These concentrations were observed at 1.30 and 1.12 h, respectively [Lefebvre et al. 1988]. Five patients with end-stage renal disease ingested 10-mg cetirizine dihydrochloride. The mean maximum plasma concentration was 285 mg/L reached at 2 h [Awni et al. 1990]. Toxicity Two cases of overdose have been reported: an adult took a 150-mg dose of cetirizine and was admitted to hospital with somnolence, but no other clinical signs or blood chemistry/haematology. An 18-month-old child overdosed on 180 mg of cetirizine and became restless, irritable and drowsy. Multiple-dose toxicity targets the liver and a single dose the central nervous system. Note For a review of the comparative safety of histamine H1 antagonists, see Simons [1994]. Half-life Elimination half-life is 7–11 h in adults; 6–7 h in children; 3.1 h in infants. Volume of Distribution In adults, volume of distribution is 0.6–0.8 l/kg; children, 0.44 L/kg. Clearance Body clearance, 0.76 mL/min; 2.13 mL/min/kg (infants). 1.27 0.80 mL/min/kg in children [Pariente-Khayat et al. 1995]. Protein Binding 93% bound. Dose Adults and children >6 years: a usual dose of 10 mg is administered daily. Children (2 to 6 years) and patients with renal impairment: usual dose 5 mg daily. Awni WM et al. (1990). Effect of haemodialysis on the pharmacokinetics of cetirizine. Eur J Clin Pharmacol 38: 67–69. Baltes E et al. (1988). Gas chromatographic method for the determination of cetirizine in plasma. J Chromatogr 430: 149–155. Lefebvre RA et al. (1988). Single dose pharmacokinetics of cetirizine in young and elderly volunteers. Int J Clin Pharmacol Res 8: 463–470. Macek J et al. (1999). Determination of cetirizine in human plasma by high-performance liquid chromatography. J Chromatogr B Biomed Sci Appl 736: 231–235. Moncrieff J (1992). Determination of cetirizine in serum using reversed-phase high-performance liquid chromatography with ultraviolet spectrophotometric detection. J Chromatogr 583: 128–130. Muscara MN, DeNucci G (1995). Comparative bioavailability of single doses of tablet formulations of cetirizine dihydrochloride in healthy male volunteers. Int J Clin Pharmacol Ther 33: 27–31. Pandya KK et al. (1996). High-performance thin-layer chromatography for the determination of cetirizine in human plasma and its use in pharmacokinetic studies. J Pharm Pharmacol 48: 510–513. Pariente-Khayat A et al. (1995). Pharmacokinetics of cetirizine in 2- to 6-year-old children. Int J Clin Pharmacol Ther 33: 340–344. Rosseel MT, Lefebvre RA (1991). Determination of cetirizine in human urine by high-performance liquid chromatography. J Chromatogr 565: 504–510. Simons FE (1994). H1-receptor antagonists. Comparative tolerability and safety. Drug Safety 10: 350–380. Spicak V et al. (1997). Pharmacokinetics and pharmacodynamics of cetirizine in infants and toddlers. Clin Pharmacol Ther 61: 325–330. Wood SG et al. (1987). The metabolism and pharmacokinetics of 14C-cetirizine in humans. Ann Allergy 59: 31–34. Zaater MF et al. (2000). RP-LC method for the determination of cetirizine in serum. J Pharm Biomed Anal 22: 739–744.
Cetoxime Antihistamine C15H17N3O = 255.3 IUPAC Name N-Benzylanilinoacetamidoxime
C
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:48:29
1068
Cetrimide
Cetoxime Hydrochloride CAS—22204-29-1 Proprietary Name Febramine Chemical Properties Mp 160 . Soluble in water.
C
Colour Tests Ammonium vanadate test—red (limit of detection, 0.1 mg); Vitali’s test—red/yellow/red-brown (limit of detection, 0.1 mg). Thin-layer Chromatography System T1—Rf 0.75 (location reagent potassium permanganate spray, positive reaction). Ultraviolet Spectrum Aqueous acid (0.1 N sulfuric acid)—242, 290 nm; aqueous alkali (0.1 N sodium hydroxide)—249, 300 nm. Disposition in the Body Toxicity LD50 (oral): in mice 300 mg/kg. Dose Up to 800 mg daily has been given.
Infrared Spectrum Principal peaks at wavenumbers 685, 1629, 776, 1174, 715, 1205 cm1. Use In concentrations of 0.01 to 1%.
Chelidonine Alkaloid C20H19NO5 = 353.4 CAS—476-32-4 Synonyms [5bR-(5ba,6b,12ba)]-5b,6,7,12b,13,14-Hexahydro-13-methyl[1,3]benzodioxolo[5,6-c]-1,3-dioxolo[4,5-i]phenanthridin-6-ol; stylophorin.
Cetrimide Cationic Disinfectant C17H38BrN = 336.4 CAS—8044-71-1 IUPAC Name Trimethyl(tetradecyl)azanium bromide Note Consists chiefly of tetradecyltrimethylammonium bromide together with smaller amounts of dodecyl- and hexadecyltrimethylammonium bromides. Proprietary Names Cetavlon; Morpan CHSA; Silquat C100. It is an ingredient of Ceanel, Cetal (liquid), Savloclens, Savlodil, Savlon (liquid), and Travasept. Chemical Properties A white to creamy-white, voluminous, free-flowing, hygroscopic powder. A solution in water foams on shaking. Mp 232 to 247 . Soluble 1 in 2 of water; freely soluble in ethanol; soluble in ether. Thin-layer Chromatography System TA—Rf 0.00; system TB—Rf 0.00; system TC—Rf 0.01; system TE—Rf 0.00; system TL—Rf 0.00; system TN—Rf 0.100; system TO—Rf 0.50; system TAE—Rf 0.00; system TAF—Rf 0.29 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 1699. High Performance Liquid Chromatography System HX—RI 56. Infrared Spectrum Principal peaks at wavenumbers 961, 909, 952, 719, 970, 724 cm1 (KC1 disk).
Chemical Properties An alkaloid obtained from the greater celandine, Chelidonium majus (Papaveraceae). A white crystalline powder. Mp 135 to 136 . Practically insoluble in water; soluble in ethanol, chloroform, and ether. Log P (octanol/water), 2.9. Colour Tests Mandelin’s test—yellow!green; Marquis test—green; sulfuric acid—brown. Thin-layer Chromatography System TA—Rf 0.72 (acidified iodoplatinate solution, positive).
Cetylpyridinium Chloride Cationic Disinfectant C21H38ClN, H2O = 358.0 CAS—7773-52-6 (cetylpyridinium); 123-03-5 (cetylpyridinium chloride, anhydrous); 6004-24-6 (cetylpyridinium chloride, monohydrate) IUPAC Name 1-Hexadecylpyridin-1-ium chloride hydrate Synonym 1-Hexadecylpyridinium chloride monohydrate Proprietary Names Ceepryn; Cepacol; Dobendan; Merocet(s). It is an ingredient of Merocaine and Tyrosolven.
Infrared Spectrum Principal peaks at wavenumbers 1035, 1257, 1222, 1497, 748, 1070 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 332, 333, 304, 335, 176, 303, 334, 162. Chemical Properties A white powder. A solution in water foams strongly when shaken. Mp 77 to 83 . Soluble 1 in 20 of water; freely soluble in ethanol and chloroform; very slightly soluble in ether and in benzene. Log P (octanol), 1.7. Thin-layer Chromatography System TA—Rf 0.20; system TL—Rf 0.00; system TAE—Rf 0.00; system TAF—Rf 0.29 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—not eluted. Ultraviolet Spectrum Aqueous acid—280 nm. No alkaline shift.
Chloral Betaine Hypnotic, Sedative C7H12Cl3NO3, H2O = 282.6 CAS—2218-68-0 IUPAC Name 2,2,2-Trichloroethane-1,1-diol; 2-(trimethylazaniumyl)acetate Chemical Properties 1-Carboxy-N,N,N-trimethanaminium inner salt compound with 2,2,2-trichloro-1,1-ethanediol (1 : 1). The adduct formed by chloral hydrate, CCl3CH(OH)2, and betaine, C5H11NO2, containing about 58% of chloral hydrate. A white crystalline powder. Mp about 124 , with decomposition. Soluble 1 in 1 of water and 1 in 4 of ethanol; practically insoluble in chloroform and ether. Ultraviolet Spectrum No significant absorption, 230–360 nm. Infrared Spectrum Principal peaks at wavenumbers 840, 1630, 1085, 1130, 730, 1500 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 81, 83, 46, 110, 82, 112, 84, 117. Dose Usually 0.87 to 2.61 g daily, but doses of up to 3.5 g may be necessary.
Chloral Hydrate Hypnotic, Sedative C2H3Cl3O2 = 165.4 CAS—302-17-0
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:48:45
Chlorambucil IUPAC Name 2,2,2-Trichloro-1,1-ethanediol Synonyms Cloral; cloral hydrate. Proprietary Names Aquachloral; Chloradorm; Chloraldurat; Chloralex; Chloralvan; Dormel; Elix-Nocte; Escre; Medianox; Noctec; Novo-chlorhydrate; Nycton; Rectules; Somnos; Welldorm. Also an ingredient of Babygencal; Dermophil Indien; Synthol.
Chemical Properties Colourless or white crystals which volatilise slowly on exposure to air and are decomposed by caustic alkalis, liberating chloroform. Mp 50 . Bp 98 . 1 mL of water dissolves the following amounts of cloral hydrate: 2.4 g at 0 , 8.3 g at 25 , 14.3 g at 140 . Soluble: 1 in 0.2 of ethanol, 1 in 1.3 of alcohol, 1 in 2 of chloroform, 1 in 1.4 olive oil, 1 in 0.5 glycerol, 1 in 68 g carbon disulfide and 1 in 1.5 of ether. Freely soluble in acetone and methylethyl ketone. Moderately or sparingly soluble in turpentine, petroleum ether, carbon tetrachloride, benzene, and toluene. Ethanolic solutions may deposit crystals of cloral ethanolate. pKa 10.0. Log P (octanol/water), 0.99. Colour Tests Fujiwara test—red; palladium chloride—black. Gas Chromatography System GA—RI 695 (tailing peak); system GI—retention time 12.5 min (tailing peak). Ultraviolet Spectrum No significant absorption, 230–360 nm. Infrared Spectrum Principal peaks at wavenumbers 835, 1083, 1300, 970, 1620 cm1. Mass Spectrum Principal peaks at m/z 82, 47, 84, 29, 111, 83, 113, 85; trichloroacetic acid 44, 83, 85, 36, 28; trichloroethanol 31, 49, 77, 113, 115, 82, 51, 117.
1069
In a fatality due to the ingestion of chloral hydrate, the following postmortem tissue concentrations of trichloroethanol were reported: blood 55 mg/L, brain 91 mg/kg, liver 200 mg/kg, urine 30 mg/L [Poklis et al. 1973]. Half-life Plasma half-life, chloral hydrate about 4 min, trichloroethanol about 7 to 11 h, urochloralic acid about 7 h, trichloroacetic acid about 4 days. Volume of Distribution Trichloroethanol, about 0.6 L/kg. Distribution in Blood Plasma : whole blood ratio, trichloroethanol, about 0.9. Protein Binding Trichloroethanol 35% and trichloroacetic acid 94%. Note For a review of the pharmacokinetics of sedatives in neonates, see JacqzAigrain, Burtin [1996]; for a review of the pharmacokinetics of hypnotic drugs, see Breimer [1977]. Dose 0.5 to 2 g daily. Children may be given 30 to 50 mg/kg body-weight with a maximum single dose of 1 g. Baselt RC, Cravey RH (1977). J Anal Toxicol 1: 81–103. Berry DJ (1975). Determination of trichloroethanol at therapeutic and overdose levels in blood and urine by electron capture gas chromatography. J Chromatogr 107: 107–114. Breimer DD et al. (1974). Gas chromatographic determination of chloral hydrate, trichloroethanol and trichloroacetic acid in blood and in urine employing head-space analysis. J Chromatogr A 88 (1): 55–63. Breimer DD (1977). Clinical pharmacokinetics of hypnotics. Clin Pharmacokinet 2: 93–109. Engelhart DA et al. (1998). Unusual death attributed to the combined effects of chloral hydrate, lidocaine, and nitrous oxide. J Anal Toxicol 22: 246–247. Jacqz-Aigrain E, Burtin P (1996). Clinical pharmacokinetics of sedatives in neonates. Clin Pharmacokinet 31: 423–443. McBay AJ et al. (1980). Spectrophotometric determination of trichloroethanol in chloral hydrate poisoning. J Anal Toxicol 4: 99–101. Meyer E et al. (1995). Determination of chloral hydrate and metabolites in a fatal intoxication. J Anal Toxicol 19: 124–126. Poklis A et al. (1973). Bull Int Assoc Forensic Toxicol 9(3–4): 8–9. Yan Z et al. (1999). Determination of chloral hydrate metabolites in human plasma by gas chromatography-mass spectrometry. J Pharm Biomed Anal 19: 309–318.
Chlorambucil Antineoplastic C14H19Cl2NO2 = 304.2 CAS—305-03-3 IUPAC Name 4-[4-[Bis(2-chloroethyl)amino]phenyl]butanoic acid Synonyms 4-[Bis(2-chloroethyl)amino]benzenebutanoic acid; chlorbutinum. Proprietary Names Chloraminophene; Leukeran. Quantification Blood UV-Vis Trichloroethanol (modified Fujiwara reaction) [McBay et al. 1980]. GC ECD [Meyer et al. 1995] ECD. Chloral hydrate, trichloroethanol, and trichloroacetic acid, head-space analysis. Limit of detection, 500 mg/L for chloral hydrate and trichloroethanol [Breimer et al. 1974]. Plasma GC ECD. Trichloroethanol [Berry 1975]. GC-MS Chloral hydrate and metabolites, see [Yan et al. 1999]. Urine UV-Vis See Blood [McBay et al. 1980]. GC See Blood [Breimer et al. 1974]. See Plasma [Berry 1975]. Disposition in the Body Readily absorbed following oral administration. It is rapidly metabolised by reduction to trichloroethanol, the major active metabolite, which is further metabolised by conjugation with glucuronic acid to give urochloralic acid and by oxidation to trichloroacetic acid, the major urinary metabolite. Trichloroethanol passes into the CSF, into breast milk, and across the placenta. About 10–30% of a dose is excreted in the urine as urochloralic acid and up to 5% as trichloroethanol in 24 h. Trichloroacetic acid is slowly excreted in urine over several days; a small amount of urochloralic acid may be excreted in the bile. Therapeutic Concentration Chloral hydrate is difficult to detect in body fluids after normal doses. The plasma concentration of trichloroethanol is usually in the range 1.5 to 15 mg/L. Trichloroacetic acid and urochloralic acid are present in plasma at concentrations similar to, or greater than, those of trichloroethanol. When alcohol has been taken, peak plasma concentrations of trichloroethanol are increased and remain elevated for about 6 h after ingestion, and those of trichloroacetic acid are decreased. Trichloroacetic acid accumulates in the plasma during chronic administration of cloral hydrate. Following a single oral dose of 825 mg to 5 subjects, peak plasma trichloroethanol concentrations of 7.6 to 12.2 mg/L (mean 9.7) were attained in 0.5 to 1 h [Berry 1975]. Toxicity Fatalities have occurred following the ingestion of 1.25 and 3 g but recovery has occurred after ingestion of 30 g. Plasma concentrations greater than 40 mg/L of trichloroethanol are likely to produce toxic effects; fatalities have been reported at blood concentrations of 20–495 mg/L (mean 155) of trichloroethanol. In a fatality caused by the admistration of chloral hydrate, lidocaine, and nitrous oxide, postmortem trichloroethanol concentrations were: plasma, 79.0 mg/L; urine, 31.0 mg/L; gastric contents, 454.0 mg/L; bile, 111.0 mg/L; vitreous humour, 40.2 mg/L; CSF, 68.3 mg/L; and liver, 164 mg/kg. Lidocaine concentrations ranged from 3.7 mg/L in urine to 19.0 mg /L in bile. Nitrous oxide was measured at 4.4 mL/L in blood [Engelhart et al. 1998]. In 4 fatalities known to involve the acute ingestion of 15 to 30 g of chloral hydrate, postmortem blood trichloroethanol concentrations ranged from 100 to 640 mg/L (mean 265) [Baselt, Cravey 1977].
CB-1348;
Chemical Properties A white crystalline or granular powder. Mp 66 . Practically insoluble in water; soluble 1 in 1.5 of ethanol, 1 in 2 of acetone, and 1 in 2.5 of chloroform; soluble in ether. pKa 5.8. Log P (octanol/pH 7.4), 1.7. Caution Chlorambucil is irritant; avoid contact with skin and mucous membranes. Thin-layer Chromatography system TD—Rf 0.33; system TE—Rf 0.06; system TF—Rf 0.40; System TAD—Rf 0.50; system TAE—Rf 0.84. Gas Chromatography System GA—chlorambucil RI 2420, chlorambucil-Me RI 2340. High Performance Liquid Chromatography System HAA—Retention time 22.4 min. Ultraviolet Spectrum Methanol—258 (A11¼642a), 303 nm. No alkaline shift.
Infrared Spectrum Principal peaks at wavenumbers 1695, 1520, 1229, 1610, 1175, 1270 cm1 (KBr disk).
C
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:48:54
1070
Chloramphenicol Chemical Properties Fine, white to greyish-white or yellowish-white crystals. Mp 149 to 153 . A solution in dehydrated alcohol is dextrorotatory and a solution in ethyl acetate is laevorotatory. Soluble 1 in 400 of water and 1 in 2.5 of ethanol; very soluble in acetone and ethyl acetate; slightly soluble in chloroform and ether. pKa 5.5. Log P (octanol/water), 1.1. Chloramphenicol Cinnamate C20H18Cl2N2O6 = 453.3 CAS—14399-14-5 Chemical Properties A white or yellowish-white crystalline powder. Mp about
C
119 . Very slightly soluble in water; soluble 1 in 25 of ethanol, 1 in 50 of chloroform, and 1 in 500 of ether. Chloramphenicol Palmitate
C27H42Cl2N2O6 = 561.5 CAS—530-43-8 Synonyms Chloramphenicol a-palmitate; palmitylchloramphenicol. Proprietary Names Chloromycetin Palmitate Suspension; Globenicol. Chemical Properties A fine, white, unctuous, crystalline powder. Mp 87 to 95 .
Very slightly soluble in water; soluble 1 in 45 of ethanol, 1 in 6 of chloroform, and 1 in 14 of ether; freely soluble in acetone; soluble in ethyl acetate. Chloramphenicol Sodium Succinate C15H15Cl2N2NaO8 = 445.2 CAS—982-57-0 Synonym Chloramphenicol a-sodium succinate Proprietary Names Chloromycetin Succinate; Globenicol; Kemicetine Succinate;
Mychel-S.
Mass Spectrum Principal ions at m/z 254, 256, 118, 255, 303, 305, 63, 45.
Quantification Plasma GC–MS Limit of detection, 1 g, and fatalities have occurred after the ingestion of >2 g and after excessive skin contamination. The maximum permissible atmospheric concentration is 0.5 mg/m3. In a non-fatal poisoning case, a 4-year-old child who ingested an unknown amount of chlordane and developed intermittent convulsions had an initial serum concentration of 3.4 mg/L which decreased to 0.14 mg/L after 72 h; the rate of decline of the serum concentration was non-linear with a terminal halflife of 88 days. Urine samples obtained during the first 3 days after ingestion showed a decrease from 1.9 mg/L to 0.05 mg/L, but increased to 0.13 mg/L on the 35th day [Aldrich, Holmes 1969]. The following postmortem concentrations were reported in a fatality due to the ingestion of chlordane: blood 4.4 mg/L, urine 0.24 mg/L [Bost 1978]. A 66-year-old man who ingested about 400 mL of a 70% commercial solution and died after 40 h had the following postmortem tissue concentrations: blood 1.7 mg/L, fat 378 mg/g, kidney 14 mg/g, liver 43 mg/g, urine 0.6 mg/L [Baselt 2000].
Tran; Disarim; Elenium; Equibral; Labican; Librium; Medilium; Mitran; Nack; Novopoxide; O.C.M.; Psichial; Psicoterina; Relaxil; Reliberan; Reposans; Seren Vita; SK-Lygen; Solium; Trilium; Tropium; Viansin. It is an ingredient of Clindex; Librax; Limbitrol; Menrium. Chemical Properties White or slightly yellowish crystalline powder. Mp 213 , with decomposition. Soluble 1 in 10 of water and 1 in 40 of ethanol; practically insoluble in chloroform and ether. pKa 4.8 [Greenblatt et al. 1978]. Log P (octanol/ water) 2.44. Colour Test Marquis test—yellow. Thin-layer Chromatography System TA—Rf 0.62; system TB—Rf 0.02; system TC—Rf 0.50; system TD—Rf 0.10; system TE—Rf 0.52; system TF—Rf 0.10; system TL—Rf 0.22; system TAD—Rf 0.53; system TAE—Rf 0.76; system TAF—Rf 0.77; system TAJ—Rf 0.48; system TAK—Rf 0.02; system TAL—Rf 0.79 (Dragendorff spray, positive; FPN reagent, yellow; acidified iodoplatinate solution, positive; Marquis reagent, yellow). Gas Chromatography System GA—chlordiazepoxide RI 2795, M (nor-) RI 2452, M (demoxepam) RI 2529, nordiazepam RI 2490, oxazepam RI 2325; system GB—chlordiazepoxide RI 2981, M (nor-) RI 2679, M (demoxepam) RI 2806; system GG—chlordiazepoxide RI 3065, nordiazepam RI 3041, oxazepam RI 2803. High Performance Liquid Chromatography System HI—chlordiazepoxide k 6.41, demoxepam k 2.42, M (nor-) k 4.47, nordiazepam k 8.00, oxazepam k 4.62; system HK—chlordiazepoxide k 2.87, demoxepam k 0.03, M (nor-) k 2.39, nordiazepam k 1.99, oxazepam k 0.73; system HX—RI 363; system HY—RI 285; system HZ—RT 3.2 min; system HAA—RT 15.2 min; system HAF—chlordiazepoxide RT 21.0 min; system HAL—chlordiazepoxide RT 7.7 min; system HAM—chlordiazepoxide RT 3.1 min; system HAX—chlordiazepoxide RT 6.9 min; system HAY—chlordiazepoxide RT 5.3 min; system HAZ—chlordiazepoxide k 1.68; system HBH—chlordiazepoxide k 6.68; system HBI—chlordiazepoxide k 1.65. Ultraviolet Spectrum Aqueous acid—246 (A11¼ 1112a), 308 nm; aqueous alkali—262 nm.
Aldrich FD, Holmes JM (1969). Acute chlordane intoxication in a child. Case report with toxicological data. Arch Environ Health 19: 129–132. Baselt RC (2000). Disposition of Toxic Drugs and Chemicals in Man, 5th edn. California: Biomedical Publications, 150–152. Bost RO, Sunshine I (1978). Bull Int Assoc Forensic Toxicol 14(1): 30. Dale WE et al. (1966). Hexane extractable chlorinated insecticides in human blood. Life Sci 5(1): 47–54.
Chlordiazepoxide Anticonvulsant, 1,4-Benzodiazepine, Tranquilliser C16H14ClN3O = 299.8 CAS—58-25-3
Infrared Spectrum Principal peaks at wavenumbers 1625, 760, 1260, 690, 1590, 850 cm1 (KBr disk).
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:49:11
Chlordiazepoxide
Mass Spectrum Principal ions at m/z 282, 299, 284, 283, 241, 56, 301, 253; 285, 286, 269, 287, 241, 242, 77, 270 demoxepam; 285, 268, 284, 77, 286, 42, 287, 233 desmethylchlordiazepoxide; 242, 269, 270, 241, 243, 271, 244, 272 desmethyldiazepam; 257, 77, 268, 239, 205, 267, 233, 259 oxazepam.
Quantification Blood GC Column: SE-54 5% phenyl methyl silicone (25 m 0.31 mm i.d., 0.17 mm). Carrier gas: He, 2–3 mL/min. Temperature programme: 200 for 1 min to 290 at 10 /min. NPD and ECD. Retention time: 6.16 min. Limit of detection, 10 mg/L ECD [Lillsunde, Seppala 1990]. Column: 3% OV-17 on Gas Chrom Q 100/120 mesh (1.2 m 3.2 mm i.d.). Carrier gas: N2, 50 mL/min. Temperature: 240 . ECD. Retention time: 9.95 min. Limit of detection, 0.2 mg/ L [Peat, Kopjak 1979]. GC-MS Column: SE-30 WCOT vitreous silica capillary (25 m 0.2 mm i.d.). Carrier gas: He, 22 psi. Temperature programme: 50 to 260 at 30 /min. EI ionisation at 70 eV. Limit of detection, 20–50 ng [Joyce et al. 1984]. HPLC Column: Bondapak C18 (30 cm 4 mm i.d.). Mobile phase: methanol : 0.025 mol/L disodium hydrogen phosphate (pH 7.5; 58 : 42) or methanol : 0.025 mol/L disodium hydrogen phosphate (pH 7.5; 73 : 37), flow rate 2.4 mL/min. UV detection. Retention time: 12 and 370 nm). Limit of quantification, 50 mg/L [Antoniewicz et al. 1992]. Column: Nova-Pak C18 (100 8.0 mm i.d., 4 mm). Mobile phase: acetonitrile : water (22 : 78) containing 0.02% isopropylamine (pH 2.4) with 0.085% phosphoric acid, flow rate 2.5 mL/ min. UV detection (l ¼ 220 nm). Limit of detection, 10 mg/L [Prunonosa et al. 1992a]. See Blood [Cuisinaud et al. 1985]. CE Capillary: fused silica (57 cm total length, 75 mm i.d.). Buffer: 100 mmol/L sodium borate buffer (pH 8.6) : 25 mmol/L SDS containing 10% acetonitrile. UV detection (l ¼ 214 nm). Limit of detection, 20 mg/L [Prunonosa et al. 1992b]. Capillary: fused silica (57 cm total length, 75 mm i.d.). Buffer: 100 mmol/L sodium borate buffer (pH 8.6) : 100 mmol/L SDS : 25 mmol/L g-cyclodextrins containing 10% acetonitrile. UV detection (l ¼ 214 nm). Limit of detection, 10 mg/L [Prunonosa et al. 1992a]. Urine HPLC See Blood. Limit of detection, 30 mg/L [Cuisinaud et al. 1985]. Oral Fluid HPLC See Blood [Cuisinaud et al. 1985]. Disposition in the Body Cicletanine undergoes glucuronidation and sulfation. Elimination is both renal and hepatic while urinary excretion of the unchanged drug is negligible. Therapeutic Concentration Ten healthy volunteers were administered a single 50 mg cicletanine dose of cicletanine. The mean peak plasma concentration was 1.78 0.586 mg/L at 0.83 0.8 h. The mean elimination half-life was 9.89 4.32 h [Prunonosa et al. 1992c]. Eight healthy volunteers were administered 50 mg orally a day for 7 days. There was no significant difference between the parameters after the first dose and after repeated doses. The mean peak plasma concentration was 1.73 ng/L after the final dose [Peraire et al. 1991]. Forty-three patients with varying degrees of chronic renal failure were administered a single oral dose of 300 or 200 mg cicletanine. Six patients with moderate renal dysfunction were administered 200 mg a day orally for 30 days. In patients with severe renal dysfunction, the pharmacokinetics of cicletanine were significantly altered, with increased elimination half-life and tissue accumulation. Only minor pharmacokinetic changes were seen in patients with mild or moderate renal impairment, even after repeated administration of the drug. Mean maximum plasma concentrations were 20 2, 14 2 and 13 2 mg/mL at 1.3 0.1, 1.3 0.2 and 0.9 0.2 h, respectively, in control subjects, patients with mild renal failure, and patients with severe renal failure, respectively [Jungers 1988]. Note For a summary of the pharmacokinetics of cicletanine under various conditions, see Fredj [1988]. For a study of the interaction of cicletanine with tolbutamide in healthy volunteers, see Bayes et al. [1996]. Half-life Approximately 5 to 18 h. Volume of Distribution In 8 subjects (weighing 75.3 6.4 kg): 35.5 to 152.4 L [Peraire et al. 1991]. Clearance Oral and renal clearance 7.3 2.5 and 0.026 0.012 L/h, respectively. Protein Binding 97.3% bound to serum proteins, 93.5% of which is to albumin [Zini et al. 1988]. Dose 50 to 100 mg orally daily. Antoniewicz SM et al. (1992). Determination of cicletanine in human plasma by high-performance liquid chromatography using automated column switching. J Chromatogr 573: 93–98. Bayes MC et al. (1996). A drug interaction study between cicletanine and tolbutamide in healthy volunteers. Eur J Clin Pharmacol 50: 381–384. Cuisinaud G et al. (1985). High-performance liquid chromatographic determination of cicletanide, a new diuretic, in plasma, red blood cells, urine and saliva. J Chromatogr 341: 97–104. Fredj G (). Clinical pharmacokinetics of cicletanine hydrochloride. Drugs Exp Clin Res 14: 181–188. Jungers P (1988). Pharmacokinetics of cicletanine in patients with impaired renal function. Drugs Exp Clin Res 14: 189–194. Peraire C et al. (1991). Multiple dose pharmacokinetic study of cicletanine in healthy volunteers. Eur J Drug Metab Pharmacokinet SpecNo3: 173–177. Prunonosa J et al. (1992). Determination of cicletanine enantiomers in plasma by high-performance capillary electrophoresis. J Chromatogr 574: 127–133. Prunonosa J et al. (1992). Comparison of high-performance liquid chromatography and highperformance capillary electrophoresis for the determination of cicletanine in plasma. J Chromatogr 581: 219–226. Prunonosa J et al. (1992). Pharmacokinetic study of cicletanine in healthy volunteers. Int J Clin Pharmacol Ther Toxicol 30: 265–270. Zini R et al. (1988). Cicletanine binding to human plasma proteins and erythrocytes, a particular HAS–drug interaction. Life Sci 43: 2103–2115.
Ciclosporin Immunosuppressant C62H111N11O12 = 1202.6 CAS—59865-13-3 IUPAC Name 30-Ethyl-33-[(Z,1S,2R)-1-hydroxy-2-methylhex-4-enyl]-1,4,7,10, 12,15,19,25,28-nonamethyl-6,9,18,24-tetrakis(2-methylpropyl)-3,21-di(propan-2yl)-1,4,7,10,13,16,19,22,25,28,31-undecazacyclotritriacontane2,5,8,11,14,17,20,23,26,29,32-undecone Synonyms [R-[RR*(E)]]-Cyclic(L-alanyl-D-alanyl-N-methyl-L-leucyl-N-methyl-Lleucyl-N-methyl-L-valyl-3-hydroxy-N,4-dimethyl-L-2-amino-6-octenoyl-L-
a-aminobutyryl-N-methylglycyl-N-methyl-L-leucyl-L-valyl-N-methyl-L-leucyl); cyclosporin; cyclosporin A; cyclosporine. Proprietary Names Neoral; Sandimmun; Sandimmune, SangCya.
Chemical Properties A white or almost white powder. Soluble in methanol, ethanol, ether, chloroform and methylene chloride. It is practically insoluble in water and saturated hydrocarbons. Quantification Blood HPLC Column: C8 (Alltima, 100 2.1 mm i.d., 5 mm) at 70 . Mobile phase: 72% methanol : 28% 50 mmol/L ammonium acetate buffer (pH 5.1), flow rate 0.3 mL/min. Electron spray tandem mass spectrometry detection. Retention time: 11.8 min. Limit of detection, 5 mg/L [Taylor et al. 1998]. Column: Spherisorb S5 CN (250-4 phasesep, 250 4.6 mm, 5 mm) at 50 . Mobile phase: hexane : isopropanol (90:10), flow rate 1.45 mL/min. UV detection (l¼212 nm). Retention times: ciclosporin, 8.9 min; metabolites, 11.0, 12.9 and 16.3 min. Limit of detection, 0.01 mg/L [Khoschsorur et al. 1997]. Column: Si 60 silica (250 4 mm, 5 mm) at 60 . Mobile phase: hexane : ethanol (85:15), flow rate 1.5 mL/min. UV detection (l¼210 nm). Retention time: 3.4 min . Limit of quantification, 25 mg/L [Poirier et al. 1994]. Column: RP-18 (RP Velosep, (400 3.2 mm, 3 mm). Mobile phase: 50% acetonitrile : 11% methanol : 39% 0.01 mol/L dipotassium hydrogen orthophosphate buffer (pH 6.5), flow rate 1.5 mL/min. UV detection (dual wavelength, 220 and 230 nm). Retention time: 2.5 min. Limit of detection, 45 mg/L [Salm et al. 1993]. Disposition in the Body After oral administration of conventional formulations absorption is very variable; absolute bioavailability can vary from 1 mg/L suggesting saturation of binding process). Note For a review of the pharmacokinetics of clarithromycin, see Fraschini et al. [1993]. For reviews of clarithromycin, see Peters, Clissold [1992] and Barradell et al. [1993]. Dose Usually 250 or 500 mg twice daily. Barradell LB et al. (1993). Clarithromycin. A review of its pharmacological properties and therapeutic use in Mycobacterium avium-intracellulare complex infection in patients with acquired immune deficiency syndrome. Drugs 46: 289–312. Chu SY et al. (1991). Simultaneous determination of clarithromycin and 14(R)-hydroxyclarithromycin in plasma and urine using high-performance liquid chromatography with electrochemical detection. J Chromatogr 571: 199–208. Chu S et al. (1993). Single- and multiple-dose pharmacokinetics of clarithromycin, a new macrolide antimicrobial. J Clin Pharmacol 33(8): 719–726. Chu SY et al. (1992). Clarithromycin pharmacokinetics in healthy young and elderly volunteers. J Clin Pharmacol 32: 1045–1049. Fish DN, Abraham E (1999). Pharmacokinetics of a clarithromycin suspension administered via nasogastric tube to seriously ill patients. Antimicrob Agents Chemother 43(5): 1277–1280. Fraschini F et al. (1993). Clarithromycin clinical pharmacokinetics. Clin Pharmacokinet 25: 189–204. Kanfer I et al. (1998). Analysis of macrolide antibiotics. J Chromatogr A 812: 255–286. Kees F et al. (1998). Determination of macrolides in biological matrices by high-performance liquid chromatography with electrochemical detection. J Chromatogr A 812: 287–293. Macek J et al. (1999). Determination of roxithromycin in human plasma by high-performance liquid chromatography with spectrophotometric detection. J Chromatogr B, Biomed Sci Appl 723: 233–238. Peters DH, Clissold SP (1992). Clarithromycin. A review of its antimicrobial activity, pharmacokinetic properties and therapeutic potential. Drugs 44: 117–164. Sastre Toran˜o J, Guchelaar HJ (1998). Quantitative determination of the macrolide antibiotics erythromycin, roxithromycin, azithromycin and clarithromycin in human serum by high-performance liquid chromatography using pre-column derivatization with 9-fluorenylmethyloxycarbonyl chloride and fluorescence detection. J Chromatogr B, Biomed Sci Appl 720: 89–97. Taninaka C et al. (2000). Determination of erythromycin, clarithromycin, roxithromycin, and azithromycin in plasma by high-performance liquid chromatography with amperometric detection. J Chromatogr B, Biomed Sci Appl 738: 405–411.
Clefamide Antiamoebic C17H16Cl2N2O5 = 399.2 CAS—3576-64-5 IUPAC Name 2,2-Dichloro-N-(2-hydroxyethyl)-N-[[4-(4-nitrophenoxy)phenyl]methyl]-acetamide Synonym Chlorophenoxamide Proprietary Name Mebinol
Chemical Properties A lemon-yellow crystalline powder. Mp 134 to 137 . Practically insoluble in water; soluble 1 in 100 of ethanol, 1 in 40 of acetone, and 1 in 80 of chloroform. Log P (octanol/water), 3.2. Colour Tests Mandelin’s test—green!brown; Marquis test—yellow; sulfuric acid—yellow. Thin-layer Chromatography System TA—Rf 0.69; system TB—Rf 0.00; system TC—Rf 0.56; system TL—Rf 0.68 (acidified iodoplatinate solution, positive). Ultraviolet Spectrum Ethanol—303 nm (A11¼310b).
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:52:29
Clemizole Infrared Spectrum Principal peaks at wavenumbers 1244, 1666, 1510, 1595, 1078, 880 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 228, 182, 363, 88, 229, 76, 276, 257.
Disposition in the Body Poorly absorbed after oral administration. Dose Clefamide has been given in doses of 1.5 to 2.25 g daily.
Clemastine Antihistamine C21H26ClNO = 343.9 CAS—15686-51-8 IUPAC Name (2R)-2-[2-[(1R)-1-(4-Chlorophenyl)-1-phenylethoxy]ethyl]-1methylpyrrolidine Synonyms Meclastine; mecloprodin.
1121
Quantification Plasma GC NPD. Limit of detection, 0.06 mg/L [Davydova et al. 2000]. ECD. Limit of detection, 1 mg/L [Tham et al. 1978]. Disposition in the Body Therapeutic Concentration Following a single oral dose equivalent to 2 mg of clemastine to 12 subjects, peak plasma concentrations of about 0.002 mg/L were attained in 3 to 5 h [Tham et al. 1978]. Nineteen healthy men and women, aged 19 to 56 years, were orally administered a single 2.68 mg clemastine fumarate dose and in addition received the same dose three times daily for 3 days in a multidose study. The mean peak plasma concentrations reached were 0.75 mg/L for the single dose and 1.75 mg/L for the multiple dose. Each observed approximately 3.4 h after dosing [Davydova et al. 2000]. Dose The equivalent of 2 to 6 mg of clemastine daily. Davydova NN et al. (2000). Determination of clemastine in human plasma by gas chromatography with nitrogen-phosphorus detection. J Chromatogr B Biomed Sci Appl 744(1): 177–181. Tham R et al. (1978). Gaschromatography of clemastine. A study of plasma kinetics and biological effect. Arzneimittelforschung 28: 1017–1020.
Clemizole Antihistamine C19H20ClN3 = 325.8 CAS—442-52-4 IUPAC Name 1-[(4-Chlorophenyl)methyl]-2-(1-pyrrolidinylmethyl)-1Hbenzimidazole
Chemical Properties Soluble in chloroform. Clemastine Fumarate C21H26ClNO, C4H4O4 = 460.0 CAS—14976-57-9 Proprietary Names Aller-eze; Alginan; Tavegil; Tavegyl; Tavist; Xolamin. Chemical Properties A white crystalline powder. Mp 177 to 178 . Slightly
soluble in dilute acetic acid; soluble in methanol. Colour Tests Liebermann’s reagent—brown; Mandelin’s test—yellow-brown; Marquis test—yellow (green rim); sulfuric acid—yellow (green rim). Thin-layer Chromatography System TA—Rf 0.46; system TB—Rf 0.49; system TC—Rf 0.25; system TE—Rf 0.58; system TL—Rf 0.09; system TAE—Rf 0.88; system TAF—Rf 0.49 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—clemastine RI 2425, M (OH-methoxycarbinol-)-H2O RI 2220; system GF—RI 2710. High Performance Liquid Chromatography System HA—k 3.7; system HX—RI 501; system HZ—retention time 14.0 min. Ultraviolet Spectrum Aqueous acid—257 nm (A11¼27b).
Chemical Properties Crystals. Mp 167 . Log P (octanol/water), 4.4. Clemizole Hydrochloride C19H20ClN3, HCl = 362.3 CAS—1163-36-6 Proprietary Name Allercur Chemical Properties A white crystalline powder. Mp about 246 . Sparingly
soluble in water; soluble in ethanol and chloroform; practically insoluble in ether. Thin-layer Chromatography System TA—Rf 0.78; system TB—Rf 0.33; system TC—Rf 0.69; system TE—Rf 0.78; system TL—Rf 0.52; system TAE—Rf 0.76; system TAF—Rf 0.73 (Dragendorff spray, positive; acidified iodoplatinate solution, positive). Gas Chromatography System GA—clemizole RI 2620, M (oxo-) RI 2965. High Performance Liquid Chromatography System HA—k 4.8 (tailing peak); system HX—RI 420. Ultraviolet Spectrum Aqueous acid—275 nm (A11¼330a); aqueous alkali— 254 (A11¼253b), 269, 275, 283 nm.
Infrared Spectrum Principal peaks at wavenumbers 1090, 1011, 700, 763, 1121, 1210 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 84, 128, 179, 42, 85, 178, 214, 98.
Infrared Spectrum Principal peaks at wavenumbers 748, 765, 740, 833, 1111, 1010 cm1 (KBr disk).
C
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:52:36
1122
Clenbuterol
Mass Spectrum Principal ions at m/z 131, 256, 125, 42, 255, 89, 258, 257.
C Dose 40 to 160 mg of clemizole hydrochloride daily.
Clenbuterol b2-Adrenoceptor Agonist, Bronchodilator C12H18Cl2N2O = 277.2 CAS—37148-27-9 IUPAC Name 1-(4-Amino-3,5-dichlorophenyl)-2-(tert-butylamino)ethanol Synonyms 4-Amino-3,5-dichloro-a-[[(1,1-dimethylethyl)amino]methyl]benzenemethanol; 4-amino-a-[(tert-butylamino)methyl]-3,5-dichlorobenzyl alcohol; NAB-365.
Infrared Spectrum Principal peaks at wavenumbers 1414, 774, 640 cm1 (KBr pellets).
Chemical Properties Log P (octanol/water) 2.47 [Prezelj et al. 2003]. Clenbuterol Hydrochloride CAS—21898-19-1 IUPAC Name 1-(4-Amino-3,
5-dichlorophenyl)-2-(tert-butylamino)ethanol hydrochloride Synonym NAB-365C Proprietary Names Broncodil; Clenasma; Contrasmina; Contraspasmin; Monores; Prontovent; Spiropent; Ventolase; Ventipulmin. Chemical Properties Colourless, microcrystalline powder. Mp 174 to 175.5 . Very soluble in water, methanol and ethanol; slightly soluble in chloroform; insoluble in benzene. Thin-layer Chromatography System TAE—Rf 0.22 (clenbuterol), Rf 0.30 (M-hydroxy), Rf 0.87 (M2), Rf 0.87 (M3); system TB—Rf 0.13 (clenbuterol), Rf 0.01 (M-hydroxy), Rf 0.00 (M2), Rf 0.00 (M3); system TE—Rf 0.58 (clenbuterol), Rf 0.43 (M-hydroxy), Rf 0.02 (M2), Rf 0.06 (M3); system TF—Rf 0.00 (M2), Rf 0.19 (M3). Plates: silica gel 60 (10 10 cm). Mobile phase: ethyl acetate : methanol : propionic acid (A, 8 : 1 : 1) and ethyl acetate : methanol : ammonia (B, 8.5 : 1 : 0.5). Developed with Ehrlich’s reagent. Rf 0.40 (A) and 0.51 (B) [Courtheyn et al. 1991]. Gas Chromatography System GAI—RT 0.57 relative to 17a-methyl-5aandrostan-3b; system GA—RI 2100 (clenbuterol); RI 1895 (Art (-H2O)) , RI 2160 (Art (-H2O)). Column: HP Ultra 1 (25 m 0.2 mm i. d., 0.11 mm). Temperature programme: 80 for 2 min, 20 /min up to 160 , then 2 /min to 190 and 30 /min to 300 for 10 min. Carrier gas: He, flow rate 1.0 ml/min. EI ionisation, SIM acquisition mode. Retention time: 15.4 min for the clenbuterol–mono-TMS (tetramethylsilane) derivative [Courtheyn et al. 1991]. Column: Equity 1701 (10 m 0.1 mm i.d., 0.1 mm). Carrier gas: He, 0.6 mL/min. Temperature programme: 100 for 1 min to 300 at 55 /min for 2.1 min. EI ionisation at 70 eV, SIM acquisition mode. Retention time: 0.06 min. Limit of detection, 1.5 mg/L [Brunelli et al. 2006]. Column: 1% phenylmethylsilicone HP 1-MS (15 m 0.25 mm i. d., 0.25 mm). Carrier gas: He, 1.1 ml/min. Temperature programme: 150 for 1 min to 215 at 15 / min to 300 at 35 /min for 2 min. FS mode. Retention time: 5.09 min [Abukhalaf et al. 2000]. High Performance Liquid Chromatography System HAW—k 6.1 (column (a)), k 5.0 (column (b)); system HAA—retention time 10.8 min; system HX— RI 326 clenbuterol-hydroxy-, RI 297 (M-hydroxy-), RI 346 ((M2)-hydroxy-), RI 419 ((M3)-hydroxy-); system HY—RI 282. Ultraviolet Spectrum Aqueous acid (0.2 mol/L NH2SO4)—242, 295 nm; basic—240, 295 nm.
Mass Spectrum Principal ions at m/z 86, 57, 41, 127, 190, 243, 90, 174.
Quantification Blood GC-MS Column: ZB5MS capillary (15 m 0.25 mm i.d., 0.25 mm). Temperature programme: 50 for 1.5 min to 300 at 20 /min for 4 min. Full scan mode. Retention time: 10.3 min. Limit of quantification, 5 mg/L, limit of detection, 2.5 mg/L [Wingert et al. 2008]. Column: DB-1 capillary (25 m 0.2 mm i.d., 0.25 mm). Carrier gas: He, 1 mL/min. Temperature programme: 120 for 1 min to 300 at 25 /min for 2 min. EI ionisation at 70 eV, SIM acquisition mode. Limit of detection, 100 mg/L [Black, Hansson 1999]. LC-MS Column: BDS Hypersil (15 cm 2 mm i.d., 3 mm). Mobile phase: 2 mmol/ L formic acid : acetonitrile (97 : 3 to 5 : 95 over 5 min), flow rate 250 mL/min. ESI, full MS. Retention time: 5.81 min. Limit of detection, 0.01 mmol/L [Yuen et al. 2005]. Plasma GC-MS Column: HP 1MS cross-linked 1% phenyl methylsilicone (15 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1.1 mL/min. Temperature programme: 150 for 1 min to 215 at 15 /min to 300 at 35 /min for 2 min. SIM acquisition mode. Limit of quantification, 1.5 mg/L, limit of detection, 0.5 mg/L [Abukhalaf et al. 2000]. Column: OV 1701 fused silica capillary (25 m 0.22 mm i.d., 0.11 mm). Carrier gas: He, 14 psi. Temperature programme: 180 to 230 at 8 /min to 290 at 20 /min. NICI at 100 eV. Limit of detection, 5 ng/L [Girault et al. 1990].
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:52:39
Clenbuterol HPLC Column: Chirobiotic T (25 cm 4.6 mm i.d., 5 mm). Mobile phase: methanol : acetonitrile (70 : 30) containing 0.3% glacial acetic acid and 0.2% triethylamine, flow rate 1.0 mL/min. UV detection (l ¼ 246 nm). Retention time: 8.38 and 9.56 min for (–)-R- and (þ)-S-clenbuterol, respectively. Limit of detection, 0.4 mg/L, respectively, may be sufficient to cause death [Gerostamoulos et al. 1996].
C
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:56:11
1158
Codeine N-oxide
In 39 deaths in which codeine was implicated, the postmortem blood concentrations for codeine ranged from 24 h, before and after 3 freeze-thaw cycles and after storage at 20 for 1 month [Beyer et al. 2007].
