A Textbook of
Clinical Pharmacology
and Therapeutics
This page intentionally left blank
A Textbook of
Clinical Pharmacology
and Therapeutics
FIFTH EDITION
JAMES M RITTER MA DP HIL FRCP FMedSciFBPHARMACOLS
Professor of Clinical Pharmacology at King’s College London School of Medicine,
Guy’s, King’s and St Thomas’ Hospitals, London, UK
LIONEL D LEWIS MA MB BCH MD FRCP
Professor of Medicine, Pharmacology and Toxicology at Dartmouth Medical School and
the Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
TIMOTHY GK MANT BSC FFPM FRCP
Senior Medical Advisor, Quintiles, Guy's Drug Research Unit, and Visiting Professor at
King’s College London School of Medicine, Guy’s, King’s and St Thomas’ Hospitals,
London, UK
ALBERT FERRO PHD FRCP FBPHAR MACOLS
Reader in Clinical Pharmacology and Honorary Consultant Physician at King’s College
London School of Medicine, Guy’s, King’s and St Thomas’ Hospitals, London, UK
PART OF HACHETTE LIVRE UK
First published in Great Britain in 1981
Second edition 1986
Third edition 1995
Fourth edition 1999
This fifth edition published in Great Britain in 2008 by
Hodder Arnold, an imprint of Hodden Education, part of Hachette Livre UK,
338 Euston Road, London NW1 3BH
http://www.hoddereducation.com
©2008 James M Ritter, Lionel D Lewis, Timothy GK Mant and Albert Ferro
All rights reserved. Apart from any use permitted under UK copyright law, this publication may
only be reproduced, stored or transmitted, in any form, or by any means with prior permission in
writing of the publishers or in the case of reprographic production in accordance with the terms
of licences issued by the Copyright Licensing Agency. In the United Kingdom such licences are
issued by the Copyright licensing Agency: Saffron House, 6–10 Kirby Street, London EC1N 8TS.
Hachette Livre’s policy is to use papers that are natural, renewable and recyclable products and
made from wood grown in sustainable forests. The logging and manufacturing processes are
expected to conform to the environmental regulations of the country of origin.
Whilst the advice and information in this book are believed to be true and accurate at the date of
going to press, neither the authors nor the publisher can accept any legal responsibility or liability
for any errors or omissions that may be made. In particular, (but without limiting the generality
of the preceding disclaimer) every effort has been made to check drug dosages; however it is
still possible that errors have been missed. Furthermore, dosage schedules are constantly being
revised and new side-effects recognized. For these reasons the reader is strongly urged to consult
the drug companies’ printed instructions before administering any of the drugs recommended in
this book.
British Library Cataloguing in Publication Data
Acatalogue record for this book is available from the British Library
Library of Congress Cataloging-in-Publication Data
Acatalog record for this book is available from the Library of Congress
ISBN 978-0-340-90046-8
1 2 3 4 5 6 7 8 9 10
Commissioning Editor: Sara Purdy
Project Editor: Jane Tod
Production Controller: Andre Sim
Cover Design: Laura de Grasse
Indexer: John Sampson
Typeset in 9/12 pt palatino by Charon Tec Ltd (A Macmillan Company) www.charontec.com
Printed and bound in Italy
What do you think about this book? Or any other Hodder Arnold title?
Please visit our website: www.hoddereducation.com
This fifth edition is dedicated to the memory of Professors Howard Rogers and John Trounce,
two of the three authors of this textbook’s first edition.
COMPANION WEBSITE
The fifth edition of A Textbook of Clinical Pharmacology and Therapeuticsis accompanied by an
exciting new website featuring the images from the book for you to download. To visit the
book’s website, please go to www
.hodderplus.com/clinicalpharmacology.
Your username is: student009
Your password is: pharma
CONTENTS
FOREWORD viii
PREFACE ix
ACKNOWLEDGEMENTS x
PART I GENERAL PRINCIPLES 1
1 Introduction to therapeutics 3
2 Mechanisms of drug action (pharmacodynamics) 6
3 Pharmacokinetics 11
4 Drug absorption and routes of administration 17
5 Drug metabolism 24
6 Renal excretion of drugs 31
7 Effects of disease on drug disposition 34
8 Therapeutic drug monitoring 41
9 Drugs in pregnancy 45
10 Drugs in infants and children 52
11 Drugs in the elderly 56
12 Adverse drug reactions 62
13 Drug interactions 71
14 Pharmacogenetics 79
15 Introduction of new drugs and clinical trials 86
16 Cell-based and recombinant DNAtherapies 92
17 Alternative medicines: herbals and
nutraceuticals 97
PART II THE NERVOUS SYSTEM 103
18 Hypnotics and anxiolytics 105
19 Schizophrenia and behavioural emergencies 110
20 Mood disorders 116
21 Movement disorders and degenerative CNS
disease 124
22 Anti-epileptics 133
23 Migraine 142
24 Anaesthetics and muscle relaxants 145
25 Analgesics and the control of pain 155
PART III THE MUSCULOSKELETAL SYSTEM 165
26 Anti-inflammatory drugs and the treatment
of arthritis 167
PART IV THE CARDIOVASCULAR SYSTEM 175
27 Prevention of atheroma: lowering plasma
cholesterol and other approaches 177
28 Hypertension 185
29 Ischaemic heart disease 196
30 Anticoagulants and antiplatelet drugs 204
31 Heart failure 211
32 Cardiac dysrhythmias 217
PART V THE RESPIRATORY SYSTEM 231
33 Therapy of asthma, chronic obstructive pulmonary
disease (COPD) and other respiratory disorders 233
PART VI THE ALIMENTARY SYSTEM 245
34 Alimentary system and liver 247
35 Vitamins and trace elements 265
PART VII FLUIDS AND ELECTROLYTES 271
36 Nephrological and related aspects 273
PART VIII THE ENDOCRINE SYSTEM 283
37 Diabetes mellitus 285
38 Thyroid 292
39 Calcium metabolism 297
40 Adrenal hormones 302
41 Reproductive endocrinology 307
42 The pituitary hormones and related drugs 316
PART IX SELECTIVE TOXICITY 321
43 Antibacterial drugs 323
44 Mycobacterial infections 334
45 Fungal and non-HIV viral infections 340
46 HIV and AIDS 351
47 Malaria and other parasitic infections 361
48 Cancer chemotherapy 367
PART X HAEMATOLOGY 387
49 Anaemia and other haematological disorders 389
PART XI IMMUNOPHARMACOLOGY 397
50 Clinical immunopharmacology 399
PART XII THE SKIN 409
51 Drugs and the skin 411
PART XIII THE EYE 421
52 Drugs and the eye 423
PART XIV CLINICAL TOXICOLOGY 431
53 Drugs and alcohol abuse 433
54 Drug overdose and poisoning 444
INDEX 451
John Trounce, who was the senior author of the first edition of this textbook, died on the
16 April 2007.
He considered a text in clinical pharmacology suitable for his undergraduate and postgradu-
ate students to be an important part of the programme he developed in his department at
Guy’s Hospital Medical School, London. It is difficult to imagine today how much resistance
from the medical and pharmacological establishments Trounce had to overcome in order to set
up an academic department, a focussed course in the medical curriculum and a separate exam
in final MB in clinical pharmacology. In other words, he helped to change a ‘non-subject’ into
one of the most important areas of study for medical students. He was also aware of the need
for a high quality textbook in clinical pharmacology that could also be used by nurses, phar-
macists, pharmacology science students and doctors preparing for higher qualifications. (For
example, it has been said that nobody knows more about acute pharmacology than an
anaesthetist.)
The present edition of the textbook reflects the advances in therapeutics since the publica-
tion of the fourth edition. It is interesting to follow in all the editions of the book, for example,
how the treatment of tumours has progressed. It was about the time of the first edition that
Trounce set up the first oncology clinic at Guy’s Hospital in which he investigated the value of
combined radiation and chemotherapy and drug cocktails in the treatment of lymphomas.
John Trounce was pleased to see his textbook (and his subject) in the expert hands of Professor
Ritter and his colleagues.
Roy Spector
Professor Emeritus in Applied Pharmacology, University of London
FOREWORD
Clinical pharmacology is the science of drug use in humans. Clinicians of all specialties pre-
scribe drugs on a daily basis, and this is both one of the most useful but also one of the most
dangerous activities of our professional lives. Understanding the principles of clinical pharma-
cology is the basis of safe and effective therapeutic practice, which is why this subject forms an
increasingly important part of the medical curriculum.
This textbook is addressed primarily to medical students and junior doctors of all special-
ties, but also to other professionals who increasingly prescribe medicines (including pharma-
cists, nurses and some other allied professionals). Clinical pharmacology is a fast moving
subject and the present edition has been completely revised and updated. It differs from the
fourth edition in that it concentrates exclusively on aspects that students should know and
understand, rather than including a lot of reference material. This has enabled us to keep its
length down. Another feature has been to include many new illustrations to aid in grasping
mechanisms and principles.
The first section deals with general principles including pharmacodynamics, pharmaco-
kinetics and the various factors that modify drug disposition and drug interaction. We have
kept algebraic formulations to a minimum. Drug metabolism is approached from a practical
viewpoint, with discussion of the exciting new concept of personalized medicine. Adverse
drug reactions and the use of drugs at the extremes of age and in pregnancy are covered, and
the introduction of new drugs is discussed from the viewpoint of students who will see many
new treatments introduced during their professional careers. Many patients use herbal or
other alternative medicines and there is a new chapter on this important topic. There is a chap-
ter on gene and cell-based therapies, which are just beginning to enter clinical practice. The
remaining sections of the book deal comprehensively with major systems (nervous, musculo-
skeletal, cardiovascular, respiratory, alimentary, renal, endocrine, blood, skin and eye) and
with multi-system issues including treatment of infections, malignancies, immune disease,
addiction and poisoning.
J
AMES M RITTER
LIONELD LEWIS
TIMOTHYGK MANT
ALBERTFERRO
PREFACE
We would like to thank many colleagues who have helped us with advice and criticism in the
revision and updating of this fifth edition. Their expertise in many specialist areas has enabled
us to emphasize those factors most relevant. For their input into this edition and/or the previ-
ous edition we are, in particular, grateful to Professor Roy Spector, Professor Alan Richens,
Dr Anne Dornhorst, Dr Michael Isaac, Dr Terry Gibson, Dr Paul Glue, Dr Mark Kinirons,
Dr Jonathan Barker, Dr Patricia McElhatton, Dr Robin Stott, Mr David Calver, Dr Jas Gill,
Dr Bev Holt, Dr Zahid Khan, Dr Beverley Hunt, Dr Piotr Bajorek, Miss Susanna Gilmour-
White, Dr Mark Edwards, Dr Michael Marsh, Mrs Joanna Tempowski. We would also like to
thank Dr Peter Lloyd and Dr John Beadle for their assistance with figures.