Beyer J et al. (2007). Detection and validated quantification of toxic alkaloids in human blood plasma–comparison of LC-APCI-MS with LC-ESI-MS/MS. J Mass Spectrom 42: 621–633. Frank AA, Reed WM (1990). Comparative toxicity of coniine, an alkaloid of Conium maculatum (poison hemlock), in chickens, quails, and turkeys. Avian Dis 34: 433–437. Frank BS et al. (1995). Ingestion of poison hemlock (Conium maculatum). West J Med 163: 573–574. Galey FD et al. (1992). Toxicosis in dairy cattle exposed to poison hemlock (Conium maculatum) in hay: isolation of Conium alkaloids in plants, hay, and urine. J Vet Diagn Invest 4: 60–64. Lee ST et al. (2008). Separation and measurement of plant alkaloid enantiomers by RP-HPLC analysis of their Fmoc-Alanine analogs. Phytochem Anal 19: 395–402. Rizzi D et al. (1989). Rhabdomyolysis and acute tubular necrosis in coniine (hemlock) poisoning. Lancet 2: 1461–1462. Rizzi D et al. (1991). Clinical spectrum of accidental hemlock poisoning: neurotoxic manifestations, rhabdomyolysis and acute tubular necrosis. Nephrol Dial Transplant 6: 939–943. Scatizzi A et al. (1993). Acute renal failure due to tubular necrosis caused by wildfowl-mediated hemlock poisoning. Ren Fail 15: 93–96.
C
Coniine Hydrobromide C8H17N, HBr = 208.1 CAS—637-49-0 Chemical Properties Colourless crystals. Mp 211 . Soluble 1 in 2 of water and
1 in 3 of ethanol; soluble in chloroform and ether. Thin-layer Chromatography System TA—Rf 0.26; system TAE—Rf 0.05; system TAF—Rf 0.70; system TL—Rf 0.05; system TB—Rf 0.39; system TC—Rf 0.13; system TE—Rf 0.37 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—not eluted. High Performance Liquid Chromatography System HX—RI 214. Ultraviolet Spectrum Aqueous acid—266 nm (A11 ¼ 6b); aqueous alkali— 262, 268 nm.
Conotoxins Neurotoxin
Infrared Spectrum Principal peaks at wavenumbers 1033, 1007, 1575, 1300, 1078, 1139 cm1 (coniine hydrobromide (Nujol mull)). Mass Spectrum Principal ions at m/z 84, 82, 80, 56, 43, 28, 30, 41.
Quantification Plasma LC-MS Column: LiChroCART (125 2 mm i.d.). Mobile phase: 50 mmol/ L aqueous ammonium formate (pH 3.5) : acetonitrile (90 : 10 for 2 min to 20 : 80 at 5 min for 2 min to 90 : 10 at 10 min), flow rate 0.4 mL/min for 2 min to 0.6 mL/min for 5 min to 0.4 mL/min for 3 min. APCI, positive ion mode, SIM acquisition mode or tandem MS, ESI, MRM acquisition mode. Limit of quantification, 50 and 0.1 mg/L for single stage MS and tandem MS, respectively [Beyer et al. 2007]. Other GC-MS Plants, Hay and Cattle Urine. Column: HP-1 capillary (12 m 0.2 mm i.d., 0.33 mm). Carrier gas: He, 2 psi. Temperature programme: 40 for 2 min to 80 at 20 /min to 90 at 5 /min to 200 at 45 /min. Limit of detection not reported [Galey et al. 1992]. Chicken, Quail and Turkey Tissue. Column: DB-17 capillary (30 m 0.25 mm i.d.). Temperature programme: 50 for 10 min to 240 at 10 /min. EI ionisation at 70 eV and CI. Limit of detection, 0.58 mg/L [Frank, Reed 1990]. HPLC Plant Extracts. Column: Betasil C18 (100 2.1 mm i.d.). Mobile phase: 20 mmol/L ammonium acetate : methanol (50 : 50), flow rate 0.5 mL/min. UV detection. Limit of detection not reported [Lee et al. 2008]. Disposition in the Body Toxicity Coniine is well absorbed from the gastrointestinal tract and is very poisonous; the estimated minimum lethal dose is 150 mg and toxic symptoms may
Chemical Properties Conotoxins are potent neurotoxins produced by the venom of the marine cone snail, genus Conus. They are peptides of 10 to 30 amino acid residues, typically with one or more disulfide bond. There have been five conotoxins identified so far:
Toxin
Target
a d
Acetylcholine receptors Inhibits the inactivation of voltage-activated calcium channels (VACC) Inhibits potassium channels Inhibits VACC in muscles Inhibits N-type VACC
Κ m v
Ziconitide Analgesic/Calcium Channel Blocker/Neuroprotective C102H172N36O32S7 = 2639.13 CAS—107452-89-1
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:56:38
Copper Synonyms v-Conotoxin MVIIA; v-conopeptide MVIAA; SNX-111; CI-1009. Proprietary Name Prialt (Elan). Chemical Properties An v-toxin derived from Conus magus. Acts as a selective
N-type VACC blocker. Use As a pharmacological tool to target VACC; licensed by FDA for the relief of intrathecal pain.
Copper Metal Cu = 63.55 CAS—7758-98-7 Chemical Properties Reddish solid. Mp 1083 . Bp 2595 . Insoluble in water. Valencies: Cu(O), Cu(þ1), Cu(þ2), Cu(þ3). Found in nature in the elemental form and also in many minerals: cuprite, malachite, azurite, chalcopyrite, chalcocite and bornite. Extensively used in all industries and many applications because of its durability, ductility, malleability, and electrical and thermal conductivity. Copper Sulfate CuSO4 = 159.6 Synonyms Blue copperas; blue stone; blue vitriol; copper (II) sulphate; cupric
sulphate; Roman vitriol; Salzburg vitriol.
Chemical Properties Blue crystals. Decomposes at 560 . Very soluble in water;
soluble in methanol; slightly soluble in ethanol. Used in metal finishing, mineral froth flotation, wood preservatives, water treatment, fungicides, algicides, petroleum refining. Colour Tests Applicable to gastric contents and scene residues. Place a small volume (0.1 mL) of sample on to a filter paper to give a spot of an approximate diameter of 1 cm. Expose the spot to concentrated ammonia fumes and add 0.1 mL of a methanolic solution of dithiooximide (10 g/L)—Copper salts give an olivegreen stain. Chromium salts also give a green stain, but usually before the dithiooximide is added. Several other metals give yellow-brown or red-brown colours. Limit of detection, 1 mg/L. Confirmatory test Place 0.1 mL of the sample in a spotting-tile well and add 0.05 mL of 0.01 mol/L hydrochloric acid. Mix 0.1 mL of ammonium mercurithiocyanate reagent (prepared by mixing 8 g of mercuric chloride and 9 g of ammonium thiocyanate in 100 mL of water) with 0.1 mL of zinc acetate solution (10 g/L) and add to the sample in the well.—Copper salts form a violet precipitate of zinc mercurithiocyanate. Limit of detection, 50 mg/L Quantification Specimen Collection Blood—5 mL, K-EDTA tube; urine—sample of 24 h urine collection (for investigation of copper-related disease). Blood DPASV Limit of detection, 2 mg/L [Moreno et al. 1999]. AAS Gas flow: 4.5 L/min. Acetylene: 1.1 L/min (l ¼ 324.8 nm). Limit of detection not reported [Piekoszewski et al. 2000]. Gas flow: 2.3 mL/min. Hollow cathode lamp (l ¼ 324.7 nm). Limit of detection not reported [Chatterjee et al. 1994]. ETAAS Dry cycle: 78 for 40 s to 82 for 10 s to 86 for 5 s. Char cycle: 600 for 20 s. Atomisation cycle: 2600 for 7 s. Carrier gas: Ar, 60 mL/min. Zeeman AA mode (l ¼ 324.8 nm). Limit of detection, 1.3 mg/L [Liska et al. 1985]. FAAS Oxidant: air, 1.60 kg/cm2. Fuel: acetylene, 0.30 kg/cm2. Zeeman AA mode (l ¼ 324.8 nm). Flame continuous aspiration, microsampler/peak area, or microsampler/peak height mode. Limit of detection, 4.1, 10.3 and 25.3 mg/L for each mode, respectively [Liska et al. 1985]. ICP-AES Limit of detection, 10 mg/kg [Dinya et al. 2005]. Meinhard or Babington nebuliser (l ¼ 324.8 nm). Limit of detection, 1.7 and 2.2 mg/L for each nebuliser, respectively [Prohaska et al. 2000]. ICP-MS Plasma gas: 15 L/min. Auxiliary gas: 1.13 L/min. Nebuliser gas: 1.0 mL/ min. Limit of detection not reported [Rainska et al. 2007]. Plasma gas: 16 L/min. Auxiliary gas: 1 L/min. Nebuliser gas: 1.15 or 1.33 mL/min. Limit of detection, 5 mg/ L [De Boer et al. 2004]. Plasma ETAAS See Blood [Liska et al. 1985]. FAAS See Blood [Liska et al. 1985]. ICP-MS Plasma gas: 15 L/min. Auxiliary gas: 0.6 L/min. Nebuliser gas: 0.93 L/ min. Limit of detection not reported [Venelinov et al. 2004]. Serum ETAAS See Blood [Liska et al. 1985]. FAAS See Blood [Liska et al. 1985]. Perkin-Elmer hollow cathode lamp (l ¼ 324.8 nm). Limit of detection, not reported [Weinstock, Uhlemann 1981]. ICP-AES Plasma gas: Ar, 12 L/min. Nebuliser gas: Ar, 0.5 L/min. Nebuliser pressure: 2.3 bar. (l ¼ 324.8nm). Limit of detection, 0.02 mmol/L [Chappuis et al. 1992]. ICP-MS Plasma gas: Ar, 11.0 L/min. Auxiliary gas: Ar, 1.4 L/min. Nebuliser gas: Ar, 0.9 to 1.0 L/min. Limit of detection, 0.03 mg/L [Gercken, Barnes 1991]. Plasma gas: 13 L/min. Auxiliary gas: 1 L/min. Nebuliser gas: 0.72 L/min. Limit of detection not reported [Vanhoe et al. 1989]. Oral Fluid ICP-MS Plasma gas: 13 L/min. Auxillary gas: 0.55 L/min. Nebuliser gas: 0.1 L/min. Limit of detection, 0.04 mg/L [Menegario et al. 2001]. Urine ETAAS Dry cycle: 110 at 1 s for 20 s to 130 in 15 s for 20 s. Char cycle: 1250 in 30 s for 15 s; change gas from Ar to 5% H2 in Ar for 5 s. Atomisation cycle: 2300 for 5 s; normal gas, Ar 250 mL/min; purge gas, 5% H2 in Ar 250 mL/min. Hollow cathode lamp (l ¼ 324.8 nm). Limit of detection, 0.08 mg/L [Lin, Huang 2001]. Dry cycle: 110 at 7 s for 13 s. Char cycle: 900 in 7 s for 23 s. Atomisation
1163
cycle: 2700 in 2 s for 10 s, gas flow: 10 mL/min (l ¼ 324.8 nm). Limit of detection not reported [Halls et al. 1981]. ICP-MS Plasma gas: 15 L/min. Auxiliary gas: 1.13 L/min. Nebuliser gas: 1.0 mL/ min. Limit of detection, not reported [Rainska et al. 2007]. See Blood. Limit of detection, 2.5 mg/L [De Boer et al. 2004]. Note For a direct spectrophotometric method for determining copper in urine, see Jero`nimo et al. [2004]. Bone FAAS Carrier gas: air-acetylene (l ¼ 324.8 nm). Limit of detection not reported [Baranowska et al. 1995]. Hair AAS Gas flow: 2.3 mL/min. Hollow cathode lamp (l ¼ 324.7 nm). Limit of detection not reported [Chatterjee et al. 1994]. ETAAS Dry cycle: 80 to 120 in 10 s. Char cycle: 300 to 400 at 10 s. Atomisation cycle: 2700 to 2800 in 5 s. Carrier gas: 200 mL/min. Hitachi model 180-50, S.N.5721-2 (l ¼ 193.8 nm). Limit of detection, not reported [Kazi et al. 2006]. ICP-MS Plasma gas: 15 L/min. Auxiliary gas: 0.8 L/min. Nebuliser gas: 0.8 L/ min. Limit of detection not reported [Samanta et al. 2004]. Note For a study following the trace element hair analysis of one man over 20 years, see Klevay et al. [2004]. Liver ETAAS Dry cycle: 120 at 30 s for 10 s, Ar, 3.0 L/min. Char cycle: 700 in 30 s for 10 s, Ar, 3.0 L/min. Atomisation cycle: 2600 in 1 s for 7 s, gas stop. Zinc hollow cathode lamp (l¼ 324.8 nm). Limit of detection not reported [Treble et al. 1998] Nail ICP-MS See Hair [Samanta et al. 2004]. Teeth ICP-AES Perkin-Elmer Plasma 40 Spectrometer (l ¼ 324.8 nm). Limit of detection not reported [Chew et al. 2000]. Synovial Fluid ICP-MS Coolant gas: Ar, 15.0 L/min. Auxiliary gas: 0.99 L/ min. Nebuliser gas: 1.0 L/min. Limit of detection, low ng/L [Krachler et al. 2000a]. Colostrum ETAAS Dry cycle: 50 to 120 at 20 s for 20 s. Char cycle: 120 to 900 in 30 s for 30 s. Atomisation cycle: 1600 for 7 s. Limit of detection, 0.71 mg/L [Turan et al. 2001]. Milk ICP-AES Cooling gas: 13 L/min. Auxiliary gas: 0.5 L/min. Meinhard nebuliser (l ¼ 324.8 nm). Limit of detection, 0.012 mg/kg [Silva et al. 1997]. ICP-MS Plasma gas: 12 to 13 L/min. Auxiliary gas: 0.9 to 1.0 L/min. Sample gas: 1.0 to 1.2 L/min. Limit of detection, 180 mg/L [Krachler et al. 2000b]. Ascitic Fluid ETAAS Dry cycle: 60 to 90 in 15 s for 5 s to 100 in 10 s for 5 s to 110 in 15 s for 5 s to 140 for 5 s, 200 mL/min. Char cycle: 140 to 800 in 10 s for 20 s, 100 mL/min. Atomisation cycle: 2400 for 4 s. Lamp current: 10.0 mA (l ¼ 324.8 nm). Limit of detection, 0.4 mg/L [Milacic, Benedik 1999; Scancar et al. 1999]. Iliac Crest ETAAS Dry cycle: 60 to 90 in 10 s for 5 s to 100 in 10 s for 5 s to 150 in 10 s, 200 mL/min. Char cycle: 150 to 1000 in 10 s for 20 s, 100 mL/min. Atomisation cycle: 2700 for 4 s. Lamp current: 10.0 mA (l ¼ 324.8 nm). Limit of detection, 0.4 mg/L [Scancar et al. 2000]. Thyroid Tissue FAAS ATI UNICAM hollow cathode lamp. Limit of quantification, 70 ppb [Yaman, Akdeniz 2004]. Other DPASV Yemeni Khat. Limit of detection, 2.8 mg/kg [Matloob 2003]. ETAAS Eggs and Chicken Feed. Dry cycle: 120 at 1 s for 50 s. Char cycle: 1400 in 1 s for 30 s to 20 in 1 s for 5 s; Ar, 300 mL/min. Atomisation cycle: 2300 for 5 s (gas stop). Limit of detection, 0.74 mg/kg [Fakayode, Olu-Owolabi 2003]. Cocaine samples. Char cycle: 1300 . Atomisation cycle: 2400 (l ¼ 324.8 nm). Limit of detection not reported [Bermejo-Barrera et al. 1999]. ICP-AES Argentine wine. Outer gas: 8.5 L/min. Auxiliary gas: 1.0 L/min. Nebuliser gas: 1.0 L/min. (l ¼ 324.7 nm). Limit of detection, 40 ng/L [Lara et al. 2005]. ICP-MS Meals from Catering Establishments. Plasma gas: 15 L/min. Auxiliary gas: 0.9 L/min. Nebuliser gas: 0.73 L/min. Limit of quantification, 46 mg/kg [Noel et al. 2003]. Note For a study characterising copper in the uterine fluid of patients using the copper T-380A intrauterine device, see Arancibia et al. [2003]. Disposition in the Body Copper is absorbed from the gastrointestinal tract bound to amino acids or as ionic copper. Average absorption efficiencies in healthy adults range from 25 to 60%, although numerous factors affect this process, including the amount of copper in the diet, competition with other metals (zinc, iron and cadmium), and age. Roughly 80% of absorbed copper is excreted into the bile. Intestinal excretion into the bowel accounts for a further 18% and 2 to 4% appears in the urine. A small proportion of the copper taken into the liver is tightly bound to ceruloplasmin and this form accounts for over 90% of the copper present in serum. Copper is an essential element required for the normal functioning of at least 30 enzymes. The ability of copper to cycle between an oxidised state and a reduced state is used by cuproenzymes involved in redox reactions. However, it is this very property which poses a potential toxicity risk, as the transitions between oxidation states can generate superoxide radicals and hydroxyl radicals. Normal Concentrations Serum—0.7 to 1.6 mg/L (11 to 25 mmol/L); urine—< 50 mg/day (0.8 mmol/day); brain—5.1 to 8.3 mg/kg; kidney—1.2 to 3.1 mg/kg; liver—3.0 to 9.5 mg/kg; muscle—0.6 to 1.0 mg/kg; spleen—0.2 to 2.1 mg/kg. Concentrations in Copper-Related Liver Disease (Wilson's Disease) Serum—100 mg/day. Toxicity Acute occupational inhalation of copper fumes or copper dust causes respiratory problems and aching muscles. In a group of workers occupationally
C
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:56:39
1164
C
Copper
exposed to copper, serum levels averaged 1.26 mg/L [Cohen 1974]. Chronic occupational copper poisoning leads to nausea, vomiting, nervous disorders and hepatomegaly, and serum copper levels of 0.8 to over 2.0 mg/L have been reported [Suciu et al. 1977]. In cases of severe copper poisoning following ingestion of soluble salts (10 to 20 g), blood copper concentrations average ~8 mg/L [Chuttani et al. 1965]. An 18-month-old boy was admitted to hospital 1 h after dinking a solution containing ~3 g cupric sulfate. His serum copper concentration was 16.5 mg/ L, which decreased to 2.3 mg/L 24 h later [Walsh et al. 1977]. In 27 patients with acute copper sulfate poisoning, 11 developed renal failure. In these 11 patients serum copper concentrations ranged from 1.15 to 3.9 mg/ L [Chugh et al. 1977]. A person ingested an unknown quantity of wood-preserving solution containing large amounts of copper sulfate and sodium dichromate, with a smaller but substantial amount of an arsenic compound. Concentrations of copper in various tissues were reported as follows:
A 25-year-old woman ingested 25 diazepam 2.5 mg tablets. She was given 2.5 g cupric sulfate in 1.75 L water as an emetic. She died 3 days later. Postmortem copper concentrations were as follows:
Tissue
Concentration (mg/kg or mg/L)
Whole blood Brain Colon wall Gastric contents Gastric wall Intestinal contents Jejunal wall Kidney Liver
5.31 1.1 0.3 4.6 1.1 12.6 1.5 8.9 19.0
[Liu et al. 2001]. Tissue
Concentration (mg/kg)
Blood Brain Heart Kidney Liver Lung Spleen Stomach Urine
5.5 63 3.7 17.5 56 11.2 3.8 33 1.5 (mg/L)
[Cross et al. 1979].
A 58-year-old white woman died 18 h after admission to hospital. Postmortem examination revealed in the stomach 275 US coins, amounting to $14.84. Kidney and liver copper concentrations were 963 and 1160 mg/kg, respectively. Concentrations of nickel, chromium,and lead were within normal ranges [Yelin et al. 1987]. A 58-year-old man was found dead. Copper concentrations in tissues at postmortem were as follows:
Tissue
Concentration (mg/kg or mg/L)
Bile Blood Kidney Liver Lung Stomach contents
2.8 13.8 41.4 35.1 33.7 2988
Concentrations of zinc and cadmium were within the normal range [Kurisaki et al. 1988]. A 48-year-old woman went to a witch-doctor (iNyanga) where she was administered a concoction of salt, vinegar, sugar, methylated ethanol and copper sulfate. She lapsed into a coma and died ~48 h later. During this time she was given additional medication that may have contained more copper sulfate. At postmortem, her blood copper concentration was 42 mg/ L [Lamont, Duflou 1988]. An 11-year-old girl accidentally drank a solution containing 29 mg/L copper sulfate. Copper concentrations in her stomach contents were 240 mg/L, antemortem serum 25 mg/L, antemortem blood 16 mg/L, and postmortem blood 66 mg/L [Gulliver 1991; Mucklow 1997]. A 47-year-old man attempted suicide by injecting copper sulfate, both IV and SC. He was given haemodialysis for acute renal failure. His serum and dialysate copper levels were as follows:
Time
Day 1 Day 3
Serum copper
Dialysate copper
Pre-dialysis (mg/L)
Post-dialysis (mg/L)
Pre-dialysis (mg/L)
Post-dialysis (mg/L)
180 165
165 168
20 28
25 23
[Oldenquist, Salem 1999].
Note For a study of copper levels in the CSF of patients with multiple sclerosis, see Melo et al. [2003]; in the blood, serum and red blood cells of patients with motor neuron disease, see Pamphlett et al. [2001].,Rahil-Khazen et al. [2002] have studied trace element levels in the postmortem tissue of 30 Norwegians. For a case of suicide by ingesting a chromated copper arsenate wood preservative, see Hay et al. [2000]. For a report warning against preparing acidic drinks in copper urns, see Gill, Bhagat [1999]. For concentrations in the urine of glass-manufacturing workers, see Apostoli et al. [1998] and Arai et al. [1994]. For a review of the detection and monitoring of disorders of copper and other trace elements, see Taylor [1996]; for a study on the environmental influences on the trace element content of teeth, see Brown et al. [2004]. For a residential exposure to copper naphthenate, see Bluhm et al. [1992]. For 4 fatal poisonings following the ingestion of ‘spiritual water’, see Akintonwa et al. [1989]. For a case study on the fatal accidental ingestion of Clinitest (20 mg copper sulfate, 300 mg citric acid, 232.5 mg sodium hydroxide and 80 mg sodium carbonate), see O’Connor et al. [1984]. Half-life 26 days. Volume of Distribution 2.0 L/kg. Distribution in Blood Serum : erythrocyte ratio, 1.2.Lamont and Duflou, 1988 Akintonwa A et al. (1989). Fatal poisonings by copper sulfate ingested from ‘spiritual water’. Vet Hum Toxicol 31: 453–454. Apostoli P et al. (1998). Multiple exposure to arsenic, antimony, and other elements in art glass manufacturing. Am J Ind Med 34: 65–72. Arai F et al. (1994). Blood and urinary levels of metals (Pb, Cr, Cd, Mn, Sb, Co and Cu) in cloisonne workers. Ind Health 32: 67–78. Arancibia V et al. (2003). Characterization of copper in uterine fluids of patients who use the copper T-380A intrauterine device. Clin Chim Acta 332: 69–78. Baranowska I et al. (1995). The analysis of lead, cadmium, zinc, copper and nickel content in human bones from the upper Silesian industrial district. Sci Total Environ 159: 155–162. Bermejo-Barrera P et al. (1999). A study of illicit cocaine seizure classification by pattern recognition techniques applied to metal data. J Forensic Sci 44: 270–274. Bluhm RE et al. (1992). Increased blood and urine copper after residential exposure to copper naphthenate. J Toxicol Clin Toxicol 30: 99–108. Brown CJ et al. (2004). Environmental influences on the trace element content of teeth: implications for disease and nutritional status. Arch Oral Biol 49: 705–717. Chappuis P et al. (1992). A sequential and simple determination of zinc, copper and aluminium in blood samples by inductively coupled plasma atomic emission spectrometry. Clin Chim Acta 206: 155–165. Chatterjee J et al. (1994). Trace metal levels of X-ray technicians’ blood and hair. Biol Trace Elem Res 46: 211–227. Chew LT et al. (2000). Zinc, lead and copper in human teeth measured by induced coupled argon plasma atomic emission spectroscopy (ICP-AES). Appl Radiat Isot 53: 633–638. Chugh KS et al. (1977). Acute renal failure following copper sulphate intoxication. Postgrad Med J 53: 18–23. Chuttani HK et al. (1965). Acute copper sulfate poisoning. Am J Med 39: 849–854. Cohen SR (1974). A review of the health hazards from copper exposure. J Occup Med 16: 621–624. Cross JD et al. (1979). A suicide by ingestion of a mixture of copper, chromium and arsenic compounds. Forensic Sci Int 13: 25–29. DeBoer JL et al. (2004). Practical and quality-control aspects of multi-element analysis with quadrupole ICP-MS with special attention to urine and whole blood. Anal Bioanal Chem 379: 872–880. Dinya M et al. (2005). Major and trace elements in whole blood of phlebotomized patients with porphyria cutanea tarda. J Trace Elem Med Biol 19: 217–220. Fakayode SO, Olu-Owolabi IB (2003). Trace metal content and estimated daily human intake from chicken eggs in Ibadan, Nigeria. Arch Environ Health 58: 245–251. Gercken B, Barnes RM (1991). Determination of lead and other trace element species in blood by size exclusion chromatography and inductively coupled plasma/mass spectrometry. Anal Chem 63: 283–287. Gill JS, Bhagat CI (1999). Acute copper poisoning from drinking lime cordial prepared and left overnight in an old urn. Med J Aust 170: 510. Gulliver JM (1991). A fatal copper sulfate poisoning. J Anal Toxicol 15: 341–342. Halls DJ et al. (1981). Determination of copper in urine by graphite furnace atomic absorption spectrometry. Clin Chim Acta 114: 21–27. Hay E et al. (2000). Suicide by ingestion of a CCA wood preservative. J Emerg Med 19: 159–163. Jero`nimo PC et al. (2004). Direct determination of copper in urine using a sol-gel optical sensor coupled to a multicommutated flow system. Anal Bioanal Chem 380: 108–114.
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:56:39
Cotarnine Kazi TG et al. (2006). Evaluation of essential and toxic metals by ultrasound-assisted acid leaching from scalp hair samples of children with macular degeneration. Clin Chim Acta 369: 52–60. Klevay LM et al. (2004). Hair as a biopsy material: trace element data on one man over two decades. Eur J Clin Nutr 58: 1359–1364. Krachler M et al. (2000a). Concentrations of trace elements in osteoarthritic knee-joint effusions. Biol Trace Elem Res 75: 253–263. Krachler M et al. (2000b). Concentrations of selected trace elements in human milk and in infant formulas determined by magnetic sector field inductively coupled plasma–mass spectrometry. Biol Trace Elem Res 76: 97–112. Kurisaki E et al. (1988). Copper-binding protein in acute copper poisoning. Forensic Sci Int 38: 3–11. Lamont DL, Duflou JA (1988). Copper sulfate. Not a harmless chemical. Am J Forensic Med Pathol 9: 226–227. Lara R et al. (2005). Trace element determination of Argentine wines using ETAAS and USN-ICPOES. Food Chem Toxicol 43: 293–297. Lin TW, Huang SD (2001). Direct and simultaneous determination of copper, chromium, aluminum, and manganese in urine with a multielement graphite furnace atomic absorption spectrometer. Anal Chem 73: 4319–4325. Liska SK et al. (1985). Determination of copper in whole blood, plasma and serum using Zeeman effect atomic absorption spectroscopy. Clin Chim Acta 150: 11–19. Liu J et al. (2001). Death following cupric sulfate emesis. J Toxicol Clin Toxicol 39: 161–163. Matloob MH (2003). Determination of cadmium, lead, copper and zinc in Yemeni khat by anodic stripping voltammetry. East Mediterr Health J 9: 28–36. Melo TM et al. (2003). Manganese, copper, and zinc in cerebrospinal fluid from patients with multiple sclerosis. Biol Trace Elem Res 93: 1–8. Menegario AA et al. (2001). Determination of Ba, Cd, Cu, Pb and Zn in saliva by isotope dilution direct injection inductively coupled plasma mass spectrometry. Analyst 126: 1363–1366. Milacic R, Benedik M (1999). Determination of trace elements in a large series of spent peritoneal dialysis fluids by atomic absorption spectrometry. J Pharm Biomed Anal 18: 1029–1035. Moreno MA et al. (1999). Trace element levels in whole blood samples from residents of the city Badajoz, Spain. Sci Total Environ 229: 209–215. Mucklow ES (1997). Chemistry set poisoning. Int J Clin Pract 51: 321–323. Noel L et al. (2003). Determination of several elements in duplicate meals from catering establishments using closed vessel microwave digestion with inductively coupled plasma mass spectrometry detection: estimation of daily dietary intake. Food Addit Contam 20: 44–56. O’Connor HJ et al. (1984). Fatal accidental ingestion of Clinitest in adult. J R Soc Med 77: 963–965. Oldenquist G, Salem M (1999). Parenteral copper sulfate poisoning causing acute renal failure. Nephrol Dial Transplant 14: 441–443. Pamphlett R et al. (2001). Blood levels of toxic and essential metals in motor neuron disease. Neurotoxicology 22: 401–410. Piekoszewski W et al. (2000). Changes in serum copper level during detoxification of acutely poisoned drug addicts. Biol Trace Elem Res 78: 1–6. Prohaska C et al. (2000). Determination of Ca, Mg, Fe, Cu, and Zn in blood fractions and whole blood of humans by ICP-OES. Fresenius J Anal Chem 367: 479–484. Rahil-Khazen R et al. (2002). Multi-element analysis of trace element levels in human autopsy tissues by using inductively coupled atomic emission spectrometry technique (ICP-AES). J Trace Elem Med Biol 16: 15–25. Rainska E et al. (2007). Evaluation of occupational exposure in a slide bearings factory on the basis of urine and blood sample analyses. Int J Environ Health Res 17: 113–122. Samanta G et al. (2004). Arsenic and other elements in hair, nails, and skin-scales of arsenic victims in West Bengal, India. Sci Total Environ 326: 33–47. Scancar J et al. (1999). Problems related to determination of trace elements in spent continuous ambulatory peritoneal dialysis fluids by electrothermal atomic absorption spectrometry. Clin Chim Acta 283: 139–150. Scancar J et al. (2000). Determination of trace elements and calcium in bone of the human iliac crest by atomic absorption spectrometry. Clin Chim Acta 293: 187–197. Silva PR et al. (1997). Multielement determination in small samples of human milk by inductively coupled plasma atomic emission spectrometry. Biol Trace Elem Res 59: 57–62. Suciu I et al. (1977). Copper poisoning in the workers from a section of copper electrolysis. In: Zaidu SH, ed. Environmental Pollution and Human Health. Lucknow: Indian Toxicology Research Centre, 211. Taylor A (1996). Detection and monitoring of disorders of essential trace elements. Ann Clin Biochem 33: 486–510. Treble RG et al. (1998). Determination of copper, manganese and zinc in human liver. Biometals 11: 49–53. Turan S et al. (2001). Determination of heavy metal contents in human colostrum samples by electrothermal atomic absorption spectrophotometry. J Trop Pediatr 47: 81–85. Vanhoe H et al. (1989). Determination of iron, cobalt, copper, zinc, rubidium, molybdenum, and cesium in human serum by inductively coupled plasma mass spectrometry. Anal Chem 61: 1851–1857. Venelinov TI et al. (2004). Dialysis–Chelex method for determination of exchangeable copper in human plasma. Anal Bioanal Chem 379: 777–780. Walsh FM et al. (1977). Acute copper intoxication. Pathophysiology and therapy with a case report. Am J Dis Child 131: 149–151. Weinstock N, Uhlemann M (1981). Automated determination of copper in undiluted serum by atomic absorption spectroscopy. Clin Chem 27: 1438–1440. Yaman M, Akdeniz I (2004). Sensitivity enhancement in flame atomic absorption spectrometry for determination of copper in human thyroid tissues. Anal Sci 20: 1363–1366. Yelin G et al. (1987). Copper toxicity following massive ingestion of coins. Am J Forensic Med Pathol 8: 78–85.
1165
C
Chemical Properties Crystals. Mp 217 to 224 , with some decomposition. Very slightly soluble in water; fairly soluble in ethanol and acetone; sparingly soluble in chloroform and ether. Log P (octanol/water), 1.5. Cortisone Acetate C23H30O6 = 402.5 CAS—50-04-4 Synonym Cortisone 21-Acetate Note The name Cortisol is also applied to hydrocortisone. Proprietary Names Adreson; Cortate; Cortelan; Cortisol; Cortison; Cortistab;
Cortisyl; Cortone; Cortone Acetate; Sterop. Chemical Properties A white crystalline powder. Mp 235 to 238 , with decomposition. Soluble 1 in 5000 of water, 1 in 300 to 1 in 350 of ethanol, and 1 in 4 of chloroform; slightly soluble in ether. Colour Tests Naphthol–sulfuric acid—orange-brown/orange; sulfuric acid— yellow (green fluorescence under UV light). Thin-layer Chromatography Cortisone acetate: system TA—Rf 0.90; system TB—Rf 0.03; system TE—Rf 0.68; system TP—Rf 0.72; system TQ—Rf 0.28; system TR—Rf 0.55; system TS—Rf 0.00; system TAE—Rf 0.87; system TAJ—Rf 0.51; system TAK—Rf 0.09; system TAL—Rf 0.83; system TAM—Rf 0.91 (DPST solution). High Performance Liquid Chromatography System HT—k 2.4; system HY—RI 372. Ultraviolet Spectrum Cortisone acetate: ethanol—240 nm (A11¼390a).
Infrared Spectrum Principal peaks at wavenumbers 1700, 1660, 1235, 1720, 1275, 1750 cm1 (cortisone acetate, KBr disk). Quantification Plasma HPLC Fluorescence detection. Limit of detection, 0.1 mg/L [Shibata et al. 1998]. Urine HPLC MS/MS. Limit of detection, 2 mg/L [Taylor et al. 2002] and see Plasma. Hair HPLC MS. Limit of detection, 0.03 to 0.17 mg/g [Cirimele et al. 2000]. Dose 25 to 50 mg of cortisone acetate daily. Cirimele V et al. (2000). Identification of ten corticosteroids in human hair by liquid chromatography-ionspray mass spectrometry. Forensic Sci Int 107: 381–388. Shibata N et al. (1998). Simultaneous determination of glucocorticoids in plasma or urine by highperformance liquid chromatography with precolumn fluorimetric derivatization by 9-anthroyl nitrile. J Chromatogr B Biomed Sci Appl 706: 191–199. Taylor RL et al. (2002). Validation of a high-throughput liquid chromatography-tandem mass spectrometry method for urinary cortisol and cortisone. Clin Chem 48: 1511–1519.
Cortisone
Cotarnine
Corticosteroid C21H28O5 = 360.4 CAS—53-06-5 IUPAC Name (8S,9S,10R,13S,14S,17R)-17-Hydroxy-17-(2-hydroxyacetyl)-10,13dimethyl-1,2,6,7,8,9,12,14,15,16-decahydrocyclopenta[a]phenanthrene-3,11-dione Synonyms Compound E; 11-dehydro-17-hydroxycorticosterone; 17,21-dihydroxypregn-4-ene-3,11,20-trione.
Haemostatic C12H15NO4 = 237.3 CAS—82-54-2 IUPAC Name 4-Methoxy-6-methyl-7,8-dihydro-5H-[1,3]dioxolo[4,5-g]isoquinolin-5-ol Synonym 5,6,7,8-Tetrahydro-4-methoxy-6-methyl-1,3-dioxolo[4,5-g]isoquinolin-5-ol
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:56:42
1166
Coumaphos Ultraviolet Spectrum
C
Chemical Properties An alkaloid obtained by oxidising noscapine with nitric acid. Crystals. Mp 132 to 137 , with decomposition. Slightly soluble in water; soluble in dilute acids, ethanol, chloroform, benzene, and ether. Log P (octanol/ water), 0.6. Cotarnine Chloride C12H14ClNO3, 2H2O = 291.7 CAS—10018-19-6 (anhydrous); 16210-52-9 (dihydrate) Synonyms Cotarnine hydrochloride; stypticine. Chemical Properties A pale yellow deliquescent powder. Mp 197 . When heated
to decomposition, highly toxic fumes are evolved. Soluble in water, ethanol, and chloroform; practically insoluble in ether. Colour Tests Liebermann’s reagent—black; Mandelin’s test—redorange!brown. Thin-layer Chromatography System TA—Rf 0.02; system TB—Rf 0.38; system TC—Rf 0.01; system TE—Rf 0.26; system TL—Rf 0.00; system TAE—Rf 0.01; system TAF—Rf 0.22 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 1808. High Performance Liquid Chromatography System HA—k 8.2 (tailing peak). Ultraviolet Spectrum Aqueous acid—253 (A11¼582b), 332 nm (A11¼666b); aqueous alkali—286, 330 nm.
Infrared Spectrum Principal peaks at wavenumbers 1605, 1086, 1490, 1655, 1505, 1300 cm1 (cotarnine chloride, KCl disk). Dose Cotarnine chloride was formerly given in doses of 20 to 100 mg.
Infrared Spectrum Principal peaks at wavenumber 1774, 1046, 1004, 888, 1609, 1151 cm1.
Mass Spectrum Principal ions at m/z 97, 109, 362, 226, 65, 210, 125, 137.
Coumaphos Ectoparasiticide, Insecticide, Nematocide C14H16ClO5PS = 362.8 CAS—56-72-4 IUPAC Name 3-Chloro-7-diethoxyphosphinothioyloxy-4-methylchromen-2one Synonyms Bayer 21/199; coumafos; ENT 17957; OMS 485; phosphorothioic acid O-(3-chloro-4-methyl-2-oxo-2H-1-benzopyran-7-yl) O,O-diethyl ester. Proprietary Names Agridip; Asunthol; Baymix; Checkmite; Co-Ral; Diolice; Meldane; Muscatox; Negasunt; Perizin; Resitox; Suntol; Umbethion. Disposition in the Body Coumaphos is readily absorbed through the skin. It is rapidly broken down in the body into non-toxic products which are eliminated via urine (the majority) and faeces. There is no evidence of bioaccumulation. Toxicity Coumaphos is highly toxic if ingested or inhaled, and is moderately toxic via skin contact and if administered via IP routes.
Chemical Properties A white to tan to grey-coloured crystalline solid. Mp 95 to 92 in the technical state). It is practically insoluble in water; limited solubility in most organic solvents; soluble in chloroform. Log P (octanol/water), 4.13. Thin-layer Chromatography System TX—Rf 0.27; system TY—Rf 0.61. Gas Chromatography System GA—RI 2573.
Coumatetralyl Rodenticide C19H16O3 = 292.3 CAS—5836-29-3
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:56:47
Cresol IUPAC Name 2-Hydroxy-3-(1,2,3,4-tetrahydronaphthalen-1-yl)chromen-4-one Synonym 4-Hydroxy-3-(1,2,3,4-tetrahydro-1-naphthyl)coumarin Proprietary Name Racumin
1167
IUPAC Name 1-[(1R,3S)-3-Hydroxycyclohexyl]-5-(2-methyloctan-2-yl)phenol
C Chemical Properties A yellowish-white crystalline powder. Mp 172 to 176 . Practically insoluble in water; soluble in ethanol and acetone; slightly soluble in ether. pKa 4.8. Log P (octanol/water), 3.5. Thin-layer Chromatography System TD—Rf 0.73; system TE—Rf 0.13; system TF—Rf 0.74; system TX—Rf 0.14; system TY—Rf 0.30 (location under UV light, pink fluorescence; acidified potassium permanganate solution, positive). Gas Chromatography System GA—coumatetralyl RI 2635, coumatetralyl-HY RI 2250, coumatetralyl-Me-HY RI 2300, coumatetralyl isomer-1-Me RI 2655, coumatetralyl isomer-2-Me RI 2690, M (OH-) isomer-1-Me RI 2910, M (OH-) isomer2-Me2 RI 2925, M (OH-) isomer-3-Me2 RI 2935, M (OH-) isomer-4-Me2 RI 2990, M (di-OH-) isomer-1-Me2 RI 3005, M (OH-methoxy-)-Me2 RI 3070, M (di-OH-) isomer-2-Me2 RI 3085, M (di-OH-) isomer-3-Me3 RI 3105, M (tri-OH-)-H2O-Me2 RI 3175. High Performance Liquid Chromatography System HY—RI 598; system HAA—retention time 5.0 min. Ultraviolet Spectrum Aqueous acid—273, 283, 309 nm; aqueous alkali— 311 nm.
Chemical Properties Scientific research tool developed by Pfizer as a potent agonist of the cannabinoid CB1 receptor. It is the main active ingredient of the herbal incense known as Spice, specifically the 1,1-dimethyloctyl homologue. It is illegal in France and Germany. Mass Spectrum Principal peak at m/z 301. Quantification Blood LC-MS MRM acquisition mode. Limit of quantification, 0.6 mg/L, limit of detection, 0.2 mg/L [Neukamm et al. 2009]. Urine LC-MS Enhanced product ion scan. Limit of detection not reported [Kraemer et al. 2009]. GC-MS EI ionisation. Limit of detection, not reported [Kraemer et al. 2009]. Hair LC-MS See Blood [Neukamm et al. 2009]. Other LC-MS Herbal Products. Column: UPLC HSS T3 (100 2.1 mm i.d., 1.8 mm). Mobile phase: 0.1% formic acid in acetonitrile : water, flow rate 0.3 mL/ min. Limit of detection not reported [Uchiyama et al. 2009]. Kraemer T et al. (2009). Studies on the metabolism of JWH-018 and of a homologue of CP 47,497, pharmacologically active ingredients of different misused incense (‘Spice’) using GC-MS and LCMS techniques. Ann Toxicol Anal 21(Suppl 1): S21–S22. Neukamm MA et al. (2009). Quantitative determination of the active ‘Spice’ ingredient JWH-018 in blood and hair by liquid chromatography–tandem mass spectrometry. Ann Toxicol Anal 21: S1–21. Uchiyama N et al. (2009). Identification of cannabinoid analogs as new type of designer drugs in herbal products. Ann Toxicol Anal 21: S1-53–S1-54.
Cresol
Infrared Spectrum Principal peaks at wavenumbers 1615, 762, 740, 1685, 1250, 1570 cm1. Mass Spectrum Principal ions at m/z 292, 121, 188, 130, 115, 91, 128, 129.
Quantification Blood TLC Limit of quantification, 0.5 mg/L. Limit of detection, 0.2 mg/L [Berny et al. 1995]. Serum HPLC Fluorescence detection (lex¼318 nm; lem¼390 nm). 1 mg/L [Chalermchaikit et al. 1993]. Liver TLC Limit of detection, 0.2 mg/g, see Blood [Berny et al. 1995]. HPLC 1 ng/g, see Serum [Chalermchaikit et al. 1993]. Berny PJ et al. (1995). Anticoagulant poisoning in animals: a simple new high-performance thinlayer chromatographic (HPTLC) method for the simultaneous determination of eight anticoagulant rodenticides in liver samples. J Anal Toxicol 19(7): 576–580. Chalermchaikit T et al. (1993). Simultaneous determination of eight anticoagulant rodenticides in blood serum and liver. J Anal Toxicol 17(1): 56–61.
CP 47,497 Analgesic, Cannabinoid Agonist C21H34O2 = 318.5 CAS—70434-82-1
Disinfectant C21H24O3 = 324.4 CAS—1319-77-3; 95-48-7 (o-cresol); 108-39-4 (m-cresol); 106-44-5 (pcresol) Synonyms Cresylic acid; tricresol. Proprietary Name It is an ingredient of Lyseptol. Note The use of the name Lysol is limited. In some countries it is a trademark applied to a product of different composition.
Chemical Properties Cresol is a mixture of o-, m-, and p-cresols (CH3C6H4OH ¼ 108.1), in which the m-isomer predominates, and of other phenols obtained from coal tar. An almost colourless to pale brownish-yellow liquid, becoming darker with age or on exposure to light. Mass per mL 1.029 to1.044 g. Bp 195 to 205 . Almost completely soluble 1 in 50 of water; miscible with ethanol, benzene, chloroform, glycerol, petroleum ether, and ether. Soluble in solutions of fixed alkali hydroxides. pKa m-cresol 10.1, o-cresol 10.3, p-cresol 10.3 (25 ). Log P (octanol/water), 2.0. Colour Tests Folin–Ciocalteu reagent—blue; Liebermann’s reagent—black; potassium dichromate—brown (o-cresol 30 s, m-cresol 2 min); heat with about an equal quantity of phthalic anhydride and a few drops of sulfuric acid until the mixture is orange-brown; cool the mixture with a few drops of water and make alkaline with sodium hydroxide solution—red with o-cresol and blue-violet with m-cresol Thin-layer Chromatography System TAJ—m-cresol RF 69, o-cresol RF 73, p-cresol RF 66; system TAK—m-cresol RF 78, o-cresol RF 82, p-cresol RF 78; system TAL—RF 96 m-, o-, p-cresol. Gas Chromatography System GA—m-cresol RI 1065, o-cresol RI 1040, pcresol RI 1060, p-cresol-AC RI 1110.
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:56:52
1168
CR Gas
Ultraviolet Spectrum Aqueous alkali—239 (A11¼949b), 290 nm (A11¼251b); ethanol—275 (A11¼163b), 279 nm (A11¼145b).
CR Gas Benzodiazepine, Riot Control Agent C13H9ON = 195.2 CAS—257-07-8 IUPAC Name Dibenz[b][1,4]oxazepine Synonyms Dibenz[b,f][1,4]oxazepine; dibenzoxazepine; tear gas.
C
Infrared Spectrum Principal peaks at wavenumbers 1209, 816, 1515, 741, 1176, 1105 cm1 (p-cresol (Nujol mull)). Mass Spectrum Principal ions at m/z 107, 108, 77, 79, 51, 39, 53, 50 (p-cresol).