ACKNOWLEDGEMENTS
PART I
GENERAL PRINCIPLES
This page intentionally left blank
Use of drugs 3
Adverse effects and risk/benefit 3
Drug history and therapeutic plan 4
Formularies and restricted lists 4
Scientific basis of use of drugs in humans 4
CHAPTER 1
INTRODUCTION TO THERAPEUTICS
USE OF DRUGS
People consult a doctor to find out what (if anything) is wrong
(the diagnosis), and what should be done about it (the treat-
ment). If they are well, they may nevertheless want to know
how future problems can be prevented. Depending on the diag-
nosis, treatment may consist of reassurance, surgery or other
interventions. Drugs are very often either the primary therapy
or an adjunct to another modality (e.g. the use of anaesthetics
in patients undergoing surgery). Sometimes contact with the
doctor is initiated because of a public health measure (e.g.
through a screening programme). Again, drug treatment is
sometimes needed. Consequently, doctors of nearly all special-
ties use drugs extensively, and need to understand the scien-
tific basis on which therapeutic use is founded.
A century ago, physicians had only a handful of effective
drugs (e.g. morphia, quinine, ether, aspirin and digitalis leaf)
at their disposal. Thousands of potent drugs have since been
introduced, and pharmaceutical chemists continue to discover
new and better drugs. With advances in genetics, cellular and
molecular science, it is likely that progress will accelerate and
huge changes in therapeutics are inevitable. Medical students
and doctors in training therefore need to learn something
of the principles of therapeutics, in order to prepare them-
selves to adapt to such change. General principles are dis-
cussed in the first part of this book, while current approaches
to treatment are dealt with in subsequent parts.
ADVERSE EFFECTS AND RISK/BENEFIT
Medicinal chemistry has contributed immeasurably to human
health, but this has been achieved at a price, necessitating a
new philosophy. Aphysician in Sir William Osler’s day in the
nineteenth century could safely adhere to the Hippocratic
principle ‘first do no harm’, because the opportunities for
doing good were so limited. The discovery of effective drugs
has transformed this situation, at the expense of very real risks
of doing harm. For example, cures of leukaemias, Hodgkin’s
disease and testicular carcinomas have been achieved through
a preparedness to accept a degree of containable harm. Similar
considerations apply in other disease areas.
All effective drugs have adverse effects, and therapeutic
judgements based on risk/benefit ratio permeate all fields of
medicine. Drugs are the physician’s prime therapeutic tools,
and just as a misplaced scalpel can spell disaster, so can a
thoughtless prescription. Some of the more dramatic instances
make for gruesome reading in the annual reports of the med-
ical defence societies, but perhaps as important is the morbid-
ity and expense caused by less dramatic but more common
errors.
How are prescribing errors to be minimized? By combining
a general knowledge of the pathogenesis of the disease to be
treated and of the drugs that may be effective for that disease
with specific knowledge about the particular patient. Dukes
and Swartz, in their valuable work Responsibility for drug-
induced injury,list eight basic duties of prescribers:
1. restrictive use – is drug therapy warranted?
2. careful choice of an appropriate drug and dose regimen
with due regard to the likely risk/benefit ratio, available
alternatives, and the patient’s needs, susceptibilities and
preferences;
3. consultation and consent;
4. prescription and recording;
5. explanation;
6. supervision (including monitoring);
7. termination, as appropriate;
8. conformity with the law relating to prescribing.
As a minimum, the following should be considered when
deciding on a therapeutic plan:
1. age;
2. coexisting disease, especially renal and or hepatic
impairment;
3. the possibility of pregnancy;
4. drug history;
5. the best that can reasonably be hoped for in this
individual patient;
6. the patient’s beliefs and goals.
DRUG HISTORY AND THERAPEUTIC PLAN
In the twenty-first century, a reliable drug history involves
questioning the patient (and sometimes family, neighbours,
other physicians, etc.). What prescription tablets, medicines,
drops, contraceptives, creams, suppositories or pessaries are
being taken? What over-the-counter remedies are being used
including herbal or ‘alternative’ therapies? Does the patient
use drugs socially or for ‘life-style’ purposes? Have they suf-
fered from drug-induced allergies or other serious reactions?
Have they been treated for anything similar in the past, and if
so with what, and did it do the job or were there any prob-
lems? Has the patient experienced any problems with anaes-
thesia? Have there been any serious drug reactions among
family members?
The prescriber must be both meticulous and humble, espe-
cially when dealing with an unfamiliar drug. Checking
contraindications, special precautions and doses in a formu-
lary such as the British National Formulary (BNF) (British
Medical Association and Royal Pharmaceutical Society of
Great Britain 2007) is the minimum requirement. The proposed
plan is discussed with the patient, including alternatives,
goals, possible adverse effects, their likelihood and measures
to be taken if these arise. The patient must understand what is
intended and be happy with the means proposed to achieve
these ends. (This will not, of course, be possible in demented
or delirious patients, where discussion will be with any
available family members.) The risks of causing harm must
be minimized. Much of the ‘art’ of medicine lies in the ability
of the prescriber to agree to compromises that are accept-
able to an individual patient, and underlies concordance
(i.e. agreement between patient and prescriber) with a thera-
peutic plan.
Prescriptions must be written clearly and legibly, conform-
ing to legal requirements. Electronic prescribing is currently
being introduced in the UK, so these are changing. Generic
names should generally be used (exceptions are mentioned
later in the book), together with dose, frequency and duration
of treatment, and paper prescriptions signed. It is prudent to
print the prescriber’s name, address and telephone number to
facilitate communication from the pharmacist should a query
arise. Appropriate follow up must be arranged.
FORMULARIES AND RESTRICTED LISTS
Historically, formularies listed the components of mixtures
prescribed until around 1950. The perceived need for hospital
formularies disappeared transiently when such mixtures
were replaced by proprietary products prepared by the
pharmaceutical industry. The BNF summarizes products
licensed in the UK. Because of the bewildering array, includ-
ing many alternatives, many hospital and primary care trusts
have reintroduced formularies that are essentially restricted
lists of the drugs stocked by the institution’s pharmacy, from
which local doctors are encouraged to prescribe. The objec-
tives are to encourage rational prescribing, to simplify pur-
chasing and storage of drugs, and to obtain the ‘best buy’
among alternative preparations. Such formularies have the
advantage of encouraging consistency, and once a decision
has been made with input from local consultant prescribers
they are usually well accepted.
SCIENTIFIC BASIS OF USE OF DRUGS IN
HUMANS
The scientific basis of drug action is provided by the discipline
of pharmacology. Clinical pharmacology deals with the effects
of drugs in humans. It entails the study of the interaction of
drugs with their receptors, the transduction (second messen-
ger) systems to which these are linked and the changes that
they bring about in cells, organs and the whole organism.
These processes (what the drug does to the body) are called
‘pharmacodynamics’. The use of drugs in society is encom-
passed by pharmacoepidemiology and pharmacoeconomics –
both highly politicized disciplines!
Man is a mammal and animal studies are essential, but
their predictive value is limited. Modern methods of molecu-
lar and cell biology permit expression of human genes, includ-
ing those that code for receptors and key signal transduction
elements, in cells and in transgenic animals, and are revolu-
tionizing these areas and hopefully improving the relevance
of preclinical pharmacology and toxicology.
Important adverse effects sometimes but not always occur
in other species. Consequently, when new drugs are used to treat
human diseases, considerable uncertainties remain. Early-phase
human studies are usually conducted in healthy volunteers,
except when toxicity is inevitable (e.g. cytotoxic drugs used
for cancer treatment, see Chapter 48).
Basic pharmacologists often use isolated preparations,
where the concentration of drug in the organ bath is controlled
precisely. Such preparations may be stable for minutes to
hours. In therapeutics, drugs are administered to the whole
organism by a route that is as convenient and safe as possible
(usually by mouth), for days if not years. Consequently, the
drug concentration in the vicinity of the receptors is usually
unknown, and long-term effects involving alterations in receptor
density or function, or the activation or modulation of homeo-
static control mechanisms may be of overriding importance.
The processes of absorption, distribution, metabolism and elim-
ination (what the body does to the drug) determine the drug
concentration–time relationships in plasma and at the recep-
tors. These processes comprise ‘pharmacokinetics’. There is
considerable inter-individual variation due to both inherited
4 INTRODUCTIONTO THERAPEUTICS
and acquired factors, notably disease of the organs responsible
for drug metabolism and excretion. Pharmacokinetic modelling
is crucial in drug development to plan a rational therapeutic
regime, and understanding pharmacokinetics is also import-
ant for prescribers individualizing therapy for a particular
patient. Pharmacokinetic principles are described in Chapter 3
from the point of view of the prescriber. Genetic influences on
pharmacodynamics and pharmacokinetics (pharmacogenet-
ics) are discussed in Chapter 14 and effects of disease are
addressed in Chapter 7, and the use of drugs in pregnancy
and at extremes of age is discussed in Chapters 9–11.
There are no good animal models of many important human
diseases. The only way to ensure that a drug with promising
pharmacological actions is effective in treating or preventing
disease is to perform a specific kind of human experiment,
called a clinical trial. Prescribing doctors must understand the
strengths and limitations of such trials, the principles of which
are described in Chapter 15, if they are to evaluate the litera-
ture on drugs introduced during their professional lifetimes.
Ignorance leaves the physician at the mercy of sources of infor-
mation that are biased by commercial interests. Sources of
unbiased drug information include Dollery’s encyclopaedic
Therapeutic drugs, 2nd edn (published by Churchill Livingstone
in 1999), which is an invaluable source of reference. Publications
such as the Adverse Reaction Bulletin, Prescribers Journal and
the succinctly argued Drug and Therapeutics Bulletin provide
up-to-date discussions of therapeutic issues of current
importance.
FURTHER READING
Dukes MNG, Swartz B. Responsibility for drug-induced injury.
Amsterdam: Elsevier, 1988.
Weatherall DJ. Scientific medicine and the art of healing. In: Warrell
DA, Cox TM, Firth JD, Benz EJ (eds), Oxford textbook of medicine,4th
edn. Oxford: Oxford University Press, 2005.
SCIENTIFIC BASIS OF USE OF DRUGS IN H UMANS 5
Key points
Drugs are prescribed by physicians of all specialties.
This carries risks as well as benefits.
Therapy is optimized by combining general knowledge
of drugs with knowledge of an individual patient.
Evidence of efficacy is based on clinical trials.
Adverse drug effects may be seen in clinical trials, but
the drug side effect profile becomes clearer only when
widely prescribed.
Rational prescribing is encouraged by local formularies.
Case history
A general practitioner reviews the medication of an
86-year-old woman with hypertension and multi-infarct
dementia, who is living in a nursing home. Her family used
to visit daily, but she no longer recognizes them, and needs
help with dressing, washing and feeding. Drugs include
bendroflumethiazide, atenolol, atorvastatin, aspirin, haloperi-
dol,imipramine, lactulose and senna. On examination, she
smells of urine and has several bruises on her head, but
otherwise seems well cared for. She is calm, but looks pale
and bewildered, and has a pulse of 48 beats/min regular,
and blood pressure 162/96mmHg lying and 122/76 mmHg
standing, during which she becomes sweaty and distressed.