Quantification Blood GC FID. Limit of detection 20 mg/L [Bruce et al. 1976]. Serum HPLC Fluorescence detection (lex¼284 nm; lem¼310 nm). Limit of detection p-cresol 0.14 mg/L [De Smet et al. 1998]. Urine GC FID [Amorim et al. 1997]. ECD [Dills et al. 1997] and see Blood. HPLC UV detection (l¼270 nm). Limit of detection, 0.8 mg [Birkett et al. 1995]. UV detection (l¼271 nm). Limit of detection, 0.2 mg/L [Schlatter, Astier 1995]. Biological Fluids GC [Yashiki et al. 1990]. Faeces HPLC See Urine [Birkett et al. 1995]. Tissues GC See Blood [Bruce et al. 1976]. Disposition in the Body Absorbed after ingestion, and through the skin and mucous membranes. It is metabolised by conjugation and oxidation; p-cresol is endogenously produced in normal subjects, and may be present in urine at concentrations of 20 to 200 mg/L (mainly in conjugated form). Toxicity The estimated minimum lethal dose is 2 g, and the maximum permissible atmospheric concentration is 5 ppm. In 2 fatalities due to the ingestion of cresol, the following postmortem tissue concentrations were reported: blood 71, 190 mg/L; brain 2.8, – mg/g; kidney 396, – mg/g; liver 900, 480 mg/g; urine –, 304 mg/L [Bruce et al. 1976]. A 65-year-old man with schizophrenia died 15 min after he ingested a large volume of saponated cresol solution. Free p-cresol was detected in heart blood at a concentration of 458.8 mg/L and m-cresol at 957.3 mg/L; and the glucuronic acid conjugated forms at 38.2 and 85.6 mg/L, respectively. 250 mL of a cresol-odour-emitting fluid was also present in the stomach [Monma-Ohtaki et al. 2002]. A 46-year-old man ingested 100 mL of saponated cresol solution and was admitted to hospital. The serum concentrations of p-cresol and m-cresol were 43.3 mg/g and 73.8 mg/g, respectively, and the total concentration of cresol was 117 mg/g. Although levels were in the fatal ranges the individual recovered after hospital treatment [Yashiki et al. 1990]. Amorim LC et al. (1997). Determination of o-cresol by gas chromatography and comparison with hippuric acid levels in urine samples of individuals exposed to toluene. J Toxicol Environ Health 50: 401–407. Birkett AM et al. (1995). Simple high-performance liquid chromatographic analysis of phenol and p-cresol in urine and feces. J Chromatogr B Biomed Appl 674(2): 187–191. Bruce AM et al. (1976). Cresol poisoning. Med Sci Law 16: 171–176. De Smet R et al. (1998). A sensitive HPLC method for the quantification of free and total p-cresol in patients with chronic renal failure. Clin Chim Acta 278(1): 1–21. Dills RL et al. (1997). Quantitation of o-, m- and p-cresol and deuterated analogs in human urine by gas chromatography with electron capture detection. J Chromatogr B Biomed Sci Appl 703(1–2): 105–113. Monma-Ohtaki J et al. (2002). An autopsy case of poisoning by massive absorption of cresol a short time before death. Forensic Sci Int 126(1): 77–81. Schlatter J, Astier A (1995). Rapid determination of O- and P-cresol isomers in urine from workers exposed to toluene by high-performance liquid chromatography using a graphitized carbon column. Biomed Chromatogr 9(6): 302–304. Yashiki M et al. (1990). Gas chromatographic determination of cresols in the biological fluids of a non-fatal case of cresol intoxication. Forensic Sci Int 47(1): 21–29. Yashiki M et al. (1990). Gas chromatographic determination of cresols in the biological fluids of a non-fatal case of cresol intoxication. Forens Sci Int 47(1): 21–29.
Chemical Properties Odourless pale yellow solid; also reported as white solid. Mp 72 . Bp 335 . Sparingly soluble in water (3.73 mg/L). Soluble in organic solvents including chlorinated organics. Chemically stable in organic solvents. Log P (octanol/water), 3.49. CR hydrolyses slowly in water. CR riot control agent formulations generally consist of 0.1% CR dissolved in a solution of 80 parts of propylene glycol and 20 parts water. It may persist for prolonged periods in the environment because of the aqueous stability of benzodiazepines in aqueous media [Meylan, Howard 1995; Olajos, Salem 2001]. CR is the parent compound of the antipsychotic drug loxapine [Blain 2003]. Disposition in the Body CR metabolism has not been elucidated in humans owing to the very high sensitivity of humans to the irritant properties of CR. Animal studies have shown that aerosols of CR are rapidly absorbed from the respiratory tract, with a plasma half-life after inhalation exposure of ~5 min. It is readily absorbed by the cornea and corneal homogenates and metabolised to a lactam derivative. Studies in rats have shown that CR is rapidly absorbed from the gastrointestinal tract, undergoes hepatic metabolism, biliary secretion, enterohepatic circulation, and renal excretion. The major metabolic pathway of CR in the rat is oxidation to its more toxic lactam form, subsequent ring hydroxylation, sulfate conjugation and urinary excretion. Toxicity CR is a potent sensory irritant of low toxicity. It is less toxic than CS or CN by all routes of exposure. Human ocular irritancy thresholds and toxicity estimates are reported as follows: irritancy threshold 0.002 mg/m3, intolerable concentration 1 mg/ m3, lethal concentration (10 min exposure) 10 000 mg/m3 [Olajos, Salem 2001]. The inhalation toxicity of chemical warfare agents, military chemicals, and riot control agents is, by convention, expressed by the notation Ct. It is defined as the product of the concentration in mg/m3 multiplied by the exposure time (t) in minutes (mgmin/ m3). The terms LCt50 and ICt50 describe the airborne dosages that are lethal (L) or incapacitating (I) to 50% of the exposed population. The minimal irritant concentration estimate in humans is 0.002 mgmin/m3, with LCt50 and ICt50 estimates reported as >100 000 mgmin/m3 and 1 mgmin/m3, respectively. Animal LCt50 (mgmin/m3) values: mouse 169 500 to 203 600; rat 139 000 to 428 400; rabbit 160 000 to 169 000; guinea pig 169 500 [Olajos, Salem 2001]. Animal LD50 (mg/kg) values: rat 5900 (oral), 766 (IP), 68 (IV); mouse 112 (IV), 4000 (oral); rabbit 1760 (oral), 47 (IV); guinea pig 629 (oral), 463 (IP) [Ballantyne, Swanston 1978]. Note For reviews of riot control agents, see Blain [2003]; Hu et al. [1989]; Olajos and Salem [2001]; and Olajos and Stopford [2004]. Ballantyne B, Swanston DW (1978). The comparative acute mammalian toxicity of 1-chloroacetophenone (CN) and 2-chlorobenzylidene malononitrile (CS). Arch Toxicol 40: 75–95. Blain PG (2003). Tear gases and irritant incapacitants. 1-chloroacetophenone, 2-chlorobenzylidene malononitrile and dibenz[b,f]-1,4-oxazepine. Toxicol Rev 22: 103–110. Hu H et al. (1989). Tear gas: harassing agent or toxic chemical weapon? JAMA 262: 660–663. Meylan WM, Howard PH (1995). Atom/fragment contribution method for estimating octanol– water partition coefficients. J Pharm Sci 84: 83–92. Olajos E, Salem JH (2001). Riot control agents: pharmacology, toxicology, biochemistry and chemistry. J Appl Toxicol 21: 355–391. Olajos EJ, Stopford W (2004). Riot Control Agents: Issues in Toxicology, Safety and Health Care. Boca Raton, FL: CRC Press.
Cropropamide Respiratory Stimulant C13H24N2O2 = 240.3 CAS—633-47-6 (cropropamide); 8015-51-8 (prethcamide) IUPAC Name 2-[[(E)-But-2-enoyl]-propylamino]-N,N-dimethylbutanamide Synonym N-[1-[(Dimethylamino)carbonyl]propyl]-N-propyl-2-butenamide Note It is an ingredient of Prethcamide, which is a mixture of equal parts (by weight of) cropropamide and crotetamide. Proprietary Names It is an ingredient of Micoren and of Respirot.
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:56:54
CS Gas Chemical Properties A liquid. Easily soluble in water and ether; miscible with ethanol. Log P (octanol/water), 1.6. Thin-layer Chromatography System TA—Rf 0.70; system TB—Rf 0.29; system TC—Rf 0.69; system TE—Rf 0.74; system TL—Rf 0.57; system TAE—Rf 0.25; system TAF—Rf 0.83 (acidified potassium permanganate solution, positive). Gas Chromatography System GA—RI 1738. High Performance Liquid Chromatography System HZ—retention time 3.3 min. Ultraviolet Spectrum No significant absorption, 230 to 360 nm.
1169
Crotetamide Respiratory Stimulant C12H22N2O2 = 226.3 CAS—6168-76-9 (crotetamide); 8015-51-8 (prethcamide) IUPAC Name 2-[[(E)-But-2-enoyl]-ethylamino]-N,N-dimethylbutanamide Synonyms Crotethamide; N-[1-[(dimethylamino)carbonyl]propyl]-N-ethyl-2butenamide. Proprietary Names It is an ingredient of Micoren and of Respirot.
Chemical Properties A liquid. Easily soluble in water and ether; miscible with ethanol. Log P (octanol/water), 1.1. Thin-layer Chromatography System TA—Rf 0.68; system TB—Rf 0.28; system TC—Rf 67; system TE—Rf 0.69; system TL—Rf 0.55; system TAE—Rf 0.83; system TAF—Rf 0.79 (acidified potassium permanganate solution, positive). Gas Chromatography System GA—RI 1688. High Performance Liquid Chromatography System HX—RI 366; system HZ—retention time 2.6 min. Ultraviolet Spectrum No significant absorption, 230 to 360 nm.
Infrared Spectrum Principal peaks at wavenumbers 1654, 1617, 1219, 1282, 1098, 1123 cm1 (KBr disk). Dose Prethcamide (cropropamide and crotetamide) is usually given in doses of 1.2 to 1.6 g daily.
Crotamiton Acaricide, Antipruritic C13H17NO = 203.3 CAS—483-63-6 IUPAC Name N-Ethyl-N-(2-methylphenyl)-2-butenamide Proprietary Names Crotamitex; Eurax; Euraxil.
Infrared Spectrum Principal peaks at wavenumbers 1655, 1617, 1234, 1098, 1282, 1123 cm1 (KBr disk). Dose See under Cropropamide.
Chemical Properties A colourless or pale yellow, oily liquid. Soluble 1 in 400 of water; miscible with ethanol, ether, and methanol. Log P (octanol/water), 2.7. Thin-layer Chromatography System TA—Rf 0.83; system TE—Rf 0.83; system TAE—Rf 0.84 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 1600. Ultraviolet Spectrum No significant absorption, 230 to 360 nm.
Cryofluorane Aerosol Propellent, Refrigerant CClF2CClF2 = 170.9 CAS—76-14-2 IUPAC Name 1,2-Dichloro-1,1,2,2-tetrafluoroethane Synonyms Dichlorotetrafluoroethane; propellent 114; tetrafluorodichloroethane. Proprietary Names Arcton 33; Arcton 114.
refrigerant
114;
Chemical Properties A colourless non-flammable gas which, when liquefied by compression, forms a clear colourless liquid. Bp about 3.5 . In the liquid state it is practically immiscible with water, but miscible with dehydrated alcohol. Log P (octanol/water), 2.8. Gas Chromatography System GA—RI 361; system GI—Retention time 2.0 min. Mass Spectrum Principal ions at m/z 85, 135, 87, 137, 31, 101, 100, 50.
CS Gas Infrared Spectrum Principal peaks at wavenumbers 1656, 1623, 1235, 1280, 1316, 1590 cm1 (thin film). Use Topically in a concentration of 10%.
Organonitrile, Riot Control Agent C10H5ClN2 = 188.6 CAS—2698-41-1
C
Clarke's Analysis of Drugs and Poisons Chapter No. C Dated: 15/3/2011 At Time: 21:57:0
1170
2C-T-2
IUPAC Name [(2-Chlorophenyl)methylene]propanedinitrile Synonyms o-Chlorobenzalmalononitrile; o-chlorobenzylidenemalononitrile; b,b-dicyano-o-chlorostyrene. Proprietary Names Paralyzer. It is also an ingredient of Sabre.
C Chemical Properties White crystalline solid with pungent pepper-like odour. Mp 95 to 96 . Bp 310 to 315 . Sparingly soluble in water; soluble in acetone, dioxane, methylene chloride, ethyl acetate, benzene [O’Neil et al. 2006]. Log P (octanol/water), 2.75 [Meylan, Howard 1995]. CS can be disseminated as a dry powder (by thermal or explosive methods), via spraying of the molten material, or in solution with inorganic solvents. CS exists in three forms: CS (pure form), CS1 and CS2 (mixtures of the crystalline agent and an aerogel). CS2 contains 95% micronised CS, 5% Cab-o-Sil and 1% hexamethyldisilazane. CS hydrolyses slowly to o-chlorobenzaldehyde and malononitrile. Chemically, CS is the most persistent of the lachrymatory agents and will adsorb on to most porous surfaces [Olajos, Salem 2001]. CS is an SN2 alkylating agent that reacts directly with nucleophilic sites [Ballantyne, Swanston 1978]. Note For a study of the thermal degradation products of CS at elevated temperatures, see Kluchinsky et al. [2002]. Mass Spectrum Principal ions at m/z 153, 183, 126, 75, 50, 161, 99, 137. Quantification Other TLC Rat Urine. Plates: aluminium coated with silica gel H-60 (20 20 cm). Solvent system: dichloromethane : methanol : acetic acid (20 : 4 : 1). UV detection (with radioactive ink). Rf values: 2-chlorobenzaldehyde, 0.91; 2-chlorohippuric acid, 0.55; 2-chlorobenzylmercapturic acid, 0.63; 2-chlorobenzoic acid, 0.76; 2chlorobenzyl alcohol, 0.85. Limit of quantification not reported [Rietveld et al. 1988]. GC-MS CS Canisters. Column: HP5-MS (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 37 cm/s. Temperature programme: 80 for 2 min to 275 at 15 / min. EI ionisation at 70 eV. Retention times: CS, 15.5 min; 2-chlorobenzaldehyde, 7.7 min; CS epoxide, 13.6 min; 3-quinolinecarbonitrile, 15.1 min; CS isomer, 16.4 min. Limit of quantification not reported [Smith et al. 2002]. Disposition in the Body Animal and human studies have shown that CS is readily absorbed from the respiratory tract and rapidly distributed by the blood throughout the body. CS and its main metabolites in blood (dihydro-CS and 2chlorobenzaldehyde) have extremely short half-lives (CS blood half-life 50 kGy) [O’Neill et al. 1993]. Thin-layer Chromatography Plates: RP-18 and KC-18. Solvent systems: (A) methanol : water; (B) acetonitrile : water; (C) tetrahydrofuran : water. Location reagent: 4-(p-nitrobenzyl)pyridine for trichothecenes; UV (l ¼ 365 nm) for aflatoxins, ochratoxin A, citrinin; aluminium trichloride for sterigmatocystin; Azoene Fast Violet B for zearalenone; 3-methyl-2-benzthiazolinone hydrazone hydrochloride for patulin and penicillic acid. Solvent composition and Rf values were reported as follows:
Mobile phase solvent ratio, Rf value
DON 3-Acetyl-DON NIV T-2 toxin HT-2 toxin Neosolaniol Fusarenone-X Diacetoxyscirpenol Aflatoxin B 1 Aflatoxin B2 Aflatoxin G1 Aflatoxin G2 Sterigmatocystin Ochratoxin A Citrinin Penicillic acid Patulin Zearalenone [Abramson et al. 1989].
A
RP-18
KC-18
B
RP-18
KC-18
C
RP-18
KC-18
65 : 35 65 : 35 65 : 35 65 : 35 65 : 35 65 : 35 65 : 35 65 : 35 70 : 30 70 : 30 70 : 30 70 : 30 90 : 10 65 : 35 60 : 40 70 : 30 60 : 40 90 : 10
0.63 0.48 0.78 0.17 0.23 0.61 0.63 0.31 0.38 0.46 0.60 0.68 0.37 0.13 0.24 0.70 0.70 0.53
0.74 0.59 0.87 0.22 0.32 0.71 0.73 0.42 0.53 0.53 0.61 0.61 0.62 0.71 0.71 0.85 0.94 0.70
65 : 35 65 : 35 65 : 35 65 : 35 65 : 35 65 : 35 65 : 35 65 : 35 70 : 30 70 : 30 70 : 30 70 : 30 90 : 10 60 : 40 60 : 40 60 : 40 60 : 40 80 : 20
0.76 0.71 0.86 0.51 0.65 0.76 0.76 0.63 0.55 0.63 0.69 0.74 0.59 N/A N/A 0.62 0.66 0.59
0.81 0.78 0.85 0.43 0.58 0.75 0.75 0.59 0.60 0.60 0.73 0.73 0.85 0.78 0.95 0.90 0.85 0.85
N/A N/A N/A N/A N/A N/A N/A N/A 60 : 40 60 : 40 60 : 40 60 : 40 70 : 30 60 : 40 N/A 60 : 40 60 : 40 65 : 35
N/A N/A N/A N/A N/A N/A N/A N/A 0.44 0.46 0.60 0.65 N/A 0.14 N/A 0.45 0.49 0.32
N/A N/A N/A N/A N/A N/A N/A N/A N/A N/A N/A N/A N/A N/A N/A 0.98 0.60 N/A
Clarke's Analysis of Drugs and Poisons Chapter No. D Dated: 17/3/2011 At Time: 21:59:26
Deoxynivalenol Gas Chromatography-Mass Spectrometry Column: HP-5MS (30 m 0.25 mm, 0.25 mm). Carrier gas: He, 2.0 mL/min. Temperature programme: 80 for 2 min to 200 at 35 /min for 5 min. EI ionisation at 70 eV, positive ion mode, SIM acquisition mode (FID also used). Retention time: 17.5 min (N,O-bis(trimethylsilyl)acetamide : trimethylchlorosilane : N-trimethylsilylimidazole derivative). Limit of quantification not reported [Krska et al. 2004]. Column: DB-5MS (30 m 0.5 mm, 0.25 mm). Temperature programme: 150 for 1 min to 280 at 3 /min. EI ionisation at 70 eV, positive ion mode, SIM acquisition mode. Retention times: DON 9.7 min, 7-acetyl-DON 9.8 min, 4-acetyl-NIV 9.8 min, 3-acetyl-NIV 9.88 min, 15-acetyl-NIV 9.94 min, NIV 10.0 min, 3,15-diacetyl-DON 10.5 min, 4,15-diacetyl-NIV 10.7 min (TMS-ether derivatives). Limit of quantification not reported [RodriguesFo et al. 2002]. Column: BP-1 methylsilicone (25 m 0.33 mm i.d., 0.5 mm). Carrier gas: He, 3.0 mL/min. Temperature programme: 150 to 300 at 15 /min. EI ionisation at 70 eV, positive ion mode, SIM acquisition mode. Retention time: 5.7 min (trifluoroacetyl ester derivative). Limit of detection, 10 pg [Wreford, Shaw 1988]. High Performance Liquid Chromatography Column: C18 (150 4.6 mm i.d., 3 mm). Mobile phase: acetonitrile : water (1 : 1), flow rate 0.4 mL/min. UV detection (l ¼ 219 nm). Limit of quantification not reported [Krska et al. 2004]. Liquid Chromatography-Mass Spectrometry Column: C8 (150 3.0 mm i.d., 5 mm). Mobile phase: methanol : water both containing 0.1 mol/L ammonium acetate (20 : 80 to 80 : 20 over 18 min), flow rate 0.5 mL/min. ESI, positive ion mode, full-scan and SIM acquisition mode, MRM acquisition mode. Limit of quantification not reported [Krska et al. 2004]. Ultraviolet Spectrum Ethanol—218 nm[Krska et al. 2004; Kuronen 1989].
Infrared Spectrum See Krska et al. [2004]. Mass Spectrum
Quantification Blood GC-MS Column: BP-5 (12 m 0.22 mm i.d., 0.25 mm). Carrier gas: He, 8 psi. Temperature programme: 90 for 2 min to 180 at 20 /min to 240 at 5 / min for 2 min. CI, negative ion mode, SIM acquisition mode. Limit of detection, 2 to 7 ppb for DON, NIV, T-2, HT-2, T-2 tetraol, diacetoxyscirpenol, scirpentriol, 15MAS (heptafluorobutyrylimidazole derivatives) [Black et al. 1986]. Urine GC-MS Column: BP-1 methylsilicone (25 m 0.2 mm i.d., 0.25 mm). Carrier gas: He, 15 psi. Temperature programme: 160 for 1 min to 275 at 10 / min, for 5 min. EI ionisation at 70 eV, positive ion mode, SIM. Limit of detection, DON, NIV, T-2, HT-2, T-2 tetraol, diacetoxyscirpenol, scirpentriol and 2 to7 ppb for15-MAS (heptafluorobutyrylimidazole derivatives) [Black et al. 1986]. LC-MS Column: C18 (250 4.6 mm i.d., 5 mm). Mobile phase: water : methanol (10 : 90 to 20 : 90 over 45 min), flow rate 1.0 mL/min. ESI, positive ion mode, SIR. Retention time: 15.8 min. Limit of detection, 4 mg/L [Meky et al. 2003]. Faeces GC Column: DB-5 (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 40 cm/s. Temperature programme: 50 for 2 min to 205 at 40 /min for 10 min to 222 at 1 /min to 280 at 10 /min for 5 min. ECD (63Ni). Limit of quantification not reported (N,O-bis(trimethylsilyl)acetamide : trimethylchlorosilane : N-trimethylsilylimidazole derivative) [Sundstol, Pettersson 2003].
1205
Other TLC Maize Samples. Plates: silica gel. Solvent system: chloroform : methanol (94 : 6). Location reagent 20% aluminium chloride in methanol. Heat for 15 min at 110 to 120 . UV detection (l ¼ 365 nm). Limit of quantification not reported [Schaafsma et al. 1998]. Wheat and Maize Samples. Plates: precoated silica gel 60 (20 20 cm), dipped in 15% aluminium chloride solution, air-dried, activated at 105 for 1 h. Solvent system: chloroform : acetone: propan-2-ol (8 : 1 : 1). Plate heated at 120 for 7 min. UV detection. Rf 0.6 (blue fluorescent spot). Limit of quantification, 40 mg/kg in wheat and 100 mg/kg in maize [Fernandez et al. 1994; Trucksess et al. 1984]. GC Pig Plasma and Urine. Column: glass packed with 3% OV-17 on 100-120 mesh Supelcoport (1.8 m 2.0 mm i.d.). Carrier gas: He, 35 mL/min. Temperature programme: 165 isothermal. ECD (Ni63). Retention times: DON 6 min, T-2 tetraol 5 min, scirpenetriol 4.5 min (trifluoroacetic acid derivatives). Limit of detection, 4000 mg/m2 dexrazoxane (on its own) but there is no evidence of cumulative toxicity. Half-life 2.1 to 2.5 h. Volume of Distribution 22.0 to 22.4 L/m2 (or 0.5 to 1.3 L/kg), tends to be higher in children compared with adults.
Clarke's Analysis of Drugs and Poisons Chapter No. D Dated: 17/3/2011 At Time: 22:0:39
1218
Dextromethorphan
Clearance Between 6.25 and 7.88 L/h/m2, tends to be higher in children than adults. Protein Binding 24; acetone 6; acetonitrile 12.5 mg/kg daily for 5 to 8.5 months suffered from pancreatitis [Faulds, Brogden 1992]. A 36-year-old man with Gilbert Syndrome, AIDS and a history of severe myelosuppression when administered with zidovudine was treated with 12 mg/ kg once daily didanosine instead. Treatment was stopped after 15 weeks owing to side effects but was resumed after 6 weeks. Three months after treatment was restarted, the man was admitted to hospital with nausea, anorexia and abdominal pain. A CT scan showed he had a fatty liver with an enlarged globular pancreas. Renal dysfunction was observed the day before he died, caused by fulminent hepatic failure (a complication of treatment) [Lai et al. 1991]. Bioavailability Bioavailability is substantially reduced by administration with or after food and varies considerably between patients; it is reported to range from 20 to 40% depending on the formulation used. Half-life Plasma, didanosine, 30 min to 4 h; dideoxyadenosine triphosphate, more than 12 h; increased in patients with renal impairment. Volume of Distribution Mean, 1.01 L/kg in adults (range 0.76 to 1.29 L/kg); 24 L/m2 in children. Clearance Plasma clearance, 600 to 800 mL/min in adults; 510 mL/min/m2 in children. Total body, 0.08 L/kg/h. Protein Binding 90%. High concentrations are found in the kidney, ventricular myocardium, liver and skeletal muscle. It is extensively metabolised by hydrolysis to digitoxigenin bisdigitoxoside, digitoxigenin monodigitoxoside and digitoxigenin, and by hydroxylation to digoxin and the corresponding digoxigenin derivatives, all of which are active. Epimerisation to the inactive metabolites epidigitoxigenin and epidigoxigenin, followed by glucuronide conjugation, also occurs; dihydrodigitoxin has also been detected in plasma. Approximately 60– 80% of a single dose is excreted in the urine over a period of 3 weeks, mainly as metabolites, and up to 20% is eliminated in the faeces. The excretion of digitoxin and its active and inactive metabolites is very variable and is independent of dose and route of administration. Approximately 20–50% of a dose appears to be excreted as unchanged drug in urine and faeces in variable proportions, and the amount of unchanged drug in the urine has been reported to be greater during maintenance treatment than in single-dose studies. Digitoxin is a metabolite of acetyldigitoxin. Therapeutic Concentration In serum, usually in the range 0.01–0.03 mg/L. After a single oral dose of 20 mg/kg, given to 6 children, peak serum concentrations of 0.02–0.05 mg/L were attained in 1.5–2.5 h [Larsen, Storstein 1983]. Following daily oral doses of 0.1 mg to 7 subjects, steady-state serum concentrations of 0.009–0.026 mg/L (mean 0.017–mg/L) were reported [Haustein 1981]. Toxicity The estimated minimum lethal dose is 3 mg. Toxic effects are usually associated with serum concentrations of ~0.03 mg/L or more. In a 65-year-old woman who died almost 15 h after ingesting 7 mg digitoxin, the plasma concentration just before death was 0.212 mg/L [Krappweis et al. 1996]. Following a massive digitoxin overdose in a 20-month-old girl, an initial serum level of 0.629 mg/L was reported. After 6 doses of digoxin-specific antibody fragments, digitoxin was undetectable in the serum but reappeared on days 6 and 7 [Schmitt et al. 1994]. A subject who ingested 10 mg digitoxin recovered following treatment with activated charcoal; a plasma concentration of 0.26 mg/L was reported 4 h after the dose, decreasing to 0.027 mg/L at 80 h [Pond et al. 1981]. Half-life Plasma half-life, 3–16 days (mean 7 days). Volume of Distribution 0.4–0.8 L/kg (mean 0.6 L/kg); increased in children. Clearance 0.04 mL/min/kg from plasma; increased in children. Distribution in Blood Plasma : whole blood ratio, ~1.7. Protein Binding 95%. Note For a review of the clinical pharmacokinetics of digitoxin see Perrier et al. [1977]. Dose Maximum initial total dose of 1.6 mg over 1–2 days; maintenance, 50 to 200 mg daily. Al Hakiem MH et al. (1982). Fluoroimmunoassay of digitoxin in serum. Clin Chem 28: 1364–1366. Faber DB (1977). Quantitation with high-performance thin-layer chromatography and programmed multiple development with high-performance micro-thin-layer material for drug analyses in biological fluids. J Chromatogr 142: 421–430. Guan Fet et al. (1999). Identification and quantification of cardiac glycosides in blood and urine samples by HPLC/MS/MS. Anal Chem 71: 4034–4043. Haustein KO (1981). Interindividual differences in the pharmacokinetics of digitoxin and digoxin during long-term treatment. Eur J Clin Pharmacol 19: 45–51. Krappweis J et al. (1996). Digitoxin intoxication with lethal outcome. Eur J Med Res 1: 551–553. Larsen A, Storstein L (1983). Digitoxin kinetics and renal excretion in children. Clin Pharmacol Ther 33: 717–726. Nore AK et al. (1980). Digitalis glycosides in serum, urine, and cerebrospinal fluid, determined with a commercial radioimmunoassay. Clin Chem 26: 321–323. Oiestad EL et al. (2009). Determination of digoxin and digitoxin in whole blood. J Anal Toxicol 33: 372–378. Perrier D et al. (1977). Clinical pharmacokinetics of digitoxin. Clin Pharmacokinet 2: 292–311. Plum J, Daldrup T (1986). Detection of digoxin, digitoxin, their cardioactive metabolites and derivatives by high-performance liquid chromatography and high-performance liquid chromatography-radioimmunoassay. J Chromatogr 377: 221–231. Pond S et al. (1981). Treatment of digitoxin overdose with oral activated charcoal. Lancet 2: 1177–1178.
Clarke's Analysis of Drugs and Poisons Chapter No. D Dated: 17/3/2011 At Time: 22:3:8
Digoxin Santos SR et al. (1987). Simultaneous analysis of digitoxin and its clinically relevant metabolites using high-performance liquid chromatography and radioimmunoassay. J Chromatogr 419: 155–164. Schmitt K et al. (1994). Massive digitoxin intoxication treated with digoxin-specific antibodies in a child. Pediatr Cardiol 15: 48–49. Tracqui A et al. (1997). High-performance liquid chromatography–ionspray mass spectrometry for the specific determination of digoxin and some related cardiac glycosides in human plasma. J Chromatogr B Biomed Sci Appl 692: 101–109.
1251
High Performance Liquid Chromatography System HM—k 11.3; system HY—RI 347; system HAA—RT 13.9 min. Ultraviolet Spectrum
Digoxin Cardiac Glycoside C41H64O14 = 780.9 CAS—20830-75-5 IUPAC Name 3-[(3S,5R,8R,9S,10S,12R,13S,14S,17R)-3-[(2R,4S,5S,6R)-5-[(2S, 4S,5S,6R)-5-[(2S,4S,5S,6R)-4,5-Dihydroxy-6-methyloxan-2-yl]oxy-4-hydroxy-6methyloxan-2-yl]oxy-4-hydroxy-6-methyloxan-2-yl]oxy-12,14-dihydroxy-10,13dimethyl-1,2,3,4,5,6,7,8,9,11,12,15,16,17-tetradecahydrocyclopenta[a]phenanthren17-yl]-2H-furan-5-one Synonyms 3b-[(O-2,6-Dideoxy-b-D-ribo-hexopyranosyl-(1!4)-O-2,6dideoxy-b-D-ribo-hexopyranosyl-(1!4)-2,6-dideoxy-b-D-ribo-hexopyranosyl) oxy]-12b,14b-dihydroxy-5b-card-20(22)-enolide, digoxinum; digoxosidum. Proprietary Names Digacin; Digomal; Digosin; Digoxin(e) Nativelle; Dilanacin; Dogoxine; Eudigox; Grexin; Hemigoxine Nativelle; Lanacordin; Lanacrist; Lanicor; Lanoxin(e); Lanoxicaps; Lenoxin; Malpluxin; Neo-Dioxanin; Novodigal; Prodigox; Purgoxin; Rougoxin; Toloxin.
D
Infrared Spectrum Principal peaks at wavenumbers 1075, 1709, 1055, 1020, 1160, 1110 cm1 (KBr disk).
Chemical Properties Crystals. A glycoside obtained from the leaves of Digitalis lanata (Scrophulariaceae). Colourless crystals or a white powder. Mp ~240 , with decomposition. Practically insoluble in water, dehydrated alcohol, ether, acetone and ethyl acetate; soluble 1 in 122 of ethanol (80%) and 1 in 4 of pyridine; slightly soluble in chloroform; freely soluble in a mixture of equal volumes of chloroform and methanol. LogP (octanol/water), 1.26. Metildigoxin C42H66O14 = 795.0 CAS—30685-43-9 IUPAC Name 3-[(3S,5R,8R,9S,10S,12R,13S,14S,17R)-12,14-Dihydroxy-3-[(2R,4S,
5S, 6R)-4-hydroxy-5-[(2S,4S,5S,6R)-4-hydroxy-5-[(2S,4S,5S, 6R)-4-hydroxy-5methoxy-6-methyloxan-2-yl]oxy-6-methyloxan-2-yl]oxy-6- methyloxan-2-yl]oxy10,13-dimethyl-1,2,3,4,5,6,7,8,9,11,12,15,16, 17-tetradecahydrocyclopenta[a]phenanthren-17-yl]-2H-furan-5-one Synonyms 3b,5b,12b)-3-{[2,6-Dideoxy-4-O-methyl-b-D-ribo-hexopyranosyl(1!4)-2,6-dideoxy-b-D-ribo-hexopyranosyl-(1!4)-2,6-dideoxy-b-D-ribo-hexopyranosyl]oxy}-12,14-dihydroxycard-20(22)-enolide; medigoxin; b-methyldigoxin, 4-O-methyldigoxin. Proprietary Names Cardiolan; Lanirapid; Lanitop; Miopat. Chemical Properties Crystals. Mp 227 to 231 . a-Acetyldigoxin
C43H66O15 = 823.0 CAS—5511-98-8 Proprietary Names Lanatilin; Sandolanid. Chemical Properties Prisms. Mp 225 with decomposition. Very sparingly sol-
uble in ethyl acetate. b-Acetyldigoxin CAS—5355-48-6 Proprietary Names Corotal; Digostad; Digotab; Digox; Gladixol N; Kardiamed;
Longdigox; Novodigal (b-acetyldigoxin or digoxin); Stillacor.
Chemical Properties Needles. Sparingly soluble in ethyl acetate.
Colour Test Antimony pentachloride—yellow!brown!black-violet. Thin-layer Chromatography System TD—Rf 0.01; system TE—Rf 0.33; system TF—Rf 0.05; system TAD—Rf 0.28; system TAE—Rf 0.85 (perchloric acid solution, followed by examination under UV light, blue fluorescence; p-anisaldehyde reagent, blue).
Mass Spectrum Principal ions at m/z 73, 58, 57, 43, 41, 39, 29, 45 (no peaks above 360).
Quantification Blood LC-MS Column: Mightysil RP-18 (150 2.0 mm i.d., 5 mm). Mobile phase: acetonitrile-2 mmol/L ammonium acetate (20 : 80) : acetonitrile-2 mmol/L ammonium acetate (80 : 20; 100 : 0 for 2 min to 0 : 100 at 12 min for 3 min to 100 : 0 at 20 min), flow rate 0.2 mL/min. ESI or APCI, positive ion mode, TIC or SIM acquisition mode. Limit of quantification, 0.05 mg/L [Guan et al. 1999]. Note For a radioimmunoassay, see Fletcher et al. [1979]. Plasma LC-MS Column: Zorbax SB-C18. Mobile phase: methanol : 0.1 % formic acid in 10 mmol/L sodium acetate (55 : 45), flow rate 1.0 mL/min. ESI. Retention time: 1.9 min. Limit of quantification, 0.5 mg/L [Vlase et al. 2009]. Column: NovaPak C18 (150 mm 2.0 mm i.d., 4 mm). Mobile phase: acetonitrile : 2 mmol/L ammonium acetate (pH 3.0, 1 : 3), flow rate 200 mL/min. Retention time: 8.08 min. Limit of detection, 0.15–0.6 mg/L [Tracqui et al. 1997]. Note For a radioimmunoassay, see Nelson et al. [1979]. Serum HPLC Column: Spherisorb (150 4.6 mm i.d., 3 mm). Mobile phase: hexane : methylene chloride : acetonitrile : methanol (36 : 6.3 : 5.2 : 0.2), flow rate
Clarke's Analysis of Drugs and Poisons Chapter No. D Dated: 17/3/2011 At Time: 22:3:21
1252
D
Dihydralazine
1.6 mL/min. Fluorescence detection (lex¼ 217 nm, lem¼ 340 nm). Limit of detection, 0.25 mg/L [Tzou et al. 1995]. Immunochemical detection. Limit of detection, 160 pg/mL [Oosterkamp et al. 1994]. Note For a radioimmunoassay, see Butler et al. [1982]. Urine HPLC Column: Partisil 10 ODS (250 4.6 mm i.d.). Mobile phase: methanol : water (35 : 65 to 45 : 55), flow rate 2.0 mL/min. UV detection (l¼ 220 nm). Retention time: 21.9 min. Limit of detection, 278 ng [Gault et al. 1980] LC-MS See Blood [Guan et al. 1999]. Disposition in the Body The absorption of digoxin after oral administration is variable and subject to bioavailability differences. Absorption occurs mainly in the small intestine and is delayed in the presence of food. Digoxin is rapidly distributed throughout the body and 90% of an administered dose. Over 12 days, 50% drug is excreted in urine and 20% in faeces. Therapeutic Concentration Ten healthy volunteers were administered dopexamine hydrochloride (total dose 180 mg/kg) over 60 min by IV infusion. There was a proportional increase in plasma concentration with a peak at 124 mg/L after 1 h. Plasma concentrations rapidly decreased when the infusion was stopped [Fitton, Benfield 1990]. Toxicity Overdose effects are short lived owing to short half-life with pharmacological actions including tachycardia, palpitations, tremors, nausea, vomiting and anginal pain. Half-life 7 min; 11 min in patients with cardiac failure. Clearance Plasma clearance, 36 mL/min/kg. Dose An initial dose of 0.5 mg/kg body weight/min is administered, which can be increased up to 1 mg/kg body weight/min, if necessary. Further increases occur in 0.5 to 1.0 mg/kg/min increments at intervals of not less than 15 min with a maximum dose of 6 mg/kg body weight/min. Baker PR et al. (1995). Determination of dopexamine hydrochloride in human blood by highperformance liquid chromatography with electrochemical detection. J Chromatogr B Biomed Appl 667(2): 283–290. Fitton A, Benfield P (1990). Dopexamine hydrochloride. A review of its pharmacodynamic and pharmacokinetic properties and therapeutic potential in acute cardiac insufficiency. Drugs 39(2): 308–330.
Clarke's Analysis of Drugs and Poisons Chapter No. D Dated: 17/3/2011 At Time: 22:7:30
Dorzolamide
Dorzolamide Antiglaucoma, Carbonic Anhydrase Inhibitor, Sulfonamide C10H16N2O4S3 = 324.4 CAS—120279-96-1 IUPAC Name (4R,6R)-4-Ethylamino-6-methyl-7,7-dioxo-5,6-dihydro-4Hthieno[5,4-b]thiopyran-2-sulfonamide Synonyms (4S,6S)-4-(Ethylamino)-5,6-dihydro-6-methyl-4H-thieno[2,3-b] thiopyran-2-sulfonamide 7,7-dioxide; L-671152.
Dorzolamide Hydrochloride C10H16N2O4S3,HCl = 360.9 CAS—130693-82-2 Synonyms MK-0507; MK-507. Proprietary Name Trusopt. It is also an ingredient of Cosopt. Chemical Properties A white to off-white crystalline powder. Mp 283 to 285 .
Soluble in water. pKa 7.8 [Remko, der Lieth 2004], 8.4 [Supuran et al. 2003]. Log P (octanol/water), 0.71 [Remko, der Lieth 2004]. Thin-layer Chromatography Ophthalmic solution. Plates: Silica gel GF254 (20 20 cm, 0.25 mm). Solvent system: methanol : 25% ammonia (100 : 1.5). UV detection (l ¼ 254 nm). Rf 0.67 [Bebawy 2002]. High Performance Liquid Chromatography Ophthalmic solution. Column: C18 (150 4.6 mm i.d., 5 mm). Mobile phase: methanol : acetonitrile : phosphate buffer (pH 2.5; 8 : 10 : 85), flow rate 1.2 mL/min. UV detection (l ¼ 250 nm). Retention time: 4.1 min. Limit of quantification, 4 mg/L [Erk 2003]. Ultraviolet Spectrum Ophthalmic solution—Principal peak at 250.3 nm. Limit of detection, 34 mg/L [Erk 2002].
Quantification Blood HPLC Column: C8 (250 4.6 mm i.d., 5 mm) and C18 (50 4.6 mm i.d., 3 mm) in series. Mobile phase: acetonitrile : water-0.085% phosphoric acid-1.6 mg/ mL sodium octanesulfonate (25 : 75), flow rate 1 mL/min. UV detection (l ¼ 252 nm). Limit of quantification, 5 mg/L [Matuszewski et al. 1994a]. Plasma HPLC Column: C8 (250 4.6 mm i.d., 5 mm) and C18 (50 4.6 mm i.d., 3 mm) in series. Mobile phase: acetonitrile : water-0.085% phosphoric acid-1.6 mg/ mL sodium octanesulfonate (25 : 75), flow rate 1 mL/min. UV detection (l ¼ 252 nm). Limit of quantification, 5 mg/L [Matuszewski et al. 1994a]. LC-MS Column: Hypersil base-deactivated cyano (100 3.0 mm i.d., 5 mm). Mobile phase: acetonitrile : 10 mmol/L ammonium acetate-0.1% trifluoracetic acid (35 : 65), flow rate 0.6 mL/min. APCI, positive ion mode, MRM acquisition mode. Limit of quantification, 0.5 mg/L [Constanzer et al. 1997]. Urine HPLC Column: C8 (250 4.6 mm i.d., 5 mm) and C18 (50 4.6 mm i.d., 3 mm) in series. Mobile phase: acetonitrile : water-0.085% phosphoric acid-1.6 mg/ mL sodium octanesulfonate (24 : 76), flow rate 1 mL/min. UV detection (l ¼ 252 nm). Limit of quantification, 5 mg/L [Matuszewski et al. 1994a].
1299
Aqueous Humour HPLC Column: C8 (125 4.0 mm i.d.). Mobile phase: acetonitrile : 0.085% phosphoric acid-2.1 mg/mL octanesulfonic acid (25 : 75). UV detection (l ¼ 252 nm). Retention time: 13.5 min. Limit of quantification, 100 mg/L, limit of detection, 50 mg/L [Schmitz et al. 1999]. Other HPLC Animal Tissues. Column: C18. Mobile phase: acetonitrile : 1% TEA solution (pH 3.5; 7 : 93), flow rate 1 mL/min. UV detection (l ¼ 254 nm). Retention time: 16.5 min. Limit of quantification not reported [Inoue et al. 2004]. Note For methods describing the chiral separation of the stereoisomers of dorzolamide, see Matuszewski et al. [1994b]; Matuszewski and Constanzer [1992]. For a Raman spectroscopy method for the detection of dorzolamide in animal tissues, see Bauer et al. [1999]. Disposition in the Body The rate and extent of drug absorption after topical application is influenced by losses caused by tear turnover and blinking, and by protein binding in tears. Only a small part of the instilled drug becomes available for absorption. Studies have reported absorption rates of 50 to 73% of the applied drug. The drug then reaches the systemic circulation via drainage through the nasolacrimal duct and is subsequently absorbed from the nasopharyngeal mucosa or goes directly through the conjuctival blood vessels. Dorzolamide is metabolised to N-deethyldorzolamide in the systemic circulation via CYP2B1/2, CYP2E1 and CYP3A2. Steady-state concentrations are achieved after 6, 12 and 18 months of treatment. It binds strongly to human erythrocytes, more precisely to the enzymes carbonic anhydrase I and II, which are mainly located in red blood cells (>90%). Dorzolamide and its metabolite are excreted predominantly via the renal route. Therapeutic Concentration Thirty-two patients scheduled for routine cataract surgery were administered a drop of a 2% solution (equivalent to 0.76 mg) of dorzolamide. Maximum aqueous humour concentrations were reached 1 to 2 h after administration. The mean concentration was 1 mg/L after ~2 h, 0.7–1.0 mg/L after 4–6 h, and ~0.2 mg/L after 12 h [Schmitz et al. 1999]. Ten patients with open-angle glaucoma or ocular hypertension were administered a drop (30 mL) of a 2% solution of dorzolamide every 8 h (~4 mg/day) for 6 months. Red cell concentrations for dorzolamide and its metabolite were 23.0 and 4.9 mmol/L, respectively. Mean daily urine concentration for dorzolamide plus metabolite was 1.7 mg/day, although large variations were observed (range 0.1 to 2.9 mg/day) [Maren et al. 1997]. Toxicity For a case report of acidosis in a neonate, see Morris et al. [2003]; for a review on dorzolamide-induced thrombocytopenia, see Martin, Danese [2001]. Half-life Approximately 130 days. Clearance Approximately 5.4 L/h. Dose As eye drops containing 2% of the base. When used as monotherapy, the usual dose is one drop three times daily; a twice daily regimen is recommended when used in conjunction with a b-blocker. Bauer NJ et al. (1999). Non-invasive assessment of ocular pharmacokinetics using confocal Raman spectroscopy. J Ocul Pharmacol Ther 15: 123–134. Bebawy LI (2002). Application of TLC-densitometry, first-derivative UV-spectrophotometry and ratio derivative spectrophotometry for the determination of dorzolamide hydrochloride and timolol maleate. J Pharm Biomed Anal 27: 737–746. Constanzer ML et al. (1997). Low level determination of dorzolamide and its de-ethylated metabolite in human plasma by liquid chromatography with atmospheric pressure chemical ionization tandem mass spectrometry. J Pharm Biomed Anal 15: 1001–1008. Erk N (2002). Simultaneous determination of dorzolamide HCl and timolol maleate in eye drops by two different spectroscopic methods. J Pharm Biomed Anal 28: 391–397. Erk N (2003). Voltammetric and HPLC determination of dorzolamide hydrochloride in eye drops. Pharmazie 58: 870–873. Inoue J et al. (2004). Effects of dorzolamide hydrochloride on ocular tissues. J Ocul Pharmacol Ther 20: 1–13. Maren TH et al. (1997). Ocular absorption, blood levels, and excretion of dorzolamide, a topically active carbonic anhydrase inhibitor. J Ocul Pharmacol Ther 13: 23–30. Martin XD, Danese M (2001). Dorzolamide-induced immune thrombocytopenia: a case report and literature review. J Glaucoma 10: 133–135. Matuszewski BK, Constanzer ML (1992). Indirect chiral separation and analyses in human biological fluids of the stereoisomers of a thienothiopyran-2-sulfonamide (TRUSOPT), a novel carbonic anhydrase inhibitor with two chiral centers in the molecule. Chirality 4: 515–519. Matuszewski BK et al. (1994). Determination of MK-507, a novel topically effective carbonic anhydrase inhibitor, and its de-ethylated metabolite in human whole blood, plasma, and urine by high-performance liquid chromatography. J Chromatogr B Biomed Appl 653: 77–85. Matuszewski BK et al. (1994). Anall chiral separation of the stereoisomers of a novel carbonic anhydrase inhibitor and its deethylated metabolite, and the assignment of absolute configuration of the human metabolite and chiral degradation products. Pharm Res 11: 449–454. Morris S et al. (2003). Topical dorzolamide and metabolic acidosis in a neonate. Br J Ophthalmol 87: 1052–1053. Remko M, der Lieth CW (2004). Theoretical study of gas-phase acidity, pKa, lipophilicity, and solubility of some biologically active sulfonamides. Bioorg Med Chem 12: 5395–5403. Schmitz K et al. (1999). Population pharmacokinetics of 2% topical dorzolamide in the aqueous humor of humans. Invest Ophthalmol Vis Sci 40: 1621–1624. Supuran CT et al. (2003). Carbonic anhydrase inhibitors. Med Res Rev 23: 146–189.
D
Clarke's Analysis of Drugs and Poisons Chapter No. D Dated: 17/3/2011 At Time: 22:7:32
1300
Dosulepin
Dosulepin Antidepressant C19H21NS = 295.4 CAS—113-53-1 IUPAC Name (3Z)-3-(6H-Benzo[c][1]benzothiepin-11-ylidene)-N,N-dimethylpropan-1-amine Synonyms 3-Dibenzo[b,e]thiepin-11(6H)-ylidene-N,N-dimethyl-1-propanamine; dothiepin.
D
Chemical Properties Mp 55 to 57 . Log P (octanol/pH 7.4), 2.8 [Ilett et al. 1992], (octanol/water) 4.49 [Hansch et al. 1995]. Extraction yield (chlorobutane), 1 [Demme et al. 2005]. Unstable under conditions of putrefaction [Pounder et al. 1994].
Mass Spectrum Principal ions at m/z 58, 236, 40, 202, 235, 203, 42, 44; desmethyldosulepin 44, 204, 203, 202, 41, 221, 57, 55; dosulepin sulfoxide 58, 44, 31, 59, 57, 42, 45, 40.
Dosulepin Hydrochloride C19H21NS,HCl = 331.9 CAS—897-15-4 Synonym Dothiepin hydrochloride Proprietary Names Dopin; Dopress; Dothapax; Dothep; Jardin; Prepadine;
Prothiaden; Protiaden(e); Thaden; Xerenal.
Chemical Properties White to faintly yellow crystalline powder. Mp 218 to
221 . Soluble 1 in 2 of water, 1 in 8 of ethanol and 1 in 2 of chloroform; practically insoluble in ether.
Colour Tests Liebermann’s reagent—red-brown; Mandelin’s test—green; Marquis test—brown; sulfuric acid—violet. Thin-layer Chromatography System TA—Rf 0.51; system TB—Rf 0.49; system TC—Rf 0.42; system TE—Rf 0.65; system TF—dosulepin-S-oxide Rf 0.00; system TL—Rf 0.16; system TAE—Rf 0.27; system TAF—Rf 0.41 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—dosulepin RI 2380, M (OH-) RI 2500, M (nor-) RI 2421, M (N-oxide) RI 2100, M (OH-N-oxide) RI 2130, M (sulfoxide) RI 2392, M (norsulfoxide) RI 2421; system GB—dosulepin RI 2486, M (nor-) RI 2507, M (sulfoxide) RI 2533, M (norsulfoxide) RI 2839; system GF—dosulepin RI 2770, dosulepin sulfoxide RI 2820; system GM—dosulepin RRT 1.259, M (nor-) RRT 1.450 (both relative to iprindole). High Performance Liquid Chromatography System HA—dosulepin k 3.2, dosulepin sulfoxide k 4.6 (tailing peak), monodesmethyldosulepin k 2.2; system HF—k 3.60; system HX—RI 428; system HY—RI 367; system HZ—RT 5.7 min. Column: Hypersil (250 4.9 mm i.d., 5 mm). Mobile phase: acetonitrile : 0.15 mol/L sodium hexane sulfonic acid (60 : 40), flow rate 2.0 mL/min. UV detection (l ¼ 260 nm). Limit of detection 1.9 mg/L [Pawlak, Clark 1989]. Column: Spherisorb (25 cm 4.6 mm i.d., 5 mm). Mobile phase ethyl acetate : methanol : 3% ammonia (85 : 15 : 1), flow rate 1.0 mL/min. UV detection (l ¼ 260 nm). Retention time: 8.54 and 9.05 min for the cis and trans-isomers, respectively. Limit of detection, 1.0 mg/L [Li Wan Po, Irwin 1979]. Ultraviolet Spectrum Aqueous acid—230 (A11 ¼ 770a), 303 nm.
Infrared Spectrum Principal peaks at wavenumbers 763, 727, 747, 1252, 963, 717 cm1 (dosulepin hydrochloride), (KBr disk).
Quantification Blood GC-MS Column: DB-5 cross-linked 5% phenyl methyl siloxane (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1.0 mL/min. Temperature programme: 50 for 2 min to 180 at 30 /min to 280 at 5 /min for 19 min. Full scan mode. Retention time: 20.1 min. Limit of quantification, 0.05 mg/L [Paterson et al. 2004]. Column: HP-5 MS fused silica 95% dimethyl 5% diphenyl polysiloxane (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1 mL/min. Temperature programme: 180 to 200 at 5 /min for 2 min to 220 at 5 /min for 5 min to 240 at 5 /min and to 320 at 50 /min. Retention time: 16.5 min. MSD, SIM acquisition mode. Limit of detection, 0.5 mg/L [Keller et al. 2000]. Column: 3% OV-101 on GasChrom W HP 80/100 mesh (2 m 2 mm i.d.). Carrier gas: He, 20 mL/min. Temperature programme: 210 to 230 at 4 /min. EI ionisation at 70 eV. Limit of detection, 1 mg/L [Maguire et al. 1981b]. HPLC Column: Novapak C18 (300 3.9 mm i.d., 4 mm). Mobile phase: methanol : tetrahydrofuran : 10 mmol/L potassium hydrogen phosphate buffer (pH 2.6; 65 : 5 : 30), flow rate 0.8 mL/min. DAD. Limit of detection not reported [Cirimele et al. 1995]. Column: Chromspher C8 (10 0.3 cm i.d., 5 mm). Mobile phase: methanol : water (30 : 70 to 75 : 25 in 5 min), flow rate 0.7,L/min. DAD (l ¼ 230 nm). Retention time: 10.5 min, 10.7 min dosulepin-sulfoxide. Limit of detection not reported [Lambert et al. 1994]. Column: Apex II ODS silica (150 4.6 mm i.d., 5 mm). Mobile phase: 0.01 mol/L sodium dihydrogen phosphate buffer (pH 3.0) : acetonitrile (60 : 40), flow rate 1.5 mL/min. UV detection (l ¼ 255 nm). Limit of detection not reported [Pounder et al. 1994]. Column: Spheri-5 RP-18 (100 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : 0.1 mol/L sodium dihydrogen phosphate : diethylamine (pH 8.0; 40 : 57.5 : 2.5), flow rate 2.0 mL/min. UV detection (l ¼ 220 or 254 nm). Retention time: 14.6 min. Limit of detection, 0.05 mg/L [McIntyre et al. 1993]. Plasma GC Column: 3% OV-17 on Chromosorb W 100/120 mesh (90 0.25" o.d.). Carrier gas: 30 mL/min. Temperature: 275 . AFID. Limit of detection, 4.0 ng [Gifford et al. 1975]. GC-MS Column: 3% OV-17 on GasChrom Q 100/120 mesh (1 m 2 mm i.d.). Carrier gas: CH4, 8-10 mL/min. Temperature: 220 . EI/CI. Limit of detection, 0.5 mg/L [Crampton et al. 1980]. HPLC Column: Ultrasphere C8 (250 4.6 mm i.d.). Mobile phase: acetonitrile : 4 mmol/L 1-octanesulfonic acid-0.5 mmol/L N,N,N,N-tetramethylethylene diame (pH 2.5; 35: 65, flow rate 2 mL/min. UV detection (l ¼ 230 nm). Retention time: 10.8 min. Limit of detection not reported [Hackett et al. 1998]. Column: mBondapak phenyl (30 cm 4 mm i.d.). Mobile phase: acetonitrile : hydrogen phosphate : sodium chloride (35 : 0.01 : 0.01), flow rate 1.8 mL/ min. UV detection (l ¼ 230 nm). Limit of quantification, 2 mg/L [Ilett et al. 1992]. Column: cyano (250 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : 0.01 mol/L dipotassium hydrogen orthophosphate buffer (pH 7.0; methanol (50 : 30 : 20), flow rate 1.7 mL/min. UV detection (l ¼ 240 nm). Retention time: 4.6 min. Limit of detection, 50 mg/L [Taylor et al. 1992]. Column: Partisil 10 ODS (25 cm 4.6 mm i.d., 10 mm). Mobile phase: acetonitrile : 0.5% aqueous potassium dihydrogen phosphate (pH 3.0; 35 : 65), flow rate 2.0 mL/min. UV detection (l ¼ 231 nm). Retention time: 9 min. Limit of detection, 10 mg/L [Brodie et al. 1977].
Clarke's Analysis of Drugs and Poisons Chapter No. D Dated: 17/3/2011 At Time: 22:7:34
Dosulepin LC-MS Column: Inertsil C8 (150 2.0 mm i.d., 5 mm). Mobile phase: methanol : 10 mmol/L ammonium acetate (pH 5.0) : acetonitrile (70 : 20 : 10), flow rate 0.1 mL/min. SSI, positive ion mode, full scan mode. Retention time: 6.5 min. Limit of quantification, 0.1 mg/L, limit of detection, 0.08 mg/L [Shinozuka et al. 2006]. Column: Symmetry C18 (150 3 mm i.d., 5 mm). Mobile phase: acetonitrile : 0.1% formic acid (28 : 72 for 4 min to 70 : 30 in 1 min for 3 min to 28 : 72 in 0.7 min), flow rate 0.6 mL/min. APCI, positive ion mode. Limit of quantification, 10 mg/L, limit of detection, 5 mg/L [Kollroser, Schober 2002]. Serum GC-MS Column: HP-5 MS (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1 mL/min. Temperature programme: 60 for 1 min to 280 at 10 /min for 20 min. EI ionisation at 70 eV. Retention time: 22.5 min. Limit of detection, 0.025 mg/L, limit of detection, 80% of the dose is excreted in urine, 40 to 45% of which remains unchanged. The drug is distributed into central spinal fluid. Therapeutic Concentration Six healthy men, aged between 21 and 31 years, were administered an oral dose of 10 and 20 mg/kg body weight eflornithine, and a 10 mg/kg IV dose. Peak plasma concentrations of 31.4 to 51.0 nmol/L were reached, within 2 to 6 h, for the 10 mg/kg oral dose and 27.4 to 137.5 nmol/L for the 20 mg/kg dose (1.5 to 4 h) [Haegele et al. 1981]. Toxicity Ototoxicity can occur and loss of hearing has been observed with a cumulative dose. Low toxicity via ingestion and IP routes. Bioavailability Oral bioavailability, 55%. Half-life 3 to 4 h. Volume of Distribution 0.33 to 0.37 L/kg. Clearance Total body clearance, 1.17 mL/min/kg. Dose A dose of 100 mg/kg body weight is administered every 6 h, IV, for at least 14 days, which may be followed by a 300 mg/kg body weight daily dose (orally) for 3 to 4 weeks. Dose is decreased for patients with impaired renal function. Cohen JL et al. (1989). High-pressure liquid chromatographic analysis of eflornithine in serum. J Pharm Sci 78(2): 114–116. Gunaratna PC et al. (1994). Pharmacokinetic studies of alpha-difluoromethylornithine in rabbits using an enzyme-linked immunosorbent assay. J Pharm Biomed Anal 12(10): 1249–1257. Haegele KD et al. (1981). Kinetics of alpha-difluoromethylornithine: an irreversible inhibitor of ornithine decarboxylase. Clin Pharmacol Ther 30: 210–215.
Quantification Serum HPLC Column: Lichrosorb C18 (240 4.0 mm i.d.). Mobile phase: acetonitrile : water : methanol (35 : 35 : 30), flow rate 1 mL/min. UV detection (l ¼ 245 nm). Retention time: 9 min. Limit of quantification and limit of detection not reported [Heikinheimo et al. 1994]. Disposition in the Body Elcometrine is poorly absorbed by the oral route and rapidly metabolised during hepatic first-pass metabolism. It has a very short halflife because of its inability to bind to sex-hormone-binding globulin or to cortisolbinding globulin, transport proteins that prolong the biological half-life of many steroid hormones. Therapeutic Concentration A group of 25 postpartum nursing women were administered an implant containing 50 mg elcometrine. Mean serum concentrations were reported as 171.5 ng/L (463 pmol/L) on day 15 and 141.2 ng/L (381 pmol/L) on day 75. Elcometrine mean serum concentrations were also measured on day 75 in breast milk and in serum from babies and were reported as 373 pmol/L and 19.3 pmol/L, respectively [Coutinho et al. 1999]. After oral administration of 100 mg of elcometrine in fasting women, the mean peak plasma concentration reached 156.7 ng/L (423 pmol/L) after 10 min. This level was reduced by 50% at 60 min [Noe et al. 1993]. Six healthy postpartum women were administered 20 mg elcometrine parenterally. After 2 weeks, mean plasma and breast milk concentrations were measured by radioimmunoassay and reported as 62 and 38 mg/L, respectively [Lahteenmaki et al. 1990]. Bioavailability Approximately 12%. Half-life Biphasic, 0.13 h and 14.6 h. Volume of Distribution Approximately 263 L. Clearance Approximately 126 L/h. Coutinho EM et al. (1999). Use of a single implant of elcometrine (ST-1435), a nonorally active progestin, as a long acting contraceptive for postpartum nursing women. Contraception 59: 115–122. Heikinheimo O et al. (1994). The progestin ST 1435—rapid metabolism in man. Contraception 50: 275–289. Lahteenmaki PL et al. (1990). Milk and plasma concentrations of the progestin ST-1435 in women treated parenterally with ST-1435. Contraception 42: 555–562. Noe G et al. (1993). Pharmacokinetics and bioavailability of ST 1435 administered by different routes. Contraception 48: 548–556.
Eletriptan 5-HT1 Receptor Agonist, Antimigraine C22H26N2O2S = 382.5 CAS—143322-58-1 IUPAC Name 3-[[(2R)-1-Methylpyrrolidin-2-yl]methyl]-5-(2-phenylsulfonylethyl)-1H-indole Synonyms 5-[2-(Benzenesulfonyl)ethyl]3-(1-methylpyrrolidin-2(R)-ylmethyl)1H-indole; 3-[[(2R)-1-methyl-2-pyrrolidinyl]methyl]-5-[2-(phenylsulfonyl)ethyl]1H-indole; 3-[(1-methylpyrrolidin-2-yl)methyl]-5-(2-phenylsulfonylethyl)-1Hindole; UK-116044.
Elcometrine Contraceptive, Progestogen, Progestational Steroid C23H30O4 = 370.5 CAS—7759-35-5 IUPAC Name [(8R,9S,10R,13S,14S,17R)-17-Acetyl-13-methyl-16-methylidene3-oxo-2,6,7,8,9,10,11,12,14,15-decahydro-1H-cyclopenta[a]phenanthren-17-yl] acetate Synonyms 17-(Acetyloxy)-16-methylene-19-norpregn-4-ene-3,20-dione; 16methylene-17a-acetoxy-19-norpregn-4-ene-3,20-dione; 16-methylene-17a-acetoxy-19 norprogesterone; ST-1435. Proprietary Names Elmetrine; Nestorone.
Chemical Properties Crystals. Mp 178 to 179 .
Chemical Properties pKa 9.2 [Cooper et al. 1999]. Log P (octanol/water), 3.58 [Wishart 2006]. Eletriptan Hydrobromide C22H26N2O2S,HBr = 463.4 CAS—177834-92-3 Proprietary Name Relpax Chemical Properties Awhite to light pale coloured powder. Readily soluble in water.
Quantification Plasma HPLC Column (100 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : 500 mmol/L potassium phosphate buffer (pH 3.5) : water (30 : 6 : 64), flow rate 1.0 mL/min. UV detection (l ¼ 225 nm). Limit of quantification, 0.5 mg/L [Cooper et al. 1999]. Oral Fluid HPLC Column (100 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : 500 mmol/L potassium phosphate buffer (pH 3.5) : water (30 : 6 : 64), flow rate 1.0 mL/min. UV detection (l ¼ 225 nm). Limit of quantification, 0.5 mg/L [Cooper et al. 1999]. Disposition in the Body Readily absorbed after oral administration, with peak plasma concentrations reached 1.5 h after dosing (2 h in patients with migraine). It is primarily metabolised by CYP3A4, with a small contribution (0.4 units/mL. Bioavailability About 92%. Half-life 4 to 5 h, 6 to 7 h in the elderly. Volume of Distribution Between 5.2 and 9.3 L. Clearance Total body clearance varies from 0.8 to 1.9 L/h. Protein Binding Enoxaparin binds endogenous plasma proteins, for example, histidine-rich glycoprotein and fibronectin. Note For a review of the pharmacokinetics of enoxaparin, see Noble et al. [1995]. For a review of low-molecular-weight heparins, see Cziraky and Spinler [1993]. Dose Dose ranges from 1 mg (100 International Units, IU)/kg body weight every 12 h with increases in 0.5 to 1 mg/kg body weight increments to a single dose of 40 mg (2000 IU) once daily. Cziraky MJ, Spinler SA (1993). Low-molecular-weight heparins for the treatment of deep-vein thrombosis. Clin Pharm 12: 892–899. Frydman AM et al. (1988). The antithrombotic activity and pharmacokinetics of enoxaparine, a low molecular weight heparin, in humans given single subcutaneous doses of 20 to 80 mg. J Clin Pharmacol 28: 609–618. Larsen ML et al. (1978). Assay of plasma heparin using thrombin and the chromogenic substrate HD-Phe-Pip-Arg-pNA (S-2238). Thromb Res 13(2): 285–288. Noble S et al. (1995). Enoxaparin. A reappraisal of its pharmacology and clinical applications in the prevention and treatment of thromboembolic disease. Drugs 49: 388–410.
Enoximone Vasodilator, Inotrope C12H12N2O2S = 248.3 CAS—77671-31-9 IUPAC Name 4-Methyl-5-(4-methylsulfanylbenzoyl)-1,3-dihydroimidazol-2one Synonyms 1,3-Dihydro-4-methyl-5-[4-(methylthio)benzoyl]-2H-imidazol-2one; fenoximone; MDL-17043; MDL-19438; RMI-17043; YMDL-17043.
Infrared Spectrum Principal peaks at wavenumbers 1700, 1610, 1435, 1325, 830, 1090 cm1.
Mass Spectrum Principal ions at m/z 42, 248, 247, 32, 151, 45, 201, 69.
Clarke's Analysis of Drugs and Poisons Chapter No. E Dated: 15/3/2011 At Time: 21:25:36
Enprostil Quantification Plasma HPLC UV detection (l¼340 nm). Limit of quantification, 0.005 and 0.01 mg/L for enoximone and the metabolites, respectively [Morita et al. 1995]. Serum HPLC UV detection (l¼335 nm). Limit of detection, 0.01 mg/L for enoximone and metabolite [Cooper, Turnell 1986]. Limit of quantification, is 0.05 mg/L for enoximone and sulfoxide metabolite [Chan et al. 1984]. Urine HPLC UV detection (l¼340 nm). Limit of quantification, 0.5 and 1.0 mg/L for enoximone and the metabolites, respectively [Morita et al. 1995]. Disposition in the Body Following IV injection or infusion it undergoes metabolism in the liver to an active sulfoxide metabolite (piroximone). It is excreted primarily via the kidney, as metabolites in urine. After an IV dose, ~70% is excreted in urine as metabolites and 1% as the unchanged drug. Therapeutic Concentration Twenty infants, aged between 0.6 and 49.7 weeks (median 6.0 weeks), were administered a loading dose of 1 mg/kg enoximone over 2 min, followed by an infusion of 10 mg/kg/min for a median time of 97 h (24 to 572 h). The infants were already anaesthetised for weaning from cardiopulmonary bypass. The maximum plasma concentration ranged between 0.708 and 6.893 mg/L (median 1.536 mg/L) [Booker et al. 2000]. Twenty-three healthy male Japanese volunteers with a mean age of 22.0 years (range, 20 to 27 years) were administered either a 0.25, 0.5 or 1.0 mg/kg single IV bolus dose at 2-weekly intervals; or a single 2.0 mg/kg dose; or a single 1.0 mg/kg bolus dose on day 1 followed by 3-hourly interval doses on day 2; or a single 10 mg/kg/min continuous infusion over a 4-h period. In the first part of the study, the peak concentrations for the 0.25, 0.5, 1.0 and 2.0 mg/ kg dose were 0.31, 1.82, 3.40 and 5.38 mg/L, respectively observed at 0.06, 0.01 and 0.0 h. The sulfoxide metabolite concentrations were 0.23, 0.53, 1.08 and 1.82 mg/L at 0.25, 0.25, 0.17 and 0.19 h for the four doses, respectively. The multiple dosing with a 1 mg/kg dose produced peak concentrations of 2.35 mg/L at 3.21 h. The single 10 mg/kg/min dose produced concentrations of 0.51 mg/L at 4.0 h [Morita et al. 1995]. Toxicity Severe supraventricular and ventricular arrhythmias can occur. Half-life In healthy volunteers, 1 to 4 h, in patients with heart failure, 6 h (bolus injections), 8 h (continuous infusion); infants (0.6 to 49.7 weeks) 1.4 to 10.9 h (median 6.4); sulfoxide, 2.9 to 17.7 h (median 7.4). Volume of Distribution 1.1 to 3.6 L/kg; 1.4 to 15.5 L/kg (median 3.8) (infants aged 0.6 to 49.7 weeks). Clearance Plasma clearance, 3.7 to 13.0 mL/min/kg; 2.4 to 18.9 mL/min/kg (median 9.2) (infants aged 0.6 to 49.7 weeks). Protein Binding 85%. Dose Maximum of 24 mg/kg IV in 24 h. Booker PD et al. (2000). Enoximone pharmacokinetics in infants. Br J Anaesth 85(2): 205–210. Chan KYet al. (1984). Simultaneous analysis of a new cardiotonic agent, MDL 17,043, and its major sulfoxide metabolite in plasma by high-performance liquid chromatography. J Chromatogr 306: 249–256. Cooper JD, Turnell DC (1986). Automatic preparation of human serum samples for analysis of the drug enoximone and its sulphoxide metabolite using high-performance liquid chromatography. J Chromatogr 380(1): 109–116. Morita S et al. (1995). Pharmacokinetics of enoximone after various intravenous administrations to healthy volunteers. J Pharm Sci 84(2): 152–157. Tarral E et al. (1990). [Determination of enoximone and its principle metabolite in serum and urine using high pressure liquid chromatography]. Therapie 45: 1–6.
Enoxolone Dermatological Agent C30H46O4 = 470.7 CAS—471-53-4 IUPAC Name (2S,4aS,6aR,6aS,6bR,8aR,10S,12aS,14bR)-10-Hydroxy-2,4a,6a, 6b,9,9,12a-heptamethyl-13-oxo-3,4,5,6,6a,7,8,8a,10,11,12, 14b-dodecahydro-1Hpicene-2-carboxylic acid Synonyms Glycyrrhetic acid; glycyrrhetinic acid; (3b,20b)-3-hydroxy-11-oxoolean-12-en-29-oic acid. Proprietary Names Arthrodont; PO 12. It is an ingredient of Gelclair.
Chemical Properties A white or faintly cream-coloured powder. Mp 296 . Very sparingly soluble in water; soluble in ethanol, ether, pyridine and acetic acid; freely soluble in chloroform and dioxane; practically insoluble in petroleum ether. Log P (octanol/water), 6.9.
1335
Colour Tests Antimony pentachloride—orange-brown!violet; naphthol–sulfuric acid—red-brown/orange; sulfuric acid—yellow Thin-layer Chromatography System TD—Rf 0.21; system TE—Rf 0.07; system TF—Rf 0.46; system TAD—Rf 0.47. Ultraviolet Spectrum Aqueous acid—254 nm; aqueous alkali—260 nm (A11¼338b).
E
Mass Spectrum Principal ions at m/z 303, 262, 135, 175, 41, 55, 43, 95.
Disposition in the Body Enoxolone is a metabolite of carbenoxolone. Use Has been used topically in a concentration of 2%.
Enprostil Antiulcerative, Prostaglandin C23H28O6 = 400.5 CAS—73121-56-9 IUPAC Name Methyl 7-[(1R,2R,3R)-3-hydroxy-2-[(E,3R)-3-hydroxy-4-(phenoxy)but-1-enyl]-5-oxocyclopentyl]hepta-4,5-dienoate Synonyms rel-7-[(1R,2R,3R)-3-Hydroxy-2-[(1E,3R)-3-hydroxy-4-phenoxy-1butenyl]-5-oxocyclopentanyl]-4,5-heptadienoic acid methyl ester; (dl)-9-keto11a,15a-dihydroxy-16-phenoxy-17,18,19,20-tetranorprosta-4,5,13-trans-trienoic acid methyl ester; methyl (E)-(11R,15R)-11,15-dihydroxy-9-oxo-16-phenoxy17,18,19,20-tetranorprosta-4,5,13-trienoate; methyl 7-{(E)-(3R*)-3-hydroxy-2[(1R*,2R*,3R*)-3-hydroxy-4-phenoxybut-1-enyl]-5-oxocyclopentyl}hepta-4,5dienoate; RS-84135. Proprietary Names Camleed; Fundyl; Gadrin(e); Gardrin.
Chemical Properties White to off-white waxy solid. Soluble in alcohol, propylene glycol and propylene carbonate. Slightly soluble in water. Log P (octanol/ water), 1.45 [ACD 2007]. High Performance Liquid Chromatography Column: Partisil 5 ODS-3 (100 4.6 mm i.d., 5 mm). Mobile phase: tetrahydrofuran : methanol : 1 mmol/L phosphate buffer (pH 6.5; 15 : 30 : 55), flow rate 1.0 mL/min. UV detection (l ¼ 220 nm). Retention time: 8 min. Limit of quantification, 4 mmol/L [Kenley et al. 1986a]. Ultraviolet Spectrum Tetrahydrofuran : methanol : phosphate buffer— 268 nm Kenley et al. [1986a].
Clarke's Analysis of Drugs and Poisons Chapter No. E Dated: 15/3/2011 At Time: 21:25:44
1336
Entacapone Ultraviolet Spectrum Neutral—306 nm.
E Infrared Spectrum Principal peaks at wavenumbers 2209, 1630, 1610, 1537 cm1. Quantification Plasma HPLC Column: Spheri-5 normal phase silica (100 4.6 mm i.d., 5 mm) followed by Column: NC2 Spheri-5 silica (220 4.6 mm i.d., 5 mm). Mobile phase: dichloromethane : acetonitrile (20 : 70); or Column: Chemcosorb 7CN reversed phase (250 4.6 mm i.d., 7 mm). Mobile phase: methanol : water : acetic acid (40 : 60 : 0.1), flow rate 1.0 mL/min. Fluorometric detection (lex ¼ 325 nm). Limit of quantification, 5 ng/L [Kiang et al. 1991]. Other HPLC Capsules. Column 1: Sphericel C18 (300 4.6 mm i.d., 10 mm). Mobile phase 1: water, flow rate 1.5 mL/min. Column 2: Partisil 10 cation exchange (250 4.6 mm i.d., 10 mm). Mobile phase 2: tetrahydrofuran : methanol : 1.0 mmol/ L ammonium phosphate buffer (pH 2.4; 15 : 35 : 50), flow rate 1.0 mL/min. UV detection (l ¼ 220 nm). Retention time: 10.8 min. Limits of quantification and detection not reported [Kenley et al. 1986b]. Disposition in the Body Eliminated predominantly via the urine, with approx. 53% excreted after 48 h. After 144 h, a further 34% is eliminated via the faeces. Therapeutic Concentration Plasma concentrations in subjects administered an oral dose of 70 mg enprostil were measured at 15, 30, 60, 90 and 120 min post-dose and reported as 24, 113, 27, 21 and 7 mg/L, respectively. The plasma level at 3 h post-dose was below the limit of quantification of the HPLC–LIFD method used [Kiang et al. 1991]. Following a single oral dose of 1 mg/kg [3H]enprostil administered to 4 healthy volunteers, plasma concentrations of 0.94 mgequiv/L (enprostil plus unidentified metabolites) were reached within 30 to 60 min [Goa, Monk 1987]. Note For a study on the efficacy of enprostil combined with cimetidine compared with cimetidine alone in treating gastric ulcers, see Murata et al. [2005]. Half-life Approximately 34 h. Dose In acute treatment of adults with duodenal or gastric ulcers, 35 mg orally, twice daily for at least 4 weeks.
Mass Spectrum Principal ions at m/z 58, 233, 29, 304, 72, 305, 159, 288.
ACD (2007) ACD/LogP DB. Toronto ON: Advanced Chemistry Development www.acdlabs.com (accessed November 2007). Goa KL, Monk JP (). Enprostil. A preliminary review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy in the treatment of peptic ulcer disease. Drugs 34: 539–559. Kenley RA et al. (1986a). Stability-specific HPLC analysis of the antiulcer prostaglandin, enprostil, in a soft elastic gelatin capsule formation. J Liq Chromatogr 9: 3577–3595. Kenley RA et al. (1986b). Multidimensional column-switching liquid chromatographic method for dissolution testing of enprostil soft elastic gelatin capsules. J Pharm Sci 75: 999–1002. Kiang CH et al. (1991). Determination of femtomole/milliliter concentrations of enprostil acid in human plasma using high-performance liquid chromatography-laser-induced fluorescence detection. J Chromatogr 567: 195–212. Murata H et al. (2005). Combination of enprostil and cimetidine is more effective than cimetidine alone in treating gastric ulcer: prospective multicenter randomized controlled trial. Hepatogastroenterology 52: 1925–1929.
Entacapone Dopaminergic Agent, Antiparkinsonian C14H15N3O5 = 305.3 CAS—130929-57-6 IUPAC Name (E)-2-Cyano-N,N-diethyl-3-(3,4-dihydroxy-5-nitrophenyl)acrylamide Proprietary Names Comtan; Comtess.
Chemical Properties Powder. Mp 153 to 156 . pKa 4.5. Extraction yield (chlorobutane), 0 [Demme et al. 2005].
Quantification Plasma HPLC Amperometric detection. Limit of quantification, 0.01 mg/L [Karlson, Wikberg 1992]. Urine HPLC Column: ODS-2 silica Spherisorb (150 4.6 mm i.d., 5 mm). Mobile phase: 25 mmol/L sodium dihydrogen phosphate buffer and 10 mmol/L citric acid buffer (pH 2.2) : methanol (55:45), flow rate 1.0 mL/min. UV detection (l¼310 nm). Retention time: entacapone, 9.5 min; (Z)-entacapone, 5.8 min. Limit of detection, 0.5 mg/L [Wikberg et al. 1993]. Disposition in the Body Entacapone is rapidly absorbed from the gastrointestinal tract and undergoes extensive first-pass metabolism. Entacapone is converted to its (cis)-isomer, (Z)-entacapone, the main metabolite in plasma, followed by direct glucuronidation to inactive glucuronide conjugates. Four metabolites have been observed—M3, M6, M8 and M10. Elimination is mainly via faeces (80 to 90%) and the remainder in urine as glucuronide conjugates and (Z)-isomer. Therapeutic Concentration Twelve healthy males were administered single oral doses of entacapone: 5, 25, 50, 100, 200, 400 and 800 mg. Peak plasma concentrations ranged from 62 to 7280 mg/L, with increasing dose, and were reached between 0.46 and 0.88 h [Keranen et al. 1994].
Clarke's Analysis of Drugs and Poisons Chapter No. E Dated: 15/3/2011 At Time: 21:25:51
Ephedrine Toxicity No special hazard to humans, anaemia observed in repeated dose toxicity studies. Bioavailability Approximately 35% (oral). Half-life Elimination half-life, t1/2 (a-phase) 0.27 to 0.37 h and (b-phase) 1.59 to 3.10 h. Volume of Distribution 181 L. Clearance Total clearance, 800 mL/min. Protein Binding Extensively binds to plasma proteins and serum albumin ~98%. Dose A usual dose of 200 mg entacapone, up to ten times a day, with a maximum of 2000 mg. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Karlsson M, Wikberg T (1992). Liquid chromatographic determination of a new catechol-Omethyltransferase inhibitor, entacapone, and its Z-isomer in human plasma and urine. J Pharm Biomed Anal 10(8): 593–600. Keranen T et al. (1994). Inhibition of soluble catechol-O-methyltransferase and single-dose pharmacokinetics after oral and intravenous administration of entacapone. Eur J Clin Pharmacol 46 (2): 151–157. Wikberg T et al. (1993). Identification of major metabolites of the catechol-O-methyltransferase inhibitor entacapone in rats and humans. Drug Metab Dispos 21: 81–92.
1337
Ephedrine Sympathomimetic C10H15NO,½H2O = 174.2 CAS—50906-05-3 IUPAC Name (1R,2S)-2-(Methylamino)-1-phenylpropan-1-ol hydrate Synonyms Hydrated ephedrine; (aR)-a-[(1S)-1-(methylamino)ethyl]benzenemethanol hemihydrate. Proprietary Names It is an ingredient of Franolyn Expectorant and Letigen.
Chemical Properties An alkaloid obtained from species of Ephedra, or prepared synthetically. Colourless crystals or white crystalline powder or granules that decompose on exposure to light. Mp 40 (hemihydrate); in warm weather it slowly volatilises. Soluble 1 in 20 of water and 1 in 90% for both ezetimibe and its glucuronide conjugate. Note For a review of ezetimibe, see Kosoglou et al. [2005]. Dose Given orally in a usual dose of 10 mg once daily. Basha SJ et al. (2007). Concurrent determination of ezetimibe and its phase I and II metabolites by HPLC with UV detection: quantitative application to various in vitro metabolic stability studies and for qualitative estimation in bile. J Chromatogr B Analyt Technol Biomed Life Sci 853: 88–96. Chaudhari BG et al. (2007). Stability-indicating reversed-phase liquid chromatographic method for simultaneous determination of simvastatin and ezetimibe from their combination drug products. J AOAC Int 90: 1242–1249. Kosoglou T et al. (2005). Ezetimibe: a review of its metabolism, pharmacokinetics and drug interactions. Clin Pharmacokinet 44: 467–494. Li S et al. (2006). Liquid chromatography–negative ion electrospray tandem mass spectrometry method for the quantification of ezetimibe in human plasma. J Pharm Biomed Anal 40: 987–992. Oswald S et al. (2006a). A LC-MS/MS method to quantify the novel cholesterol lowering drug ezetimibe in human serum, urine and feces in healthy subjects genotyped for SLCO1B1. J Chromatogr B Analyt Technol Biomed Life Sci 830: 143–150. Oswald S et al. (2006b). Disposition and sterol-lowering effect of ezetimibe are influenced by singledose coadministration of rifampin, an inhibitor of multidrug transport proteins. Clin Pharmacol Ther 80: 477–485. Patrick JE et al. (2002). Disposition of the selective cholesterol absorption inhibitor ezetimibe in healthy male subjects. Drug Metab Dispos 30: 430–437. Singh S et al. (2006). Stress degradation studies on ezetimibe and development of a validated stability-indicating HPLC assay. J Pharm Biomed Anal 41: 1037–1040. Sistla R et al. (2005). Development and validation of a reversed-phase HPLC method for the determination of ezetimibe in pharmaceutical dosage forms. J Pharm Biomed Anal 39: 517–522.
E
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:44:54
F Famciclovir
Mass Spectrum Principal ions at m/z 43, 262, 136, 202, 135, 321, 83, 148.
Antiviral C14H19H5O4 = 321.3 CAS—104227-87-4 IUPAC Name [2-(Acetyloxymethyl)-4-(2-aminopurin-9-yl)butyl] acetate Synonyms 9-[4-Acetoxy-3-(acetoxymethyl)but-1-yl]-2-aminopurine; BRL-42810; FCV. Proprietary Names Famvir; Vectavir.
F
Chemical Properties A white to pale-yellow solid. Mp 102 to 104 . It is soluble in water (>25% at 25 ), acetone and methanol; sparingly soluble in ethanol and isopropanol. Log P (octanol/water, pH 4.8), 1.09; log P (octanol/buffer pH 7.4), 2.08. Gas Chromatography Column: methyl silicone (HP1, 0.2 mm i.d., 0.33 mm). Temperature: 280 . Carrier gas: He, flow rate 0.9 mL/min. MS detection. Retention index: 2552 [Mills, Roberson 1993]. High Performance Liquid Chromatography Column: ODS Apex 1 (100 4.6 mm i.d., 3 mm). Mobile phase: (A)-methanol : 0.01 mol/L disodium hydrogen orthophosphate (pH 7.0) (7:93); (B)-methanol : 0.01 mol/L disodium hydrogen orthophosphate (pH 7.0) (35:65). Elution programme: (A:B) (100:0) to (0:100) in 4 min, held for 1.5 min, back to initial conditions in 1 min, flow rate 2.0 mL/min. UV detection: dual wavelength (l1¼254 nm, l2¼305 nm). Retention time: penciclovir (metabolite), 1.6 min; 6-deoxyfamciclovir precursor, 2.6 min; famciclovir, 6.0 min [Winton et al. 1990]. Ultraviolet Spectrum Aqueous acid—224, 246, 310 nm; basic—304 nm.
Infrared Spectrum Principal peaks at wavenumbers 1730, 1616, 1216 cm1 (KBr disk).
Quantification Plasma HPLC Column: ODS Apex 1 (100 4.6 mm i.d., 3 mm). Mobile phase: methanol : 0.01 mol/L sodium phosphate buffer (pH 7.0) (5:95), flow rate 1.0 mL/ min. UV detection: dual wavelength (l1¼254 nm, l2¼305 nm). Retention time: penciclovir, 7.8 min; 6-deoxyfamciclovir precursor, 14.0 min. Limit of quantification, 0.2 mg/L [McMeekin et al. 1992]. Urine HPLC See Plasma. Limit of quantification, 10 mg/L [McMeekin et al. 1992]. Disposition in the Body Famciclovir is rapidly absorbed after oral administration and converted, by deacetylation, in blood, and oxidation (aldehyde oxidase), in the liver, to penciclovir (BR-39123) and several inactive metabolites. Virtually no famciclovir is detected in plasma or urine. Penciclovir is converted by intracellular virus-induced thymine kinase to penciclovir triphosphate which is responsible for the antiviral activity. The drug is excreted, primarily, in urine as the metabolite, penciclovir, and the 6-deoxy precursor, BRL-42359. 73% of penciclovir can be detected in urine over 24 h and the remainder appears in faeces. Elimination is reduced in patients with renal impairment. Therapeutic Concentration Twenty healthy male volunteers, with a mean age of 36 years, were administered single oral doses of 125, 250, 500 and 750 mg famciclovir, after 10 h fasting. The peak plasma concentrations of penciclovir: 0.84, 1.59, 3.34 and 5.09 mg/L, respectively, were reached within 0.5 to 0.75 h. Concentrations reached below 0.2 mg/L within 6 h for the 125 and 250 mg doses, and within 10 h for the higher doses [Pue et al. 1994]. Bioavailability Approximately 77%. Half-life 2 to 3 h. Volume of Distribution Steady state, 0.98 to 1.08 L/kg; metabolite, penciclovir, 1.5 L/kg. Clearance Approximately 0.48 L/h/kg. Distribution in Blood The blood : plasma ratio for the metabolite, penciclovir is ~1.0. Protein Binding The metabolite, penciclovir is 90% of a dose is excreted in the urine in the form of conjugates of the drug and hydroxylated metabolites. Therapeutic Concentration A single oral dose of 600 mg to 9 subjects produced a mean peak plasma concentration of 63 mg/L in 3 to 4 h. Daily oral doses of 1200 mg to 5 subjects produced a mean steady-state plasma concentration of 87 mg/L [Henson et al. 1980]. Following daily oral doses of 10 to 25 mg/kg to 18 children for 3 weeks, maximum steady-state plasma concentrations of 52 to 372 mg/L (mean 120) were reported 2 to 8 h after a dose [M€akel€a et al. 1983]. Toxicity Fenclofenac was used in rheumatic disorders but it had a high incidence of adverse effects, especially skin reactions, and was withdrawn from the market. Half-life Plasma half-life, 15 to 40 h (mean 26). Volume of Distribution About 0.2 L/kg. Distribution in Blood Plasma : whole blood ratio, about 1.7. Protein Binding About 99%. Dose 0.6 to 1.2 g daily. Henson R et al. (1980). Pharmacokinetics of fenclofenac following single and multiple doses. Eur J Drug Metab Pharmacokinet 5(4): 217–223. M€akel€a AL et al. (1983). Pharmacokinetics of fenclofenac in children with juvenile rheumatoid arthritis. Eur J Clin Pharmacol 25(3): 381–388.
Fendosal Analgesic C25H19NO3 = 381.4 CAS—53597-27-6 IUPAC Name 2-Hydroxy-5-(2-phenyl-4,5-dihydrobenzo[e]indol-3-yl)benzoic acid Synonym 5-(4,5-Dihydro-2-phenyl-3H-benz[e]indol-3-yl)-2-hydroxybenzoic acid Proprietary Name Alnovin
Infrared Spectrum Principal peaks at wavenumbers 1242, 1258, 1707, 1215, 1100, 772 cm1 (KBr disk). Chemical Properties A yellow powder. Mp 239 to 241 . Practically insoluble in water; soluble 1 in 50 of ethanol; slightly soluble in alkaline solutions and in propylene glycol. pKa 3.1. Thin-layer Chromatography System TA—Rf 0.95; system TE—Rf 0.22; system TAJ—Rf 0.05; system TAK—Rf 0.68; system TAL—Rf 0.83. Ultraviolet Spectrum Aqueous alkali—292 nm (A11¼564b). Infrared Spectrum Principal peaks at wavenumbers 1675, 1242, 1500, 1615, 1298, 765 cm1 (KBr disk). Quantification Plasma Fluorescence spectrophotometry Limit of detection, 0.1 mg/L [Hill et al. 1980]. Dose Fendosal has been given in doses of 200 to 400 mg. Hill HM et al. (1980). Rapid fluorimetric procedure for the analysis of fendosal in plasma and data following oral dosing. Biopharm Drug Dispos 1: 97–102.
Fenetylline
Mass Spectrum Principal ions at m/z 296, 215, 217, 298, 216, 152, 181, 251.
Quantification Plasma GC ECD. Limit of detection, 300 mg/L [Henson et al. 1980].
Xanthine Stimulant C18H23N5O2 = 341.4 CAS—3736-08-1 IUPAC Name 1,3-Dimethyl-7-[2-(1-phenylpropan-2-ylamino)ethyl]purine2,6-dione Synonyms Amfetyline; 3,7-dihydro-1,3-dimethyl-7-[2-[(1-methyl-2-phenylethyl) amino]ethyl]-1H-purine-2,6-dione; 7-ethyltheophylline amfetamine; fenethylline.
Chemical Properties Practically insoluble in water; soluble in chloroform. Extraction yield (chlorobutane), 0.9 [Demme et al. 2005].
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:47:1
Fenfluramine Fenetylline Hydrochloride C18H23N5O2,HCl = 377.9 CAS—1892-80-4 Synonyms Fenethylline hydrochloride; H814; R-720-11. Proprietary Names Captagon; Fitton. Chemical Properties A white crystalline powder. Mp 227 to 229 and 237 to
239 . Soluble in water. Colour Tests Amalic acid test—pink-orange/violet; Marquis test—orange. Thin-layer Chromatography System TA—Rf 0.55; system TB—Rf 0.03; system TC—Rf 0.45; system TE—Rf 0.54; system TL—Rf 0.14; system TAE—Rf 0.44 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—fenetylline RI 2830, fenetylline-AC RI 3110, M (N-desalkyl)-AC RI 2480, M (amfetamine) RI 1125, M (etofylline) RI 2125; system GB—fenetylline RI 2900. High Performance Liquid Chromatography System HC—k 0.27; system HX—RI 336; system HY—RI 277. Ultraviolet Spectrum Aqueous acid—275 nm (A11¼242a). No alkaline shift.
Infrared Spectrum Principal peaks at wavenumbers 1664, 1705, 1600, 1546, 746, 1219 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 250, 70, 207, 91, 251, 119, 148, 56.
1393
Proprietary Names Dima-Fen; Pesos; Ponderal; Ponderax; Pondimin. Chemical Properties A white crystalline powder. Mp 168 to 172 . Soluble 1 in
20 of water, 1 in 10 of ethanol and 1 in 10 of chloroform; practically insoluble in ether. Colour Test Liebermann’s reagent (100 )—yellow. Thin-layer Chromatography System TA—Rf 0.48; system TB—Rf 0.42; system TC—Rf 0.16; system TE—Rf 0.61; system TL—Rf 0.11; system TAE—Rf 0.20; system TAJ—Rf 0.07; system TAK—Rf 0.25; system TAL—Rf 0.68 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—fenfluramine RI 1230, norfenfluramine RI 1133, fenfluramine-TFA RI 1455, fenfluramine-PFP RI 1455, fenfluramineAC RI 1580, norfenfluramine-AC RI 1510; system GB—fenfluramine RI 1252, norfenfluramine RI 1157; system GC—fenfluramine RI 1621, norfenfluramine RI 1470. High Performance Liquid Chromatography System HA—fenfluramine k 1.3, norfenfluramine k 1.0; system HC—k 0.88; system HX—RI 371; system HY— RI 315; system HZ—retention time 3.9 min; system HAA—retention time 13.1 min. Ultraviolet Spectrum Aqueous acid—264 (A11¼22a), 271 nm.
Infrared Spectrum Principal peaks at wavenumbers 1165, 1116, 1070, 698, 793, 1202 cm1 (fenfluramine hydrochloride, KBr disk).
Quantification Hair MS–GC For method of quantification, see Kikura and Nakahara [1997]. Dose Fenetylline hydrochloride has been given in doses of 25 to 100 mg daily to treat narcolepsy. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Kikura R, Nakahara Y (1997). Hair analysis for drugs of abuse. XVI. Disposition of fenethylline and its metabolite into hair and discrimination between fenethylline use and amphetamine use by hair analysis. J Anal Toxicol 21: 291–296.
Fenfluramine Anorectic C12H16F3N = 231.3 CAS—458-24-2 IUPAC Name N-Ethyl-1-[3-(trifluoromethyl)phenyl]propan-2-amine Synonym N-Ethyl-a-methyl-3-(trifluoromethyl)benzeneethanamine
Chemical Properties Practically insoluble in water; soluble in chloroform. pKa 9.1 (25 ). Log P (octanol/water), 3.4. Fenfluramine Hydrochloride C12H16F3N,HCl = 267.7 CAS—404-82-0
Mass Spectrum Principal ions at m/z 72, 44, 159, 73, 58, 42, 109, 56; norfenfluramine 44, 42, 159, 43, 45, 184, 41, 109.
Quantification Blood GC–MS Fenfluramine, amfetamine and metamfetamine. Limit of detection, 5 mg/L for fenfluramine [Namera et al. 2000]. Plasma GC Nitrogen-specific detection. For method of quantification for D-fenfluramine and D-norfenfluramine, see Richards et al. [1989]. ECD. For method of quantification for enantiomers of fenfluramine and norfenfluramine see, Srinivas et al. [1988]. ECD. Limit of detection, 5 mg/L for fenfluramine, 100 ng/L for norfenfluramine [Midha et al. 1979].
F
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:47:11
1394
F
Fenimide
HPLC Fluorescence detection. For method of quantification for DL-fenfluramine, DL-norfenfluramine and phentermine, see Kaddoumi et al. [2001]. Urine GC See Plasma [Richards et al. 1989]. ECD. For method of quantification for fenfluramine and norfenfluramine, see Midha et al. [1979]. GC–MS For method of quantification for fenfluramine and amfetamine-related drugs, see Namera et al. [2002]. Disposition in the Body Readily absorbed after oral administration and accumulates in the tissues. The major metabolite in the blood is the N-desethyl derivative, norfenfluramine, which is active. It is also metabolised by oxidation to m-trifluoromethylbenzoic acid which is conjugated with glycine to form m-trifluoromethylhippuric acid. The rate of elimination is influenced by urinary pH and urinary flow. In acidic urine about 23% of a dose is excreted unchanged and about 17% as norfenfluramine in 48 h; the remainder consists of m-trifluoromethylhippuric acid; in alkaline urine about 2% is excreted as unchanged drug and norfenfluramine; when the urinary pH is not controlled, 3 to 10% may be excreted as unchanged drug and 3 to 14% as norfenfluramine. Up to 5% of a dose is eliminated in the faeces as fenfluramine and norfenfluramine. Therapeutic Concentration In plasma, usually in the range 0.05 to 0.15 mg/L. There is considerable inter-subject variation in plasma concentrations and it has been reported that the therapeutic effect (weight loss) is greater in those patients who can tolerate higher plasma concentrations (more than 0.2 mg/L) [Innes et al. 1977]. A single oral dose of 60 mg administered to 5 subjects, resulted in a mean plasma concentration of 0.06 mg/L of fenfluramine in 2 to 4 h and 0.016 mg/L of norfenfluramine in 4 to 6 h. Steady-state plasma concentrations of 0.04 to 0.12 mg/L of fenfluramine were attained in 3 to 4 days after daily administration of 60 mg, in divided doses, to 6 subjects; concentrations of norfenfluramine were similar [Campbell 1971]. Toxicity In adults the minimum lethal dose is probably in excess of 2 g but for young children as little as 200 mg may cause death. Toxic effects may be produced when the plasma concentration is greater than about 0.5 mg/L, and death has occurred at concentrations above 6 mg/L. Three children aged 6 years, 3 years, and 1 year 9 months ingested between them about 4 g of fenfluramine hydrochloride. The youngest and the oldest child died; postmortem blood concentrations were 6.5 and 16 mg/L respectively and liver concentrations were 48 and 136 mg/g; a urine concentration of 60 mg/L was found in the younger child [Gold et al. 1969]. A 13-year-old boy died after ingesting 2 g of fenfluramine hydrochloride; postmortem concentrations, mg/L or mg/g, were: [Fleisher, Campbell 1969].