Her rectum is loaded with hard stool. Imipramine was started
three years previously. Urine culture showed only a light
mixed growth. All of the medications were stopped and
manual evacuation of faeces performed. Stool was nega-
tive for occult blood and the full blood count was normal.
Two weeks later, the patient was brighter and more mobile.
She remained incontinent of urine at night, but no longer
during the day, her heart rate was 76 beats/min and her
blood pressure was 208/108mmHg lying and standing.
Comment
It is seldom helpful to give drugs in order to prevent some-
thing that has already happened (in this case multi-infarct
dementia), and any benefit in preventing further ischaemic
events has to be balanced against the harm done by the
polypharmacy. In this case, drug-related problems probably
include postural hypotension (due to imipramine, ben-
droflumethiazide and haloperidol), reduced mobility (due to
haloperidol), constipation (due to imipramine and haloperi-
dol), urinary incontinence (worsened by bendroflumethi-
azide and drugs causing constipation) and bradycardia (due
to atenolol). Drug-induced torsades de pointes (a form of
ventricular tachycardia, see Chapter 32) is another issue.
Despite her pallor, the patient was not bleeding into the
gastro-intestinal tract, but aspirin could have caused this.
Introduction 6
Receptors and signal transduction 6
Agonists 7
Antagonism 8
Partial agonists 9
Slow processes 9
Non-receptor mechanisms 10
CHAPTER 2
MECHANISMS OF DRUG ACTION
(PHARMACODYNAMICS)
INTRODUCTION
Pharmacodynamics is the study of effects of drugs on biological
processes. An example is shown in Figure 2.1, demonstrating
and comparing the effects of a proton pump inhibitor and of a
histamine H
2
receptor antagonist (both drugs used for the treat-
ment of peptic ulceration and other disorders related to gastric
hyperacidity) on gastric pH. Many mediators exert their effects
as a result of high-affinity binding to specific receptors in
plasma membranes or cell cytoplasm/nuclei, and many thera-
peutically important drugs exert their effects by combining with
these receptors and either mimicking the effect of the natural
mediator (in which case they are called ‘agonists’) or blocking it
(in which case they are termed ‘antagonists’). Examples include
oestrogens (used in contraception, Chapter 41) and anti-
oestrogens (used in treating breast cancer, Chapter 48), alpha-
and beta-adrenoceptor agonists and antagonists (Chapters 29
and 33) and opioids (Chapter 25).
Not all drugs work via receptors for endogenous medi-
ators: many therapeutic drugs exert their effects by combining
with an enzyme or transport protein and interfering with its
function. Examples include inhibitors of angiotensin convert-
ing enzyme and serotonin reuptake. These sites of drug action
are not ‘receptors’ in the sense of being sites of action of
endogenous mediators.
Whether the site of action of a drug is a receptor or another
macromolecule, binding is usually highly specific, with precise
steric recognition between the small molecular ligand and the
binding site on its macromolecular target. Binding is usually
reversible. Occasionally, however, covalent bonds are formed
with irreversible loss of function, e.g. aspirin binding to cyclo-
oxygenase (Chapter 30).
Most drugs produce graded concentration-/dose-related
effects which can be plotted as a dose–response curve. Such
curves are often approximately hyperbolic (Figure 2.2a). If plot-
ted semi-logarithmically this gives an S-shaped (‘sigmoidal’)
shape (Figure 2.2b). This method of plotting dose–response
curves facilitates quantitative analysis (see below) of full agonists
(which produce graded responses up to a maximum value),
antagonists (which produce no response on their own, but
reduce the response to an agonist) and partial agonists (which
produce some response, but to a lower maximum value than that
of a full agonist, and antagonize full agonists) (Figure 2.3).
RECEPTORS AND SIGNAL TRANSDUCTION
Drugs are often potent (i.e. they produce effects at low concen-
tration) and specific (i.e. small changes in structure lead to pro-
found changes in potency). High potency is a consequence of
high binding affinity for specific macromolecular receptors.
0
1
2
3
4
5
6
7
8
9
10
112233
Median gastric pH
Predose Postdose
5.0
4.1-5.0
2.0-4.0
< 2.0 21
15
25
37
41
38
37
9090
n
Predose Postdose Predose Postdose
P
re
R
x
pH
Figure 2.1:Effect of omeprazole and cimetidine on gastric pH in a
group of critically ill patients. This was a study comparing the
effect of immediate-release omeprazole with a loading dose of
40mg, a second dose six to eight hours later, followed by 40mg
daily, with a continuous i.v. infusion of cimetidine. pH monitoring
of the gastric aspirate was undertaken every two hours and
immediately before and one hour after each dose. Red,
omeprazole; blue, cimetidine. (Redrawn with permission from
Horn JR, Hermes-DeSantis ER, Small, RE ‘New Perspectives in the
Management of Acid-Related Disorders: The Latest Advances in
PPI Therapy’. Medscape Today
http://www.medscape.com/viewarticle/503473_9 17 May 2005.)
AGONISTS 7
Receptors were originally classified by reference to the relative
potencies of agonists and antagonists on preparations contain-
ing different receptors. The order of potency of isoprenaline
adrenaline noradrenaline on tissues rich in β-receptors, such
as the heart, contrasts with the reverse order in α-receptor-
mediated responses, such as vasoconstriction in resistance
arteries supplying the skin. Quantitative potency data are best
obtained from comparisons of different competitive antag-
onists, as explained below. Such data are supplemented, but not
replaced, by radiolabelled ligand-binding studies. In this way,
adrenoceptors were divided first into αand β, then subdivided
intoα
1
/α
2
andβ
1
/β
2
. Many other useful receptor classifications,
including those of cholinoceptors, histamine receptors, sero-
tonin receptors, benzodiazepine receptors, glutamate receptors
and others have been proposed on a similar basis. Labelling
with irreversible antagonists permitted receptor solubilization
and purification. Oligonucleotide probes based on the deduced
sequence were then used to extract the full-length DNA
sequence coding different receptors. As receptors are cloned
and expressed in cells in culture, the original functional classifi-
cations have been supported and extended. Different receptor
subtypes are analogous to different forms of isoenzymes, and a
rich variety has been uncovered – especially in the central ner-
vous system – raising hopes for novel drugs targeting these.
Despite this complexity, it turns out that receptors fall into
only four ‘superfamilies’ each linked to distinct types of signal
transduction mechanism (i.e. the events that link receptor acti-
vation with cellular response) (Figure 2.4). Three families are
located in the cell membrane, while the fourth is intracellular
(e.g. steroid hormone receptors). They comprise:
Fast (millisecond responses) neurotransmitters (e.g.
nicotinic receptors), linked directly to a transmembrane
ion channel.
Slower neurotransmitters and hormones (e.g. muscarinic
receptors) linked to an intracellular G-protein (‘GPCR’).
Receptors linked to an enzyme on the inner membrane
(e.g. insulin receptors) are slower still.
Intranuclear receptors (e.g. gonadal and glucocorticosteroid
hormones): ligands bind to their receptor in cytoplasm and
the complex then migrates to the nucleus and binds to
specific DNAsites, producing alterations in gene
transcription and altered protein synthesis. Such effects
occur over a time-course of minutes to hours.
AGONISTS
Agonists activate receptors for endogenous mediators – e.g.
salbutamol is an agonist at β
2
-adrenoceptors (Chapter 33).
The consequent effect may be excitatory (e.g. increased
heart rate) or inhibitory (e.g. relaxation of airway smooth
muscle). Agonists at nicotinic acetylcholine receptors (e.g.
suxamethonium, Chapter 24) exert an inhibitory effect
(neuromuscular blockade) by causing long-lasting depolariza-
tion at the neuromuscular junction, and hence inactivation of
the voltage-dependent sodium channels that initiate the action
potential.
Endogenous ligands have sometimes been discovered long
after the drugs that act on their receptors. Endorphins and
enkephalins (endogenous ligands of morphine receptors)
were discovered many years after morphine. Anandamide is a
central transmitter that activates CB (cannabis) receptors
(Chapter 53).
0
0
510
100
Effect (%)
[Drug]
(a)
1 10 100
100
Effect (%)
[Drug]
(b)
Figure 2.2:Concentration/dose–response curves plotted (a) arithmetically and (b) semi-logarithmically.
1
0
10 100
100
Effect (%)
[Drug]
A
B
C
Figure 2.3:Concentration/dose–response curves of two full
agonists (A, B) of different potency, and of a partial agonist (C).
8 MECHANISMSOF DRUG ACTION (PHARMACODYNAMIC S)
ANTAGONISM
Competitive antagonists combine with the same receptor as an
endogenous agonist (e.g. ranitidineat histamine H
2
-receptors),
but fail to activate it. When combined with the receptor, they
prevent access of the endogenous mediator. The complex
between competitive antagonist and receptor is reversible.
Provided that the dose of agonist is increased sufficiently, a
maximal effect can still be obtained, i.e. the antagonism is sur-
mountable. If a dose (C) of agonist causes a defined effect when
administered alone, then the dose (C) needed to produce the
same effect in the presence of antagonist is a multiple (C/C)
known as the dose ratio (r). This results in the familiar parallel
shift to the right of the log dose–response curve, since the add-
ition of a constant length on a logarithmic scale corresponds to
multiplication by a constant factor (Figure 2.5a). β-Adrenoceptor
antagonists are examples of reversible competitive antagonists.
By contrast, antagonists that do not combine with the same
receptor (non-competitive antagonists) or drugs that combine
irreversibly with their receptors, reduce the slope of the log
dose–response curve and depress its maximum (Figure 2.5b).
Physiological antagonism describes the situation where two
drugs have opposing effects (e.g. adrenaline relaxes bronchial
smooth muscle, whereas histamine contracts it).
Fast (ms)
neurotransmitter
(e.g. glutamate)
Slow (s)
neurotransmitter
or hormone
(e.g.-adrenoceptor)
Ion channel
Direct effect (min)
on protein
phosphorylation
(e.g. insulin)
Control (hours)
of DNA/new
protein synthesis
(e.g. steroid hormones)
Cell
membrane
Cytoplasm
Second messengers
Protein
phosphorylation
Ca
2
release
Cellular effects
Nucleus
Change in
membrane
potential
GE
E
Figure 2.4:Receptors and signal transduction. G, G-protein; E, enzyme; Ca, calcium.
1 10 100
0
100
Effect (%)
AA[B]
1
A[B]
2
[Agonist](a)
10 100
1
0
100
A
A[C]
1
A[C]
2
[Agonist](b)
Figure 2.5:Drug antagonism. Control concentration/dose–response curves for an agonist A together with curves in the presence of (a) a
competitive antagonist B and (b) a non-competitive antagonist C. Increasing concentrations of the competitive antagonist ([B]
1
, [B]
2
)
cause a parallel shift to the right of the log dose–effect curve (a), while the non-competitive antagonist ([C]
1
, [C]
2
) flattens the curve
and reduces its maximum (b).
SLOW PROCESSES 9
The relationship between the concentration of a competi-
tive antagonist [B], and the dose ratio (r) was worked out by
Gaddum and by Schildt, and is:
r1 [B]/K
B
,
where K
B
is the dissociation equilibrium constant of the
reversible reaction of the antagonist with its receptor. K
B
has
units of concentration and is the concentration of antagonist
needed to occupy half the receptors in the absence of agonist.
The lower the value of K
B
, the more potent is the drug. If sev-
eral concentrations of a competitive antagonist are studied
and the dose ratio is measured at each concentration, a plot of
(r1) against [B] yields a straight line through the origin with
a slope of 1/K
B
(Figure 2.6a). Such measurements provided
the means of classifying and subdividing receptors in terms of
the relative potencies of different antagonists.
PARTIAL AGONISTS
Some drugs combine with receptors and activate them, but are
incapable of eliciting a maximal response, no matter how high
their concentration may be. These are known as partial agonists,
and are said to have low efficacy. Several partial agonists are
used in therapeutics, including buprenorphine(a partial agonist
at morphine μ-receptors, Chapter 25) and oxprenolol (partial
agonist at β-adrenoceptors).
Full agonists can elicit a maximal response when only a
small proportion of the receptors is occupied (underlying the
concept of ‘spare’ receptors), but this is not the case with par-
tial agonists, where a substantial proportion of the receptors
need to be occupied to cause a response. This has two clinical
consequences. First, partial agonists antagonize the effect of a
full agonist, because most of the receptors are occupied with
low-efficacy partial agonist with which the full agonist must
compete. Second, it is more difficult to reverse the effects of a
partial agonist, such as buprenorphine, with a competitive
antagonist such as naloxone, than it is to reverse the effects of
a full agonist such as morphine. Alarger fraction of the recep-
tors is occupied by buprenorphine than by morphine, and a
much higher concentration of naloxoneis required to compete
successfully and displace buprenorphinefrom the receptors.
SLOW PROCESSES
Prolonged exposure of receptors to agonists, as frequently
occurs in therapeutic use, can cause down-regulation or
desensitization. Desensitization is sometimes specific for a
particular agonist (when it is referred to as ‘homologous
desensitization’), or there may be cross-desensitization to dif-
ferent agonists (‘heterologous desensitization’). Membrane
receptors may become internalized. Alternatively, G-protein-
mediated linkage between receptors and effector enzymes
(e.g. adenylyl cyclase) may be disrupted. Since G-proteins link
several distinct receptors to the same effector molecule, this
can give rise to heterologous desensitization. Desensitization
is probably involved in the tolerance that occurs during
prolonged administration of drugs, such as morphine or
benzodiazepines (see Chapters 18 and 25).
Therapeutic effects sometimes depend on induction of tol-
erance. For example, analogues of gonadotrophin-releasing
hormone (GnRH), such as goserelin or buserelin, are used to
treat patients with metastatic prostate cancer (Chapter 48).
Gonadotrophin-releasing hormone is released physiologically
in a pulsatile manner. During continuous treatment with
buserelin, there is initial stimulation of luteinizing hormone
(LH) and follicle-stimulating hormone (FSH) release, followed
by receptor desensitization and suppression of LH and FSH
release. This results in regression of the hormone-sensitive
tumour.
100
50
0
10
9
10
8
510
9
[Antagonist]
Slope1/K
B
Dose ratio –1
(a)
log[Antagonist]
9 8 7
pA
2
Slope1
log (dose ratio) –1
2
1
0
(b)
Figure 2.6:Competitive antagonism. (a) A plot of antagonist concentration vs. (dose ratio 1) gives a straight line through the origin.
(b) A log–log plot (a Schildt plot) gives a straight line of unit slope. The potency of the antagonist (pA
2
) is determined from the intercept
of the Schildt plot.
10 MECHANISMS OF DRUG ACTION (PHARMACODYNAMICS)
Conversely, reduced exposure of a cell or tissue to an agon-
ist (e.g. by denervation) results in increased receptor numbers
and supersensitivity. Prolonged use of antagonists may pro-
duce an analogous effect. One example of clinical importance
is increased β-adrenoceptor numbers following prolonged use
of beta-blockers. Abrupt drug withdrawal can lead to tachy-
cardia and worsening angina in patients who are being treated
for ischaemic heart disease.
NON-RECEPTOR MECHANISMS
In contrast to high-potency/high-selectivity drugs which com-
bine with specific receptors, some drugs exert their effects via
simple physical properties or chemical reactions due to their
presence in some body compartment. Examples include antacids
(which neutralize gastric acid), osmotic diuretics (which increase
the osmolality of renal tubular fluid), and bulk and lubricating
laxatives. These agents are of low potency and specificity, and
hardly qualify as ‘drugs’ in the usual sense at all, although some
of them are useful medicines. Oxygen is an example of a highly
specific therapeutic agent that is used in high concentrations
(Chapter 33). Metal chelating agents, used for example in the
treatment of poisoning with ferrous sulphate, are examples of
drugs that exert their effects through interaction with small
molecular species rather than with macromolecules, yet which
possess significant specificity.
General anaesthetics (Chapter 24) have low molar poten-
cies determined by their oil/water partition coefficients, and
low specificity.
Key points
Most drugs are potent and specific; they combine with
receptors for endogenous mediators or with high affinity
sites on enzymes or other proteins, e.g. ion-transport
mechanisms.
There are four superfamilies of receptors; three are
membrane bound:
directly linked to ion channel (e.g. nicotinic
acetylcholine receptor);
linked via G-proteins to an enzyme, often adenylyl
cyclase (e.g. β
2
-receptors);
directly coupled to the catalytic domain of an
enzyme (e.g. insulin)
The fourth superfamily is intracellular, binds to DNA
and controls gene transcription and protein synthesis
(e.g. steroid receptors).
Many drugs work by antagonizing agonists. Drug
antagonism can be:
competitive;
non-competitive;
physiological.
Partial agonists produce an effect that is less than the
maximum effect of a full agonist. They antagonize full
agonists.
Tolerance can be important during chronic
administration of drugs acting on receptors, e.g.
central nervous system (CNS) active agents.
Case history
A young man is brought unconscious into the Accident and
Emergency Department. He is unresponsive, hypoventilat-
ing, has needle tracks on his arms and pinpoint pupils.
Naloxone is administered intravenously and within 30
seconds the patient is fully awake and breathing normally.
He is extremely abusive and leaves hospital having
attempted to assault the doctor.
Comment
The clinical picture is of opioid overdose, and this was con-
firmed by the response to naloxone, a competitive antag-
onist of opioids at μ-receptors (Chapter 25). It would have
been wise to have restrained the patient before adminis-
tering naloxone, which can precipitate withdrawal symp-
toms. He will probably become comatose again shortly
after discharging himself, as naloxone has a much shorter
elimination half-life than opioids such as morphine or
diacetyl-morphine (heroin), so the agonist effect of the
overdose will be reasserted as the concentration of the
opiate antagonist falls.
FURTHER READING
Rang HP. The receptor concept: pharmacology’s big idea. British
Journal of Pharmacology2006; 147 (Suppl. 1): 9–16.
Rang HP, Dale MM, Ritter JM, Flower RD. Chapter 2, How drugs act:
general principles. Chapter 3, How drugs act: molecular aspects.
In: Rang and Dale’s pharmacology, 6th edn. London: Churchill
Livingstone, 2007.
CONSTANT-RATE INFUSION
If a drug is administered intravenously via a constant-rate
pump, and blood sampled from a distant vein for measure-
ment of drug concentration, a plot of plasma concentration
versus time can be constructed (Figure 3.1). The concentration
rises from zero, rapidly at first and then more slowly until a
plateau (representing steady state) is approached. At steady
state, the rate of input of drug to the body equals the rate of
elimination. The concentration at plateau is the steady-state
concentration (C
SS
). This depends on the rate of drug infusion
and on its ‘clearance’. The clearance is defined as the volume
of fluid (usually plasma) from which the drug is totally elimi-
nated (i.e. ‘cleared’) per unit time. At steady state,
administration rateelimination rate
elimination rateC
SS
clearance
so
clearanceadministration rate/C
SS
Introduction 11
Constant-rate infusion 11
Single-bolus dose 12
Repeated (multiple) dosing 13
Deviations from the one-compartment model
with first-order elimination 14
Non-linear (‘dose-dependent’) pharmacokinetics 15
CHAPTER 3
PHARMACOKINETICS
INTRODUCTION
Pharmacokinetics is the study of drug absorption, distribu-
tion, metabolism and excretion (ADME) – ‘what the body does
to the drug’. Understanding pharmacokinetic principles, com-
bined with specific information regarding an individual drug
and patient, underlies the individualized optimal use of the
drug (e.g. choice of drug, route of administration, dose and
dosing interval).
Pharmacokinetic modelling is based on drastically simplif-
ying assumptions; but even so, it can be mathematically cum-
bersome, sadly rendering this important area unintelligible to
many clinicians. In this chapter, we introduce the basic con-
cepts by considering three clinical dosing situations:
constant-rate intravenous infusion;
bolus-dose injection;
repeated dosing.
Bulk flow in the bloodstream is rapid, as is diffusion over
short distances after drugs have penetrated phospholipid mem-
branes, so the rate-limiting step in drug distribution is usually
penetration of these membrane barriers. Permeability is deter-
mined mainly by the lipid solubility of the drug, polar water-
soluble drugs being transferred slowly, whereas lipid-soluble,
non-polar drugs diffuse rapidly across lipid-rich membranes.
In addition, some drugs are actively transported by specific
carriers.
The simplest pharmacokinetic model treats the body as a
well-stirred single compartment in which an administered
drug distributes instantaneously, and from which it is elimi-
nated. Many drugs are eliminated at a rate proportional to
their concentration – ‘first-order’ elimination. A single (one)-
compartment model with first-order elimination often approx-
imates the clinical situation surprisingly well once absorption
and distribution have occurred. We start by considering this,
and then describe some important deviations from it.
[Drug] in plasma
Constant infusion of drug
Time
Figure 3.1:Plasma concentration of a drug during and after a
constant intravenous infusion as indicated by the bar.
12 PHARMACOKINETICS
Clearance is the best measure of the efficiency with which a
drug is eliminated from the body, whether by renal excretion,
metabolism or a combination of both. The concept will be
familiar from physiology, where clearances of substances with
particular properties are used as measures of physiologically
important processes, including glomerular filtration rate and
renal or hepatic plasma flow. For therapeutic drugs, knowing
the clearance in an individual patient enables the physician
to adjust the maintenance dose to achieve a desired target
steady-state concentration, since
required administration rate desired C
SS
clearance
This is useful in drug development. It is also useful in clinical
practice when therapy is guided by plasma drug concentrations.