Blood Bile Brain Kidney Liver Urine
Fenfluramine
Norfenfluramine
6.5 64.5 42 27.1 49 89
0.75 10.2 5.3 1.5 8.5 10
In a fatality involving the suicidal ingestion of fenfluramine, a blood concentration of 7.46 mg/L was reported. Chronic fenfluramine use was demonstrated by the presence of the drug in hair (14.1 mg/g) [Kintz, Mangin 1992]. Half-life Plasma half-life, 11 to 30 h (mean 20). Distribution in Blood Plasma : whole blood ratio, about 0.74. Protein Binding About 30%. Note For reviews of fenfluramine, see Pinder et al. [1975] and Vivero et al. [1998]. For a review of fenfluramine poisoning, see Von M€ uhlendahl and Krienke [1979]. Dose Initially 40 mg of fenfluramine hydrochloride daily, increasing to 60 to 120 mg daily. Campbell DB, Turner P (1971). Plasma concentrations of fenfluramine and its metabolite, norfenfluramine, after single and repeated oral administration. Br J Pharmacol 43(2): 465P–466P. Fleisher MR, Campbell DB (1969). Fenfluramine overdosage. Lancet 2: 1306–1307. Gold RG et al. (1969). Fenfluramine overdosage. Lancet 2: 1306. Innes JA et al. (1977). Plasma fenfluramine levels, weight loss, and side effects. Br Med J 2: 1322–1325. Kaddoumi A et al. (2001). Fluorometric determination of DL-fenfluramine, DL-norfenfluramine and phentermine in plasma by achiral and chiral high-performance liquid chromatography. J Chromatogr B Biomed Sci Appl 763: 79–90. Kintz P, Mangin P (1992). Toxicological findings after fatal fenfluramine self-poisoning. Hum Exp Toxicol 11(1): 51–52. Midha KK et al. (1979). Can J Pharm Sci 14: 18–21. Namera A et al. (2000). Simple and simultaneous analysis of fenfluramine, amphetamine and methamphetamine in whole blood by gas chromatography-mass spectrometry after headspace-solid phase microextraction and derivatization. Forensic Sci Int 109: 215–223. Namera A et al. (2002). Automated headspace solid-phase microextraction and in-matrix derivatization for the determination of amphetamine-related drugs in human urine by gas chromatography-mass spectrometry. J Chromatogr Sci 40: 19–25. Pinder RM et al. (1975). Fenfluramine: a review of its pharmacological properties and therapeutic efficacy in obesity. Drugs 10(4): 241–323.
Richards RP et al. (1989). The measurement of d-fenfluramine and its metabolite, d-norfenfluramine in plasma and urine with an application of the method to pharmacokinetic studies. Xenobiotica 19: 547–553. Srinivas NR et al. (1988). Enantioselective gas chromatographic assay with electron-capture detection for dl-fenfluramine and dl-norfenfluramine in plasma. J Chromatogr 433: 105–117. Vivero LE et al. (1998). A close look at fenfluramine and dexfenfluramine. J Emerg Med 16(2): 197–205. Von M€ uhlendahl KE, Krienke EG (1979). Fenfluramine poisoning. Clin Toxicol 14(1): 97–106.
Fenimide Succinimide, Antipsychotic C13H15NO2 = 217.3 CAS—60-45-7 IUPAC Name 4-Ethyl-3-methyl-3-phenylpyrrolidine-2,5-dione Synonyms CI-419; a-ethyl-a0 -methyl-a0 -phenylsuccinimide; fenimid.
fenetimide;
Chemical Properties White crystalline powder. Insoluble in water; soluble in ethanol and chloroform. Extracted by organic solvents from aqueous acid or alkaline solutions. Thin-layer Chromatography System T1—Rf 0.75 (location reagents: strongly acid iodoplatinate spray, positive reaction; iodine-carbon tetrachloride spray, positive reaction). Ultraviolet Spectrum Ethanol—253, 259, 248 and 265 nm.
Fenitrothion Organophosphate, Cholinesterase Inhibitor, Insecticide C9H12NO5PS = 277.2 CAS—122-14-5 IUPAC Name Dimethoxy-(3-methyl-4-nitrophenoxy)-sulfanylidene-l5phosphane Synonyms AC-47300; Bayer 41831; Bayer S 5660; ENT-25715; MEP; OMS-45; OMS-223; phosphorothioic acid O,O-dimethyl O-(3-methyl-4-nitrophenyl) ester; S-5660; S-1102-A; metathion. Proprietary Names Accothion; Agrothion; Cyfen; Cytel; Cyten; Dicofen; Etalene; Fenitox; Folithion; Micromite; Novathion; Nuvanol; Pestroy; Sumithion; Verthion.
Chemical Properties A yellow to brown oil. Mp 3.4 . Bp 118 at 0.05 mmHg. It is practically insoluble in water (14 mg/L at 30 ); has low solubility in aliphatic hydrocarbons; soluble in most organic solvents including alcohol esters, ketones, aromatic hydrocarbons and chlorinated hydrocarbons. It has a solubility in hexane of 42 g/kg at 20 to 25 ; dichloromethane, methanol and xylene >1000 g/kg; propan2-ol 193 g/kg. Log P (octanol/water), 3.43. Thin-layer Chromatography System TX—Rf 0.32; system TY—Rf 0.76; system TZ—Rf 0.82; system TAA—Rf 0.65; system TAB—Rf 0.50; system TAC—Rf 0.17. Gas Chromatography System GA—RI 1905; system GK—RRT 1.01 (relative to caffeine); system GKA—RI 1944; system GKB—RI 2112; system GKC—RI 2278. High Performance Liquid Chromatography System HAP—k 2.86; system HAO—k 0.22. Ultraviolet Spectrum Aqueous acid (acetonitrile)—268 nm.
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:47:12
Fenitrothion
Infrared Spectrum Principal peaks at wavenumber 1055, 830, 971, 1243, 1359, 1539 cm1.
Mass Spectrum Principal ions at m/z 125, 109, 79, 277, 47, 93, 63, 260.
Quantification Blood GC Column: Shimadzu HiCap-CBP 1 (10 m 0.53 mm i.d., 1.0 mm). Carrier gas: N2, 20 mL/min. Temperature: 160 . FID or FPD. Limit of detection not reported [Kojima et al. 1989]. Column: 3% OV-17 on Chromosorb G 60/80 mesh (1 m 2.6 cm i.d.). Carrier gas: N2, 40 mL/min. Temperature: 240 . FPD. Limit of detection, 0.1 ng [Yashiki et al. 1986]. GC-MS Column: Shimadzu HiCap-CBP 1 (12 m 0.53 mm i.d.). Carrier gas: He, 20 mL/min. Temperature programme: 100 to 280 at 8 /min. EI ionisation at 70 eV or CI at 150 eV. Limit of detection not reported [Kojima et al. 1989]. HPLC Column: Novapak C18 (150 3.9 mm i.d., 4 mm). Mobile phase: acetonitirile : water (4 : 6), flow rate 1.0 mL/min. UV detection (l ¼ 210 nm). Limit of detection, 0.1 mg/L [Ageda et al. 2006]. Serum Note For a HPTLC method for the detection of fenirothion, see Futagami et al. [1997]. GC-MS Column: DB-5MS 5% phenyl methyl siloxane (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1.0 mL/min. Temperature programme: 90 for 1 min to 180 at 30 /min to 300 at 4 /min for 2 min. EI ionisation at 70 eV. Retention time:
1395
10.6 min. Limit of quantification, 3.3 mg/L, limit of detection, 1.1 mg/L [Pitarch et al. 2003]. HPLC Column: Nucleosil 5C18 (15 cm 4 mm i.d.). Mobile phase: acetonitrile : water (50 : 50), flow rate 1.0 mL/min. DAD (l ¼ 230 nm). Retention time: 9.54 min. Limit of quantification, 0.15 mg/L, limit of detection, 0.15 mg/L [Cho et al. 1997]. LC-MS Column: XTerra MS C18 (20 2.1 mm i.d., 3.5 mm). Mobile phase: 10 mmol/L ammonium formate : methanol (100 : 0 to 0 : 100 in 3 min for 6.5 min to 100 : 0 at 10 min), flow rate 0.3 mL/min. APCI, positive or negative ion mode. Retention time: 5.61 min. Limit of quantification, 0.25 mg/L, limit of detection, 0.125 mg/L [Inoue et al. 2007]. Urine GC See Blood [Kojima et al. 1989]. See Blood [Yashiki et al. 1986]. HPLC See Serum [Cho et al. 1997]. Column: Chemcosorb ODS (15 cm 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : water (44 : 56) containing 0.1% acetic acid, flow rate 0.5 mL/min. UV detection (l ¼ 315 nm). Limit of detection, 3.4 g/L. [Chang, Lin 1995]. LC-MS Column: Discovery C18 (50 2.1 mm i.d., 5 mm) followed by Supelco ABZþ (100 2.1 mm i.d., 5 mm). Mobile phase: acetonitrile : 0.01% formic acid in water followed by acetonitrile: 0.0025% formic acid, flow rate 300 mL/min. ESI, MRM acquisition mode, negative ion mode. Retention time: 2.2 min for 3-methyl4-nitrophenol. Limit of detection, 66 h for patches releasing 25 mg/h fentanyl. Peak plasma concentrations of 0.91 mg/L were obtained with 25 mg//h patches and 39.3 mg//L with 200 mg/h patches [Collins et al. 1999]. Administration of fentanyl (10 to 15 mg/kg) via an oral transmucosal delivery system to 17 children resulted in peak plasma concentrations of 1.03 0.31 mg/L. The children were thought to have swallowed a large fraction of the dose, leading to a relatively late and variable peak concentration time of 53 40 min [Wheeler et al. 2002]. Toxicity The estimated minimum lethal dose is 2 mg. The following postmortem tissue concentrations were reported in a fatality resulting from the self-administration of fentanyl (in mg/L or mg/g): serum 17.7, blood 27.5, urine 92.7, bile 58.2, liver 0.0775, kidney 0.0415, brain 0.0302, lung 0.0834, stomach 31.6. A partly filled syringe containing 2800 mg/L fentanyl was found near the scene of death [Chaturvedi et al. 1990]. In a review of 25 fatalities involving fentanyl transdermal patches, the following post-mortem tissue concentrations were reported (in mg/L or mg/ g): heart blood 1.89–139 (23 cases), femoral blood 3.1–43 (13 cases), vitreous humour 2–20 (4 cases), liver 0.0058–0.613 (22 cases), bile 3.5–262 (15 cases), urine 2.9–895 (19 cases), gastric 0–122 mg total (17 cases), spleen 0.0078–0.079 (3 cases), kidney 0.011 (1 case), lung 0.031 (1 case) [Anderson, Muto 2000]. An 83-year-old female who was found dead with three 100 mg/h fentanyl patches on her chest had the following tissue concentrations at postmortem: blood 25 mg/L, brain 0.054 mg/g, heart 0.094 mg/g, kidney 0.069 mg/g, liver 0.104 mg/g [Edinboro et al. 1997]. A postmortem serum fentanyl concentration of 2 mg/L was reported in a 35year-old woman who had intravenously injected the contents of a trandermal fentanyl patch (5 mg, shared with another person who survived); 0.16 g/L ethanol was also found in the serum [Reeves, Ginifer 2002]. A 63-year-old man who was found dead with 20 fentanyl patches of different strengths with a total dose of 1350 mg/h had the following tissue concentrations at postmortem: femoral blood 0.0949 mg/g, heart blood (left) 0.0459 mg/g, heart blood (right) 0.0748 mg/g, urine 101 mg/L, bile 468 mg/L, gastric contents 745 mg/L, CSF 78.4 mg/L, vitreous humour 133 mg/L [Wiesbrock et al. 2008]. Half-life Plasma half-life, ~3.7 h (dose dependent: also increased in the elderly and premature infants and during cardiopulmonary by-pass surgery). Volume of Distribution Approximately 4 L/kg. Clearance Plasma clearance, ~13 mL/min/kg. Protein Binding ~80%. Dose For analgesia in surgery, initially, the equivalent of 50 to 200 mg fentanyl. IV; supplementary doses of 50 mg. With assisted ventilation, an initial dose of 300 to 3500 mg may be given. Anderson DT, Muto JJ (2000). Duragesic transdermal patch: postmortem tissue distribution of fentanyl in 25 cases. J Anal Toxicol 24: 627–634.
F
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:48:11
1402
F
Fenthion
Bagheri H et al. (2007). Determination of fentanyl in human plasma by head-space solid-phase microextraction and gas chromatography–mass spectrometry. J Pharm Biomed Anal 43: 1763–1768. Bovill JG, Sebel PS (1980). Pharmacokinetics of high-dose fentanyl: a study in patients undergoing cardiac surgery. Br J Anaesth 52: 795–801. Chaturvedi AK et al. (1990). A death due to self-administered fentanyl. J Anal Toxicol 14: 385–387. Collins JJ et al. (1999). Transdermal fentanyl in children with cancer pain: feasibility, tolerability, and pharmacokinetic correlates. J Pediatr 134: 319–323. Coopman V et al. (2007). LC-MS/MS analysis of fentanyl and norfentanyl in a fatality due to application of multiple Durogesic transdermal therapeutic systems. Forensic Sci Int 169: 223–227. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid–liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Ebrahimzadeh H et al. (2008). Determination of fentanyl in biological and water samples using single-drop liquid–liquid–liquid microextraction coupled with high-performance liquid chromatography. Anal Chim Acta 626: 193–199. Edinboro LE et al. (1997). Fatal fentanyl intoxication following excessive transdermal application. J Forensic Sci 42: 741–743. Fryirs B et al. (1997). Determination of subnanogram concentrations of fentanyl in plasma by gas chromatography–mass spectrometry: comparison with standard radioimmunoassay. J Chromatogr B Biomed Sci Appl 688: 79–85. Gergov M et al. (2009). Simultaneous screening and quantification of 25 opioid drugs in postmortem blood and urine by liquid chromatography–tandem mass spectrometry. Forensic Sci Int 186: 36–43. Gunnar T et al. (2005). Validated toxicological determination of 30 drugs of abuse as optimized derivatives in oral fluid by long column fast gas chromatography/electron impact mass spectrometry. J Mass Spectrom 40: 739–753. Hung OR et al. (1995). Pharmacokinetics of inhaled liposome-encapsulated fentanyl. Anesthesiology 83: 277–284. Huynh NH et al. (2005). Determination of fentanyl in human plasma and fentanyl and norfentanyl in human urine using LC-MS/MS. J Pharm Biomed Anal 37: 1095–1100. Kintz P et al. (2005). Evidence of addiction by anesthesiologists as documented by hair analysis. Forensic Sci Int 153: 81–84. Klinke HB, Linnet K (2007). Performance of four mixed-mode solid-phase extraction columns applied to basic drugs in urine. Scand J Clin Lab Invest 67: 778–782. Kowalski SR et al. (1987). Sensitive gas liquid chromatography method for the determination of fentanyl concentrations in blood. J Pharmacol Methods 18: 347–355. Kumar K et al. (1996). A sensitive assay for the simultaneous measurement of alfentanil and fentanyl in plasma. J Pharm Biomed Anal 14: 667–673. Martin TL et al. (2006). Fentanyl-related deaths in Ontario, Canada: toxicological findings and circumstances of death in 112 cases (2002-2004). J Anal Toxicol 30: 603–610. Michiels M et al. (1977). A sensitive radioimmunoassay for fentanyl: plasma level in dogs and man. Eur J Clin Pharmacol 12: 153–158. Moises EC et al. (2005). Pharmacokinetics and transplacental distribution of fentanyl in epidural anesthesia for normal pregnant women. Eur J Clin Pharmacol 61: 517–522. Moore C et al. (2008). Analysis of pain management drugs, specifically fentanyl, in hair: application to forensic specimens. Forensic Sci Int 176: 47–50. Musshoff F et al. (2007). Determination of opioid analgesics in hair samples using liquid chromatography/tandem mass spectrometry and application to patients under palliative care. Ther Drug Monit 29: 655–661. Naidong W et al. (2002). Simultaneous development of six LC-MS-MS methods for the determination of multiple analytes in human plasma. J Pharm Biomed Anal 28: 1115–1126. Nitsun M et al. (2006). Pharmacokinetics of midazolam, propofol, and fentanyl transfer to human breast milk. Clin Pharmacol Ther 79: 549–557. Poklis A, Backer R (2004). Urine concentrations of fentanyl and norfentanyl during application of Duragesic transdermal patches. J Anal Toxicol 28: 422–425. Portier EJ et al. (1999). Simultaneous determination of fentanyl and midazolam using high-performance liquid chromatography with ultraviolet detection. J Chromatogr B Biomed Sci Appl 723: 313–318. Reeves MD, Ginifer CJ (2002). Fatal intravenous misuse of transdermal fentanyl. Med J Aust 177: 552–553. Schneider S et al. (2008). Determination of fentanyl in sweat and hair of a patient using transdermal patches. J Anal Toxicol 32: 260–264. Skulska A et al. (2007). [Determination of fentanyl, atropine and scopolamine in biological material using LC-MS/APCI methods]. Przegl Lek 64: 263–267. Valaer AK et al. (1997). Development of a gas chromatographic-mass spectrometric drug screening method for the N-dealkylated metabolites of fentanyl, sufentanil, and alfentanil. J Chromatogr Sci 35: 461–466. Van Nimmen NF et al. (2004). Highly sensitive gas chromatographic-mass spectrometric screening method for the determination of picogram levels of fentanyl, sufentanil and alfentanil and their major metabolites in urine of opioid exposed workers. J Chromatogr B Analyt Technol Biomed Life Sci 804: 375–387. Van Rooy HH et al. (1981). The assay of fentanyl and its metabolites in plasma of patients using gas chromatography with alkali flame ionisation detection and gas chromatography–mass spectrometry. J Chromatogr 223: 85–93. Wheeler M et al. (2002). Uptake pharmacokinetics of the Fentanyl Oralet in children scheduled for central venous access removal: implications for the timing of initiating painful procedures. Paediatr Anaesth 12: 594–599. Wiesbrock UO et al. (2008). [Excessive use of fentanyl patches as the only means of suicide]. Arch Kriminol 222: 23–30. Woestenborghs RJ et al. (1987). Assay methods for fentanyl in serum: gas–liquid chromatography versus radioimmunoassay. Anesthesiology 67: 85–90. Wong SC et al. (2008). Concurrent detection of heroin, fentanyl, and xylazine in seven drug-related deaths reported from the Philadelphia Medical Examiner’s Office. J Forensic Sci 53: 495–498.
Fenthion Cholinesterase Inhibitor, Acaricide, Insecticide C10H15O3PS2 = 278.3 CAS—55-38-9 IUPAC Name Dimethoxy-(3-methyl-4-methylsulfanylphenoxy)-sulfanylidenel5-phosphane Synonyms Bay 29493; bayer 29493; O,O-dimethyl-o-(4-methylthio)-m-tolyl] phosphorothioate; DMTP; ENT25540; MPP; mercaptophos; OMS2; phosphorothioic acid O,O-dimethyl O-[3-methyl-4-(methylthio)phenyl] ester; S-1752.
Proprietary Names Baycid; Baytex; Entex; Lebaycid; Lysoff; Queletox; Spotten; Talodex; Tiguvon.
Chemical Properties Pure fenthion is a colourless liquid. The technical form is an oily yellowish-brown liquid. Mp 7.5 . Bp 87 at 0.01 mmHg. It is practically insoluble in water; readily soluble in methanol, ethanol, acetone, 2-propanol, dichloromethane, toluene and most organic solvents, including alcohol, ethers, esters and halogenated aromatics; slightly soluble in hexane. Log P (octanol/water), 4.09 [Hansch et al. 1995]. Extraction yield (chlorobutane), 1 [Demme et al. 2005]. Stable through 3 freeze-thaw cycles and after 3 weeks at –30 [Inoue et al. 2007]. Thin-layer Chromatography System TX—Rf 0.41; system TY—Rf 0.81; system TZ—Rf 0.90; system TAA—Rf 0.68. Plates: silica gel. Solvent system: benzene : methanol (9 : 1). Developed by spraying with 0.2% palladium chloride solution. Rf 0.9 [Meyer et al. 1998]. Gas Chromatography System GA—RI 1938. Ultraviolet Spectrum Methanol—229 nm; aqueous acid—252 nm.
Mass Spectrum Principal ions at m/z 278, 125, 109, 169, 93.
Quantification Blood GC Column: DB-5 (30 m 0.32 mm i.d., 0.25 mm). Carrier gas: He, 2.4 mL/ min. Temperature programme: 180 for 3 min to 230 at 10 /min for 35 min to 270 at 5 /min for 5 min. Retention time: 33.6 min. Limit of detection, 1 ppb [Tsatsakis et al. 2002]. Column: HP-1 fused silica (10 m 0.53 mm i.d., 2.65 mm). Carrier gas: He, 15 mL/min. Temperature programme: 60 for 2 min to 250 at 10 /min for 8 min. NPD. Retention time: 14.16 min. Limit of quantification, 0.5 mg/L, limit of detection, 0.15 mg/L [Garcia-Repetto et al. 2001]. Column: DB-5 (15 m 0.32 mm i.d., 1.5 mm). Carrier gas: He, 6.1 mL/min. Temperature programme: 120 to 240 at 10 /min. FID. Limit of detection not reported [Tsatsakis et al. 1996]. Column: Chromosorb W/HP 8/100 mesh 3% OV-17 (1/2" o.d. 2 mm i.d.) Carrier gas: N2, 2.5 mL/min. Temperature: 200 . FPD. Limit of detection, 0.1 mg/L [Brunetto et al. 1992]. GC-MS Column: HP-5MS fused silica capillary (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1.0 mL/min. Temperature programme: 120 for 1 min to 290 at 10 /min for 1 min. EI ionisation, SIM acquisition mode. Limit of detection, 0.01 mg/ g [Musshoff et al. 2002]. Column: SGE BPX5 (25 m 0.22 mm i.d., 0.25 mm). Carrier gas: He, 0.8 mL/min. Column temperature: 60 to 120 at 30 /min to 220 at 10 /min to 290 at 5 /min to 300 for 3 min. EI ionisation, SIM acquisition mode. Retention time: 14.3 min. Limit of quantification, 0.1 mg/L [Meyer et al. 1998]. HPLC Column: Aluspher RP-select B (125 4.0 mm i.d., 5 mm). Mobile phase: 0.0125 mol/L sodium hydroxide in methanol : 0.0125 mol/L sodium hydroxide in water (10 : 90 for 5 min to 90 : 10 over 15 min for 5 min), flow rate 1.0 mL/min. UV detection (l ¼ 250 nm). Retention time: 18.3 min. Limit of quantification, 0.25 mg/ L [Meyer et al. 1998]. LC-MS Column: Luna C18 reversed phase (30 2.0 mm i.d., 3 mm). Mobile phase: 2 mmol/L ammonium acetate buffer containing 1 mL/L formic acid in water (pH 2.8) : 2 mmol/L ammonium acetate buffer containing 1 mL/L formic acid in methanol (60 : 40 to 20 : 80 at 1.5 min to 60 : 40 at 6 min), flow rate 0.4 mL/min. ESI,
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:48:15
Fenyramidol positive ion mode, SRM acquisition mode. Limit of quantification, 0.5 mg/L [Salm et al. 2009]. Plasma GC Column: 3% OV-17 on Chromosorb WAWDMCS 100/120 mesh (2 m 1/8" i.d.). Carrier gas: N2, 50 mL/min. Temperature: 230 . Limit of detection, 3 mg/L [Mahieu et al. 1982]. Serum Note For a HPTLC method for the detection of fenthion, see Futagami et al. [1997]. HPLC Column: Nucleosil 5C18 (15 cm 4 mm i.d.). Mobile phase: acetonitrile : water (50 : 50), flow rate 1.0 mL/min. DAD (l ¼ 230 nm). Retention time: 14.1 min. Limit of quantification, 0.25 mg/L, limit of detection, 0.13 mg/L [Cho et al. 1997]. LC-MS Column: XTerra MS C18 (20 2.1 mm i.d., 3.5 mm). Mobile phase: 10 mmol/L ammonium formate : methanol (100 : 0 to 0 : 100 in 3 min for 6.5 min to 100 : 0 at 10 min), flow rate 0.3 mL/min. APCI, positive or negative ion mode. Retention time: 5.61 min. Limit of quantification, 1.25 mg/L, limit of detection, 1 mg/L [Inoue et al. 2007]. Urine GC See Blood [Tsatsakis et al. 2002]. HPLC See Serum [Cho et al. 1997]. Vitreous Humour GC See Blood [Tsatsakis et al. 1996]. Adipose Tissue GC See Blood [Tsatsakis et al. 1996]. Brain GC See Blood [Tsatsakis et al. 1996]. Kidney GC See Blood [Tsatsakis et al. 1996]. Liver GC See Blood [Tsatsakis et al. 1996]. Orchis GC See Blood [Tsatsakis et al. 1996]. Thyroid GC See Blood [Tsatsakis et al. 1996]. Disposition in the Body Fenthion is readily absorbed into the bloodstream via the digestive tract, lungs and skin, and is systematically distributed. It is rapidly metabolised to weak active products by several pathways, the major pathway being by hepatic hydrolysis. It is eliminated through urine and faeces as the hydrolysis products. Fenthion is stored in fat and its release is delayed for metabolism. Toxicity Fenthion is moderately toxic after ingestion and dermal exposure, and is slightly toxic if inhaled. The allowed daily intake is 1 mg/kg. A lethal dose of 50 mg/ kg has been established, but the route of administration is not known. Acute respiratory failure defined as intermediate syndrome begins 24–96 h after the cholinergic crisis [Karademir et al. 1990; Sedgwick, Senanayake 1997; van den Neucker et al. 1991]. Delayed neuropathy can follow 2 to 3 weeks later with the inhibition of the neuropathy target esterase [Serrano, Fedriani 1997]. A 67-year-old male was admitted to hospital. His blood and urine fenthion concentrations were 2.7 and 0.5 mg/L, respectively, at the time of hospital admission. He was discharged 43 days later. A 13-year-old male was spraying crops with fenthion. On admission to hospital his blood concentration was 0.95 mg/L. After 5 days he was breathing by himself and he was discharged from hospital 4 days later [Tsatsakis et al. 2002]. A 66-year-old female was found dead in her bathroom, a suicide case that was not suspected to be fenthion poisoning. After toxicological analysis, a fenthion blood concentration of 3.8 mg/L was detected, and it was determined that fenthion ingestion was the cause of death. A total amount of 17 g of fenthion was found in the stomach and 203 g/g in the liver [Meyer et al. 1998]. A 69-year-old farmer ingested 200 mL Lebaycid. Postmortem concentrations of fenthion were 1.7, 4.8, 23.1, 16.8, 135.2 5.8, 7.1, or 13.8 mg/L or mg/g in the vitreous humour, blood, kidney, liver, fat, orchis, thyroid and brain, respectively [Tsatsakis et al. 1996]. A 41-year-old male was admitted to hospital having ingested an unknown quantity of fenthion. On admission, 20 h after the ingestion, he had a blood fenthion concentration of 0.27 mg/L. This increased on the first day to 0.78 mg/L. He died on day 7 from a refractory cardiovascular collapse [Brunetto et al. 1992]. A 43-year-old male ingested 30 mL of Lebaycid (–18 g fenthion). On admission to hospital his blood fenthion concentration was 71 mg/L, and 3 h later it was 102 mg/L [Mahieu et al. 1982]. Note Several cases in which extrapyramidal manifestations complicated the organophosphorus intoxication are reported in Senanayake and Sanmuganathan [1995]. For a case of prolonged toxicity after fenthion ingestion, see Merrill and Mihm [1982] or Borowitz [1988]. For a case of fenthion taken during pregnancy, see Karalliedde et al. [1988]. Cases of SC injection of fenthion are reported in Bala et al. [2008], Hadimioglu et al. [2002], Premaratna et al. [2001], Serrano and Fedriani [1997]. Bala I et al. (2008). Prolonged cholinergic crisis and compartment syndrome following subcutaneous injection of an organophosphate compound for suicide attempt. J Forensic Leg Med 15: 256–258. Borowitz SM (1988). Prolonged organophosphate toxicity in a twenty-six-month-old child. J Pediatr 112: 302–304. Brunetto MR et al. (1992). Observation on a human intentional poisoning case by the organophosphorus insecticide fenthion. Invest Clin 33: 89–94. Cho Y et al. (1997). Determination of organophosphorous pesticides in biological samples of acute poisoning by HPLC with diode-array detector. Chem Pharm Bull (Tokyo) 45: 737–740. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Futagami K et al. (1997). Application of high-performance thin-layer chromatography for the detection of organophosphorus insecticides in human serum after acute poisoning. J Chromatogr B Biomed Sci Appl 704: 369–373. Garcia-Repetto R et al. (2001). New method for determination of ten pesticides in human blood. J AOAC Int 84: 342–349. Hadimioglu N et al. (2002). Systemic organophosphate poisoning following the percutaneous injection of insecticide. Case report. Skin Pharmacol Appl Skin Physiol 15: 195–199.
1403
Hansch C et al. (1995). The expanding role of quantitative structure-activity relationships (QSAR) in toxicology. Toxicol Lett 79: 45–53. Inoue S et al. (2007). Rapid simultaneous determination for organophosphorus pesticides in human serum by LC-MS. J Pharm Biomed Anal 44: 258–264. Karademir M et al. (1990). Two cases of organophosphate poisoning with development of intermediate syndrome. Hum Exp Toxicol 9: 187–189. Karalliedde L et al. (1988). Acute organophosphorus insecticide poisoning during pregnancy. Hum Toxicol 7: 363–364. Mahieu P et al. (1982). Severe and prolonged poisoning by fenthion. Significance of the determination of the anticholinesterase capacity of plasma. J Toxicol Clin Toxicol 19: 425–432. Merrill DG, Mihm FG (1982). Prolonged toxicity of organophosphate poisoning. Crit Care Med 10: 550–551. Meyer E et al. (1998). Analysis of fenthion in postmortem samples by HPLC with diode-array detection and GC-MS using solid-phase extraction. J Anal Toxicol 22: 248–252. Musshoff F et al. (2002). Simple determination of 22 organophosphorous pesticides in human blood using headspace solid-phase microextraction and gas chromatography with mass spectrometric detection. J Chromatogr Sci 40: 29–34. Premaratna R et al. (2001). Parasuicide by self-injection of an organophosphate insecticide. Hum Exp Toxicol 20: 377–378. Salm P et al. (2009). Liquid chromatography-tandem mass spectrometry method for the simultaneous quantitative determination of the organophosphorus pesticides dimethoate, fenthion, diazinon and chlorpyrifos in human blood. J Chromatogr B Analyt Technol Biomed Life Sci 877: 568–574. Sedgwick EM, Senanayake N (1997). Pathophysiology of the intermediate syndrome of organophosphorus poisoning. J Neurol Neurosurg Psychiatry 62: 201–202. Senanayake N, Sanmuganathan PS (1995). Extrapyramidal manifestations complicating organophosphorus insecticide poisoning. Hum Exp Toxicol 14: 600–604. Serrano N, Fedriani J (1997). Fenthion suicide poisoning by subcutaneous injection. Intensive Care Med 23: 129. Tsatsakis AM et al. (1996). Experiences with acute organophosphate poisonings in Crete. Vet Hum Toxicol 38: 101–107. Tsatsakis AM et al. (2002). Severe fenthion intoxications due to ingestion and inhalation with survival outcome. Hum Exp Toxicol 21: 49–54. Van denNeucker K et al. (1991). The neurophysiologic examination in organophosphate ester poisoning. Case report and review of the literature. Electromyogr Clin Neurophysiol 31: 507–511.
Fenticlor Antifungal C12H8Cl2O2S = 287.2 CAS—97-24-5 IUPAC Name 4-Chloro-2-(5-chloro-2-hydroxyphenyl)sulfanylphenol Synonym 2,20 -Thiobis[4-chlorophenol] Proprietary Names Novex. It is an ingredient of Dermisdin.
Chemical Properties A white crystalline powder. Mp 176 . Practically insoluble in water; freely soluble in ethanol; soluble in aqueous solutions of sodium hydroxide. Log P (octanol/water), 4.6. Thin-layer Chromatography System TAE—Rf 0.91. Ultraviolet Spectrum Principal peak at 300 nm.
Infrared Spectrum Principal peaks at wavenumbers 1269, 825, 1209, 818, 889, 1104 cm1. Use Fenticlor has been applied topically in concentrations of up to 2%.
Fenyramidol Analgesic, Muscle Relaxant C13H14N2O = 214.3 CAS—553-69-5 IUPAC Name 1-Phenyl-2-(pyridin-2-ylamino)ethanol
F
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:48:20
1404
Feprazone
Synonyms Phenyramidol; a-[(2-pyridinylamino)methyl]benzenemethanol.
Chemical Properties Crystals. Mp 82 to 85 . pKa 5.9. Log P (octanol/water), 1.7. Fenyramidol Hydrochloride C13H14N2O,HCl = 250.7 CAS—326-43-2 Synonyms Phenyramidol hydrochloride; IN-511; MJ-505; NSC-17777. Proprietary Name Cabral Chemical Properties A white crystalline powder. Mp 140 to 142 . Freely soluble
Colour Tests p-Dimethylaminobenzaldehyde—red; Liebermann’s reagent— brown-orange (!brown at 100 ). Thin-layer Chromatography System TE—Rf 0.19; system TG—Rf 0.45; system TAE—Rf 0.92 (chromic acid solution, yellow; Ludy Tenger reagent, orange; mercurous nitrate spray, positive). Gas Chromatography System GA—RI 2380; system GD—methyl derivative RRT 1.81 (relative to n-C16H34); system GF—RI 2800. High Performance Liquid Chromatography System HV—RRT 0.92 (relative to meclofenamic acid). Ultraviolet Spectrum Methanolic alkali—266 nm (A11¼700b). Infrared Spectrum Principal peaks at wavenumbers 1715, 1300, 767, 1745, 715, 703 cm1 (KBr disk).
in water; soluble in ethanol.
F
Colour Tests Cyanogen bromide—orange-pink; Mandelin’s test—blue; Marquis test—yellow. Thin-layer Chromatography System TA—Rf 0.69; system TB—Rf 0.08; system TC—Rf 0.52; system TE—Rf 0.76; system TL—Rf 0.59; system TAE—Rf 0.80; system TAF—Rf 0.86 (Dragendorff spray, positive; acidified iodoplatinate solution, positive; acidified potassium permanganate solution, positive). Gas Chromatography System GA—RI 1960. High Performance Liquid Chromatography System HX—RI 282. Ultraviolet Spectrum Aqueous acid—237 (A11¼666a), 309 nm.
Mass Spectrum Principal ions at m/z 183, 77, 252, 320, 184, 41, 69, 51.
Infrared Spectrum Principal peaks at wavenumbers 1613, 1524, 700, 768, 1058, 752 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 107, 108, 78, 79, 80, 77, 51, 52.
Dose Up to 3.2 g of fenyramidol hydrochloride daily.
Feprazone Analgesic C20H20N2O2 = 320.4 CAS—30748-29-9 IUPAC Name 4-(3-Methyl-2-butenyl)-1,2-diphenyl-3,5-pyrazolidinedione Synonyms Phenylprenazone; prenazone. Proprietary Names Brotazona; Methrazone; Reuflodol; Zepelan; Zepelin.
Quantification Plasma HPLC UV detection. Limit of detection, 100 mg/L feprazone, 200 mg/L 4(3-hydroxymethyl)feprazone [Spahn, Mutschler 1982]. Disposition in the Body Absorbed after oral administration. Metabolised by hydroxylation to 4-(3-hydroxymethyl)feprazone. 0.7 mg/L for efficacy reported no adverse effects [Duff et al. 1981]. A young woman attempted to commit suicide and was found with no effective blood pressure and in extreme cardiac suffering. A flecainide concentration of 5.4 mg/L was observed (therapeutic range 0.2 to 1.0 mg/L) and during 10 h of cardiopulmonary bypass support, this reduced to 1.4 mg/L. Blood pressure and effective cardiac rhythm eventually returned and treatment was continued until drug levels decreased further. Unfortunately, the woman died, however, owing to severe neurological damage which occurred at the time of overdose [Yasui et al. 1997]. Bioavailability 90 to 95%. Half-life Plasma half-life, 7 to 23 h; 19 h (patients with arrhythmia); 26 to 49 h (patients with renal/hepatic dysfunction); 50 h (patients with congestive heart failure). Volume of Distribution Steady state, 5 to 13.4 L/kg; average 5.5 to 8.7 L/kg for IV administration and 10 L/kg for oral. Clearance Plasma, 4.6 to 12.1 mL/min/kg after an IV dose of 0.6 to 1.7 mg/kg; 4.1 to 17 mL/min/kg after an oral dose of 60 to 240 mg. Protein Binding 32 to 58%. Note For reviews of flecainide, see Holmes and Heel [1985] and Conard and Ober [1984].
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:49:4
Flosequinan
1409
Dose IV administration: initially 2 mg/kg body weight is administered for the first 30 min, followed by 1.5 mg/kg/h for the first hour and maintained at 0.1 to 0.25 mg/ kg/h for subsequent hours. The infusion period should not exceed 24 h and the maximum dose is 600 mg. For patients with severe renal impairment (creatinine clearance 0.2 mg/L flurazepam or 0.5 mg/L N1-desalkylflurazepam may be toxic, and blood concentrations >0.5 mg/L flurazepam may be fatal. In a fatality attributed to flurazepam overdose, the following postmortem concentrations were reported for flurazepam, N1-desalkylflurazepam, and N1-(2-hydroxyethyl)flurazepam, respectively: blood 0.51, 0.14, 9 mg/L; urine 7, 3.9, 98 mg/L. The amount ingested was estimated to be more than 2.4 g [Aderjan, Mattern 1979]. In a 5-year-old child who died following the ingestion of flurazepam and phenobarbital, the following postmortem tissue concentrations were reported for flurazepam, N1-desalkylflurazepam and N1-(2-hydroxyethyl)flurazepam, respectively: blood 3.2, 1.8 and 2.5 mg/L; brain 0.8, 0.7 and 0.7 mg/g; kidney 0.9, 0.6 and 1.1 mg/g; and liver 2.7, 3.1 and 3.5 mg/g. Phenobarbital concentrations were consistent with a therapeutic dose and low concentrations of phenytoin were also detected [Ferrara et al. 1979]. In a 52-year-old female whose death was attributed to suicidal ingestion of up to 2.2 g flurazepam, the following postmortem tissue concentrations were reported: femoral blood 5.5 mg/L, liver 130 mg/g, bile 33 mg/L, vitreous humour 1.3 mg/L, urine 3.3 mg/L, gastric contents 600 mg (total); desalkylfurazepam was also detected in blood, liver, bile and vitreous humour, but at concentrations much lower than the parent compound [McIntyre et al. 1994]. A 68-year-old woman was found dead at home with the following flurazepam concentrations: heart blood 2.8 mg/L, bile 323 mg/L and urine 172 mg/L [Martello et al. 2006]. Half-life Plasma : flurazepam 2–3 h, N1-desalkylflurazepam 2–5 days, N1-(2hydroxyethyl)flurazepam 10–20 h. Protein Binding Flurazepam ~97%, N1-desalkylflurazepam ~98% and N1-(2hydroxyethyl)flurazepam ~90%. Dose The equivalent of 15 to 30 mg of flurazepam, as a hypnotic. Aderjan R, Mattern R (1979). [A fatal monointoxication by flurazepam (Dalmadorm). Problems of the toxicological interpretation (author’s trans.)]. Arch Toxicol 43: 69–75. Borrey D et al. (2001). Simultaneous determination of fifteen low-dosed benzodiazepines in human urine by solid-phase extraction and gas chromatography–mass spectrometry. J Chromatogr B Biomed Sci Appl 765: 187–197. Burstein ES et al. (1988). Quantitation of flurazepam and three metabolites by electron capture gas liquid chromatography. J Anal Toxicol 12: 122–125. Cooper SF, Drolet D (1982). Gas–liquid chromatographic determination of flurazepam and its major metabolites in plasma with electron-capture detection. J Chromatogr 231: 321–331. De Silva JA et al. (1974). Spectrofluorodensitometric determination of flurazepam and its major metabolites in blood. J Pharm Sci 63: 1837–1841. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid–liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Dussy FE et al. (2006). Quantification of benzodiazepines in whole blood and serum. Int J Legal Med 120: 323–330. Ferrara SD et al. (1979). Concentrations of phenobarbital, flurazepam, and flurazepam metabolites in autopsy cases. J Forensic Sci 24: 61–69.
F
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:50:56
1430
F
Flurbiprofen
Gjerde H et al. (1992). Simultaneous determination of common benzodiazepines in blood using capillary gas chromatography. J Pharm Biomed Anal 10: 317–322. Greenblatt DJ et al. (1981). Kinetics and clinical effects of flurazepam in young and elderly noninsomniacs. Clin Pharmacol Ther 30: 475–486. Gunnar Tet al. (2004). Validated semiquantitative/quantitative screening of 51 drugs in whole blood as silylated derivatives by gas chromatography–selected ion monitoring mass spectrometry and gas chromatography electron capture detection. J Chromatogr B Analyt Technol Biomed Life Sci 806: 205–219. Hasegawa M, Matsubara I (1975). Metabolic fates of flurazepam. I. Gas chromatographic determination of flurazepam and its metabolites in human urine and blood using electron capture detector. Chem Pharm Bull (Tokyo) 23: 1826–1833. Kratzsch C et al. (2004). Screening, library-assisted identification and validated quantification of 23 benzodiazepines, flumazenil, zaleplone, zolpidem and zopiclone in plasma by liquid chromatography/mass spectrometry with atmospheric pressure chemical ionization. J Mass Spectrom 39: 856–872. Lillsunde P, Sepp€al€a T (1990). Simultaneous screening and quantitative analysis of benzodiazepines by dual-channel gas chromatography using electron-capture and nitrogen–phosphorus detection. J Chromatogr 533: 97–110. Martello S et al. (2006). Acute flurazepam intoxication: a case report. Am J Forensic Med Pathol 27: 55–57. McIntyre IM et al. (1994). A fatality due to flurazepam. J Forensic Sci 39: 1571–1574. Moore C et al. (2007). Determination of benzodiazepines in oral fluid using LC-MS-MS. J Anal Toxicol 31: 596–600. Nakamura M et al. (2009). Simultaneous determination of benzodiazepines and their metabolites in human serum by liquid chromatography–tandem mass spectrometry using a high-resolution octadecyl silica column compatible with aqueous compounds. Biomed Chromatogr 23: 357–364. Papoutsis II et al. (2010). Development and validation of an EI-GC-MS method for the determination of benzodiazepine drugs and their metabolites in blood: applications in clinical and forensic toxicology. J Pharm Biomed Anal 52: 609–614. Pirnay S et al. (2002). Sensitive method for the detection of 22 benzodiazepines by gas chromatography–ion trap tandem mass spectrometry. J Chromatogr A 954: 235–245. Pujadas M et al. (2007). A simple and reliable procedure for the determination of psychoactive drugs in oral fluid by gas chromatography–mass spectrometry. J Pharm Biomed Anal 44: 594–601. Salama Z et al. (1988). Determination of flurazepam and its major metabolites N-1-hydroxyethyland N-1-desalkylflurazepam in plasma by capillary gas chromatography. Arzneimittelforschung 38: 400–403. Smink BE et al. (2004). Quantitative analysis of 33 benzodiazepines, metabolites and benzodiazepine-like substances in whole blood by liquid chromatography–(tandem) mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 811: 13–20. Tiscione NB et al. (2008). Quantitation of benzodiazepines in whole blood by electron impact-gas chromatography–mass spectrometry. J Anal Toxicol 32: 644–652.
Infrared Spectrum Principal peaks at wavenumbers 1695, 1220, 707, 930, 773, 960 cm1 (KBr disk).
Mass Spectrum Principal ions at m/z 199, 244, 200, 178, 179, 184, 183, 245.
Flurbiprofen Analgesic C15H13FO2 = 244.3 CAS—5104-49-4 IUPAC Name 2-(3-Fluoro-4-phenylphenyl)propanoic acid Synonym 2-Fluoro-a-methyl-[1,10 -biphenyl]-4-acetic acid Proprietary Names Ansaid; Antadys; Benactiv; Cebutid; Edolfene; Evril; Fenomel; Flurofen; Froben; Ocufen; Ocuflur; Reupax; Strefen; Strepfen; Transact.
Chemical Properties A colourless crystalline solid. Mp about 110 . Slightly soluble in water; freely soluble in most organic solvents. Log P (octanol/water), 4.2. Colour Tests Liebermann’s reagent—brown; Marquis test—red. Thin-layer Chromatography System TD—Rf 0.30; system TE—Rf 0.06; system TF—Rf 0.30; system TG—Rf 0.16; system TAD—Rf 0.45; system TAJ—Rf 0.47; system TAK—Rf 0.69; system TAL—Rf 0.91 (Ludy Tenger reagent, orange). Gas Chromatography System GA—flurbiprofen RI 1900, flurbiprofen-Me RI 1885, M (OH-)-Me2 RI 2180, M (OH-methoxy)-Me2 RI 2310; system GD—methyl derivative RRT 1.30 (relative to n-C16H34); system GL—flurbiprofen-Me RI 1880, M (OH-)-Me2 RI 2180. High Performance Liquid Chromatography System HV—RRT 0.89 (relative to meclofenamic acid); system HX—RI 585; system HZ—retention time 11.8 min; system HAA—retention time 21.3 min; system HAX—retention time 8.0 min; system HAY—retention time 8.9 min. Ultraviolet Spectrum Aqueous acid—247 nm (A11¼787b). No alkaline shift.