However, such situations are limited (Chapter 8). Furthermore,
some chemical pathology laboratories report plasma concentra-
tions of drugs in molar terms, whereas drug doses are usually
expressed in units of mass. Consequently, one needs to know the
molecular weight of the drug to calculate the rate of administra-
tion required to achieve a desired plasma concentration.
When drug infusion is stopped, the plasma concentration
declines towards zero. The time taken for plasma concentration
to halve is the half-life (t
1/2
). A one-compartment model with
first-order elimination predicts an exponential decline in con-
centration when the infusion is discontinued, as shown in
Figure 3.1. After a second half-life has elapsed, the concentration
will have halved again (i.e. a 75% drop in concentration to 25%
of the original concentration), and so on. The increase in drug
concentration when the infusion is started is also exponential,
being the inverse of the decay curve. This has a very important
clinical implication, namely that t
1/2
not only determines the
time-course of disappearance when administration is stopped,
but also predicts the time-course of its accumulation to steady
state when administration is started.
Half-life is a very useful concept, as explained below.
However, it is not a direct measure of drug elimination, since
differences in t
1/2
can be caused either by differences in the effi-
ciency of elimination (i.e. the clearance) or differences in another
important parameter, the apparent volume of distribution (V
d
).
Clearance and not t
1/2
must therefore be used when a measure
of the efficiency with which a drug is eliminated is required.
SINGLE-BOLUS DOSE
The apparent volume of distribution (V
d
) defines the relation-
ship between the mass of a bolus dose of a drug and the
plasma concentration that results. V
d
is a multiplying factor
relating the amount of drug in the body to the plasma concen-
tration, C
p
(i.e. the amount of drug in the bodyC
p
V
d
).
Consider a very simple physical analogy. By definition, con-
centration (c) is equal to mass (m) divided by volume (v):
Thus if a known mass (say 300mg) of a substance is dissolved
in a beaker containing an unknown volume (v) of water, vcan
be estimated by measuring the concentration of substance in a
sample of solution. For instance, if the concentration is
0.1mg/mL, we would calculate that v 3000 mL (v m/c).
This is valid unless a fraction of the substance has become
adsorbed onto the surface of the beaker, in which case the
solution will be less concentrated than if all of the substance
had been present dissolved in the water. If 90% of the sub-
stance is adsorbed in this way, then the concentration in
solution will be 0.01mg/mL, and the volume will be corre-
spondingly overestimated, as 30000 mLin this example. Based
on the mass of substance dissolved and the measured concen-
tration, we might say that it is ‘as if’ the substance were dis-
solved in 30L of water, whereas the real volume of water in
the beaker is only 3L.
Now consider the parallel situation in which a known
mass of a drug (say 300mg) is injected intravenously into a
human. Suppose that distribution within the body occurs
instantaneously before any drug is eliminated, and that blood
is sampled and the concentration of drug measured in the
plasma is 0.1mg/mL. We could infer that it is as if the drug
has distributed in 3L, and we would say that this is the appar-
ent volume of distribution. If the measured plasma concen-
tration was 0.01mg/mL, we would say that the apparent
volume of distribution was 30L, and if the measured concen-
tration was 0.001mg/mL, the apparent volume of distribution
would be 300L.
What does V
d
mean? From these examples it is obvious that
it is not necessarily the real volume of a body compartment,
since it may be greater than the volume of the whole body. At the
lower end, V
d
is limited by the plasma volume (approximately
3L in an adult). This is the smallest volume in which a drug
could distribute following intravenous injection, but there is no
theoretical upper limit on V
d
, with very large values occurring
when very little of the injected dose remains in the plasma, most
being taken up into fat or bound to tissues.
c
m
v
Key points
Pharmacokinetics deals with how drugs are handled by
the body, and includes drug absorption, distribution,
metabolism and excretion.
Clearance (Cl ) is the volume of fluid (usually plasma)
from which a drug is totally removed (by metabolism
excretion) per unit time.
During constant i.v. infusion, the plasma drug
concentration rises to a steady state (C
SS
) determined by
the administration rate (A) and clearance (C
SS
A/Cl ).
The rate at which C
SS
is approached, as well as the rate
of decline in plasma concentration when infusion is
stopped are determined by the half-life (t
1/2
).
The volume of distribution (V
d
) is an apparent volume
that relates dose (D) to plasma concentration (C): it is
‘as if’ dose Dmg was dissolved in V
d
L to give a
concentration of Cmg /L.
The loading dose is C
p
V
d
whereC
p
is the desired
plasma concentration.
The maintenance dose C
SS
Cl, where C
SS
is the
steady-state concentration.
In reality, processes of elimination begin as soon as the
bolus dose (d) of drug is administered, the drug being cleared
at a rate Cl
s
(total systemic clearance). In practice, blood is
sampled at intervals starting shortly after administration
of the dose. Cl
s
is determined from a plot of plasma concentra-
tion vs. time by measuring the area under the plasma concen-
tration vs. time curve (AUC). (This is estimated mathematically
using a method called the trapezoidal rule – important in drug
development, but not in clinical practice.)
If the one-compartment, first-order elimination model holds,
there is an exponential decline in plasma drug concentration,
just as at the end of the constant rate infusion (Figure 3.2a). If
the data are plotted on semi-logarithmic graph paper, with
time on the abscissa, this yields a straight line with a negative
slope (Figure 3.2b). Extrapolation back to zero time gives the
concentration (c
0
) that would have occurred at time zero, and
this is used to calculate V
d
:
Half-life can be read off the graph as the time between any
point (c
1
) and the point at which the concentration c
2
has
decreased by 50%, i.e. c
1
/c
2
2. The slope of the line is the
elimination rate constant, k
el
:
t
1/2
andk
el
are related as follows:
V
d
is related partly to characteristics of the drug (e.g. lipid sol-
ubility) and partly to patient characteristics (e.g. body size,
t
l
kk
12
2
0 693
/
n
el el

.
k
Cl
V
el
s
d
V
d
c
d
0
Cl
d
s
AUC
plasma protein concentration, body water and fat content). In
general, highly lipid-soluble compounds that are able to pen-
etrate cells and fatty tissues have a larger V
d
than more polar
water-soluble compounds.
V
d
determines the peak plasma concentration after a bolus
dose, so factors that influence V
d
, such as body mass, need to
be taken into account when deciding on dose (e.g. by express-
ing dose per kg body weight). Body composition varies from
the usual adult values in infants or the elderly, and this also
needs to be taken into account in dosing such patients (see
Chapters 10 and 11).
V
d
identifies the peak plasma concentration expected
following a bolus dose. It is also useful to know V
d
when
considering dialysis as a means of accelerating drug
elimination in poisoned patients (Chapter 54). Drugs with a
large V
d
(e.g. many tricyclic antidepressants) are not removed
efficiently by haemodialysis because only a small fraction of
the total drug in the body is present in plasma, which is the
fluid compartment accessible to the artificial kidney.
If both V
d
andt
1/2
are known, they can be used to estimate
the systemic clearance of the drug using the expression:
Note that clearance has units of volume/unit time (e.g.
mL/min),V
d
has units of volume (e.g. mLor L ), t
1/2
has units
of time (e.g. minutes) and 0.693 is a constant arising because
ln(0.5) ln 2 0.693. This expression relates clearance to V
d
andt
1/2
, but unlike the steady-state situation referred to above
during constant-rate infusion, or using the AUC method fol-
lowing a bolus, it applies only when a single-compartment
model with first-order elimination kinetics is applicable.
Cl
V
t
s
d
/
0693
12
.
REPEATED(MULTIPLE) DOSING 13
Key points
The ‘one-compartment’ model treats the body as a
single, well-stirred compartment. Immediately
following a bolus dose D, the plasma concentration
rises to a peak (C
0
) theoretically equal to D/V
d
and then
declines exponentially.
The rate constant of this process (k
el
) is given by Cl/V
d
.
k
el
is inversely related to t
1/2
, which is given by 0.693/k
el
.
Thus,Cl 0.693 V
d
/t
1/2
.
Repeated bolus dosing gives rise to accumulation
similar to that observed with constant-rate infusion,
but with oscillations in plasma concentration rather
than a smooth rise. The size of the oscillations is
determined by the dose interval and by t
1/2
. The steady
state concentration is approached (87.5%) after three
half-lives have elapsed.
REPEATED (MULTIPLE) DOSING
If repeated doses are administered at dosing intervals much
greater than the drug’s elimination half-life, little if any accu-
mulation occurs (Figure 3.3a). Drugs are occasionally used in
[Drug] in plasma
Time(a)
Log [Drug] in plasma
Time(b)
Figure 3.2:One-compartment model. Plasma concentration–time
curve following a bolus dose of drug plotted (a) arithmetically
and (b) semi-logarithmically. This drug fits a one-compartment
model, i.e. its concentration falls exponentially with time.
this way (e.g. penicillinto treat a mild infection), but a steady
state concentration greater than some threshold value is often
needed to produce a consistent effect throughout the dose
interval. Figure 3.3b shows the plasma concentration–time
curve when a bolus is administered repeatedly at an interval
less than t
1/2
. The mean concentration rises toward a plateau,
as if the drug were being administered by constant-rate infu-
sion. That is, after one half-life the mean concentration is 50%
of the plateau (steady-state) concentration, after two half-lives
it is 75%, after three half-lives it is 87.5%, and after four
half-lives it is 93.75%. However, unlike the constant-rate infu-
sion situation, the actual plasma concentration at any time
swings above or below the mean level. Increasing the dosing
frequency smoothes out the peaks and troughs between doses,
while decreasing the frequency has the opposite effect. If the
peaks are too high, toxicity may result, while if the troughs are
too low there may be a loss of efficacy. If a drug is adminis-
tered once every half-life, the peak plasma concentration (C
max
)
will be double the trough concentration (C
min
). In practice, this
amount of variation is tolerable in many therapeutic situa-
tions, so a dosing interval approximately equal to the half-life
is often acceptable.
Knowing the half-life alerts the prescriber to the likely
time-course over which a drug will accumulate to steady
state. Drug clearance, especially renal clearance, declines with
age (see Chapter 11). A further pitfall is that several drugs
have active metabolites that are eliminated more slowly than
the parent drug. This is the case with several of the benzodi-
azepines (Chapter 18), which have active metabolites with
half-lives of many days. Consequently, adverse effects (e.g. con-
fusion) may appear only when the steady state is approached
after several weeks of treatment. Such delayed effects may
incorrectly be ascribed to cognitive decline associated with
ageing, but resolve when the drug is stopped.
Knowing the half-life helps a prescriber to decide whether
or not to initiate treatment with a loading dose. Consider
digoxin (half-life approximately 40 hours). This is usually
prescribed once daily, resulting in a less than two-fold varia-
tion in maximum and minimum plasma concentrations, and
reaching 90% of the mean steady-state concentration in
approximately one week (i.e. four half-lives). In many clinical
situations, such a time-course is acceptable. In more urgent
situations a more rapid response can be achieved by using a
loading dose. The loading dose (LD) can be estimated by mul-
tiplying the desired concentration by the volume of distribu-
tion (LDC
p
V
d
).