Quantification Plasma GC ECD. Limit of detection, 50 mg/L [Kaiser et al. 1974]. GC-MS Limit of detection, 2 mg, over a prolonged period of time, leads to temporary adrenal suppression. Half-life 7.8 h. Volume of Distribution 4.2 L/kg (range 2.3 to 16.7 L/kg), also reported as 318 L. Clearance Total blood clearance, 0.0182 L/h (range 0.0103 to 0.0284 L/h). Plasma clearance, 1.1 L/min. Protein Binding 91%. Dose Adults (by inhalation): usual dose of 100 to 250 mg twice a day up to 500 to 1000 mg twice a day. Children >4 years old: 50 to 100 mg twice a day. Oral dose: 5 mg four times a day. Krishnaswami S et al. (2000). A sensitive LC-MS/MS method for the quantification of fluticasone propionate in human plasma. J Pharm Biomed Anal 22: 123–129. Laugher L et al. (1999). An improved method for the determination of fluticasone propionate in human plasma. J Pharm Biomed Anal 21(4): 749–758. Li YN et al. (1997). A sensitive method for the quantification of fluticasone propionate in human plasma by high-performance liquid chromatography/atmospheric pressure chemical ionisation mass spectrometry. J Pharm Biomed Anal 16: 447–452. Mackie AE et al. (1996). Pharmacokinetics of intravenous fluticasone propionate in healthy subjects. Br J Clin Pharmacol 41: 539–542. M€ ollmann H et al. (1998). Pharmacokinetic and pharmacodynamic evaluation of fluticasone propionate after inhaled administration. Eur J Clin Pharmacol 53(6): 459–467.
Flutoprazepam Anxiolytic C19H16ClFN2O = 342.8 CAS—25967-29-7 IUPAC Name 7-Chloro-1-(cyclopropylmethyl)-5-(2-fluorophenyl)-1,3-dihydro-2H-1,4-benzodiazepin-2-one Synonyms KB-509; ID-1937. Proprietary Names Restar; Restas.
Mass Spectrum Principal ions at m/z 57, 139, 351, 134, 121, 333, 313, 109.
Quantification Plasma HPLC Limit of detection, 0.01 mg/L [Krishnaswami et al. 2000]. MS–MS detection. Limit of quantification, 0.025 mg/L [Laugher et al. 1999]. Disposition in the Body Fluticasone is poorly absorbed and undergoes extensive first-pass metabolism. The only known metabolite in humans is a 17b-carboxylic acid metabolite. Less than 5% of a dose is excreted in urine as the metabolite with the remainder being excreted in faeces as the parent drug (up to 75%) and the metabolite. Oral bioavailability is 1%. After IV administration, the drug is extensively administered in the body.
Chemical Properties Crystals. Mp 118 to 122 . Quantification Plasma HPLC Column: C18 mBondapak (10 mm, 300 3.9 mm i.d.). Mobile phase: 0.01 mol/L potassium dihydrogen phosphate : acetonitrile (pH 4.5, 50:50), flow rate 1.2 mL/min. UV detection (l¼235 nm). Retention time: flutoprazepam, 16 min; norflutoprazepam, 6 min; 3-hydroxynorflutoprazepam, 4 min; 3-hydroxyflutoprazepam, 12 min. Limit of detection of flutoprazepam 25 mg/L and 10 mg/L for norflutoprazepam [Conti et al. 1991]. Serum GC-MS Column: cross-linked methylsilicone (12 0.2 m i.d.). Temperature programme: 170 (1 min), increased to 270 at 8 /min, held for 10 min. Carrier gas: He, 14.4 psi (99.3 kPa). Detection: mass spectrometer (SIM, m/z 313 for flutoprazepam and 256 for internal standard (IS)). IS: diazepam. Retention time: flutoprazepam, 10.6 min; IS, 8.6 min. Limit of detection, 1 mg/L [Barzaghi et al. 1989]. HPLC UV detection (l¼235 nm). Limit of detection of norflutoprazepam, 3hydroxyflutoprazepam and N-desalkyl-3-hydroxyflutoprazepam, 2 mg/L [Barzaghi et al. 1989]. Disposition in the Body Flutoprazepam is well absorbed after oral administration and undergoes extensive hepatic metabolism, by N-dealkylation, to an active metabolite, norflutoprazepam. Other possible metabolites include 3-hydroxyflutoprazepam and N-desalkyl-3-hydroxyflutoprazepam (both found in urine), and glucuronic acid conjugates of these. Extremely low bioavailability. Therapeutic Concentration Eight young, healthy males between 20 and 28 years were administered a single oral dose of flutoprazepam (2 mg), after overnight fasting. Peak plasma concentrations of norflutoprazepam ranged between 10.6 and 32.4 mg/L and were reached within 2 to 12 h [Barzaghi et al. 1989]. Toxicity Flutoprazepam is moderately toxic by ingestion and intraperitoneal routes.
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:51:16
Fluvoxamine Half-life Norflutoprazepam, 87 22 h. Dose Usual dose of 2 to 4 mg flutoprazepam once daily. Barzaghi N et al. (1989). Pharmacokinetics of flutoprazepam, a novel benzodiazepine drug, in normal subjects. Eur J Drug Metab Pharmacokinet 14: 293–298. Conti I et al. (1991). Propranolol does not alter flutoprazepam kinetics and metabolism in the rat. Eur J Drug Metab Pharmacokinet 16: 53–58.
Fluvastatin Antihyperlipoproteinaemic C24H26FNO4 = 411.5 CAS—93957-54-1 IUPAC Name (3R,5S,6E)-rel-7-[3-(4-Fluorophenyl)-1-(1-methylethyl)-1Hindol-2-yl]-3,5-dihydroxy-6-heptenoic acid
Chemical Properties pKa 4.6. Log P (octanol/water), 4.85. Fluvastatin Sodium C24H25FNNaO4 = 433.5 CAS—93957-55-2 Synonyms XU-62-320; Fluidostatin. Proprietary Names Canef; Cranoc; Fractal; Lescol; Lipaxan; LOCOL; Lymetel;
Primesin; Vastin.
Chemical Properties A white to pale yellow, hygroscopic powder. Mp 194 to
1433
Quantification Plasma HPLC Fluorescence detection (lex¼305 nm, lem¼390 nm). Limit of detection, 1.0 mg/L [Kalafsky, Smith 1993]. Disposition in the Body Fluvastatin is rapidly and completely absorbed after oral administration and undergoes extensive first pass metabolism in the liver, the primary site of action. Hydroxylation of the indole rings at the 5- and 6- positions as well as N-dealkylation and b-oxidation of the side chains occurs. The main metabolite, N-desisopropylpropionic acid, is inactive. The active metabolite, fluvastatin lactone, is rapidly eliminated and not in significant amounts in plasma. Excretion is mainly via faeces, 90% (98% bound. Dose A usual dose of 20 to 40 mg fluvastatin sodium is taken once daily and can be increased at intervals of up to 4 weeks, to a maximum of 40 mg twice daily. Tse FL et al. (1992). Pharmacokinetics of fluvastatin after single and multiple doses in normal volunteers. J Clin Pharmacol 32: 630–638. Kalafsky G, Smith HT (1993). High-performance liquid chromatographic method for the determination of fluvastatin in human plasma. J Chromatogr 614: 307–313. Toreson H, Eriksson BM (1996). Determination of fluvastatin enantiomers and the racemate in human blood plasma by liquid chromatography and fluorometric detection. J Chromatogr A 729: 13–18.
197 . Soluble in water, ethanol and methanol.
High Performance Liquid Chromatography Column: Rx-C8 Zorbax (150 4.6 mm i.d., 5 mm). Temperature: 40 . Mobile phase: aqueous TBAF (tetrabutylammonium fluoride) : 0.1 mol/L phosphate buffer (pH 6.0) : methanol (15:25:60), flow rate 1.0 mL/min. Fluorescence detection (lex¼305 nm, lem¼390 nm). Retention time: 15 min [Toreson, Eriksson 1996]. Ultraviolet Spectrum Aqueous acid—235, 305 nm.
Fluvoxamine Antidepressant, Selective Serotonin Reuptake Inhibitor (SSRI) C15H21F3N2O2 = 318.3 CAS—54739-18-3 IUPAC Name 2-[(E)-[5-Methoxy-1-[4-(trifluoromethyl)phenyl]pentylidene] amino]oxyethanamine Synonym (E)-5-Methoxy-1-[4-(trifluoromethyl)phenyl]-1-pentanone O-(2aminoethyl)oxime
Chemical Properties pKa 8.7. Log P (n-heptane/water), 0.04, (octanol/ pH 7.2), 1.34 [Kristensen et al. 2002]. Extraction yield (chlorobutane), 0.8 [Demme et al. 2005]. Stable in plasma and oral fluid after 3 freeze-thaw cycles [De Castro et al. 2008]. Stock solutions were stable for 15 days when stored at 4 . Stable in plasma and urine stored at 20 after 3 freeze-thaw cycles or at room temperature for 24 h. Stable in plasma and urine for 3 and 2 months, respectively [Ulu 2007]. Mass Spectrum Principal ions at m/z 236, 235, 264, 235, 307, 278, 321, 248, 207.
Fluvoxamine Maleate C15H21F3N2O2,C4H4O4 = 434.4 CAS—61718-82-9 Synonyms DU-23000; MK-264. Proprietary Names Avoxin; Depromel; Dumirox; Dumyrox; Faverin; Fevarin;
Floxyfral; Luvoxe; Maveral; Servox.
Chemical Properties White or off-white odourless crystalline powder. Mp 120
to 121.5 . Sparingly soluble in water; freely soluble in ethanol, methanol and in chloroform; practically insoluble in diethyl ether. Thin-layer Chromatography System TAE—Rf 0.18; system TB—Rf 0.12; system TE—Rf 0.46. Gas Chromatography System GA—RI 1885; system GB—RI 1911; system GM—RRT 0.295. High Performance Liquid Chromatography System HAA—retention time 15.3 min; system HAX—retention time 10.0 min; system HAY—retention time 5.9 min; system HX—RI 430; system HY—RI 363; system HZ—retention time 5.6 min. Liquid Chromatography-Mass Spectrometry Column: Phenomenex Synergi Fusion (20 2 mm i.d., 2.5 mm). Mobile phase: water-acetonitrile-formic acid (95 : 5 : 0.1) : acetonitrile with 0.1% formic acid (97 : 3 to 10 : 90 at 0.6 min), flow rate 1 mL/min. MRM acquisition mode [Youdim et al. 2008].
F
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:51:24
1434
Fluvoxamine
Ultraviolet Spectrum Aqueous acid—254 nm (A11 ¼ 0.310).
F Infrared Spectrum
Mass Spectrum Principal ions at m/z 187, 71, 45, 276, 43, 172, 145, 200.
Quantification Blood GC Column: HP cross-linked methylsilicone (25 m 0.2 mm i.d., 0.11 mm). Carrier gas: He, 195 kPa. Temperature programme: 180 for 1 min to 300 at 10 /min for 3 min. NPD. Limit of quantification, 90 mg/L, limit of detection, 27 mg/L [Martinez et al. 2004]. GC-MS Column: HP-1 cross linked methylsilicone (12.5 m 0.2 mm i.d., 0.33 mm). Carrier gas: He, 1.2 mL/min. Temperature programme: 125 to 290 at 20 /min for 12.7 min. FID, MSD, EI ionisation, full scan mode. Limit of quantification, 0.125 mg/L [Kunsman et al. 1999]. LC-MS Column: XTerra RP18 (100 2.1 mm i.d.). Mobile phase: acetonitrile : 4 mmol/L ammonium formate buffer (pH 3.2; 15 : 85 for 1 min to 35 : 65 over 12 min for 1 min), flow rate 1mL/min. ESI, positive ion mode, MRM acquisition mode. Limit of quantification, 5 mg/L [Castaing et al. 2007]. Plasma For a TLC method see Schweitzer et al. [1986]. GC-MS Column: J & W-5 MS (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1.3 mL/min. Temperature programme: 90 for 1 min to 180 at 50 /min for 10 min to 300 at 10 /min for 2.5 min. EI ionisation at 70 eV, MSD, SIM acquisition mode. Limit of quantification, 5 mg/L [Wille et al. 2007]. Column: Varian factor FOUR VF5ms (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1.3 mL/min. Temperature programme: 90 for 0.5 min to 180 at 50 /min for 10 min to 300 at 10 /min for
10 min. EI ionisation at 70 eV, MSD, SIM acquisition mode. Limit of detection not reported [Wille et al. 2005]. Column: Optima 5 fused silica capillary (15 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 50 mL/min. Temperature programme: 145 for 0.5 min to 207 at 30 /min for 7 min to 221 at 1 /min to 290 at 30 /min. Injector temperature: 250 . EI, SIM acquisition mode. Retention time: 20.0 min. Limit of quantification, 2 mg/L [Eap et al. 1996]. HPLC Column: Phenomenex C18 (250 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : water (80 : 20), flow rate 1 mL/min. UV detection (l ¼ 450 nm). Retention time: 4.8 min. Limit of quantification, 5 mg/L [Ulu 2007]. Column: Varian ResElut C8 reversed phase (150 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : 10.5 mmol/L phosphate buffer containing 0.12% triethylamine (pH 3.5; 30 : 70), flow rate 1.2 mL/min. UV detection (l ¼ 245 nm). Limit of quantification, 15 mg/L, limit of detection, 5.0 mg/L [Saracino et al. 2006]. Column: Hypurity C18 (250 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : phosphate buffer (pH 3.8; 25 : 75 to 40 : 60 over 10 min to 44 : 56 in 8 min), flow rate 1.0 mL/min. DAD (l ¼ 240 nm). Retention time: 17.3 min. Limit of quantification 25 mg/L, limit of detection, 5 mg/L [Duverneuil et al. 2003]. Column: Grand Pack C4-5 (150 4.6 mm i.d., 5 mm). Mobile phase: 0.5% potassium dihydrogen phosphate : acetonitrile (pH 2.5; 75 : 25), flow rate 1.0 mL/min. UV detection (l ¼ 254 nm). Retention time: 21.7 min. Limit of detection, 5 mg/L [Gerstenberg et al. 2003]. Column: Grand Pack C4-5 (150 4.6 mm i.d., 5 mm). Mobile phase: 0.5% potassium dihydrogen phosphate (pH 2.5) : acetonitrile (75 : 25), flow rate 1.0 mL/min. UV detection (l ¼ 254 nm). Retention time: 21.7 min. Limit of detection, 10 mg/L [Ohkubo et al. 2003]. See also Titier et al. [2003], Spigset et al. [2001], Palego et al. [2000], Lucca et al. [2000], H€artter et al. [1992],Pullen & Fatmi [1992], van der Meersch-Mougeot and Diquet [1991], Foglia et al. [1989], and Schweitzer et al. [1986]. LC-MS Column: Sunfire C18 (20 2.1 mm i.d., 3.5 mm). Mobile phase: 2 mmol/ L ammonium formate buffer (pH 3.0) : acetonitrile (85 : 15 for 0.5 min to 50 : 50 at 4 min to 30 : 70 at 5 min for 3 min), flow rate 0.4 mL/min. ESI, positive ion mode, MRM acquisition mode. Retention time: 3.8 min. Limit of quantification, 10 mg/L [De Castro et al. 2008]. Column: Inertsil C8 (150 2.0 mm i.d., 5 mm). Mobile phase: methanol : 10 mmol/L ammonium acetate (pH 5.0) : acetonitrile (70 : 20 : 10), flow rate 0.1 mL/min. SSI, positive ion mode, full scan mode. Retention time: 6.5 min. Limit of quantification, 0.2 mg/L, limit of detection, 0.13 mg/L [Shinozuka et al. 2006]. Serum HPLC Column: Shimpack CLC-ODS (150 4.6 mm i.d., 5 mm). Mobile phase: methanol : 0.05 mol/L sodium phosphate buffer (pH 2.8; 72 : 28) containing 1 mL/L triethylamine, flow rate 2.0 mL/min. Fluorescence detection (lex ¼ 470 nm, lem ¼ 537 nm). Retention time: 4.1 min. Limit of quantification, 0.5 mg/L, limit of detection, 0.2 mg/L [Bahrami, Mohammadi 2007]. Column: Nucleosil 100-5Protect 1 (250 4.6 mm i.d., 5 mm). Mobile phase: 25 mmol/L potassium dihydrogenphosphate (pH 7.0) : acetonitrile (60 : 40), flow rate, 1.0 mL/min. UV detection (l ¼ 230 nm). Retention time: 11.6 min. Limit of detection not reported [Frahnert et al. 2003]. Column: ODS C18 (25 cm 4.6 mm i.d.). Mobile phase: acetonitrile : 0.05 mol/L sodium phosphate buffer (pH 3.8; 50 : 50), flow rate 1.0 mL/min. UV detection (l ¼ 22.4 nm). Retention time: 9.51 min. Limit of detection, 50 ng [Tournel et al. 2001]. LC-MS Column: Chromolith Speed ROD C18 (50 4.6 mm i.d., 5 mm). Mobile phase: methanol : 5 mmol/L acetic acid (pH 3.9) in ammonia (20 : 80 to 70 : 30 at 4 min for 1 min to 20 : 80), flow rate 1.0 mL/min. ESI, positive ion mode, MRM acquisition mode. Limit of quantification, 1.17 mg/L [Kirchherr, K€ uhn-Velten 2006]. Column: Uptisphere (12.5 cm 2 mm i.d., 5 mm). Mobile phase: 50 mmol/ L ammonium acetate (pH 4.0)-acetonitrile: acetonitrile (100 : 0), flow rate 200 mL/ min for 5 min to 300 mL/min in 1 min for 3 min to 200 mL/minin 0.5 min for 0.5 min. ESI, positive ion mode, SIM acquisition mode. Limit of quantification, 19 nmol/L [Gutteck, Rentsch 2003]. Urine HPLC See Plasma. Limit of quantification, 2 mg/L [Ulu 2007]. GC-MS Column: CP-SIL 8 CB (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1.2 mL/min. Temperature programme: 60 for 2 min to 200 at 20 /min to 280 at 5 /min. EI ionisation at 70 eV. Limit of detection, 0.38 mg/lL [Salgado-Petinal et al. 2005]. Column: HP cross-linked methylsilicane (12 m 0.2 mm i.d., 330 nm). Carrier gas: He, 1 mL/min. Temperature programme: 100 for 3 min to 310 at 30 /min for 8 min. EI ionisation at 70 eV, full scan mode. Limit of detection, 100 mg/ L [Maurer, Bickeboeller-Friedrich 2000]. Milk HPLC Column: MOS- Hypersil C8 reversed phase (100 2 mm i.d., 3 mm). Mobile phase: 0.02 mol/L monobasic potassium phosphate : N, N-dimethyloctylamine (pH 6.0) : acetonitrile (64 : 0.11 : 30), flow rate 0.5 mL/min. UV detection (l ¼ 225 nm). Retention time: 6.74 min. Limit of quantification, 2.0 mg/L, limit of detection, 1.25 mg/L [Hostetter et al. 2004]. Column: Select B C18 (250 4.6 mm i.d.). Mobile phase: 45 mmol/L phosphate buffer (pH 3) : acetonitrile (55 : 45), flow rate 1.5 mL/min. UV detection (l ¼ 210 nm) Limit of detection, 2 mg/L [Kristensen et al. 2002]. Oral Fluid LC-MS See Plasma. Limit of quantification, 2 mg/L [De Castro et al. 2008]. Disposition in the Body Fluvoxamine is readily and completely absorbed after oral administration with peak plasma concentrations occurring after about 2 to 8 h (prolonged with enteric-coated tablets to 4 to 12 h). However, it undergoes significant first-pass metabolism and the bioavailability is about 53%. It is widely distributed. Fluvoxamine undergoes extensive metabolism in the liver, mainly by oxidative deamination, oxidative demethylation and N-acetylation to produce several inactive metabolites. The parent and all 11 metabolites have been recovered in urine; 80%) in maize-based foods [Jackson et al. 1996]. o-Phthaldialdehyde derivatives of fumonisins are unstable at 24 but the stability is much improved at 4 [Williams et al. 2004]. Decomposition of FB1 and FB2 occurs in methanolic solutions (percentage decreases after 6-week storage at 4 , 25 and 40 are 5%, 35% and 60%, respectively), whereas they are stable in acetonitrile : water (1 : 1) for up to 6 months at –18 , 4 and 25 [Visconti et al. 1994]. Gamma irradiation of maize flour at 15 kGy was shown to reduce FB1 and FB2 content by ~ 20% [Visconti et al. 1996]. The extraction efficiency of fumonisins from corn-based products is dependent on solvent composition and temperature and is most efficient with an ethanol: water (3 : 7) extraction solvent at 80 [Lawrence et al. 2000]. Studies have shown that some essential oils (eugenol, cinnamic aldehyde, thymol, carvacol and myristin) are effective in inhibiting the growth of F. moniliforme [Juglal et al. 2002] and that reaction with D-glucose may detoxify FB1 [Lu et al. 2002]. Fumonisin B1 C34H59NO15 = 721.8 CAS—116355-83-0 IUPAC Name 1,2,3-Propanetricarboxylic acid 1,10 -[1-(12-amino-4,9,11-trihy-
droxy-2-methyltridecyl)-2-(1-methylpentyl)-1,2-ethanediyl] ester Quantification Plasma HPLC Column: C18 (150 3.9 mm i.d., 5 mm). Mobile phase: acetonitrile (20%) in deionised water containing 0.005 mol/L tetrabutylammonium sulfate (TBA), (pH 2.5), flow rate 2.0 mL/min. Retention time: 8.8 min. Limit of quantification, 0.4 mg/L, limit of detection, 0.1 mg/L [Cwik et al. 1997]. Formulations HPLC Column: C18 Resolve (150 3.9 mm i.d., 5 mm). Mobile phase: acetonitrile : water : phosphoric acid (85%) (57:75:0.09) containing 5 mmol/L TBA, flow rate 2 mL/min. Retention time: fosphenytoin, 9.1 min. Limit of quantification, 15 mg/L [Fischer et al. 1997]. Disposition in the Body Fosphenytoin sodium is rapidly and completely metabolised to phenytoin and other metabolites including phosphate and formaldehyde which are subsequently converted to formate and further metabolites. Phenytoin is metabolised to 5-(p-hydroxyphenyl)-5-phenylhydantoin. The metabolites are excreted in urine. It is distributed in CSF, saliva, semen, gastrointestinal fluids, bile and breast milk. Therapeutic Concentration Twenty healthy, male volunteers (Caucasian, Black, Hispanic and Asian) were administered with a single IV dose of 150, 300, 600 and 1200 mg fosphenytoin sodium over 30 min. Peak plasma concentrations of fosphenytoin were observed half an hour after the beginning of the infusion and reached mean levels of 20, 36, 75 and 129 mg/L, respectively. Peak phenytoin plasma concentrations were reached at 2.2, 4.0, 7.4 and 17.2 mg/L, respectively, 51 to 109 min from the end of infusion [Gerber et al. 1988]. Toxicity The blood toxic range is 30 to 50 mg/L and a concentration of >100 mg/L has been associated with fatalities. Acute hepatotoxicity and hypersensitivity reactions have been observed and phenytoin and the other metabolites may result in toxicity after overdosing, including hypotension, coma and respiratory or circularity depression which may be fatal. Bioavailability About 90%. Half-life (IM administration) 33 min, (IV administration) 8 min. Volume of Distribution Adults and children: 0.6 to 0.7 L/kg, infants: 0.7 to 0.7 L/ kg, full term neonates: 0.8 to 0.9 L/kg and premature neonates: 1 to 1.2 L/kg. Clearance 19.8 L/h. Protein Binding About 90 to 99%. Dose An initial dose of 10 to 15 mg phenytoin sodium equivalent (PSE)/kg body weight is administered at a rate of 50 to 150 mg/min. A daily maintenance dose of 4 to 5 mg/kg body weight is then administered. The dose is reduced in the elderly and patients with renal or hepatic impairment. Up to 450 mg has been administered intramuscularly in adults and IV doses of up to 3000 mg (38 mg/kg body weight).
Synonyms FB1; macrofusine.
Chemical Properties Most prevalent toxin produced from F. moniliforme, a
common mould associated with maize. The maximum yield of FB1 produced by F. moniliforme in maize cultures is at temperatures of 20 to 25 and incubation periods of between 7 and 13 weeks [Alberts et al. 1990]. Powder, very hygroscopic. Fumonisin B2 C34H59NO14 = 691 CAS—116355-84-1 IUPAC Name 1,2,3-Propanetricarboxylic
acid 1,10 -[1-(12-amino-9,11-dihydroxy-2-methyltridecyl)-2-(1-methylpentyl)-1,2-ethanediyl] ester Chemical Properties Powder, very hygroscopic. Thin-layer Chromatography Plates: C18. Solvent system: methanol : 1% aqueous potassium chloride (3 : 2). Locating reagent fluorescamine solution, buffered acetonitrile and UV detection. Rf values: FB1 0.10, FB2 0.50. Limit of quantification not reported [Dutton 1996] Plates: method 1, normal-phase silica; method 2, C18 silica (activated at 110 for 10 min). Solvent systems: method 1, chloroform : methanol : acetic acid (60 : 35 : 10); method 2, methanol : water (80 : 20). Locating reagent, acidic anisaldehyde. Rf values: method 1, FB1 0.32, FB2 0.52; method 2, FB1 0.61, FB2 0.47. Limit of quantification not reported [Ackermann 1991]. High Performance Liquid Chromatography Column: C18 (250 x 4.0 mm i.d., 5 mm). Mobile phase: methanol-0.05 mol/L sodium dihydrogen phosphate (pH 5.0; 50 : 50) : acetonitrile-water (80 : 20; 100 : 0 for 5 min to 50 : 50 for 15 min), flow rate 1.0 mL/min. Fluorescence detection (o-phthalaldehyde [OPA] derivatives lex ¼ 335 nm, lem ¼ 440 nm; 4-fluoro-7-nitro-2,1,3-benzoxadiazole [NBD-F] derivatives lex ¼ 440 nm, lem ¼ 535 nm). Limit of detection, 0.01 mg/kg for FB1 and FB2 (OPA derivatives), 0.08 mg/kg (NBD-F derivatives) [Hinojo et al. 2006]. Column: C8 (250 x 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile-watertrifluoroacetic acid (pH 2.7; 5 : 95 : 0.025) : acetonitrile-water-trifluoroacetic acid (90 : 10 : 0.025; 80 : 20 to 40 : 60 over 30 min to 20 : 80 over 10 min to 0 : 100 over 2 min), flow rate 1.0 mL/min. ELS detection. Retention times: FB1 16.3 min, FB2
Clarke's Analysis of Drugs and Poisons Chapter No. F Dated: 15/3/2011 At Time: 21:52:0
Fumonisins 21.4 min, FB3 19.1 min, FB4 24.4 min (underivatised samples). Limit of quantification not reported [Wilkes et al. 1995]. Column: C18 (250 x 3.2 mm i.d., 5 mm). Mobile phase: methanol : 0.1 mol/L sodium dihydrogen phosphate (pH 3.35; 75 : 25), flow rate 1.0 mL/min. Fluorescence detection (lex ¼ 335 nm, lem ¼ 440 nm). Retention times: FB1 ~5 min, FB2 ~ 12 min (OPA derivatives). Limit of quantification not reported [Visconti et al. 1994]. Liquid Chromatography-Mass Spectrometry Column: C18 (150 x 4.6 mm i.d., 4 mm). Mobile phase: 0.2% formic acid : acetonitrile containing 0.2% formic acid (75 : 25 for 5 min to 60 : 40 over 30 min for 2 min to 40 : 60 over 3 min for 5 min), flow rate 1.0 mL/min. ESI, positive ion mode. Limit of quantification not reported [Josephs 1996]. Mass Spectrum
Quantification Urine LC-MS Column: C18 (50 x 4.6 mm i.d., 5 mm). Mobile phase: water-acetonitrile-formic acid (90 : 10 : 0.1) : water-acetonitrile-formic acid (10 : 90 : 0.1; 75 : 25 to 25 : 75 over 11 min), flow rate 1.0 mL/min. ESI, positive ion mode. Retention time: FB1 8.0 min. Limit of quantification, 20 ng/L [Gong et al. 2008]. Hair LC-MS Column: C18 (150 x 4.6 mm i.d., 5 mm). Mobile phase: water-acetonitrile-formic acid (90 : 10 : 0.1) : water-acetonitrile-formic acid (10 : 90 : 0.1; 80 : 20 to 72 : 28 over 24 min for 1 min to 80 : 20 over 2 min for 8 min), flow rate 0.7 mL/ min. ESI, positive ion mode. Retention times: FB1 11.5 min, FB2 26.5 min, FB3 20.5 min. Limit of quantification, 60 pg on-column; limit of detection, 25 pg oncolumn [Sewram et al. 2003]. Other TLC Maize Samples. Plates: C18. Solvent system: methanol : 4% aqueous potassium chloride (70 : 30). Locating reagent or derivatisation reagent, fluorescamine. UV detection. Rf value: FB1 0.35. Limit of quantification, 0.5 mg/kg [Shephard, Sewram 2004]. Plates: C18. Solvent system: 4% aqueous potassium chloride : methanol (2 : 3). Derivatisation spray agents (sequentially): 0.1 mol/L sodium tetraborate, 0.40 g/L fluorescamine in acetonitrile; dry for 1 to 5 min and then spray with 0.01 mol/L boric acid : acetonitrile (2 : 3); dry plate at 55 to 60 for 15 min. UV detection (l ¼ 366 nm). Rf values: FB1 0.5, FB2 0.25. Limit of detection, FB10.1 mg/kg [Preis, Vargas 2000]. HPLC Maize Samples and Laboratory Cultures. Column: C18 (250 x 4.6 mm, 5 mm). Mobile phase: acetonitrile-acetic acid (99 : 1) : water-acetic acid (99 : 1; 60 : 40 for 8 min to 80 : 20 over 16 min for 4 min), flow rate 1.0 mL/min. Fluorescence detection (lex ¼ 420 nm, lem ¼ 500 nm). Limit of quantification, FB1 and FB2 (2,3-naphthalene dicarboxaldehyde derivatives) 25 mg/kg [Arino et al. 2007]. Maize Samples. C18 (250 x 4.6 mm i.d., 5 mm). Mobile phase: acetonitrile : water : acetic acid (61 : 38 : 1), flow rate 1.0 mL/min. Fluorescence detection (lex ¼ 420 nm, lem ¼ 500 nm). Retention times: FB1 7.4 min, FB2 13.3 min (naphthalene-2,3-dicarboxaldehyde derivatives). Limit of detection, FB1 20 mg/kg, FB2 15 mg/kg [Lino et al. 2006]. Swine Liver Samples. Column: phenylhexyl (250 x 4.6 mm i.d., 5 mm). Mobile phase: aqueous buffer (pH 3.4)-2% glacial acetic acid-0.1% TEA : acetonitrile (30 : 70 to 50 : 50 over 50 min), flow rate 1.0 mL/min. Pre-column derivatisation with OPA. Fluorescence detection (lex ¼ 334 nm, lem ¼ 440 nm). Retention times: FB1 ~ 45 min, aminopentol-1 ~ 41 min. Limit of quantification, FB1 75 mg/kg, aminopentol-1 42 mg/kg; limit of detection, FB1 20 mg/kg, aminopentol-1 10 mg/kg [Pagliuca et al. 2005]. Herbal Tea and Medicinal Plant Samples. Column: C18 (250 x 4.6 mm i.d., 5 mm). Mobile phase: methanol : 0.1 mol/L sodium dihydrogen phosphate (pH 3.3; 8 : 2), flow rate 0.7 mL/min. Fluorescence detection (lex ¼ 338 nm, lem ¼ 455 nm). Retention times: FB1 10.9 min, FB2 25.6 min (OPA derivatives). Limit of quantification, FB1 103 mg/kg, FB2 1562 mg/kg; limit of detection, FB1 31 mg/kg, FB2 468 mg/kg [Martins et al. 2001; Omurtag, Yazicioglu 2004]. Maize Kernels, Tortillas, Masa samples. Column: C18 (250 x 4.6 mm i.d., 5 mm). Mobile phase: methanol : 0.1 mol/L phosphate buffer (pH 3.35; 77 : 23), flow rate 1.0 mL/min. Fluorescence detection (lex ¼ 335 nm, lem ¼ 440 nm). Limit of detection, 25 mg/kg [De La Campa et al. 2004; Dilkin et al. 2001; Duncan et al. 1998; Sydenham et al. 1996; Visconti et al. 2001]. Maize Silage Samples. Column: C18 (250 x 4.0 mm i.d., 5 mm). Mobile phase: methanol-acetonitrile-water-propan-2-ol-acetic acid (100 : 52 : 73 : 20 : 4) : propan2-ol (100 : 0 for 30 min to 70 : 30 over 30 min), flow rate 1.0 mL/min. Retention times: FB1 20.9 min, FB3 40.7 min, FB2 43.3 min (naphthalene dicarboxaldehyde derivatives). Fluorescence detection (lex ¼ 268 nm, lem ¼ 470 nm). Limit of detection, FB1 50 mg/kg, FB2 and FB3 500 mg. Serious nephrotoxicity can also occur. Half-life 25–111 h. Dose The usual dose is 100 to 200 mg/m2 body surface administered once daily. Webster LK et al. (2000). A pharmacokinetic and phase II study of gallium nitrate in patients with non-small cell lung cancer. Cancer Chemother Pharmacol 45(1): 55–58.
g-Butyrolactone Depressant C4H6O2 = 86.1 CAS—96-48-0 IUPAC Name Oxolan-2-one Synonyms gBL; 1,2-Butanolide; butyrylactone; dihydro-2(3H)-furanone; gamma hydroxybutyric acid lactone; gamma lactone; 2-Oxanolone; tetrahydro-2-furanone. Proprietary Names Blue Nitro; Blue Nitro Vitality; Firewater; Gamma G; GH Revitalizer; Insom-X; Invigorate; Longevity; Remforce; Renewtrient; Revivarant; Revivarant G.
Chemical Properties Colourless liquid. Mp 43 ; Bp 204 . It is miscible with water, alcohol, ketones, esters and aromatic hydrocarbons; limited solubility in aliphatic and cycloaliphatic hydrocarbons. Log P (octanol/water), 0.64 [Hansch et al. 1995]. No significant instability [Wood et al. 2004]. Stock standard solutions, internal standard solutions and prepared solutions were stable for 3 months at 4 . Stable in plasma for 3 months [Fukui et al. 2003]. (S)-3-Hydroxy-g Butyrolactone
Chemical Properties A white, or faintly cream-coloured, hygroscopic powder. Mp about 235 , with decomposition. Soluble 1 in 0.6 of water, 1 in 115 of ethanol and 1 in 1500 of chloroform; freely soluble in dilute acetone; sparingly soluble in anhydrous acetone, ether and benzene. Log P (octanol/water), 6.4. Thin-layer Chromatography System TA—Rf 0.00; system TN—Rf 0.34; system TO—Rf 0.05 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 2625. Ultraviolet Spectrum Aqueous acid—225 (A11¼525a), 266 nm (A11¼9a). Infrared Spectrum Principal peaks at wavenumbers 1100, 1120, 1260, 1000, 780, 1600 cm1 (KBr disk). Quantification Plasma Spectrofluorimetry Limit of detection, 50 ng [Ramzan et al. 1980]. Disposition in the Body Slowly and incompletely absorbed after oral administration; absorbed after IM administration but is generally given by the IV route. Small amounts enter the CSF. Almost 100% of a dose is excreted unchanged in the urine in 24 to 30 h. Negligible amounts are excreted in the bile.
C4H6O3 = 102.0 CAS—7331-52-4 Synonym HGB Chemical Properties Pale-yellow liquid. Bp 98 to 100 .
Colour Test Specific tests for GBL using mixtures of several common reagents are given in the Colour Tests chapter. Gas Chromatography System GAO—GBL RT 4.0 min;GHB-TMS2 RT 5.6 min. Gas Chromatography-Mass Spectrometry Column: HP Ultra 1 bonded phase capillary (12 m 0.2 mm i.d., 0.33 mm). Carrier gas: He, 0.2 kg/cm2. Temperature programme: 50 for 0.6 min to 275 at 15 /min. SIM acquisition mode (m/z: 41, 42, 56, 86, 100). Retention time: 2.6 min [Ferrara et al. 1993]. High Performance Liquid Chromatography System HBG—GBL RT 4.0 min; GHB RT 3.5 min. Column: C18 mBondapak (300 3.9 mm i.d., 10 mm). Mobile phase: 10 mmol/L potassium dihydrogen phosphate buffer (pH 3.0): methanol (70 : 30), flow rate 1.0 mL/min. DAD (l ¼ 215 nm). Retention time: 3.96 min for GBL; 3.45 min for GHB [Mesmer, Satzger 1998].
Clarke's Analysis of Drugs and Poisons Chapter No. G Dated: 16/3/2011 At Time: 7:21:30
g-Butyrolactone Infrared Spectrum Principal peaks at wavenumbers 1820, 1163, 1047, 2993, 2909, 874 cm1.
Mass Spectrum Principal ions at m/z 42, 28, 41, 29, 27, 86, 56, 39.
Quantification Blood GC Column: DB-624 capillary (30 m 0.25 mm i.d., 1.4 mm). Temperature programme: 50 for 3 min to 150 at 20 /min for 7 min. Carrier gas: N2, 34 cm/s. IS: g butyrolactone-d6. FID, SIM acquisition mode (m/z: 86 for GBL and 92 for IS). Retention time: 11.2 min for GBL; 12.7 min for IS. Limit of detection, 0.5 mg/L for GHB [LeBeau et al. 2000]. GC-MS Column: ZB-FFAP (30 m). Carrier gas: He. EI ionisation. Limit of detection not reported [Strickland et al. 2005]. Column: DB5-MS (30 m 0.25 mm i.d., 5 mm).Carrier gas: He, 15 psi. Temperature programme: 60 for 1 min to 85 for 1 min to 300 for 15 min all at 30 /min. MID. Limit of detection, 10 mg/L [Duer et al. 2001]. Column: 5% phenylmethylsilicone (30 m 0.25 mm i.d., 0.33 mm). Carrier gas: He. Temperature programme: 60 for 2 min to 180 at 20 /min to 250 at 35 /min for 4 min. SIM acquisition mode. Limit of quantification, 1 mg/ L, limit of detection, 0.5 mg/L [Couper, Logan 2000]. EI ionisation, full scan mode. Retention time: 10.9 min. Limit of detection, 0.5 mg/L for GHB [LeBeau et al. 2000]. Plasma GC-MS Column: DB-5 (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He. Temperature programme: 60 to 115 at 10 /min up to 300 at 30 /min. Retention time: 4.54 min. Limit of quantification, 8 mg/L [Jones et al. 2008]. Column: Varian Factor IV (15 m 0.25 mm i.d., 0.25 mm). Temperature programme: 60 for 2 min to 180 at 20 /min to 250 at 50 /min for 9.4 min. EI ionisation, MRM acquisition mode. Limit of quantification, 100 mg/L [Paul et al. 2006]. Column: DB-5 MS (25 m 0.2 mm i.d., 0.33 mm). Carrier gas: He, 38 cm/s. Temperature programme: 60 for 1 min to 90 at 10 /min to 270 at 35/ min for 1 min. SIM acquisition mode. Retention time: 4.35 min. Limit of quantification, 0.5 mg/L, limit of detection, 0.2 mg/L [Brenneisen et al. 2004]. Column: DB-WAX capillary (30 m 0.32 mm i.d., 0.25 mm). Temperature programme: 50 for 1 min to 190 at 20 /min to 250 at 40 /min for 2 min. NICI at 70 eV, SIM acquisition mode. Retention time: 5.89 min. Limit of detection, 10 mg/L [Fukui et al. 2003]. Column: FFAP (acid-modified polyethylene glycol) capillary (25 m 0.2 mm i.d., 0.3 mm). Carrier gas: He, 0.8 mL/min. Temperature programme: 50 for 1.5 min to 240 at 30 /min for 2 min. IS: g-butyrolactone-d6. PICI, SIM acquisition mode (m/z: 86, 87, 88 for GBL and 92,93,94 for IS). Retention time: 6.54 min for GBL; 6.53 min, IS. Limit of detection, 0.05 mg/L [Frison et al. 2000].
1453
Serum GC-MS Macherey-Nagel Optima 5MS 5% phenyl 95% methylsiloxane (30 m 0.25 mm i.d., 1 mm). Carrier gas: He, 1.0 mL/min. Temperature programme: 35 for 1 min to 100 at 10 /min to 200 at 50 /min for 1 min. EI ionisation at 70 eVor PICI. Limit of quantification, 0.2 mg/L, limit of detection, 0.16 mg/L [Lenz et al. 2009]. Column: Macherey-Nagel Optima 5MS GC capillary (30 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1 mL/min. Temperature programme: 35 for 1 min to 100 at 10 /min to 200 at 50 /min for 1 min. PICI, full scan mode. Limit of quantification, 2 mg/L, limit of detection, 0.5 mg/L [Lenz et al. 2008]. Urine GC See Blood [Duer et al. 2001]. See Blood [LeBeau et al. 2000]. GC-MS See Serum. Limit of detection, 0.17 mg/L [Lenz et al. 2009]. See Serum. Limit of quantification, 4 mg/L [Lenz et al. 2008]. See Blood. Limit of quantification, 0.2 mg/L, limit of detection, 0.1 mg/L [Brenneisen et al. 2004]. Column: DB5MS. Carrier gas: He, 12 psi. Temperature programme: 60 to 270 at 9 /min for 23.3 min in total. EI ionisation, SIM acquisition mode. Limit of detection, 2 mg/L [Blair et al. 2001]. See Blood [LeBeau et al. 2000]. See Plasma. Limit of detection, 0.1 mg/L [Frison et al. 2000]. Column: HP-1MS cross-linked methylsiloxane capillary (12 m 0.25 mm i.d., 0.25 mm). Carrier gas: He, 1.6 mL/min. Temperature programme: 65 for 0.5 min to 105 at 15 /min to 300 at 25 /min. SIM acquisition mode. Limit of detection, 5 mg/L [McCusker et al. 1999]. LC-MS Column: Atlantis dC18 (100 3 mm i.d., 5 mm). Mobile phase: 0.1% aqueous formic acid: methanol (90 : 10), flow rate 0.2 mL/min. ESI, positive ion mode, MRM acquisition mode. Retention time: 5.1 min for b-hydroxybutyric acid. Limit of quantification, 1.0 mg/L [Wood et al. 2004]. Ocular Fluid GC See Blood [Duer et al. 2001]. Brain GC See Blood [Duer et al. 2001]. Other GC-MS Commercial Beverages. Column: HP-5 MS (30 m 0.25 mm i.d., 0.25 mm). Temperature programme: 45 for 2 min to 110 at 5 K/min to 300 at 30 K/min. Full scan mode. Limit of quantification, 150 ppb, limit of detection, 50 ppb [Sabucedo, Furton 2004]. Wine. Column: DB-5 (20 m 0.18 mm). Carrier gas: He, 10.2 psi. Temperature programme: 60 for 1 min to 300 at 15 / min. Retention time: 3.6 min. Limit of detection, 5 mg/L [Vose et al. 2001]. CE Beverages. Column: Polymicro capillary (total/effective length 80/72 cm, 50 mm). Mobile phase: 20 mmol/L SDS (pH 9.2) with 7.0% acetonitrile. DAD. Limit of detection, 152 mg/L [Bishop et al. 2004]. Note For the positive detection of GBL in vermouth, sherry, port, and red and white wine see Elliott, Burgess [2005]. Disposition in the Body g-butyrolactone (GBL) is well absorbed after administration and is biotransformed in the body to g- hydroxybutyrate (GHB), which is a potent CNS depressant. This conversion can occur within minutes. GBL has a greater bioavailability than GHB. Toxicity Signs and symptoms of toxicity can include prolonged unconsciousness and coma, respiratory depression (including respiratory arrest and very dangerously slow breathing), nausea/vomiting, confusion, anxiety/nervousness, cardiac arrest and seizures. A 44-year-old male tourist collapsed after consuming ‘Furamax Revitaliser’ – 2.5 g of 2(3H) furanone dihydro (GBL) per fluid ounce (8 g/100 mL). He subsequently made a full recovery [Dupont, Thornton 2001]. A 19-year-old female was offered a drink from a plastic container at a party. After consuming some of the drink, she had no recollection of the rest of the evening. Her friends described her as being ‘drunker than she should have been’. A sexual assault examination indicated that intercourse had occurred. A urine sample had a concentration of GBL of 4.4 mg/L, 9 h after the drink was consumed. GHB was detected at concentrations consistent with endogenous levels. A 25-year-old male was found sleeping behind the wheel of his car in the middle of a busy street. In the car there was a container of RenewTrient, a GBLcontaining product. Blood analysis showed a concentration of GHB of 157 mg/L but GBL was not detected. [LeBeau et al. 2000]. Note See Lenz et al. [2008] for more case studies. Half-life 20–30 min. Bishop SC et al. (2004). Micellar electrokinetic chromatographic screening method for common sexual assault drugs administered in beverages. Forensic Sci Int 141: 7–15. Blair S et al. (2001). Determination of gamma-hydroxybutyrate in water and human urine by solid phase microextraction-gas chromatography/quadrupole ion trap spectrometry. J Forensic Sci 46: 688–693. Brenneisen R et al. (2004). Pharmacokinetics and excretion of gamma-hydroxybutyrate (GHB) in healthy subjects. J Anal Toxicol 28: 625–630. Couper FJ, Logan BK (2000). Determination of gamma-hydroxybutyrate (GHB) in biological specimens by gas chromatography–mass spectrometry. J Anal Toxicol 24: 1–7. Duer WC et al. (2001). Application of a convenient extraction procedure to analyze gammahydroxybutyric acid in fatalities involving gamma-hydroxybutyric acid, gamma-butyrolactone, and 1,4-butanediol. J Anal Toxicol 25: 576–582. Dupont P, Thornton J (2001). Near-fatal gamma-butyrolactone intoxication–first report in the UK. Hum Exp Toxicol 20: 19–22. Elliott SP, Burgess V (2005). Clinical urinalysis of drugs and alcohol in instances of suspected surreptitious administration ("spiked drinks"). Sci Justice 45: 129–134. Ferrara SD et al. (1993). Therapeutic gamma-hydroxybutyric acid monitoring in plasma and urine by gas chromatography-mass spectrometry. J Pharm Biomed Anal 11: 483–487. Frison G et al. (2000). Determination of gamma-hydroxybutyric acid (GHB) in plasma and urine by headspace solid-phase microextraction and gas chromatography/positive ion chemical ionization mass spectrometry. Rapid Commun Mass Spectrom 14: 2401–2407. Fukui Yet al. (2003). Validation of a simple gas chromatographic-mass spectrometric method for the determination of gamma-butyrolactone in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 785: 73–80. Hansch C, et al. (1995). Exploring QSAR: Hydrophobic, Electronic, and Steric Constants. Washington DC: American Chemical Society. Jones AW et al. (2008). Driving under the influence of gamma-hydroxybutyrate (GHB). Forensic Sci Med Pathol 4: 205–211.