DEVIATIONS FROM THE
ONE-COMPARTMENT MODEL WITH
FIRST-ORDER ELIMINATION
TWO-COMPARTMENT MODEL
Following an intravenous bolus a biphasic decline in plasma
concentration is often observed (Figure 3.4), rather than a sim-
ple exponential decline. The two-compartment model (Figure
3.5) is appropriate in this situation. This treats the body as a
smaller central plus a larger peripheral compartment. Again,
these compartments have no precise anatomical meaning,
although the central compartment is assumed to consist of
14 PHARMACOKINETICS
[Drug] in plasma
(a)
[Drug] in plasma
Time
(b)
Figure 3.3:Repeated bolus dose injections (at arrows) at (a)
intervals much greater than t
1/2
and (b) intervals less than t
1/2
.
Figure 3.4:Two-compartment model. Plasma concentration–time
curve (semi-logarithmic) following a bolus dose of a drug that fits
a two-compartment model.
blood (from which samples are taken for analysis) plus the
extracellular spaces of some well-perfused tissues. The periph-
eral compartment consists of less well-perfused tissues into
which drug permeates more slowly.
The initial rapid fall is called the α phase, and mainly
reflects distribution from the central to the peripheral com-
partment. The second, slower phase reflects drug elimination.
It is called the βphase, and the corresponding t
1/2
is known as
t
1/2
. This is the appropriate value for clinical use.
NON-LINEAR (‘DOSE-DE PENDENT’)
PHARMACOKINETICS
Although many drugs are eliminated at a rate that is approxi-
mately proportional to their concentration (‘first-order’ kinet-
ics), there are several therapeutically important exceptions.
Consider a drug that is eliminated by conversion to an
inactive metabolite by an enzyme. At high concentrations, the
enzyme becomes saturated. The drug concentration and reac-
tion velocity are related by the Michaelis–Menten equation
(Figure 3.6). At low concentrations, the rate is linearly related
to concentration, whereas at saturating concentrations the rate
is independent of concentration (‘zero-order’ kinetics). The
same applies when a drug is eliminated by a saturable trans-
port process. In clinical practice, drugs that exhibit non-linear
kinetics are the exception rather than the rule. This is because
most drugs are used therapeutically at doses that give rise to
concentrations that are well below the Michaelis constant
(K
m
), and so operate on the lower, approximately linear, part
of the Michaelis–Menten curve relating elimination velocity to
plasma concentration.
Drugs that show non-linear kinetics in the therapeutic
range include heparin, phenytoin and ethanol. Some drugs
(e.g. barbiturates) show non-linearity in the part of the toxic
range that is encountered clinically. Implications of non-linear
pharmacokinetics include:
1. The decline in concentration vs. time following a bolus
dose of such a drug is not exponential. Instead, elimination
NON-LINEAR (‘DOSE-DEPENDENT’) PHARMACOKINETI CS 15
Drug
Central
compartment
Peripheral (tissue)
compartment
Elimination
Blood
sample
Figure 3.5:Schematic representation of a two-compartment
model.
[S]
Velocity (V)
K
m
V
max
Figure 3.6:Michaelis–Menten relationship between the velocity
(V) of an enzyme reaction and the substrate concentration ([S]).
[S] at 50% V
max
is equal to K
m
, the Michaelis–Menten constant.
[Drug] in plasma
1
10
100
Time
Figure 3.7:Non-linear kinetics: plasma concentration–time curve
following administration of a bolus dose of a drug eliminated by
Michaelis–Menten kinetics.
y
Figure 3.8:Non-linear kinetics: steady-state plasma concentration
of a drug following repeated dosing as a function of dose.
FURTHER READING
Rowland M, Tozer TN. Therapeutic regimens. In: Clinical pharmacoki-
netics: concepts and applications, 3rd edn. Baltimore, MD: Williams
and Wilkins, 1995: 53–105.
Birkett DJ. Pharmacokinetics made easy (revised), 2nd edn. Sydney:
McGraw-Hill, 2002. (Lives up to the promise of its title!)
begins slowly and accelerates as plasma concentration
falls (Figure 3.7).
2. The time required to eliminate 50% of a dose increases
with increasing dose, so half-life is not constant.
3. Amodest increase in dose of such a drug disproportionately
increases the amount of drug in the body once the drug-
elimination process is saturated (Figure 3.8). This is very
important clinically when using plasma concentrations of,
for example, phenytoinas a guide to dosing.
16 PHARMACOKINETICS
Case history
A young man develops idiopathic epilepsy and treatment
is started with phenytoin, 200mg daily, given as a single
dose last thing at night. After a week, the patient’s serum
phenytoin concentration is 25
μmol/L. (Therapeutic range is
40–80
μmol/L.) The dose is increased to 300mg/day. One
week later he is complaining of unsteadiness, there is nys-
tagmus and the serum concentration is 125
μmol/L. The
dose is reduced to 250mg/day. The patient’s symptoms
slowly improve and the serum phenytoin concentration
falls to 60
μmol/L (within the therapeutic range).
Comment
Phenytoin shows dose-dependent kinetics; the serum con-
centration at the lower dose was below the therapeutic
range, so the dose was increased. Despite the apparently
modest increase (to 150% of the original dose), the plasma
concentration rose disproportionately, causing symptoms
and signs of toxicity (see Chapter 22).
Key points
Two-compartment model. Following a bolus dose the
plasma concentration falls bi-exponentially, instead
of a single exponential as in the one-compartment
model. The first () phase mainly represents
distribution; the second () phase mainly represents
elimination.
Non-linear (‘dose-dependent’) kinetics. If the
elimination process (e.g. drug-metabolizing enzyme)
becomes saturated, the clearance rate falls.
Consequently, increasing the dose causes a
disproportionate increase in plasma concentration.
Drugs which exhibit such properties (e.g. phenytoin)
are often difficult to use in clinical practice.
Introduction 17
Bioavailability, bioequivalence and generic vs.
proprietary prescribing 17
Prodrugs 18
Routes of administration 19
CHAPTER 4
DRUG ABSORPTION AND ROUTES
OF ADMINISTRATION
INTRODUCTION
Drug absorption, and hence the routes by which a particular
drug may usefully be administered, is determined by the rate
and extent of penetration of biological phospholipid mem-
branes. These are permeable to lipid-soluble drugs, whilst pre-
senting a barrier to more water-soluble drugs. The most
convenient route of drug administration is usually by mouth,
and absorption processes in the gastro-intestinal tract are
among the best understood.
BIOAVAILABILITY, BIOEQUIVALENCE AND
GENERIC VS. PROPRIETARY PRESCRIBING
Drugs must enter the circulation if they are to exert a systemic
effect. Unless administered intravenously, most drugs are
absorbed incompletely (Figure 4.1). There are three reasons
for this:
1. the drug is inactivated within the gut lumen by gastric
acid, digestive enzymes or bacteria;
2. absorption is incomplete; and
3. presystemic (‘first-pass’) metabolism occurs in the gut
wall and liver.
Together, these processes explain why the bioavailability of
an orally administered drug is typically less than 100%.
Bioavailability of a drug formulation can be measured experi-
mentally (Figure 4.2) by measuring concentration vs. time
curves following administration of the preparation via its
intended route (e.g. orally) and of the same dose given intra-
venously (i.v.).
BioavailabilityAUCoral/AUCi.v. 100%
Many factors in the manufacture of the drug formulation influ-
ence its disintegration, dispersion and dissolution in the gastro-
intestinal tract. Pharmaceutical factors are therefore important
in determining bioavailability. It is important to distinguish
statistically significant from clinically important differences in
this regard. The former are common, whereas the latter are not.
However, differences in bioavailability did account for an epi-
demic of phenytoin intoxication in Australia in 1968–69.
Affected patients were found to be taking one brand of pheny-
toin: the excipient had been changed from calcium sulphate to
lactose, increasing phenytoin bioavailability and thereby pre-
cipitating toxicity. An apparently minor change in the manufac-
turing process of digoxin in the UK resulted in reduced potency
due to poor bioavailability. Restoring the original manufactur-
ing conditions restored potency but led to some confusion, with
both toxicity and underdosing.
These examples raise the question of whether prescribing
should be by generic name or by proprietary (brand) name.
When a new preparation is marketed, it has a proprietary name
Systemic
circulation
Oral
administration
Inactivation
in liver
Inactivation
in gut lumen
Inactivation
in stomach
Inactivation
in gut wall
Portal blood
Incomplete
absorption
Figure 4.1:Drug bioavailability following oral administration may
be incomplete for several reasons.
supplied by the pharmaceutical company, and a non-proprietary
(generic) name. It is usually available only from the company
that introduced it until the patent expires. After this, other com-
panies can manufacture and market the product, sometimes
under its generic name. At this time, pharmacists usually shop
around for the best buy. If a hospital doctor prescribes by propri-
etary name, the same drug produced by another company may
be substituted. This saves considerable amounts of money. The
attractions of generic prescribing in terms of minimizing costs
are therefore obvious, but there are counterarguments, the
strongestof which relates to the bioequivalence or otherwise of
the proprietary product with its generic competitors. The for-
mulation of a drug (i.e. excipients, etc.) differs between different
manufacturers’ products of the same drug, sometimes affecting
bioavailability. This is a particular concern with slow-release or
sustained-release preparations, or preparations to be adminis-
tered by different routes. Drug regulatory bodies have strict cri-
teria to assess whether such products can be licensed without
the full dataset that would be required for a completely new
product (i.e. one based on a new chemical entity).
It should be noted that the absolute bioavailability of two
preparations may be the same (i.e. the same AUC), but that the
kinetics may be very different (e.g. one may have a much
higher peak plasma concentration than the other, but a shorter
duration). The rate at which a drug enters the body determines
the onset of its pharmacological action, and also influences the
intensity and sometimes the duration of its action, and is
important in addition to the completeness of absorption.
Prescribers need to be confident that different preparations
(brand named or generic) are sufficiently similar for their sub-
stitution to be unlikely to lead to clinically important alter-
ations in therapeutic outcome. Regulatory authorities have
responded to this need by requiring companies who are seek-
ing to introduce generic equivalents to present evidence that
their product behaves similarly to the innovator product that
is already marketed. If evidence is presented that a new
generic product can be treated as therapeutically equivalent to
the current ‘market leader’, this is accepted as ‘bioequiva-
lence’. This does not imply that all possible pharmacokinetic
parameters are identical between the two products, but that
any such differences are unlikely to be clinically important.
It is impossible to give a universal answer to the generic vs.
proprietary issue. However, substitution of generic for brand-
name products seldom causes obvious problems, and excep-
tions (e.g. different formulations of the calcium antagonist
diltiazem, see Chapter 29) are easily flagged up in formularies.