G
Clarke's Analysis of Drugs and Poisons Chapter No. G Dated: 16/3/2011 At Time: 7:21:30
1454
g-Hydroxybutyrate
LeBeau MA et al. (2000). Analysis of biofluids for gamma-hydroxybutyrate (GHB) and gammabutyrolactone (GBL) by headspace GC-FID and GC-MS. J Anal Toxicol 24: 421–428. Lenz D et al. (2009). Determination of gamma-hydroxybutyric acid in serum and urine by headspace solid-phase dynamic extraction combined with gas chromatography-positive chemical ionization mass spectrometry. J Chromatogr A 1216: 4090–4096. Lenz D et al. (2008). Intoxications due to ingestion of gamma-butyrolactone: organ distribution of gamma-hydroxybutyric acid and gamma-butyrolactone. Ther Drug Monit 30: 755–761. McCusker RR et al. (1999). Analysis of gamma-hydroxybutyrate (GHB) in urine by gas chromatography-mass spectrometry. J Anal Toxicol 23: 301–305. Mesmer MZ, Satzger RD (1998). Determination of gamma-hydroxybutyrate (GHB) and gammabutyrolactone (GBL) by HPLC/UV-VIS spectrophotometry and HPLC/thermospray mass spetrometry. J Forensic Sci 43: 489–492. Paul R et al. (2006). GC-MS-MS determination of gamma-hydroxybutyrate in blood and urine. J Anal Toxicol 30: 375–379. Sabucedo AJ, Furton KG (2004). Extractionless GC/MS analysis of gamma-hydroxybutyrate and gamma-butyrolactone with trifluoroacetic anhydride and heptafluoro-1-butanol from aqueous samples. J Sep Sci 27: 703–709. Strickland RM et al. (2005). Survival of massive gamma-hydroxybutyrate/1,4-butanediol overdose. Emerg Med Australas 17: 281–283. Vose J et al. (2001). Detection of gamma-butyrolactone (GBL) as a natural component in wine. J Forensic Sci 46: 1164–1167. Wood M et al. (2004). Simultaneous analysis of gamma-hydroxybutyric acid and its precursors in urine using liquid chromatography-tandem mass spectrometry. J Chromatogr A 1056: 83–90.
G
g-Hydroxybutyrate Anaesthetic C4H8O3 = 104.1 CAS—591-81-1 IUPAC Name 4-Hydroxybutanoic Synonyms BRN-1720582; gamma hydroxybutyric acid; GHB; 4-hydroxybutyric acid. Street Names Georgia home boy; grievous bodily harm; liquid ecstasy; scoop; fantasy.
Chemical Properties Crystals. It is water soluble and produces solutions that are mildly saline. Extraction yield (chlorobutane), 0 [Demme et al. 2005]. Sodium Gamma Hydroxybutyrate Synonyms NSC-84223; sodium gamma hydroxybutyric acid; sodium oxybate; sodium oxybutyrate; Wy3478. Proprietary Names Alcover; Gamma-OH; Somatomax PM; Somsanit; Xyrem.
Sodium-4-hydroxybutyrate C4H8NaO3 = 127.1 CAS—502-85-2 Chemical Properties A colourless, odourless powder, capsules or liquid.
Colour Test Specific tests using mixtures of several common reagents are given in the Colour Tests chapter. Gas Chromatography System GAO—g-hydroxybutyrate (GHB)-TMS2 RT 5.6 min, g-butyrolactone (GBL) RT 4.0 min. High Performance Liquid Chromatography System HBG—GHB-TMS2 RT 3.5 min, GBL RT 4.0 min. Column: mBondapak C18 (300 3.9 mm i.d., 10 mm). Mobile phase: 10 mmol/L potassium dihydrogen phosphate buffer (pH 3.0) : methanol (70 : 30), flow rate 1.0 mL/min. Retention time: GHB 3.45 min, GBL 3.96 min [Mesmer, Satzger 1998]. Mass Spectrum Principal ions at m/z 147, 73, 75, 233, 117, 148, 77, 59 (GHB-TMS2).
Quantification Blood GC Column: DB-624 (30 m 0.25 mm i.d., 1.4 mm). Carrier gas: N2, 34 cm/s. Temperature programme: 50 for 3 min to 150 at 20 /min for 7 min. FID. Limit of detection, 0.5 mg/L [LeBeau et al. 2000]. GC-MS See GC. EI ionisation, full scan mode. Retention time: 10.9 min. Limit of detection, 0.5 mg/L [LeBeau et al. 2000]. Plasma GC-MS Column: ULTRA-1 (12 m 0.2 mm i.d., 0.33 mm). Carrier gas: He, 0.2 kg/cm2. Temperature programme: 50 for 0.6 min to 275 at 15 /min. EI
ionisation, SIM acquisition mode. Limit of detection, 0.2 mg/L [Ferrara et al. 1993]. Column: HP ULTRA 1 bonded phase (12 m 0.2 mm i.d., 0.3 mm). EI ionisation, SIM acquisition mode. Limit of detection, 1 mg/L [Ferrara et al. 1992]. Urine GC See Blood [LeBeau et al. 2000]. GC-MS Column: HP-1 (25 m 0.32 mm i.d., 0.5 mm). Carrier gas: He, 1.0 mL/ min. Temperature programme: 50 for 1 min to 300 at 10 /min. Limit of detection, 2 mg/L [Kavanagh et al. 2001]. Column: HP-1 100% polydimethylsiloxane (12 m 0.2 mm i.d., 0.33 mm). Temperature programme: 60 for 2 min to 260 at 30 /min for 1 min. Carrier gas: He, 0.5 mL/min. I.S.: GHB-d6. SIM acquisition mode, m/z 233, 234, 235 for GHB and 239, 240, 241 for IS. Retention time: GHB 4.43 min, IS 4.41 min. Limit of detection, 2 mg/L [Elian 2000]. See Blood [LeBeau et al. 2000]. Column: HP-1MS cross-linked methylsiloxane (12 m 0.25 mm i.d., 0.25 mm). Temperature programme: 65 for 0.5 min to 105 at 15 /min to 300 at 25 /min. Carrier gas: He, 1.6 mL/min. SIM acquisition mode, m/z as above. I.S.: GHB-d6. Retention time: GHB 3.7 min. Limit of detection, 5 mg/L [McCusker et al. 1999]. See Plasma. Limit of detection, 0.1 mg/L [Ferrara et al. 1993]. See Plasma. Limit of detection, 0.2 mg/L [Ferrara et al. 1992] Hair LC-MS Limit of quantitation 0.4 ng/mL, limit of detection, 0.2 ng/mL [Stout et al. 2010]. Disposition in the Body GHB is readily absorbed after oral administration and rapidly metabolised in the liver by oxidative enzymes. Conversion to GBL can occur and elimination is rapid, via the kidneys, with urine recovery virtually complete within 8 h of administration. Only negligible amounts of the parent drug are recovered unchanged in urine (75 mg/L may be associated with toxic effects. It has a high incidence of anaphylactic reactions and has been withdrawn from the market in most countries. Half-life About 1 to 2 h. Dose Up to 1.2 g daily. Ennachachibi A et al. (1988). Effective high-performance liquid chromatographic determination of glafenine in plasma: pharmacokinetic application. J Chromatogr 427: 307–314. Tournet MC et al. (1981). J Chromatogr 224: 348–352. Tracqui A et al. (1988). Simultaneous determination of glafenine and floctafenine in human plasma using high-performance liquid chromatography. Ann Biol Clin (Paris) 46: 665–667. Vermerie N et al. (1992). Pharmacokinetics of glafenine and glafenic acid in patients with cirrhosis, compared to healthy volunteers. Fundam Clin Pharmacol 6: 197–203.
Glaucine Cough Suppressant, Drug of Abuse, Quinoline C21H25NO4 = 355.4 CAS—475-81-0 (d-glaucine); 5630-11-5 (dl-glaucine). Synonyms Boldine dimethyl ether; (6aS)-5,6,6a-7-tetrahydro-1,2,9,10-tetramethoxy-6-methyl-4H-dibenzo[de,g]quinoline; 1,2,9,10-tetramethoxyaporphine. Proprietary Name Glauvent. It is also an ingredient in Bronchitussin; Bronchocin; Broncholytin.
Clarke's Analysis of Drugs and Poisons Chapter No. G Dated: 16/3/2011 At Time: 7:21:33
Glibenclamide
Chemical Properties Found in Glaucium flavum Crantz (G. luteum Scop.), Papaveraceae, in Dicentra and Corydalis species, Fumariaceae and several more species. The d-form is prevalent in nature. Orthorhombic plates, prisms from ethyl acetate or ether. Mp 120 . Soluble in acetone, alcohol, chloroform, ethyl acetate; moderately soluble in ether, petroleum ether; practically insoluble in water and benzene. Dimethoxy analogue of boldine. Glaucine Hydrochloride Trihydrate C21H25NO4,HCl, 3H2O = 445.9 Chemical Properties Mp 232 (anhydrous). Soluble in water, alcohol and
1459
Temperature programme: 200 for 4 min to 250 at 10 /min for 15 min. EI ionisation at 70 eV, positive ion mode, SIM acquisition mode. Retention times: glaucine 8.0 min, sarcophylline 6.2 min, sarcocapnine 6.5 min, cularicine 6.6 min, O-methylcularicine 6.8 min, cularidine 7.2 min, celtisine 7.3 min, cularine 7.5 min, sarcocapnidine 7.5 min, celtine 7.6 min, protopine 8.8 min, ribasine 9.5 min, dihydrosanguinarine 10.9 min, chelidonine 11.7 min. Limit of quantification, not reported [Suau et al. 2005]. HPLC Plant MaterialSamples (Critesion lechleri). Column: C18 (250 4.6 mm i. d., 5 mm). Mobile phase: (leaf samples) acetonitrile : 0.1 mol/L phosphate (pH 2.5)10 mmol/L SDS-0.08% TEA (43 : 57); (latex samples) acetonitrile : 0.1 mol/L phosphate (pH 2.5)-10 mmol/L SDS-0.1% TEA (45 : 55), flow rate 2 mL/min. UV detection (l ¼ 254 nm). Retention times: (leaf samples) glaucine 9.8 min, norisoboldine 4.2 min, isoboldine 4.6 min, magnoflorine 5.2 min, thaliporphine 6.5 min, taspine 9.0 min. Limit of quantification not reported [Milanowski et al. 2002]. Therapeutic Concentration Three healthy volunteers were administered a single 60 mg oral dose of d-glaucine. Peak plasma concentrations were reported as follows:
Cmax (mg/L) Time (h) Urinary excretion 0–72 h (% of dose)
Subject 1
Subject 2
Subject 3
200 1.5 0.10
285 2 0.27
255 0.75 0.13
[Fels et al. 1984].
chloroform. Glaucine Hydrobromide C21H25NO4,HBr = 436.1 CAS—5996-06-5 (d-form) Chemical Properties Mp 235 . Less soluble than the hydrochloride.
Glaucine Phosphate (C21H25NO4)2,3H3PO4 = 1004.8 CAS—73239-87-9 (dl-form) Synonym DL-832 Chemical Properties Crystalline powder.
Ultraviolet Spectrum Principal peaks at 290, 310 nm.
Toxicity LD50 in mice 98 mg/kg (IV), 401 mg/kg (oral). A 23-year-old woman presented to an emergency department following the ingestion of 2 tablets of ‘head candy’, marketed as a 1-benzylpiperazine (BZP)free ‘herbal high’. She developed nausea and vomiting within 30 min of ingestion. Serum and urine samples were collected at admission and both contained glaucine. The serum glaucine concentration was ~0.7 mg/L. She was discharged the following day her symptoms had resolved [Dargan et al. 2008]. Note For a study of the mechanism of action of glaucine in the rat, see Orallo et al. [1993]; for a study of the abuse potential of glaucine in monkeys, see Schuster et al. [1982]. Dargan PI et al. (2008). Detection of the pharmaceutical agent glaucine as a recreational drug. Eur J Clin Pharmacol 64: 553–554. Fels JP et al. (1984). Determination of glaucine in plasma and urine by high-performance liquid chromatography. J Chromatogr 308: 273–281. Milanowski DJ et al. (2002). Geographic distribution of three alkaloid chemotypes of Croton lechleri. J Nat Prod 65: 814–819. Orallo F et al. (1993). Study of the mechanism of the relaxant action of (þ)-glaucine in rat vas deferens. Br J Pharmacol 110: 943–948. Schuster CR et al. (1982). Experimental studies of the abuse potential of d,l-glaucine 1,5-phosphate in rhesus monkeys. Pharmacol Biochem Behav 16: 851–854. Suau R et al. (2005). Identification and quantification of isoquinoline alkaloids in the genus Sarcocapnos by GC-MS. Phytochem Anal 16: 322–327. Velcheva M et al. (1992). The alkaloids of the roots of Thalictrum flavum L. Acta Pharm Nord 4: 57–58.
Glibenclamide Mass Spectrum Principal ions at m/z 354, 355, 80. Quantification Plasma HPLC Column: LiChrosorb Si 60 (125 4.0 mm i.d., 5 mm). Mobile phase: n-hexane : methanol : tetrahydrofuran : diethylamine (88.5 : 7.5 : 4 : 0.15), flow rate 1.5 mL/min. Fluorescence detection (lex ¼ 310 nm, lem ¼ 340 nm). Retention time: 3.4 min. Limit of quantification, 5 mg/L [Fels et al. 1984]. Serum GC-MS Column: (5%-phenyl)-methylpolysiloxane capillary (30 m 0.25 mm, 0.5 mm). Carrier gas: He, 1.0 mL/min. Temperature programme: 80 for 4 min to 290 at 25 /min for 9.6 min. EI ionisation at 70 eV, positive ion mode, SIM acquisition mode. Retention time: 19.1 min (methyl tert-butyl ether derivative). Limit of quantification not reported [Dargan et al. 2008]. Urine HPLC Column: LiChrosorb Si 60 (125 4.0 mm i.d., 5 mm). Mobile phase: n-hexane : methanol : tetrahydrofuran : diethylamine (88.5 : 7.5 : 4 : 0.15), flow rate 1.5 mL/min. Fluorescence detection (lex ¼ 310 nm, lem ¼ 340 nm). Retention time: 3.4 min. Limit of quantification, 2 mg/L [Fels et al. 1984]. GC-MS Column: (5%-phenyl)-methylpolysiloxane capillary (30 m 0.25 mm, 0.5 mm). Carrier gas: He, 1.0 mL/min. Temperature programme: 80 for 4 min to 290 at 25 /min for 9.6 min. EI ionisation at 70 eV, positive ion mode, SIM acquisition mode. Retention time: 19.1 min (methyl tert-butyl ether derivative). Limit of quantification not reported [Dargan et al. 2008]. Other TLC Plant Samples (Thalictrum flavum). Plates: silica gel GF254. Solvent system: petroleum ether : chloroform : acetone : methanol (4 : 4 : 1 : 1). Location reagent Dragendorff’s reagent. Rf values: glaucine 0.71, thalidazine 0.38, thaliglucine 0.35. Limit of quantification not reported [Velcheva et al. 1992]. GC-MS Plant Samples (members of the genus Sarcocapnos). Column: HP-1 fused silica capillary (12 m 0.2 mm i.d., 0.33 mm). Carrier gas: He, 1.0 mL/min.
Antidiabetic C23H28ClN3O5S = 494.0 CAS—10238-21-8 IUPAC Name 5-Chloro-N-[2-[4-[[[(cyclohexylamino)carbonyl]amino]sulfonyl]-phenyl]ethyl]-2-methoxybenzamide Synonyms Glybenclamide; glyburide; glycbenzcyclamide. Proprietary Names Azuglucon; Basstiverit; Calabren; Daonil; DiaBeta; Diabetamide; Diacon; Euglucan; Euglucon; Glib; Gliben; Glibenbeta; Gluconorm; Glucoremed; Glynase; Hemi-Doanil; Libanil; Malix; Maninil; Micronase; Miglucan; Semi-Daonil; Semi-Euglucon.
Chemical Properties A white crystalline powder. Mp 172 to 174 . Sparingly soluble in water; practically insoluble in ether; soluble 1 in 330 of ethanol, 1 in 36 of chloroform and 1 in 250 of methanol. pKa 5.3. Log P (octanol/water), 4.8. Extraction yield (chlorobutane), 0.2 [Demme et al. 2005]. Colour Tests Aromaticity (method 2)—yellow/orange; Liebermann’s reagent (100 )—orange (15 s).
G
Clarke's Analysis of Drugs and Poisons Chapter No. G Dated: 16/3/2011 At Time: 7:21:35
1460
Glibornuride
Thin-layer Chromatography System TA—Rf 0.80; system TB—Rf 0.00; system TD—Rf 0.30; system TE—Rf 0.11; system TF—Rf 0.30; system TAD—Rf 0.57; system TAE—Rf 0.90 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—glibenclamide-Me RI 3800, glibencamide-Me2 RI 3840. High Performance Liquid Chromatography System HX—RI 637; system HY—RI 571; system HZ—retention time 14.4 min; system HAA—retention time 22.0 min; system HAX—retention time 8.5 min; system HAY—retention time 9.8 min. Ultraviolet Spectrum Methanol—275, 300 nm (A11¼63a).
G
Infrared Spectrum Principal peaks at wavenumbers 1524, 1160, 1623, 1718, 1276, 823 cm1 (KBr disk).
Dose 5 to 15 mg daily. Abdel-Hamid ME et al. (1989). A rapid high-performance liquid chromatography assay of glibenclamide in serum. J Clin Pharm Ther 14: 181–188. Adams WJ et al. (1982). Determination of glyburide in human serum by liquid chromatography with fluorescence detection. Anal Chem 54: 1287–1291. Balant L (1981). Clinical pharmacokinetics of sulphonylurea hypoglycaemic drugs. Clin Pharmacokinet 6: 215–241. Balant L et al. (1975). Comparison of the pharmacokinetics of glipizide and glibenclamide in man. Eur J Clin Pharmacol 8: 63–69. Berger M et al. (1977). [Attempted suicide using glibenclamide:course of glucose, insulin, glibenclamide and C-peptide blood levels]. Dtsch Med Wochenschr 102: 586–587. Castoldi D, Tofanetti O (1979). Gas chromatographic determination of glibenclamide in plasma. Clin Chim Acta 93(2): 195–198. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Emilsson H et al. (1986). High-performance liquid chromatographic determination of glibenclamide in human plasma and urine. J Chromatogr 383: 93–102. Jackson JE, Bressler R (1981). Clinical pharmacology of sulphonylurea hypoglycaemic agents: part 1. Drugs 22: 211–245. Khatri J et al. (2001). A novel extractionless hplc fluorescence method for the determination of glyburide in the human plasma: application to a bioequivalence study. J Pharm Pharm Sci 4: 201–206. Niopas I, Daftsios AC (2002). A validated high-performance liquid chromatographic method for the determination of glibenclamide in human plasma and its application to pharmacokinetic studies. J Pharm Biomed Anal 28: 653–657. Rydberg T et al. (1991). Determination of glibenclamide and its two major metabolites in human serum and urine by column liquid chromatography. J Chromatogr 564: 223–233. Sener A et al. (1995). Standardized procedure for the assay and identification of hypoglycemic sulfonylureas in human plasma. Acta Diabetol 32: 64–68. Valdes Santurio JR, Gonzalez Porto E (1996). Determination of glibenclamide in human plasma by solid-phase extraction and high-performance liquid chromatography. J Chromatogr B Biomed Appl 682: 364–370.
Glibornuride Antidiabetic C18H26N2O4S = 366.5 CAS—26944-48-9 IUPAC Name 1-(3-Hydroxy-4,7, 7-trimethyl-2-bicyclo[2.2.1]heptanyl)-3-(4methylphenyl)sulfonylurea Synonym [1S-(endo,endo)]-N-[[(3-Hydroxy-4,7,7-trimethylbicyclo[2.2.1]hept2-yl)amino]carbonyl]-4-methylbenzenesulfonamide Proprietary Names Gluborid; Glutril.
Quantification Plasma GC ECD. Limit of detection, 5 mg/L [Castoldi, Tofanetti 1979]. HPLC Limit of detection, 99.5%. Note For a review of glimepiride, see Langtry and Balfour [1998]. Dose Usually up to 4 mg daily. Langtry HD, Balfour JA (1998). Glimepiride. A review of its use in the management of type 2 diabetes mellitus. Drugs 55: 563–584. Lehr KH, Damm P (1990). Simultaneous determination of the sulphonylurea glimepiride and its metabolites in human serum and urine by high-performance liquid chromatography after precolumn derivatization. J Chromatogr 526: 497–505. Malerczyk V et al. (1994). Dose linearity assessment of glimepiride (Amaryl) tablets in healthy volunteers. Drug Metab Drug Interact 11(4): 341–357. Ratheiser K et al. (1993). Dose relationship of stimulated insulin production following intravenous application of glimepiride in healthy man. Arzneimittelforschung 43: 856–858.
Glipizide
Infrared Spectrum
Antidiabetic C21H27N5O4S = 445.5 CAS—29094-61-9 IUPAC Name N-[2-[4-[[[(Cyclohexylamino)carbonyl]amino]sulfonyl]phenyl] ethyl]-5-methylpyrazinecarboxamide Synonyms Glipizidum; glydiazinamide; K-4024; CP-28720. Proprietary Names Apamid; Diasef; Dipazide; Glibenese; Glidiab; Glipid; Glipiscand; Gluco-Rite; Glucotrol; Glupital; Glygen; Melizid(e); Mindiab; Minidiab; Minodiab; Ozidia.
Chemical Properties A white powder. Mp about 205 . Practically insoluble in water and ethanol; soluble in chloroform, dimethylformamide and dilute solutions of alkali hydroxides; sparingly soluble in acetone. pKa 5.9. Log P (octanol/ water), 1.9. Colour Test Mercurous nitrate—black. Thin-layer Chromatography System TA—Rf 0.87; system TB—Rf 0.00; system TC—Rf 0.41; system TE—Rf 0.07; system TL—Rf 0.05; system TAE— Rf 0.86. Gas Chromatography System GA—glipizide-Me RI 3420, glipizide-Me2 RI 3455. High Performance Liquid Chromatography System HX—RI 478; system HY—RI 423; system HZ—retention time 4.5 min; system HAA—retention time 17.6 min. Ultraviolet Spectrum Aqueous acid—276 nm (A11¼231a). No alkaline shift.
Clarke's Analysis of Drugs and Poisons Chapter No. G Dated: 16/3/2011 At Time: 7:21:36
Gliquidone
1463
Maggi E et al. (1981). Radioimmunoassay of glipizide in human plasma. Eur J Clin Pharmacol 21: 251–255. Sener A et al. (1995). Standardized procedure for the assay and identification of hypoglycemic sulfonylureas in human plasma. Acta Diabetol 32: 64–68. Wahlin-Boll E, Melander A (1979). High-performance liquid chromatographic determination of glipizide and some other sulfonylurea drugs in serum. J Chromatogr 164: 541–546. Wahlin-Boll E et al. (1982). Bioavailability, pharmacokinetics and effects of glipizide in type 2 diabetics. Clin Pharmacokinet 7: 363–372.
Gliquidone Antidiabetic C27H33N3O6S = 527.6 CAS—33342-05-1 IUPAC Name 1-Cyclohexyl-3-[4-[2-(7-methoxy-4,4-dimethyl-1,3-dioxoisoquinolin-2-yl)ethyl]phenyl]sulfonylurea Synonym N-[(Cyclohexylamino)carbonyl]-4-[2-(3,4-dihydro-7-methoxy-4,4dimethyl-1,3-dioxo-2(1H)-isoquinolinyl)ethyl]benzenesulfonamide Proprietary Names Glurenor; Glurenorm. Infrared Spectrum Principal peaks at wavenumbers 1528, 1690, 1650, 1159, 1032, 900 cm1 (KBr disk).
G
Chemical Properties A white or slightly yellow crystalline substance. Mp about 178 . Practically insoluble in water; slightly soluble in ethanol and methanol; soluble in acetone and chloroform. Log P (octanol/water), 4.6. Colour Test Koppanyi–Zwikker test—violet. Thin-layer Chromatography System TAE—Rf 0.93. Gas Chromatography System GA—gliquidone RI 2024, gliquidone-Me RI 3850. High Performance Liquid Chromatography System HY—RI 667. Ultraviolet Spectrum Methanol—311 nm (A11¼50a); aqueous alkali—276 nm.
Mass Spectrum Principal ions at m/z 150, 121, 56, 93, 39, 151, 66, 94.
Quantification Plasma HPLC Glipizide and other sulfonylureas. Limit of detection, 10 to 40 mg/L [Sener et al. 1995]. UV detection. Limit of detection, 5 to 10 mg/L [Emilsson 1987]. Radioimmunoassay Limit of detection, 1 mg/L [Maggi et al. 1981]. Serum HPLC UV detection. Limit of detection, 20 mg/L [Wahlin-Boll, Melander 1979]. Urine HPLC See Plasma [Emilsson 1987]. Disposition in the Body Readily absorbed after oral administration. Metabolised by hydroxylation to form a number of inactive metabolites, principally the 4-transhydroxycyclohexyl and 3-cis-hydroxycyclohexyl derivatives. About 65 to 85% of a dose is excreted in the urine in 24 h, with about 3 to 10% as unchanged drug, up to 80% as hydroxylated metabolites, mainly the 4-trans-hydroxycyclohexyl derivative, and about 1 to 2% as an N-acetamido metabolite; about 11% of a dose is eliminated in the faeces. Therapeutic Concentration After a single oral dose of 5 mg given to 6 subjects, peak serum concentrations of 0.11 to 0.49 mg/L (mean 0.33) were attained in about 1.6 h [Wahlin-Boll et al. 1982]. Bioavailability Almost 100%. Half-life Plasma half-life, 2 to 4 h. Volume of Distribution About 0.2 L/kg. Clearance Plasma clearance, about 0.6 mL/min/kg. Protein Binding About 98%. Note For a review of the pharmacokinetics of glipizide, see Brogden et al. [1979]. Dose Usually 2.5 to 20 mg daily; up to 40 mg have been used. Brogden RN et al. (1979). Glipizide: a review of its pharmacological properties and therapeutic use. Drugs 18: 329–353. Emilsson H (1987). High-performance liquid chromatographic determination of glipizide in human plasma and urine. J Chromatogr 421: 319–326.
Infrared Spectrum Principal peaks at wavenumbers 1700, 1652, 1160, 1285, 1295, 1530 cm1 (KBr disk).
Clarke's Analysis of Drugs and Poisons Chapter No. G Dated: 16/3/2011 At Time: 7:21:37
1464
Gloxazone
Quantification Plasma HPLC Gliquidone and other sulfonylureas. Limit of detection, 10 to 40 mg/L [Sener et al. 1995]. UV detection. Limit of detection, 30 mg/L [Guo et al. 1992]. Radioimmunoassay Limit of detection, 1 mg/L [Kopitar, Kompa 1975]. Disposition in the Body Readily absorbed after oral administration. Extensively metabolised by hydroxylation and demethylation to inactive metabolites. Less than 5% of a dose is excreted in the urine and about 95% is eliminated in the faeces, via the bile. Therapeutic Concentration After a single oral dose of 15 mg to 10 subjects, peak plasma concentrations of about 0.7 mg/L, and peak blood concentrations of about 0.37 mg/L were reported [Kopitar 1975]. In 20 subjects, a single 30-mg oral dose produced peak plasma concentrations of 0.12 to 2.14 mg/L (mean 0.65) at 1.25 to 4.75 h (2.25) [von Nicolai et al. 1997]. Half-life Plasma half-life, about 1.5 h. Distribution in Blood Plasma:whole blood ratio, about 1.9. Protein Binding About 99%. Dose 15 to 180 mg daily.
G
Guo P et al. (1992). [Direct injection of plasma to determine gliquidone in plasma using HPLC column switching technique]. Yao Xue Xue Bao 27: 452–455. Kopitar Z (1975). [Human pharmacokinetics and metabolism of 14C-labeled gliquidone (AR-DF 26)]. Arzneimittelforschung 25: 1455–1460. Kopitar Z, Kompa HE (1975). [Radioimmuno assay of the sulfonylurea AR-DF 26 (author’s transl)]. Arzneimittelforschung 25: 1469–1472. Sener A et al. (1995). Standardized procedure for the assay and identification of hypoglycemic sulfonylureas in human plasma. Acta Diabetol 32: 64–68. von Nicholai H et al. (1997). Duration of action and pharmacokinetics of the oral antidiabetic drug gliquidone in patients with non-insulin-dependent (type 2) diabetes mellitus. Arzneimittelforschung 47: 247–252.
Gloxazone
Glutethimide Hypnotic C13H15NO2 = 217.3 CAS—77-21-4 IUPAC Name 3-Ethyl-3-phenyl-2,6-piperidinedione Synonyms Glutethimidum; glutetimide. Proprietary Name Doriden(e)
Chemical Properties Colourless crystals or white crystalline powder. Mp 85 to 89 . Practically insoluble in water; soluble 1 in 5 of ethanol, 1 in less than 1 of chloroform and 1 in 12 of ether; freely soluble in acetone. pKa 9.2. Log P (octanol/ water), 1.9. Extraction yield (chlorobutane), 1 [Demme et al. 2005]. Colour Tests Koppanyi–Zwikker test—violet; Liebermann’s reagent—redorange; mercurous nitrate—black. Thin-layer Chromatography System TA—Rf 0.75; system TB—Rf 0.31; system TD—Rf 0.63; system TE—Rf 0.80; system TF—Rf 0.62; system TAD—Rf 0.70; system TAE—Rf 0.86; system TAF—Rf 0.89 (Dragendorff spray, weak reaction; acidified iodoplatinate solution, positive; mercuric chloride–diphenylcarbazone reagent, positive; mercurous nitrate spray, black). Gas Chromatography System GA—glutethimide RI 1830, M (OH-ethyl-) RI 1865, M (OH-phenyl-) RI 1875, M (OH-ethyl-)-AC RI 2060, M (OH-phenyl-)-AC RI 2250; system GB—glutethimide RI 1910, M (OH-ethyl-) RI 1958, M (OHphenyl-) RI 2040; system GF—RI 2315. High Performance Liquid Chromatography System HE—k 7.97; system HX—RI 436; system HY—RI 401; system HZ—retention time 4.8 min; system HAX—retention time 6.6 min; system HAY—retention time 6.2 min. Ultraviolet Spectrum Ethanol—252, 258 (A11¼18a), 264 nm.
Anaplasmodastat (Veterinary) C8H16N6OS2 = 276.4 CAS—2507-91-7 IUPAC Name [(E)-[(1E)-1-(Carbamothioylhydrazinylidene)-3-ethoxybutan-2ylidene] amino]thiourea Synonym 2,20 -[1-(1-Ethoxyethyl)-1,2-ethanediylidene] bishydrazinecarbothioamide
Chemical Properties A yellow powder. Practically insoluble in water and ether; slightly soluble in chloroform. Colour Test Palladium chloride—red. Thin-layer Chromatography System TA—Rf 0.77 (acidified iodoplatinate solution, positive). Ultraviolet Spectrum Aqueous acid—329 nm; aqueous alkali—303 nm.
Infrared Spectrum Principal peaks at wavenumbers 1587, 1079, 1228, 1259, 1575, 1022 cm1 (KBr disk).
Infrared Spectrum Principal peaks at wavenumbers 1686, 1710, 1200, 1270, 1281, 704 cm1 (KBr disk).
Mass Spectrum Principal ions at m/z 189, 132, 117, 160, 91, 115, 103, 77; 4hydroxyglutethimide 146, 233, 103, 133, 91, 117, 115, 77; 2-phenylglutarimide 104, 189, 103, 117, 78, 91, 51, 146.
Clarke's Analysis of Drugs and Poisons Chapter No. G Dated: 16/3/2011 At Time: 7:21:37
Glyceryl Trinitrate
1465
Proprietary Names Coro-Nitro; Deponit; Glytrin; Minitran; Nitrek; Nitro-Bid; Nitrocine; Nitro-Derm; Nitrodisc; Nitro-Dur; Nitrogard; Nitroglyn; Nitrol; Nitrolingual; Nitromin; Nitrotime; Nitronal; Nitrong; NitroQuick; Nitrostat; NitroTab; NTS; Percutol; Suscard; Sustac; Transdermal-NTG; Transderm-Nitro; Transiderm-Nitro; Tridil.
Quantification Plasma GC FID. Glutethimide and 4-hydroxyglutethimide. Limit of detection, 200 mg/L for glutethimide and 500 mg/L for 4-hydroxyglutethimide [Hansen, Fischer 1974]. GC-MS Glutethimide and six metabolites. Limit of detection, 50 mg/L [Kennedy et al. 1978]. Serum GC FID. Limit of detection, 250 mg/L [Kadar, Kalow 1972]. Urine GC See Plasma [Hansen, Fischer 1974]. GC-MS See Plasma [Kennedy et al. 1978]. Tissues GC See Plasma [Hansen, Fischer 1974]. Disposition in the Body Irregularly absorbed from the gastrointestinal tract; absorption is enhanced if alcohol is taken concomitantly. Widely distributed in body tissues and fat. Glutethimide is a racemate; the (þ)-isomer is metabolised in the liver to 4-hydroxyglutethimide and the ()-isomer to 2-(1-hydroxyethyl)-2-phenylglutarimide. 4-Hydroxyglutethimide is twice as active as glutethimide in animals but does not appear to contribute to the effects of single therapeutic doses in humans; however it tends to accumulate in the plasma during intoxication and may contribute to the central nervous depression in overdose cases. Both hydroxylated metabolites are converted to glucuronides and undergo enterohepatic circulation; they are still being excreted in the urine after >48 h. 2-Ethyl-2-(4hydroxyphenyl)glutarimide is a major metabolite during chronic administration. Other metabolites include 2-phenylglutarimide, a-phenyl-g-butyrolactone and 2ethyl-2-phenylglutaconimide, which are pharmacologically active; numerous other mono- and dihydroxyphenyl metabolites have been isolated from human urine. 2 mg/L, although recovery has occurred after development of plasma concentrations up to 90 mg/L.
Disposition in the Body Hexaconazole is rapidly excreted from the body with no significant retention of the substance in the body. Toxicity Hexaconazole is moderately toxic by ingestion and via skin exposure.
Hexamethonium Bromide Antihypertensive C12H30Br2N2 = 362.2 CAS—60-26-4 (hexamethonium); 55-97-0 (dibromide) IUPAC Name N,N,N,N0 ,N0 ,N0 -Hexamethyl-1,6-hexanediaminium dibromide Synonyms Hexamethone bromide; hexonium bromide. Proprietary Names Esametina; Gangliostat; Simpatoblock; Vegolysen; Vegolysin.
Chemical Properties A white or creamy-white hygroscopic powder. Mp 274 to 276 . Soluble 1 in less than 1 of water and 1 in 60 of ethanol; insoluble in acetone, chloroform and ether.
H
Clarke's Analysis of Drugs and Poisons Chapter No. H Dated: 16/3/2011 At Time: 7:37:37
1482
Hexestrol
Hexamethonium Iodide
Hexethal
C12H30I2N2 = 456.2 CAS—870-62-2 Synonym Hexonium iodide Proprietary Name Hexathide Chemical Properties A white, slightly hygroscopic, crystalline powder. Soluble 1
Barbiturate C12H20N2O3 = 240.3 CAS—77-30-5 IUPAC Name 5-Ethyl-5-hexyl-2,4,6(1H,3H,5H)-pyrimidinetrione
in 2 of water; practically insoluble in ethanol. Thin-layer Chromatography System TA—Rf 0.00; system TN—Rf 0.36; system TO—Rf 0.10 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—not eluted. Ultraviolet Spectrum No significant absorption, 230 to 360 nm. Infrared Spectrum Principal peaks at wavenumbers 913, 970, 1630, 945, 1063, 1000 cm1 (KBr disk). Disposition in the Body Toxicity A healthy subject taking part in a pharmacological trial died after receiving about 1 g of hexamethonium by inhalation (a non-standard route of administration) [Ramsay 2001]. Dose Hexamethonium bromide has been given parenterally in doses of up to 500 mg daily. Ramsay S (2001). Johns Hopkins takes responsibility for volunteer’s death. Lancet 358: 213.
H
Hexestrol Oestrogen C18H22O2 = 270.4 CAS—5635-50-7; 84-16-2 (meso) IUPAC Name 4-[4-(4-Hydroxyphenyl)hexan-3-yl]phenol Synonyms Dihydrostilboestrol; 4,40 -(1,2-diethyl-1,2-ethanediyl)bisphenol; hexanoestrol; hexoestrol; synestrol; synoestrol. Proprietary Names Cycloestrol; Hormoestrol (tablets); Synthovo; Syntrogene.
Chemical Properties pKa 8.0. Log P (octanol/water), 2.5. Hexethal Sodium C12H19N2NaO3 = 262.3 CAS—144-00-3 Proprietary Names Hebarel; Ortal Sodium. Chemical Properties A white or slightly yellowish powder. Mp 126 . Very soluble
in water; soluble in ethanol; insoluble in ether and benzene. Aqueous solutions are unstable and decompose on standing. Thin-layer Chromatography System TD—Rf 0.53; system TE—Rf 0.44; system TF—Rf 0.67; system TH—Rf 0.74; system TAD—Rf 0.60. Gas Chromatography System GA—hexethal RI 1850, hexethal-Me2 RI 1745. High Performance Liquid Chromatography System HG—k 34.28; system HH—k 20.39; system HY—RI 451. Ultraviolet Spectrum Borax buffer 0.05 mol/L (pH 9.2)—238 (A11¼423b); 1 mol/L sodium hydroxide (pH 13)—253 nm (A11¼323b).
Chemical Properties Colourless crystals or white crystalline powder. Mp 185 to 188 . Practically insoluble in water and dilute mineral acids; freely soluble in ether; soluble in ethanol and acetone; slightly soluble in benzene and chloroform. Log P (octanol/water), 5.6. Hexestrol Dipropionate C24H30O4 = 382.5 CAS—4825-53-0 Proprietary Names Hormoestrol (injection); Retalon Oleosum. Chemical Properties A white crystalline powder. Mp 127 to 128 . Sparingly
soluble in water; soluble in warm ethanol and ether. Thin-layer Chromatography System TB—Rf 0.02; system TE—Rf 0.73; system TF—Rf 0.70; system TAE—Rf 0.88. Gas Chromatography System GA—RI 2402. High Performance Liquid Chromatography System HX—RI 618. Ultraviolet Spectrum Ethanol—230 (A11¼775b), 280 nm (A11¼135a); aqueous alkali—242 (A11¼965b), 297 nm (A11¼175b).
Infrared Spectrum Principal peaks at wavenumbers 1175, 1523, 1220, 840, 857, 1615 cm1 (KBr disk). Dose Hexestrol has been given in doses of 1 to 5 mg daily.
Infrared Spectrum Principal peaks at wavenumbers 1698, 1720, 1757, 1316, 1230, 850 cm1 (hexethal sodium).
Clarke's Analysis of Drugs and Poisons Chapter No. H Dated: 16/3/2011 At Time: 7:37:38
Hexobarbital Mass Spectrum Principal ions at m/z 156, 141, 55, 41, 157, 43, 98, 39.
1483
(mercuric chloride–diphenylcarbazone reagent, positive; mercurous nitrate spray, black; acidified potassium permanganate solution, yellow-brown on violet). Gas Chromatography System GA—hexobarbital RI 1855, M (1,5-dimethyl-5(3-oxo-1-cyclohexen-1-yl)barbituric acid) RI 2050, M (nor-) RI 1980, M (30 -oxo-) RI 2055, M (OH-)-H2O RI 1970, hexobarbital-Me RI 1800, M (oxo-)-Me RI 2020; system GF—RI 2380; system GAJ—hexobarbital RRT 0.940 (relative to methylphenobarbital). High Performance Liquid Chromatography System HG—k 7.37; system HH—k 5.67; system HX—RI 419; system HY—RI 242; system HZ—retention time 4.3 min; system HAL—retention time 2.4 min. Ultraviolet Spectrum Borax buffer 0.05 mol/L (pH 9.2)—243 nm (A11¼331a); 1 mol/L sodium hydroxide (pH 13) —243 nm (A11¼301b).
Hexetidine Antimicrobial C21H45N3 = 339.6 CAS—141-94-6 IUPAC Name 1,3-Bis(2-ethylhexyl)hexahydro-5-methyl-5-pyrimidinamine Proprietary Names Bactidol; Collu-Hextril; Doreperol N; Drossadin; Gurfix; Hexatin; Hexifluor; Hexigel; Hexoral; Hextril; Isozid-H; Kleenosept; Oraldene; Oraldine; Oraseptic; Oralspray; Steri/Sol; Vagi-Hex.
H Chemical Properties A viscous oil. Mass per mL about 0.87 g. Refractive index 1.466. Practically insoluble in water; soluble in ethanol, acetone, chloroform, petroleum ether and benzene. pKa 8.3. Log P (octanol/water), 5.3. Thin-layer Chromatography System TA—Rf 0.70; system TB—Rf 0.48; system TC—Rf 0.40; system TE—Rf 0.79; system TL—Rf 0.20; system TAE—Rf 0.30; system TAF—Rf 0.91 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—RI 2093. Ultraviolet Spectrum No significant absorption, 230 to 360 nm. Infrared Spectrum Principal peaks at wavenumbers 1093, 854, 1300, 910, 1176, 1234 cm1 (thin film). Mass Spectrum Principal ions at m/z 142, 57, 42, 197, 185, 339, 240, 226. Quantification Saliva HPLC [McCoy et al. 2000]. Use As a 0.1% solution.
Infrared Spectrum Principal peaks at wavenumbers 1720, 1665, 1748, 1200, 1275, 1045 cm1 (KBr disk).
McCoy CP et al. (2000). Determination of the salivary retention of hexetidine in-vivo by highperformance liquid chromatography. J Pharm Pharmacol 52: 1355–1359.
Hexobarbital Sedative, Barbiturate C12H16N2O3 = 236.3 CAS—56-29-1 IUPAC Name 5-(Cyclohexen-1-yl)-1,5-dimethyl-1,3-diazinane-2,4,6-trione Synonyms Ciclobarbital; 5-(1-cyclohexen-1-yl)-1,5-dimethyl-2,4,6(1H,3H,5H)pyrimidinetrione; enhexymalum; enimal; hexobarbitalum; hexobarbitone; methexenyl; methyl-cyclohexenylmethyl-barbiturs€aure; methylhexabarbital. Proprietary Names Noctivane; Sombulex. Mass Spectrum Principal ions at m/z 221, 81, 157, 80, 79, 155, 41, 77; 30 oxohexobarbital 250, 95, 39, 235, 66, 207, 41, 193.
Chemical Properties Colourless crystals or a white crystalline powder. Mp 145 to 147 . Practically insoluble in water; soluble 1 in 45 of ethanol, 1 in 4 of chloroform and 1 in 80 of ether; soluble in acetone, benzene and methanol. pKa 8.2 (20 ). Log P (octanol/water), 2.0. Hexobarbital Sodium C12H15N2NaO3 = 258.3 CAS—50-09-9 Synonyms Enhexymalnatrium; hexenalum; sodium hexobarbitone; soluble
hexobarbital. Proprietary Names Evipan-Natrium; Noctivane Sodium. Chemical Properties A white, very hygroscopic powder. Discolours on exposure to
air. Very soluble in water; freely soluble in ethanol, acetone and methanol; practically insoluble in chloroform, ether and benzene. A solution in water slowly decomposes. Colour Tests Koppanyi–Zwikker test—violet; mercurous nitrate—black; vanillin reagent—brown/violet. Thin-layer Chromatography System TD—Rf 0.65; system TE—Rf 0.53; system TF—Rf 0.65; system TH—Rf 0.85; system TAD—Rf 0.69; system TAE—Rf 0.85
Quantification See also under Amobarbital. Plasma GC-MS Hexobarbital enantiomers and metabolites [Prakash et al. 1991]. Urine GC-MS See Plasma [Prakash et al. 1991]. Disposition in the Body Readily absorbed after oral administration. The sodium salt has a very short duration of action and is usually administered intravenously. Hexobarbital is inactivated in the liver by N-demethylation and oxidation. About 32% of a dose is excreted in the urine in 24 h as 30 -oxohexobarbital, 5% as 30 hydroxyhexobarbital and 18% as 1,5-dimethylbarbituric acid; 0.1 mg/L. In 2 deaths caused by the ingestion of hydrocodone and phenyltoloxamine, the following postmortem concentrations were reported for the 2 cases, respectively: hydrocodone 0.3 mg/L in blood (both cases); hydrocodone 14.3 mg/L and none in bile, hydromorphone 98 and 48 mg/L in bile; in the first case, phenyltoloxamine was present in bile at a concentration of 0.4 mg/L [Park et al. 1982]. Half-life Plasma half-life, ~4 h. Dose Usually 5 to 10 mg hydrocodone tartrate by mouth every 4 to 6 h. Balikova M et al. (2000). [Evaluation of methods of trace analysis of various opiates including hydrocodone and hydromorphone in the blood and urine using gas chromatography–mass spectrometry]. Soud Lek 45: 11–16. Barnhart JW, Caldwell WJ (1977). Gas chromatographic determination of hydrocodone in serum. J Chromatogr 130: 243–249. Broussard LA et al. (1997). Simultaneous identification and quantitation of codeine, morphine, hydrocodone, and hydromorphone in urine as trimethylsilyl and oxime derivatives by gas chromatography–mass spectrometry. Clin Chem 43: 1029–1032. Cimbura G, Koves E (1981). Radioimmunological screening and gas chromatographic determination of morphine and related narcotic analgesics in post mortem blood. J Anal Toxicol 5: 296–299. Coles R et al. (2007). Simultaneous determination of codeine, morphine, hydrocodone, hydromorphone, oxycodone, and 6-acetylmorphine in urine, serum, plasma, whole blood, and meconium by LC-MS-MS. J Anal Toxicol 31: 1–14. Cone EJ, Darwin WD (1978). Simultaneous determination of hydromorphone, hydrocodone and their 6alpha- and 6beta-hydroxy metabolites in urine using selected ion recording with methane chemical ionization. Biomed Mass Spectrom 5: 291. Dahn T et al. (2010). Quantitation of morphine, codeine, hydrocodone, hydromorphone, oxycodone, oxymorphone, and 6-monoacetylmorphine (6-MAM) in urine, blood, serum, or plasma using liquid chromatography with tandem mass spectrometry detection. Methods Mol Biol 603: 411–422. Goldberger BA et al. (2010). Quantitation of opioids in blood and urine using gas chromatography– mass spectrometry (GC-MS). Methods Mol Biol 603: 399–410. Honigberg IL, Stewart JT (1980). Radioimmunoassay of hydromorphone and hydrocodone in human plasma. J Pharm Sci 69: 1171–1173. Jones J et al. (2002). The simultaneous determination of codeine, morphine, hydrocodone, hydromorphone, 6-acetylmorphine, and oxycodone in hair and oral fluid. J Anal Toxicol 26: 171–175. Meatherall R (1999). GC-MS confirmation of codeine, morphine, 6-acetylmorphine, hydrocodone, hydromorphone, oxycodone, and oxymorphone in urine. J Anal Toxicol 23: 177–186. Park JI et al. (1982). Hydromorphone detected in bile following hydrocodone ingestion. J Forensic Sci 27: 223–224. Sweetman S, ed. (2007). Martindale: The Complete Drug Reference, 35 edn. London: Pharmaceutical Press. Zhang R et al. (2009). Determination and pharmacokinetic study of hydrocodone in human plasma by liquid chromatography coupled with tandem mass spectrometry. Artif Cells Blood Substit Immobil Biotechnol 37: 203–207.