PRODRUGS
One approach to improving absorption or distribution to a rel-
atively inaccessible tissue (e.g. brain) is to modify the drug
molecule chemically to form a compound that is better
absorbed and from which active drug is liberated after absorp-
tion. Such modified drugs are termed prodrugs (Figure 4.3).
Examples are shown in Table 4.1.
18 DRUG ABSORPTION AND ROUTES OF ADMINISTRATION
i.v.dosing
Oral dosing
[Drug] in plasma
100
10
1
Time
Figure 4.2:Oral vs. intravenous dosing: plasma concentration–time
curves following administration of a drug i.v. or by mouth (oral).
Key points
Drugs must cross phospholipid membranes to reach the
systemic circulation, unless they are administered
intravenously. This is determined by the lipid solubility of
the drug and the area of membrane available for
absorption, which is very large in the case of the ileum,
because of the villi and microvilli. Sometimes polar drugs
can be absorbed via specific transport processes (carriers).
Even if absorption is complete, not all of the dose may
reach the systemic circulation if the drug is metabolized
by the epithelium of the intestine, or transported
back into lumen of the intestine or metabolized in the
liver, which can extract drug from the portal blood
before it reaches the systemic circulation via the
hepatic vein. This is called presystemic (or ‘first-pass’)
metabolism.
‘Bioavailability’ describes the completeness of
absorption into the systemic circulation. The amount of
drug absorbed is determined by measuring the plasma
concentration at intervals after dosing and integrating
by estimating the area under the plasma
concentration/time curve (AUC). This AUC is expressed as
a percentage of the AUC when the drug is administered
intravenously (100% absorption). Zero per cent
bioavailability implies that no drug enters the systemic
circulation, whereas 100% bioavailability means that all
of the dose is absorbed into the systemic circulation.
Bioavailability may vary not only between different
drugs and different pharmaceutical formulations of the
same drug, but also from one individual to another
,
depending on factors such as dose, whether the dose
is taken on an empty stomach, and the presence of
gastro-intestinal disease, or other drugs.
The rate of absorption is also important (as well as the
completeness), and is expressed as the time to peak
plasma concentration (T
max
). Sometimes it is desirable
to formulate drugs in slow-release preparations to
permit once daily dosing and/or to avoid transient
adverse effects corresponding to peak plasma
concentrations. Substitution of one such preparation
for another may give rise to clinical problems unless the
preparations are ‘bioequivalent’. Regulatory authorities
therefore require evidence of bioequivalence before
licensing generic versions of existing products.
Prodrugs are metabolized to pharmacologically active
products. They provide an approach to improving
absorption and distribution.
ROUTES OF ADMINISTRATION
ORAL ROUTE
FOR LOCAL EFFECT
Oral drug administration may be used to produce local effects
within the gastro-intestinal tract. Examples include antacids,
and sulphasalazine, which delivers 5-amino salicylic acid
(5-ASA) to the colon, thereby prolonging remission in patients
with ulcerative colitis (Chapter 34). Mesalazine has a pH-
dependent acrylic coat that degrades at alkaline pH as in the
colon and distal part of the ileum. Olsalazine is a prodrug con-
sisting of a dimer of two 5-ASAmoieties joined by a bond that is
cleaved by colonic bacteria.
FOR SYSTEMIC EFFECT
Oral administration of drugs is safer and more convenient for
the patient than injection. There are two main mechanisms of
drug absorption by the gut (Figure 4.4).
Passive diffusion
This is the most important mechanism. Non-polar lipid-soluble
agents are well absorbed from the gut, mainly from the small
intestine, because of the enormous absorptive surface area
provided by villi and microvilli.
Active transport
This requires a specific carrier. Naturally occurring polar
substances, including sugars, amino acids and vitamins, are
absorbed by active or facilitated transport mechanisms. Drugs
that are analogues of such molecules compete with them for
transport via the carrier. Examples include L-dopa, methotrex-
ate, 5-fluorouracil and lithium (which competes with sodium
ions for absorption).
Other factors that influence absorption include:
1. surgical interference with gastric function – gastrectomy
reduces absorption of several drugs;
2. disease of the gastro-intestinal tract (e.g. coeliac disease,
cystic fibrosis) – the effects of such disease are
unpredictable, but often surprisingly minor (see
Chapter 7);
3. the presence of food – the timing of drug administration in
relation to meal times can be important. Food and drink
dilute the drug and can bind it, alter gastric emptying and
increase mesenteric and portal blood flow;
ROUTES OF ADMINISTRATION 19
Passive diffusion of a
water-soluble drug
through an aquas
channel or pore
Passive diffusion
of a lipid-soluble
drug
Carrier-mediated
active transport
of drug
Lumen
Drug
Epithelial
cell
membrane
ATP
D
D
D
D
D
D
D
D
D
D
D
D
D
D
Figure 4.4:Modes of absorption of drugs
from the gut.
PRODRUG
DRUG
Relatively well-
absorbed and/or
good tissue
penetration
INACTIVE
Various
enzymes
in body
Relatively poorly
absorbed and/or
poor tissue
penetration
ACTIVE
Figure 4.3:Clinical use of prodrugs.
Table 4.1:Prodrugs
Prodrug Product
Enalapril Enalaprilat
Benorylate Aspirin and paracetamol
Levodopa Dopamine
Minoxidil Minoxidil sulphate
Carbimazole Methimazole
Vanciclovir Aciclovir
4. drug metabolism by intestinal flora – this may affect drug
absorption. Alteration of bowel flora (e.g. by concomitant
use of antibiotics) can interrupt enterohepatic recycling and
cause loss of efficacy of oral contraceptives (Chapter 13);
5. drug metabolism by enzymes (e.g. cytochrome P450 family
3A(CYP3A)) in the gastro-intestinal epithelium
(Chapter 5);
6. drug efflux back into the gut lumen by drug transport
proteins (e.g. P-glycoprotein (P-gp), ABCB1).
Prolonged action and sustained-release preparations
Some drugs with short elimination half-lives need to be adminis-
tered frequently, at inconveniently short intervals, making adher-
ence to the prescribed regimen difficult for the patient. Adrug
with similar actions, but a longer half-life, may need to be substi-
tuted. Alternatively, there are various pharmaceutical means of
slowing absorption of a rapidly eliminated drug. The aim of such
sustained-release preparations is to release a steady ‘infusion’ of
drug into the gut lumen for absorption during transit through
the small intestine. Reduced dosing frequency may improve
compliance and, in the case of some drugs (e.g. carbamazepine),
reduce adverse effects linked to high peak plasma concentra-
tions. Absorption of such preparations is often incomplete, so it is
especially important that bioavailability is established and sub-
stitution of one preparation for another may lead to clinical prob-
lems. Other limitations of slow-release preparations are:
1. Transit time through the small intestine is about six hours,
so once daily dosing may lead to unacceptably low trough
concentrations.
2. If the gut lumen is narrowed or intestinal transit is slow,
as in the elderly, or due to other drugs (tricyclic
antidepressants, opiates), there is a danger of high local
drug concentrations causing mucosal damage.
Osmosin™, an osmotically released formulation of
indometacin, had to be withdrawn because it caused
bleeding and ulceration of the small intestine.
3. Overdose with sustained-release preparations is difficult
to treat because of delayed drug absorption.
4. Sustained-release tablets should not be divided.
5. Expense.
BUCCAL AND SUBLI NGUAL ROUTE
Drugs are administered to be retained in the mouth for local
disorders of the pharynx or buccal mucosa, such as aphthous
ulcers (hydrocortisonelozenges or carbenoxolone granules).
Sublingual administration has distinct advantages over oral
administration (i.e. the drug to be swallowed) for drugs with
pronounced presystemic metabolism, providing direct and
rapid access to the systemic circulation, bypassing the intestine
and liver. Glyceryl trinitrate,buprenorphine and fentanyl are
given sublingually for this reason. Glyceryl trinitrateis taken
either as a sublingual tablet or as a spray. Sublingual adminis-
tration provides short-term effects which can be terminated by
swallowing the tablet. Tablets for buccal absorption provide
more sustained plasma concentrations, and are held in one
spot between the lip and the gum until they have dissolved.
RECTAL ROUTE
Drugs may be given rectally for local effects (e.g. to treat proc-
titis). The following advantages have been claimed for the rec-
tal route of administration of systemically active drugs:
1. Exposure to the acidity of the gastric juice and to digestive
enzymes is avoided.
2. The portal circulation is partly bypassed, reducing
presystemic (first pass) metabolism.
3. For patients who are unable to swallow or who are
vomiting.
Rectaldiazepam is useful for controlling status epilepticus in
children. Metronidazoleis well absorbed when administered
rectally, and is less expensive than intravenous preparations.
However, there are usually more reliable alternatives, and
drugs that are given rectally can cause severe local irritation.
SKIN
Drugs are applied topically to treat skin disease (Chapter 51).
Systemic absorption via the skin can cause undesirable effects,
for example in the case of potent glucocorticoids, but the
application of drugs to skin can also be used to achieve a sys-
temic therapeutic effect (e.g. fentanyl patches for analgesia).
The skin has evolved as an impermeable integument, so the
problems of getting drugs through it are completely different
from transport through an absorptive surface such as the gut.
Factors affecting percutaneous drug absorption include:
1. skin condition – injury and disease;
2. age – infant skin is more permeable than adult skin;
3. region –plantar forearm scalp scrotum posterior
auricular skin;
4. hydration of the stratum corneum – this is very important.
Increased hydration increases permeability. Plastic-film
occlusion (sometimes employed by dermatologists)
increases hydration. Penetration of glucocorticosteroids is
increased up to 100-fold, and systemic side effects are
more common;
5. vehicle – little is known about the importance of the
various substances which over the years have been
empirically included in skin creams and ointments. The
physical chemistry of these mixtures may be very complex
and change during an application;
6. physical properties of the drug – penetration increases with
increasing lipid solubility. Reduction of particle size
enhances absorption, and solutions penetrate best of all;
7. surface area to which the drug is applied – this is especially
important when treating infants who have a relatively
large surface area to volume ratio.
20 DRUG ABSORPTION AND ROUTES OF ADMINISTRATION
caused by timolol eyedrops given for open-angle glaucoma.
However, such absorption is not sufficiently reliable to make
use of these routes for therapeutic ends.
INTRAMUSCULAR INJECTION
Many drugs are well absorbed when administered intramus-
cularly. The rate of absorption is increased when the solution is
distributed throughout a large volume of muscle. Dispersion is
enhanced by massage of the injection site. Transport away from
the injection site is governed by muscle blood flow, and this
varies from site to site (deltoidvastus lateralis gluteus max-
imus). Blood flow to muscle is increased by exercise and absorp-
tion rates are increased in all sites after exercise. Conversely,
shock, heart failure or other conditions that decrease muscle
blood flow reduce absorption.