Hydrocortisone Corticosteroid C21H30O5 = 362.5 CAS—50-23-7 IUPAC Name (8S,9S,10R,11S,13S,14S,17R)-11,17-Dihydroxy-17-(2-hydroxyacetyl)-10,13-dimethyl-2,6,7,8,9,11,12,14,15, 16-decahydro-1H-cyclopenta[a]phenanthren-3-one Synonyms Compound F; cortisol; 17-hydroxycorticosterone; (11b)-11,17,21trihydroxypregn-4-ene-3,20-dione. Note Cortisol is also used as a proprietary name for cortisone acetate. Proprietary Names Hydrocortisone and its esters are ingredients of many proprietary preparations—see Sweetman [2009].
Chemical Properties A white crystalline powder. Mp about 214 , with decomposition. A solution in dioxan is dextrorotatory. Practically insoluble in water and
1495
ether; soluble 1 in 40 of ethanol; slightly soluble in chloroform; very soluble in dioxan. Hydrocortisone sodium succinate, pKa 5.1. Log P (octanol/water), 1.6. Hydrocortisone Acetate C23H32O6 = 404.5 CAS—50-03-3 Synonym Hydrocortisone 21-acetate Chemical Properties A white crystalline powder. Mp about 220 , with decom-
position. Practically insoluble in water and ether; soluble 1 in 230 of ethanol; slightly soluble in chloroform; soluble in dioxane. Hydrocortisone Butyrate C25H36O6 = 432.6 CAS—13609-67-1 Synonym Hydrocortisone 17-butyrate
Hydrocortisone Cipionate C29H42O6 = 486.6 CAS—508-99-6 Synonyms Hydrocortisone cypionate; hydrocortisone cyclopentylpropionate. Chemical Properties A white crystalline powder. Practically insoluble in water;
soluble in ethanol; very soluble in chloroform; slightly soluble in ether. Hydrocortisone Hydrogen Succinate C25H34O8 = 462.5 CAS—2203-97-6 Synonyms Cortisol hemisuccinate; hydrocortisone hemisuccinate. Chemical Properties A white crystalline powder. Mp 170 to 173 or 210 to
214 . Practically insoluble in water; soluble 1 in 40 of ethanol and 1 in 7 of dehydrated alcohol. Hydrocortisone Sodium Phosphate
C21H29Na2O8P = 486.4 CAS—6000-74-4 Chemical Properties A white or light yellow hygroscopic powder. Soluble 1 in 4
of water; slightly soluble in ethanol; practically insoluble in chloroform and ether. Hydrocortisone Sodium Succinate C25H35NaO8 = 484.5 CAS—125-04-2 Synonym Hydrocortisone 21-sodium succinate Chemical Properties A white, hygroscopic, crystalline powder or amorphous
solid. Mp 169.0 to 171.2 . Soluble 1 in 3 of water and 1 in 34 of ethanol; practically insoluble in chloroform and ether. It is unstable in aqueous solution. Hydrocortisone Valerate C26H38O6 = 446.6 CAS—57524-89-7 Synonym Hydrocortisone 17-valerate
Colour Tests Antimony pentachloride—orange; naphthol–sulfuric acid—yellow-brown/yellow-brown; sulfuric acid—green, orange dichroism (green fluorescence under ultraviolet light). Thin-layer Chromatography Hydrocortisone: system TA—Rf 0.96; system TB—Rf 0.00; system TE—Rf 0.45; system TF—Rf 0.28; system TP—Rf 0.27; system TQ—Rf 0.02; system TR—Rf 0.08; system TS—Rf 0.00; system TAE—Rf 0.86; system TAJ—Rf 0.36; system TAK—Rf 0.05; system TAL—Rf 0.74; system TAM— Rf 0.58. Hydrocortisone acetate: system TP—Rf 0.51; system TQ—Rf 0.11; system TR—Rf 0.38; system TS—Rf 0.00. Hydrocortisone hydrogen succinate: system TP— Rf 0.08; system TQ—Rf 0.00; system TR—Rf 0.00; system TS—Rf 0.00. Hydrocortisone sodium phosphate: system TP—Rf 0.00; system TQ—Rf 0.00; system TR—Rf 0.00; system TS—Rf 0.00 (DPST solution). Gas Chromatography System GA—RI 2740. High Performance Liquid Chromatography System HT—k 5.8; system HX—RI 403; system HY—RI 349; system HAA—retention time 17.7 min. Ultraviolet Spectrum Ethanol—240 nm (A11¼435a).
Infrared Spectrum Principal peaks at wavenumbers 1640, 1702, 1610, 1232, 1042, 1115 cm1 (KBr disk).
H
Clarke's Analysis of Drugs and Poisons Chapter No. H Dated: 16/3/2011 At Time: 7:37:47
1496
Hydroflumethiazide IUPAC Name 3,4-Dihydro-6-(trifluoromethyl)-2H-1,2,4-benzothiadiazine-7sulfonamide 1,1-dioxide Synonym Trifluoromethylhydrothiazide Proprietary Names Diucardin; Elodrine; Finuret; Hydol; Hydrenox; Leodrine; Rodiuran; Rontyl; Saluron; Sisuril. It is an ingredient of Aldactide, Protensin-M, Rautrax, Salutensin and Spio-Co.
H Quantification Plasma HPLC Fluorescence. Hydrocortisone and other glucocorticoids. Limit of detection, 0.1 mg/L for hydrocortisone [Shibata et al. 1998]. UV detection. Hydrocortisone, prednisone and prednisolone. Limit of detection, 0.1 mg/L may be toxic. In a fatality attributed to hydromorphone overdose, the following tissue concentrations were reported: blood 1.2 mg/L, brain a trace, kidney 1.2 mg/ g, liver 0.4 mg/g and urine 1.1 mg/L; diazepam was also detected [Baselt 1978]. The following postmortem tissue concentrations were reported in a fatality caused by an injected overdose of hydromorphone: blood 0.17 mg/L, bile 8.6 mg/L, kidney 0.13 mg/g and liver 0.07 mg/g [Walls 1976]. Half-life Plasma half-life, ~2.5 h. Bioavailability Approximately 50% but there is considerable intersubject variation. Volume of Distribution ~3 L/kg. Protein Binding ~7%. Dose Hydromorphone hydrochloride 1.3 to 4 mg orally every 4 to 6 h. Al Asmari AI, Anderson RA (2007). Method for quantification of opioids and their metabolites in autopsy blood by liquid chromatography–tandem mass spectrometry. J Anal Toxicol 31: 394–408. Angst MS et al. (2001). Pharmacodynamics of orally administered sustained- release hydromorphone in humans. Anesthesiology 94: 63–73. Balikova M et al. (2000). [Evaluation of methods of trace analysis of various opiates including hydrocodone and hydromorphone in the blood and urine using gas chromatography–mass spectrometry]. Soud Lek 45: 11–16. Baselt RC (1978). TIAFT Bull 1420. Bouquillon AI et al. (1992). Simultaneous solid-phase extraction and chromatographic analysis of morphine and hydromorphone in plasma by high-performance liquid chromatography with electrochemical detection. J Chromatogr 577: 354–357. Broussard LA et al. (1997). Simultaneous identification and quantitation of codeine, morphine, hydrocodone, and hydromorphone in urine as trimethylsilyl and oxime derivatives by gas chromatography–mass spectrometry. Clin Chem 43: 1029–1032. Cimbura G, Koves E (1981). Radioimmunological screening and gas chromatographic determination of morphine and related narcotic analgesics in post mortem blood. J Anal Toxicol 5: 296–299.
Clarke's Analysis of Drugs and Poisons Chapter No. H Dated: 16/3/2011 At Time: 7:37:50
Hydroquinine Cone EJ et al. (1977). Urinary excretion of hydromorphone and metabolites in humans, rats, dogs, guinea pigs, and rabbits. J Pharm Sci 66: 1709–1713. Dahn T et al. (2010). Quantitation of morphine, codeine, hydrocodone, hydromorphone, oxycodone, oxymorphone, and 6-monoacetylmorphine (6-MAM) in urine, blood, serum, or plasma using liquid chromatography with tandem mass spectrometry detection. Meth Mol Biol 603: 411–422. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid–liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Honigberg IL, Stewart JT (1980). Radioimmunoassay of hydromorphone and hydrocodone in human plasma. J Pharm Sci 69: 1171–1173. Jones J et al. (2002). The simultaneous determination of codeine, morphine, hydrocodone, hydromorphone, 6-acetylmorphine, and oxycodone in hair and oral fluid. J Anal Toxicol 26: 171–175. Lee JW et al. (1991). Sensitive and specific radioimmunoassays for opiates using commercially available materials. I: Methods for the determinations of morphine and hydromorphone. J Pharm Sci 80: 284–288. Reidenberg MM et al. (1988). Hydromorphone levels and pain control in patients with severe chronic pain. Clin Pharmacol Ther 44: 376–382. Saady JJ et al. (1982). Rapid, simultaneous quantification of morphine, codeine, and hydromorphone by GC/MS. J Anal Toxicol 6: 235–237. Sawyer WR et al. (1988). Heroin, morphine, and hydromorphone determination in postmortem material by high performance liquid chromatography. J Forensic Sci 33: 1146–1155. Vallner JJ et al. (1981). Pharmacokinetics and bioavailability of hydromorphone following IV and oral administration to human subjects. J Clin Pharmacol 21: 152–156. Walls HC (1976). Bull TIAFT 12: 7–8.
Hydroquinidine Antiarrhythmic C20H26N2O2 = 326.4 CAS—1435-55-8 IUPAC Name (S)-[(2R, 5R)-5-Ethyl-1-azabicyclo[2.2.2]octan-2-yl]-(6-methoxyquinolin-4-yl) methanol Synonyms Dihydrochinidin; (9S)-10,11-dihydro-60 -methoxycinchonan-9-ol; dihydroquinidine; hydroconchinine.
1499
Mass Spectrum Principal ions at m/z 138, 326, 55, 110, 189, 82, 160, 139.
Quantification Plasma HPLC Fluorescence detection. Hydroquinidine, quinidine, hydroxyquinidine, and quinidine N-oxide. Limit of detection, 1 mg/g, the median ketobemidone concentrations were: blood 0.1 mg/g; liver 0.4 mg/g (after both oral and IV injection) [Steentoft, Worm 1994]. Note For fatal intoxications in the Nordic population in 1984 and 1985, see Steentoft et al. [1989b]and Steentoft et al. [1989a]. Bioavailability 40%. Half-life 2 h. Volume of Distribution 2 to 3 L/kg. Clearance 10 mL/min/kg. Dose Ketobemidone has been given in doses of 5 to 10 mg. Al-Shurbaji A, Tokics L (2002). The pharmacokinetics of ketobemidone in critically ill patients. Br J Clin Pharmacol 54: 583–586. Bondesson U et al. (1983). Simultaneous determination of ketobemidone and its N-demethylated metabolite in patient plasma samples by gas chromatography mass spectrometry with selected ion monitoring. Biomed Mass Spectrom 10: 283–286. Breindahl T, Andreasen K (1999). Validation of urine drug-of-abuse testing methods for ketobemidone using thin-layer chromatography and liquid chromatography-electrospray mass spectrometry. J Chromatogr B Biomed Sci Appl 736: 103–113. J€ onsson A et al. (2004). Fatal intoxications in a Swedish forensic autopsy material during 1992-2002. Forensic Sci Int 143: 53–59. Klinke HB, Linnet K (2007). Performance of four mixed-mode solid-phase extraction columns applied to basic drugs in urine. Scand J Clin Lab Invest 67: 778–782. Steentoft A, Worm K (1994). Cases of fatal intoxication with Ketogan. J Forensic Sci Soc 34: 181–185. Steentoft A et al. (1989a). Fatal intoxications in the age group 15-34 years in Denmark in 1984 and 1985. A forensic study with special reference to drug addicts. Z Rechtsmed 103: 93–100. Steentoft A et al. (1989b). Fatal intoxications in the Nordic countries. A forensic toxicological study with special reference to young drug addicts. Z Rechtsmed 102: 355–365. Sundstr€ om I et al. (2001). Identification of phase I and phase II metabolites of ketobemidone in patient urine using liquid chromatography-electrospray tandem mass spectrometry. J Chromatogr B Biomed Sci Appl 763: 121–131. Sundstr€ om I et al. (2002). Identification of glucuronide conjugates of ketobemidone and its phase I metabolites in human urine utilizing accurate mass and tandem time-of-flight mass spectrometry. J Mass Spectrom 37: 414–420. Svensson JO et al. (2001). Determination of ketobemidone and its metabolites in plasma and urine using solid-phase extraction and liquid chromatography-mass spectrometry. Ther Drug Monit 23: 399–405.
1543
Ketoconazole Antifungal C26H28Cl2N4O4 = 531.4 CAS—65277-42-1 IUPAC Name cis-1-Acetyl-4-[4-[[2-(2,4-dichlorophenyl)-2-(1H-imidazol-1ylmethyl)-1,3-dioxolan-4-yl]methoxy]phenyl]piperazine Synonym R-41400 Proprietary Names Daktarin Gold; Fungarest; Fungoral; Ketoderm; Ketoisdin; Nizoral; Orifungal M; Panfungol; Sebizole; Terzolin.
Chemical Properties Crystals. Mp 146 . Practically insoluble in water; soluble 1 in 54 of ethanol, 1 in about 2 of chloroform and 1 in 9 of methanol; very slightly soluble in ether. Log P (octanol/water), 4.4. Thin-layer Chromatography System TB—Rf 00; system TE—Rf 0.50; system TAE—Rf 0.68. High Performance Liquid Chromatography System HX—RI 439; system HY—RI 464; system HZ—retention time 5.2 min; system HAA—retention time 15.7 min. Ultraviolet Spectrum Aqueous acid—269 nm (A11 ¼ 26a); aqueous alkali— 287 nm (A11 ¼ 29b); methanol—244 (A11 ¼ 280b), 296 nm (A11 ¼ 32b).
K
Infrared Spectrum Principal peaks at wavenumbers 1507, 1640, 1240, 1258, 1200, 1221 cm1 (KBr disk).
Quantification Plasma HPLC Fluorescence detection. Limit of detection, 40 mg/L [Yuen, Peh 1998]. Electrochemical detection. For method, see Hoffman et al. [1988]. Fluorescence detection. Limit of detection, 40 mg/L [Pascucci et al. 1983]. UV detection. Limit of detection, 6 mg/L, and fatalities with blood concentrations >14 mg/L. In 3 deaths involving accidental ingestion of 25 g, the following postmortem tissue concentrations were reported: blood 44, 92 and 11 mg/L; brain 17, 32 and 7 mg/g; kidney 66, – and 68 mg/g; liver 70, 96 and 20 mg/g; lung 94, 130 and 49 mg/g; and urine 59, – and – mg/L; in the first 2 cases, death occurred within a few minutes of ingestion, whereas the third subject survived for 24 h [Borkowski, Dluzneiwska, 1976]. In 33 patients with cardiac disorder and given lidocaine infusions for more than 1 day, serum levels above 8 mg/L occurred in 6 of 27 patients who had no toxicity; 5 of 6 patients with toxicity had levels below 8 mg/L [Drayer et al. 1983]. In a death cause by accidental IV injection of 2 g lidocaine, the following postmortem tissue concentrations were reported: blood 30 mg/L, brain 135 mg/g, heart 106 mg/g, kidney 204 mg/g, lung 87 mg/g and skeletal muscle 20 mg/g [Poklis et al. 1984]. In 2 deaths from deliberate self-poisoning with lidocaine, the first by oral ingestion and the second by IV injection, postmortem blood analysis revealed concentrations of 40 and 53 mg/L, respectively; urine contained 49 mg/L lidocaine in the first victim [Dawling et al. 1989]. In the case of a 1-month-old infant who inadvertently received an IV injection of 50 mg lidocaine, the calculated maximum level was 5.39 mg/L [Jonville et al. 1990]. In a 32-year-old hospital patient whose death was attributed to an overdose of lidocaine (~1500 mg) administered with homicidal intent, postmortem revealed the following lidocaine concentrations: blood, 22.2 mg/L, liver 43.6 mg/g, kidney 28.3 mg/g, brain 23.1 mg/g and heart 13.1 mg/g [Kalin, Brissie 2002]. In a fatality in a patient with paroxysmal ventricular arrhythmia given an injection of 5 mL of 10% lidocaine hydrochloride (500 mg) instead of 2.5 mL of 2% (50 mg), the following concentrations were reported in postmortem tissues that had been fixed in formalin for 40 days: parietal lobe 308 ng/g, occipital lobe 208.7 ng/g, temporal lobe 318 ng/g, frontal lobe, 223.2 ng/g, cerebellum 200.9 ng/g, pons 285.7 ng/g, liver 109.5 ng/g, kidney 109.5 ng/g and skeletal muscle 127 ng/g; formalin solution contained 8.4 mg/L. Animal studies found levels in these tissues decreased to 25–33% of the original value on storage in formalin [Kudo et al. 2004]. In the case of suicide by a 31-year-old woman through oral ingestion of lidocaine, the following postmortem levels were reported: blood 31 mg/L, gastric contents 2.5 g, liver 10 mg/g, kidney 12 mg/g, brain 9 mg/g, spleen 24 mg/ g, lung 84 mg/g, heart 9 mg/g, urine 9 mg/L and bile 6 mg/L [Centini et al. 2007]. Half-life Plasma half-life, lidocaine 1–2 h, increased in subjects with liver disease or after acute myocardial infarction; monoethylglycinexylidide ~1–2 h, glycinexylidide ~10 h. Volume of Distribution Approximately 1–2 L/kg. Clearance Plasma clearance, ~5–20 mL/min/kg. Protein Binding Approximately 70% at therapeutic concentrations but there is considerable intersubject variation and binding appears to be concentrationdependent Note For a review of the pharmacokinetics of lidocaine, see Benowitz, Meister [1978]; for a review of the pharmacokinetics and pharmacodynamics of IV agents for ventricular arrhythmias, see Nolan [1997]; for a study of the pharmacokinetics of epidural lidocaine and bupivacaine during caesarean section, see Downing et al. [1997].
1575
Dose For local anaesthesia, the maximum single dose of lidocaine hydrochloride by injection should not exceed 300 mg (4.5 mg/kg), unless administered with adrenaline. For ventricular arrhythmias, initially 50–100 mg IV, followed by an infusion. Abdel-Rehim M et al. (2000). High-performance liquid chromatography–tandem electrospray mass spectrometry for the determination of lidocaine and its metabolites in human plasma and urine. J Chromatogr B Biomed Sci Appl 741: 175–188. Abraham I et al. (1997). Simultaneous analysis of lignocaine and bupivacaine enantiomers in plasma by high-performance liquid chromatography. J Chromatogr B Biomed Sci Appl 703: 203–208. Altun Z et al. (2004). New trends in sample preparation: on-line microextraction in packed syringe (MEPS) for LC and GC applications. Part III: Determination and validation of local anaesthetics in human plasma samples using a cation-exchange sorbent, and MEPS-LC-MS-MS. J Chromatogr B Analyt Technol Biomed Life Sci 813: 129–135. Baniceru M et al. (2004). Determination of some local anesthetics in human serum by gas chromatography with solid-phase extraction. J Pharm Biomed Anal 35: 593–598. Benowitz NL, Meister W (1978). Clinical pharmacokinetics of lignocaine. Clin Pharmacokinet 3: 177–201. Bo LD et al. (1999). Highly sensitive bioassay of lidocaine in human plasma by high-performance liquid chromatography–tandem mass spectrometry. J Chromatogr A 854: 3–11. Borkowski T, Dluzneiwska A (1976). A fatal case involving lidocaine. J Forensic Sci 12: 17–18. Centini F et al. (2007). Suicide due to oral ingestion of lidocaine: a case report and review of the literature. Forensic Sci Int 171: 57–62. Chen Y et al. (1992a). A quick, sensitive high-performance liquid chromatography assay for monoethylglycinexylidide and lignocaine in serum/plasma using solid-phase extraction. Ther Drug Monit 14: 317–321. Chen Y et al. (1992b). High-performance liquid chromatographic method for the simultaneous determination of monoethylglycinexylidide and lignocaine. J Chromatogr 574: 361–364. Chen L et al. (2007). Simultaneous determination of nikethamide and lidocaine in human blood and cerebrospinal fluid by high performance liquid chromatography. J Pharm Biomed Anal 43: 1757–1762. Chik Z et al. (2006). Validation of high-performance liquid chromatographic-mass spectrometric method for the analysis of lidocaine in human plasma. J Chromatogr Sci 44: 262–265. Dawling S et al. (1989). Fatal lignocaine poisoning: report of two cases and review of the literature. Hum Toxicol 8: 389–392. Demme U et al. (2005). Systematic evaluation of 1-chlorobutane for liquid-liquid extraction of drugs. Proceedings of the 12th TIAFT, Seoul: 481–486. Downing JW et al. (1997). The pharmacokinetics of epidural lidocaine and bupivacaine during cesarean section. Anesth Analg 84: 527–532. Drayer DE et al. (1983). Plasma levels, protein binding, and elimination data of lidocaine and active metabolites in cardiac patients of various ages. Clin Pharmacol Ther 34: 14–22. Fukuda T et al. (2003). Free lidocaine concentrations during continuous epidural anesthesia in geriatric patients. Reg Anesth Pain Med 28: 215–220. Gammaitoni AR, Davis MW (2002). Pharmacokinetics and tolerability of lidocaine patch 5% with extended dosing. Ann Pharmacother 36: 236–240. Gammaitoni AR et al. (2002). Pharmacokinetics and safety of continuously applied lidocaine patches 5%. Am J Health Syst Pharm 59: 2215–2220. Groeben H et al. (2000). Lidocaine inhalation for local anaesthesia and attenuation of bronchial hyper-reactivity with least airway irritation. Effect of three different dose regimens. Eur J Anaesthesiol 17: 672–679. Grouls RJ et al. (1995). Capillary gas chromatographic method for the determination of n-butyl-paminobenzoate and lidocaine in plasma samples. J Chromatogr B Biomed Appl 673: 51–57. Hattori H et al. (1991). Determination of local anaesthetics in body fluids by gas chromatography with surface ionization detection. J Chromatogr 564: 278–282. Hawkins JD et al. (1982). A single-step assay for lidocaine and its major metabolite, monoethylglycinexylidide, in plasma by gas–liquid chromatography and nitrogen phosphorus detection. Ther Drug Monit 4: 103–106. Jonville AP et al. (1990). Accidental lidocaine overdosage in an infant. J Toxicol Clin Toxicol 28: 101–106. Kakiuchi Yet al. (2002). Chromatographic determination of free lidocaine and its active metabolites in plasma from patients under epidural anesthesia. Int J Clin Pharmacol Ther 40: 493–498. Kalin JR, Brissie RM (2002). A case of homicide by lethal injection with lidocaine. J Forensic Sci 47: 1135–1138. Koster EH et al. (2000). Determination of lidocaine in plasma by direct solid-phase microextraction combined with gas chromatography. J Chromatogr B Biomed Sci Appl 739: 175–182. Kudo K et al. (2004). A fatal case of poisoning by lidocaine overdosage: analysis of lidocaine in formalin-fixed tissues: a case report. Med Sci Law 44: 266–271. Laroche N et al. (1998). Capillary gas chromatographic method for the measurement of small concentrations of monoethylglycinexylidide and lidocaine in plasma. J Chromatogr B Biomed Sci Appl 716: 375–381. Lau OW et al. (1991). Gas–liquid chromatographic determination and pharmacological studies of six clinically-used local anesthetics. Meth Find Exp Clin Pharmacol 13: 475–481. Levine B et al. (1983). Gas chromatographic analysis of lidocaine in blood and tissues. J Anal Toxicol 7: 123–124. Lindberg R et al. (1983). Improved liquid-chromatographic determination of lidocaine and its desethylated metabolites in serum. Clin Chem 29: 1572–1573. Lorec AM et al. (1994). Rapid simultaneous determination of lidocaine, bupivacaine, and their two main metabolites using capillary gas–liquid chromatography with nitrogen phosphorus detector. Ther Drug Monit 16: 592–595. Lotfi H et al. (1997). Simultaneous determination of lidocaine and bupivacaine in human plasma: application to pharmacokinetics. Ther Drug Monit 19: 160–164. Ma M et al. (2006). Liquid-phase microextraction combined with high-performance liquid chromatography for the determination of local anaesthetics in human urine. J Pharm Biomed Anal 40: 128–135. Manna L et al. (2002). Development and validation of a reversed-phase liquid chromatographic method for the assay of lidocaine in aqueous humour samples. J Pharm Biomed Anal 29: 1121–1126. Moises EC et al. (2008). Pharmacokinetics of lidocaine and its metabolite in peridural anesthesia administered to pregnant women with gestational diabetes mellitus. Eur J Clin Pharmacol 64: 1189–1196. Nolan PEJr (1997). Pharmacokinetics and pharmacodynamics of IV agents for ventricular arrhythmias. Pharmacotherapy 17: 65S–75S. Ohshima T, Takayasu T (1999). Simultaneous determination of local anesthetics including estertype anesthetics in human plasma and urine by gas chromatography–mass spectrometry with solid-phase extraction. J Chromatogr B Biomed Sci Appl 726: 185–194.
L
Clarke's Analysis of Drugs and Poisons Chapter No. L Dated: 17/3/2011 At Time: 22:14:42
1576
Lidoflazine
Perrotti P et al. (2006). Serum levels and possible haemodynamic effects following anorectal application of an ointment containing nifedipine and lignocaine: a study in healthy volunteers. Clin Drug Invest 26: 459–467. Piwowarska J et al. (2004). Liquid chromatographic method for the determination of lidocaine and monoethylglycine xylidide in human serum containing various concentrations of bilirubin for the assessment of liver function. J Chromatogr B Analyt Technol Biomed Life Sci 805: 1–5. Poklis A et al. (1984). Tissue distribution of lidocaine after fatal accidental injection. J Forensic Sci 29: 1229–1236. Puente NW, Josephy PD (2001). Analysis of the lidocaine metabolite 2,6-dimethylaniline in bovine and human milk. J Anal Toxicol 25: 711–715. Raikos N et al. (2009). Analysis of anaesthetics and analgesics in human urine by headspace SPME and GC. J Sep Sci 32: 1018–1026. Rodovnichenko MS et al. (1998). [The detection and quantitative determination of lidocaine and novocainamide in biological fluids]. Sud Med Ekspert 41: 24–27. Rousseau GF et al. (2002). Plasma lidocaine concentrations following insertion of 2% lidocaine gel into the uterine cavity after uterine balloon thermal ablation. Br J Anaesth 89: 846–848. Sangster J (1997). Octanol-Water Partition Coefficients: Fundamentals and Physical Chemistry.. Sattler A et al. (1995). Development of a HPLC-system for quantitative measurement of lidocaine and bupivacaine in patient’s plasma during postoperative epidural pain therapy. Pharmazie 50: 741–744. Sporkert F, Pragst F (2000). Determination of lidocaine in hair of drug fatalities by headspace solidphase microextraction. J Anal Toxicol 24: 316–322. Stoliarov EE et al. (2009). [Detection of certain local anesthetics in biological fluids by chemotoxicological analysis]. Sud Med Ekspert 52: 24–27. Stymne B, Lillieborg S (2001). Plasma concentrations of lignocaine and prilocaine after a 24-h application of analgesic cream (EMLA) to leg ulcers. Br J Dermatol 145: 530–534. Sweetman S, ed. (2009). Martindale, The Complete Drug Reference, 36 edn. London: Pharmaceutical Press. Tam YK et al. (1987). High-performance liquid chromatography of lidocaine and nine of its metabolites in human plasma and urine. J Chromatogr 423: 199–206. Teatino Barbaro A (2004). About an unusual case: GC-MS detection of lidocaine. Forensic Sci Int 146 (Suppl): S93–S94. Watanabe T et al. (1998). Simple analysis of local anaesthetics in human blood using headspace solid-phase microextraction and gas chromatography–mass spectrometry-electron impact ionization selected ion monitoring. J Chromatogr B Biomed Sci Appl 709: 225–232. Williamson KM et al. (1997). Intraperitoneal lignocaine for pain relief after total abdominal hysterectomy. Br J Anaesth 78: 675–677.
Infrared Spectrum Principal peaks at wavenumbers 1505, 1645, 1220, 1155, 835, 775 cm1 (KBr disk). Mass Spectrum Principal ions at m/z 343, 70, 344, 109, 42, 113, 491, 56.
Disposition in the Body Lidoflazine is well absorbed after oral administration. It is excreted mainly as metabolites, the major metabolites being bis(4-fluorophenyl)butyric acid and the glucuronide conjugate of bis(4-fluorophenyl)butan1-ol. In 7 days, about 40% of an oral dose is excreted in the urine and about 40% is eliminated in the faeces. Therapeutic Concentration After a single oral dose of 120 mg given with food, peak plasma concentrations of about 0.06 mg/L were attained in about 1 h. Following repeated oral administration of 120 mg three times a day, a mean steady-state plasma concentration of 0.12 mg/L was reported [Vanhoutte, Van Nueten 1973]. Half-life Plasma half-life, about 1 day. Dose 120 to 360 mg daily. Vanhoutte PM, Van Nueten JM (1973). Lidoflazine. In: Scriabine A, ed. New Drugs Annual: Cardiovascular Drugs. New York: Raven Press, 203–226.
Limaprost Lidoflazine
L
Antianginal C30H35F2N3O = 491.6 CAS—3416-26-0 IUPAC Name 4-[4,4-Bis(4-fluorophenyl)butyl]-N-(2,6-dimethylphenyl)-1-piperazineacetamide Synonyms Ordiflazine; R-7904. Proprietary Names Clinium; Corflazine; Klinium.
Chemical Properties A white or slightly-yellow amorphous powder. Mp 159 to 161 . Almost insoluble in water; soluble 1 in 90 of ethanol and 1 in 220 of ether; very soluble in chloroform. Log P (octanol/water), 5.6. Colour Tests Mandelin’s test—yellow; Marquis test—yellow. Thin-layer Chromatography System TA—Rf 0.70; system TB—Rf 0.11; system TC—Rf 0.63; system TE—Rf 0.70; system TL—Rf 0.36; system TAE—Rf 0.70; system TAF—Rf 0.77 (acidified iodoplatinate solution, positive). Gas Chromatography System GA—lidoflazine RI 3870, M (desaminocarboxy-) RI 2230, M (desaminocarboxy-)-Me RI 2125; system GB—lidoflazine, not eluted. High Performance Liquid Chromatography System HA—k 0.6; system HX—RI 530. Ultraviolet Spectrum Acid isopropyl alcohol—266 (A11¼47b), 272 nm.
Antianginal, Prostaglandin C22H36O5 = 380.5 CAS—74397-12-9 IUPAC Name (E)-7-[(1R,2R,3R)-3-Hydroxy-2-[(E,3S,5S)-3-hydroxy-5-methylnon-1-enyl]-5-oxocyclopentyl]hept-2-enoic acid Synonyms (2E,11a,13E,15S,17S)-11,15-Dihydroxy-17,20-dimethyl-9-oxoprosta2,13-dien-1-oic acid; 17S,20-dimethyl-trans-2,3-didehydro-PGE1; (E)-7-{(1R,2R,3R)3-hydroxy-2-[(E)-(3S,5S)-3-hydroxy-5-methyl-1-nonenyl]-5-oxocyclopentyl}-2-heptenoic acid; 17S-methyl-v-homo-trans-D2-PGE1; ONO-1206; OP-1206; 9-oxo11a,15a-dihydroxy-17S,20-dimethylprosta-trans-2,trans-13-deenoic acid.
Chemical Properties White crystals. Mp 97 to 100 . Under high humidity conditions, limaprost degrades rapidly; aqueous solutions of limaprost are most stable at pH 3 to 4 [Moribe et al. 2007]. Limaprost is stable in plasma for up to 73 days at –80 , after three freeze–thaw cycles, up to 4 h at room temperature, in whole blood for 4 h at 4 , and in processed samples for 96 h at 4 [Komaba et al., 2007]. Limaprost Alfadex CAS—88852-12-4 Proprietary Names Opalmon; Prorenal. Chemical Properties a-Cyclodextrin inclusion compound of limaprost.
High Performance Liquid Chromatography Column: Inertsil ODS-2 (150 4.6 mm i.d., 5 mm). Mobile phase: water : acetonitrile (linear gradient, not described), flow rate 1.0 mL/min. Samples derivatised with 4-(N,N-dimethylaminosulfonyl)-7-(1-piperazinyl)-2,1,3-benzoadiazole. Fluorescence detection (lex ¼ 440 nm, lem ¼ 569 nm). Limit of detection, 1.7 to 5.0 fmol/L [Toyo’oka et al. 1992]. Column: ODS (150 4.6 mm i.d.). Mobile phase: 0.02 mol/L potassium dihydrogen phosphate : acetonitrile : isopropyl alcohol (9 : 5 : 2), flow rate 0.8 mL/min. UV detection (l ¼ 215 nm). Limit of detection not reported [Moribe et al. 2007]. Mass Spectrum
Clarke's Analysis of Drugs and Poisons Chapter No. L Dated: 17/3/2011 At Time: 22:14:50
Lindane Quantification Plasma LC-MS Column: Capcell PAK phenyl UG120 (150 2.0 mm, 5 mm). ESI, negative ion mode, MRM acquisition mode. Limit of quantification, 0.1 ng/L [Komaba et al., 2007] Disposition in the Body Rapidly absorbed following oral administration, with peak plasma concentrations reached within 1 h. Studies in rats have shown that 70% of an orally administered radiolabelled dose is excreted in the faeces, with the majority of the remainder excreted in urine over a 96-h post-dose period. At the time of writing, the extent or site of metabolism and route of excretion has not yet been investigated in humans. Therapeutic Concentration Twenty-four healthy volunteers were administered either 5 or 10 mg limaprost orally and monitored for 4 h. Mean peak plasma concentrations for the 2 doses were 1.02 and 1.93 ng/L, respectively, reached at 1.0 h [Komaba et al., 2007]. Twelve healthy adult volunteers were administered either 5 or 10 mg limaprost orally. Mean peak plasma concentrations for the 2 doses were 1.18 and 2.06 ng/L reached at 0.75 h and 0.5 h, respectively [Tsuboshima et al., 1992]. Half-life Approximately 1 h. Clearance Approximately 3110 L/h. Dose Given by mouth as limaprost alfadex, in a dose equivalent to limaprost 15 to 30 mg daily in three divided doses. Komaba J et al. (2007). Ultra sensitive determination of limaprost, a prostaglandin E1 analogue, in human plasma using on-line two-dimensional reversed-phase liquid chromatography–tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 852: 590–597. Moribe K et al. (2007). Stabilization mechanism of limaprost in solid dosage form. Int J Pharm 338: 1–6. Toyo’oka T et al. (1992). Sensitive fluorometric detection of prostaglandins by high performance liquid chromatography after precolumn labelling with 4-(N,N-dimethylaminosulfonyl)-7-(1piperazinyl)-2,1,3-benzoxadiazole (DBD-PZ). Biomed Chromatogr 6: 143–148. Tsuboshima M et al. (1992). Prostaglandins: synthetic and pharmacological studies and development. Yakugaku Zasshi 112: 447–469.
Lincomycin Antibiotic C18H34N2O6S = 406.5 CAS—154-21-2 IUPAC Name (2S,4R)-N-[(1R,2R)-2-Hydroxy-1-[(2R,3R,4S,5R,6R)-3,4,5-trihydroxy-6-methylsulfanyloxan-2-yl]propyl]-1-methyl-4-propylpyrrolidine-2-carboxamide Synonym (2S-trans)-Methyl 6,8–dideoxy-6-[[(1-methyl-4–propyl-2-pyrrolidinyl)carbonyl]amino]-1-thio-D-erythro-a-D-galacto-octopyranoside; NSC70731.
1577
Infrared Spectrum Principal peaks at wavenumbers 1655, 1104, 1075, 1564, 1040, 1262 cm1 (lincomycin hydrochloride, KBr disk). Dose The equivalent of 1.5 to 2 g of lincomycin daily.
Lindane Insecticide C6H6Cl6 = 290.8 CAS—58-89-9 IUPAC Name (1a,2a,3b,4a,5a,6b)-1,2,3,4,5,6-Hexachlorocyclohexane Synonyms 666; Benhexachlor; gamma benzene hexachloride; gamma-BHC; gamma-HCH; HCH; hexicide. Proprietary Names Aphtiria; Delitex N; Elentol; Escabin; Esoderm; Gambex; Gamene; GBH; G-Well; Hexicid; Hexit; Jacutin; Kwell; Kwellada; Lendianon; Lindanoxil; Lorexane; Pediletan; Pilensar; Piodrex; Pioletal; Pionax; Plurisan; Pruritrat; Quellada; Sarnapin; Sarpiol; Scabecid; Scabene.
Chemical Properties A white crystalline powder. Mp 112.5 . Practically insoluble in water; soluble 1 in 19 of dehydrated alcohol, 1 in 2 of acetone, 1 in 3.5 of chloroform and 1 in 5.5 of ether. Log P (octanol/water), 3.7. Thin-layer Chromatography System TE—Rf 0.86; system TF—Rf 0.75; system TX—Rf 0.51; system TY—Rf 0.92. Gas Chromatography System GA—RI 1745; system GK—RRT 0.76(relative to caffeine). Infrared Spectrum Principal peaks at wavenumbers 680, 665, 695, 778, 840, 950 cm1 (KBr disk).
Chemical Properties Slightly soluble in water; soluble in methanol, lower alcohols, ethyl acetate, acetone and chloroform. pKa 7.5. Log P (octanol/water), 0.6. Lincomycin Hydrochloride C18H34N2O6S,HCl,H2O = 461.0 CAS—859-18-7 (anhydrous); 7179-49-9 (monohydrate) Proprietary Names Albiotic; Anbycin; Cillimicina; Frademicina; Fredcina; Linco;
Lincocin(e); Lincocina; Lincogin; Lincolan; Lincomy; Linco-Plus; Lincorex; Lingo; Linmycin; Macrolin; Mycivin; Princol; Rimsalin. Chemical Properties A white crystalline powder. Soluble 1 in 1 of water, 1 in 40 of ethanol and 1 in 20 of dimethylformamide; soluble in methanol; practically insoluble in chloroform and ether. Colour Test Sodium nitroprusside (method 3)—violet. Thin-layer Chromatography System TA—Rf 0.67; system TAE—Rf 0.75 (acidified potassium permanganate solution, positive). Ultraviolet Spectrum No significant absorption, 230 to 360 nm.
Mass Spectrum Principal ions at m/z 181, 183, 109, 219, 111, 217, 51, 221.
L
Clarke's Analysis of Drugs and Poisons Chapter No. L Dated: 17/3/2011 At Time: 22:14:56
1578
L
Linuron
Quantification Blood GC ECD. Limit of detection, 1 mg/L [Radomski, Ray 1970]. ECD. For method, see Jain et al. [1965]. Serum GC ECD. Limit of detection, 0.02 mg/L have been associated with toxic effects. The estimated minimum oral lethal dose is 200 mg/kg and the maximum permissible atmospheric concentration is 0.5 mg/m3. Toxic doses or long-term exposure may cause liver necrosis. The maximum acceptable daily intake is 10 mg/kg. A young girl who ingested about 1.6 g of lindane was found to have a serum concentration of 0.84 mg/L after 2 h, following convulsions; the concentration decreased to 0.49 mg/L after 4 h; urinary concentrations of individual free phenolic metabolites determined 5.5 h after ingestion ranged from 0.04 to 0.74 mg/L [Starr, Clifford 1972]. A fat concentration of 343 mg/g was reported in a fatality caused by lindane [Hayes, Vaughn 1977]. Half-life Derived from urinary excretion data, about 26 h. Use Topically in concentrations of 0.1 to 1%. Baumann K et al. (1980). Occupational exposure to hexachlorocyclohexane. I. Body burden of HCH-isomers. Int Arch Occup Environ Health 47: 119–127. Feldmann RJ, Maibach HJ (1974). Percutaneous penetration of some pesticides and herbicides in man. Toxicol Appl Pharmacol 28: 126–132. Hayes WJ, Vaughn WK (1977). Mortality from pesticides in the United States in 1973 and 1974. Toxicol Appl Pharmacol 42: 235–252. Jain N et al. (1965). Simplified gas chromatographic analysis of pesticides from blood. J Pharm Pharmacol 17: 362–367. Milby TH et al. (1968). Humane exposure to lindane; blood lindane levels as a function of exposure. J Occup Med 10: 584–587. Peper M et al. (1999). Long-term exposure to wood-preserving chemicals containing pentachlorophenol and lindane is related to neurobehavioral performance in women. Am J Ind Med 35: 632–641. Radomski JL, Ray A (1970). J Chromatogr Sci 8: 108–114. Saady JJ, Poklis A (1990). Determination of chlorinated hydrocarbon pesticides by solid-phase extraction and capillary GC with electron capture detection. J Anal Toxicol 14: 301–401. Starr HG, Clifford NJ (1972). Acute lindane intoxication: a case study. Arch Environ Health 25: 374–375.
Linuron Herbicide C9H10Cl2N2O2 = 249.1 CAS—330-55-2 IUPAC Name N0 -(3,4-Dichlorophenyl)-N-methoxy-N-methylurea Synonyms Du Pont herbicide 326; HOE-2810. Proprietary Names Afalon; Garnitan; Linex; Linorox; Linurex; Lorox; Premalin; Rotalin; Sarclex; Sinuron; Siolcid.
Chemical Properties A white crystalline solid (fine flakes or coarse powder). Mp 93 to 94 . Soluble in water (81 mg/L at 25 ) and soluble in acetone (500 g/kg at 25 ), ethanol (150 g/kg at 25 ), benzene (150 g/kg at 25 ), toluene, xylene (130 g/kg at 25 ), and heptane (15 g/kg at 25 ); readily soluble in dimethylformamide, chloroform and diethyl ether; moderately soluble in aromatic hydrocarbons; sparingly soluble in aliphatic hydrocarbons. Log P (octanol/water), 3.00. Thin-layer Chromatography System TX—Rf 0.22; system TY—Rf 0.31. Gas Chromatography System GA—linuron RI 1927, linuron-Me RI 1785, 3,4dichloroaniline RI 1323; system GK—linuron RRT 0.97, 3,4-dichloroaniline RRT 0.36, 3,4-dichlorophenyl-isocyanateRRT 0.13 (all relative to caffeine). High Performance Liquid Chromatography System HY—RI 506; system HAA—retention time 21.3 min. Infrared Spectrum Principal peaks at wavenumber 2269, 1507, 1729, 1594, 1132, 1282 cm1.
Mass Spectrum Principal ions at m/z 61, 160, 132, 124, 46, 248, 73, 109.
Disposition in the Body Linuron is broken down completely after passing through the liver. It is unlikely to bioaccumulate. Metabolites include 3,4-dichloroaniline and 3,4-dichlorophenylisocyanate.
Liothyronine Thyroid Agent C15H12I3NO4 = 651.0 CAS—6893-02-3 IUPAC Name (2S)-2-Amino-3-[4-(4-hydroxy-3-iodophenoxy)-3,5-diiodophenyl]propanoic acid Synonyms O-(4-Hydroxy-3-iodophenyl)-3,5-diiodo-L-tyrosine; L-tri-iodothyronine. Note The abbreviation T3 is often used for tri-iodothyronine.
Clarke's Analysis of Drugs and Poisons Chapter No. L Dated: 17/3/2011 At Time: 22:15:0
Lisinopril
1579
Chemical Properties A white to off-white crystalline powder. It is soluble in water (1 in 10), methyl alcohol (1 in 70); practically insoluble in alcohol, acetone, chloroform and in ether. pKa 2.5, 4.0, 6.7, 10.1 (25 ). Log P (octanol/water), 1.22; log P (phosphate buffer (0.1 mol/L, pH 7)/octanol), 10.2 0.5 (room temperature). Extraction yield (chlorobutane), 0 [Demme et al. 2005]. Lisinopril Dihydrate Chemical Properties Crystals. Mp 236 to 237 , with decomposition. Insoluble in water, alcohol and propylene glycol; soluble in dilute alkalis. pKa 8.5. Log P (octanol/water), 3.0. Liothyronine Hydrochloride C15H12I3NO4,HCl = 687.4 CAS—6138-47-2 Proprietary Name Thybon Chemical Properties Long birefringent needles. Mp 202 to 203 with
decomposition. Liothyronine Sodium C15H11I3NNaO4 = 673.0 CAS—55-06-1 Synonym Sodium liothyronine Proprietary Names Cynomel; Cytomel; Dispon; Neo-Tiroimade; Ro-Thyronine;
Tertroxin; Thybon; Thyrotardin; Ti-Tre; Triostat; Triyodisan; Triyotex. It is an ingredient of Euthroid, Novothyral, and Thyrolar. Chemical Properties A white to buff-coloured solid or crystalline powder. Practically insoluble in water, chloroform, ether and most other organic solvents; soluble 1 in 500 of ethanol; soluble in solutions of alkali hydroxides. Ultraviolet Spectrum Liothyronine sodium: aqueous alkali—319 nm (A11¼65a).
C21H31N3O3,2H2O = 441.5 CAS—83915-83-7 Proprietary Names Acemin; Acerbon; Alapril; Carace; Coric; Doneka; Prinil;
Prinivil; Novatec; Tensopril; Vivatec; Zestril. Also an ingredient of Novazyd; Prinzide; Zestoretic. Chemical Properties An off-white crystalline powder. Solubility: water (97 g/L), methanol (14 g/L), ethanol, acetone, acetonitrile, chloroform and N,N-dimethylformamide (99%. Dose Up to 100 mg of liothyronine sodium daily.
Infrared Spectrum Principal peaks at wavenumbers 1655, 1570, 1388, 741, 732 cm1 (broad peak around 3000 cm1) (KBr).
Lisinopril Antihypertensive C21H31N3O5 = 405.5 CAS—76547-98-3 IUPAC Name (2S)-1-[(2S)-6-Amino-2-[[(2S)-1-hydroxy-1-oxo-4-phenylbutan-2-yl]amino] hexanoyl]pyrrolidine-2-carboxylic acid Synonyms (S)-1-[N2-(1-Carboxy-3-phenylpropyl]-L-lysyl]-L-proline; L-154,826000T; lisinoprilum; MK-521.
Mass Spectrum Principal peaks at m/z 70, 91, 84, 113, 245, 41, 28, 224.
Clarke's Analysis of Drugs and Poisons Chapter No. L Dated: 17/3/2011 At Time: 22:15:8
1580
Lithium Carbonate
Quantification Plasma GC–MS Limit of detection 0.5 mg/L [Shioya et al. 1989]. Urine GC–MS Limit of detection 5 mg/L [Shioya et al. 1989]. Disposition in the Body Lisinopril is slowly and incompletely absorbed after oral administration. Absorption varies between 6 and 60% depending on the individual, but on average is 25% of the dose. Metabolism does not occur as it is already an active diacid (a lysine derivative of enalaprilat). The absorbed drug is excreted unchanged in urine via the kidneys and the unabsorbed drug is excreted in faeces. Therapeutic Concentration The serum therapeutic concentration is 20 to 70 mg/L. Twelve healthy male volunteers were administered with an oral dose of 10 mg lisinopril. Peak serum concentrations of 0.095 0.055 mg/L were observed in 7 1 h [Ulm et al. 1982]. Bioavailability 25 to 50%. Half-life 12 h. Volume of Distribution Mean 124 L. Clearance 6.36 L/h (following a 10 mg oral dose in healthy subjects); reduced in renal impairment and the elderly. Clearance reduced in impaired renal function (glomerular filtration rate