The drug must be sufficiently water soluble to remain in
solution at the injection site until absorption occurs. This is a
problem for some drugs, including phenytoin, diazepamand
digoxin, as crystallization and/or poor absorption occur when
these are given by intramuscular injection, which should there-
fore be avoided. Slow absorption is useful in some circum-
stances where appreciable concentrations of drug are required
for prolonged periods. Depot intramuscular injections are
used to improve compliance in psychiatric patients (e.g. the
decanoate ester of fluphenazinewhich is slowly hydrolysed to
release active free drug).
Intramuscular injection has a number of disadvantages:
1. pain – distension with large volumes is painful, and
injected volumes should usually be no greater than 5mL;
2. sciatic nerve palsy following injection into the buttock –
this is avoided by injecting into the upper outer gluteal
quadrant;
3. sterile abscesses at the injection site (e.g. paraldehyde);
4. elevated serum creatine phosphokinase due to enzyme
release from muscle can cause diagnostic confusion;
5. severe adverse reactions may be protracted because there
is no way of stopping absorption of the drug;
6. for some drugs, intramuscular injection is less effective
than the oral route;
7. haematoma formation.
SUBCUTANEOUS INJECTION
This is influenced by the same factors that affect intramuscular
injections. Cutaneous blood flow is lower than in muscle so
absorption is slower. Absorption is retarded by immobiliza-
tion, reduction of blood flow by a tourniquet and local cooling.
Adrenaline incorporated into an injection (e.g. of local anaes-
thetic) reduces the absorption rate by causing vasoconstriction.
Sustained effects from subcutaneous injections are extremely
important clinically, most notably in the treatment of insulin-
dependent diabetics, different rates of absorption being
achieved by different insulin preparations (see Chapter 37).
Transdermal absorption is sufficiently reliable to enable system-
ically active drugs (e.g. estradiol,nicotine, scopolamine) to be
administered by this route in the form of patches. Transdermal
administration bypasses presystemic metabolism. Patches are
more expensive than alternative preparations.
LUNGS
Drugs, notably steroids, β
2
-adrenoceptor agonists and mus-
carinic receptor antagonists, are inhaled as aerosols or particles
for their local effects on bronchioles. Nebulized antibiotics are
also sometimes used in children with cystic fibrosis and recur-
rent Pseudomonas infections. Physical properties that limit sys-
temic absorption are desirable. For example, ipratropium is a
quaternary ammonium ion analogue of atropine which is
highly polar, and is consequently poorly absorbed and has
reduced atropine-like side effects. A large fraction of an
‘inhaled’ dose of salbutamol is in fact swallowed. However,
the bioavailability of swallowed salbutamolis low due to inac-
tivation in the gut wall, so systemic effects such as tremor are
minimized in comparison to effects on the bronchioles.
The lungs are ideally suited for absorption from the gas
phase, since the total respiratory surface area is about 60m
2
,
through which only 60mL blood are percolating in the capil-
laries. This is exploited in the case of volatile anaesthetics, as
discussed in Chapter 24. Anasal/inhaled preparation of insulin
was introduced for type 2 diabetes (Chapter 37), but was not
commercially successful.
NOSE
Glucocorticoids and sympathomimetic amines may be admin-
istered intranasally for their local effects on the nasal mucosa.
Systemic absorption may result in undesirable effects, such as
hypertension.
Nasal mucosal epithelium has remarkable absorptive
properties, notably the capacity to absorb intact complex pep-
tides that cannot be administered by mouth because they
would be digested. This has opened up an area of therapeutics
that was previously limited by the inconvenience of repeated
injections. Drugs administered by this route include desmo-
pressin (DDAVP, an analogue of antidiuretic hormone) for
diabetes insipidus and buserelin(an analogue of gonadotrophin
releasing hormone) for prostate cancer.
EYE, EAR AND VAGINA
Drugs are administered topically to these sites for their local
effects (e.g. gentamicinor ciprofloxacin eyedrops for bacterial
conjunctivitis, sodium bicarbonate eardrops for softening wax,
and nystatin pessaries for Candida infections). Occasionally,
they are absorbed in sufficient quantity to have undesirable sys-
temic effects, such as worsening of bronchospasm in asthmatics
ROUTES OF ADMINISTRATION 21
Sustained effects have also been obtained from subcutaneous
injections by using oily suspensions or by implanting a pellet
subcutaneously (e.g. oestrogen or testosterone for hormone
replacement therapy).
INTRAVENOUS INJECTION
This has the following advantages:
1. rapid action (e.g. morphine for analgesia and furosemide
in pulmonary oedema);
2. presystemic metabolism is avoided (e.g. glyceryl trinitrate
infusion in patients with unstable angina);
3. intravenous injection is used for drugs that are not
absorbed by mouth (e.g. aminoglycosides (gentamicin)
and heparins). It is also used for drugs that are too painful
or toxic to be given intramuscularly. Cytotoxic drugs must
not be allowed to leak from the vein or considerable local
damage and pain will result as many of them are severe
vesicants (e.g. vincristine,doxorubicin);
4. intravenous infusion is easily controlled, enabling
precise titration of drugs with short half-lives. This is
essential for drugs such as sodium nitroprussideand
epoprostenol.
The main drawbacks of intravenous administration are as
follows:
1. Once injected, drugs cannot be recalled.
2. High concentrations result if the drug is given too rapidly –
the right heart receives the highest concentration.
3. Embolism of foreign particles or air, sepsis or
thrombosis.
4. Accidental extravascular injection or leakage of toxic drugs
(e.g.doxorubicin) produce severe local tissue necrosis.
5. Inadvertent intra-arterial injection can cause arterial
spasm and peripheral gangrene.
INTRATHECAL INJECTION
This route provides access to the central nervous system for
drugs that are normally excluded by the blood–brain barrier.
This inevitably involves very high risks of neurotoxicity, and
this route should never be used without adequate training. (In
the UK, junior doctors who have made mistakes of this kind
have been held criminally, as well as professionally, negligent.)
The possibility of causing death or permanent neurological
disability is such that extra care must be taken in checking that
both the drug and the dose are correct. Examples of drugs used
in this way include methotrexate and local anaesthetics (e.g.
levobupivacaine) or opiates, such as morphine and fentanyl.
(More commonly anaesthetists use the extradural route to
administer local anaesthetic drugs to produce regional analge-
sia without depressing respiration, e.g. in women during
labour.) Aminoglycosides are sometimes administered by
neuro-surgeons via a cisternal reservoir to patients with Gram-
negative infections of the brain. The antispasmodic baclofenis
sometimes administered by this route.
Penicillinused to be administered intrathecally to patients
with pneumococcal meningitis, because of the belief that it
penetrated the blood–brain barrier inadequately. However,
when the meninges are inflamed (as in meningitis), high-dose
intravenous penicillin results in adequate concentrations in
the cerebrospinal fluid. Intravenous penicillin should now
always be used for meningitis, since penicillinis a predictable
neurotoxin (it was formerly used to produce an animal model
of seizures), and seizures, encephalopathy and death have
been caused by injecting a dose intrathecally that would have
been appropriate for intravenous administration.points
22 DRUG ABSORPTION AND ROUTES OF ADMINISTRATION
Key points
Oral – generally safe and convenient
Buccal/sublingual – circumvents presystemic metabolism
Rectal – useful in patients who are vomiting
Transdermal – limited utility, avoids presystemic
metabolism
Lungs – volatile anaesthetics
Nasal – useful absorption of some peptides (e.g.
DDAVP; see Chapter 42)
Intramuscular – useful in some urgent situations (e.g.
behavioural emergencies)
Subcutaneous – useful for insulin and heparin in
particular
Intravenous – useful in emergencies for most rapid and
predictable action, but too rapid administration is
potentially very dangerous, as a high concentration
reaches the heart as a bolus
Intrathecal – specialized use by anaesthetists
Case history
The health visitor is concerned about an eight-month-old
girl who is failing to grow. The child’s mother tells you that
she has been well apart from a recurrent nappy rash, but
on examination there are features of Cushing’s syndrome.
On further enquiry, the mother tells you that she has been
applying clobetasone, which she had been prescribed her-
self for eczema, to the baby’s napkin area. There is no bio-
chemical evidence of endogenous over-production of
glucocorticoids. The mother stops using the clobetasone
cream on her daughter, on your advice. The features of
Cushing’s syndrome regress and growth returns to normal.
Comment
Clobetasone is an extremely potent steroid (see Chapter
50). It is prescribed for its top-ical effect, but can penetrate
skin, especially of an infant. The amount prescribed that is
appropriate for an adult would readily cover a large frac-
tion of an infant’s body surface area. If plastic pants are
used around the nappy this may increase penetration
through the skin (just like an occlusive dressing, which is
often deliberately used to increase the potency of topical
steroids; see Chapter 50), leading to excessive absorption
and systemic effects as in this case.
FURTHER READING
Fix JA. Strategies for delivery of peptides utilizing absorption-
enhancing agents. Journal of Pharmaceutical Sciences 1996; 85:
1282–5.
Goldberg M, Gomez-Orellana I. Challenges for the oral delivery
of macromolecules. Nature Reviews Drug Discovery 2003; 2:
289–95.
Mahato RI, Narang AS, Thoma L, Miller DD. Emerging trends in oral
delivery of peptide and protein drugs. Critical Reviews in
Therapeutic Drug Carrier Systems2003; 20: 153–2.
Mathiovitz E, Jacobs JS, Jong NS et al. Biologically erodable micros-
pheres as potential oral drug delivery systems. Nature1997; 386:
410–14.
Rowland M, Tozer TN. Clinical pharmacokinetics: concepts and applica-
tions, 3rd edn. Baltimore, MD: Williams and Wilkins, 1995: 11–50.
Skyler JS, Cefalu WT, Kourides I Aet al. Efficacy of inhaled human
insulin in type 1 diabetes mellitus: a randomized proof-of-concept
study. Lancet2001; 357: 324–5.
Varde NK, Pack DW. Microspheres for controlled release drug deliv-
ery. Expert Opinion on Biological Therapy2004; 4: 35–51.
ROUTES OF ADMINISTRATION 23
4-hydroxyphenytoin-glucuronide, which is readily excreted
viathe kidney.
PHASE I METABOLISM
The liver is the most important site of drug metabolism.
Hepatocyte endoplasmic reticulum is particularly important,
but the cytosol and mitochondria are also involved.
ENDOPLASMIC RETICULUM
Hepatic smooth endoplasmic reticulum contains the cytochrome
P450 (CYP450) enzyme superfamily (more than 50 different
CYPs have been found in humans) that metabolize foreign
substances – ‘xenobiotics’, i.e. drugs as well as pesticides, fertil-
izers and other chemicals ingested by humans. These metabolic
reactions include oxidation, reduction and hydrolysis.
OXIDATION
Microsomal oxidation causes aromatic or aliphatic hydroxyla-
tion, deamination, dealkylation or S-oxidation. These reac-
tions all involve reduced nicotinamide adenine dinucleotide
phosphate (NADP), molecular oxygen, and one or more of a
group of CYP450 haemoproteins which act as a terminal oxi-
dase in the oxidation reaction (or can involve other mixed
function oxidases, e.g. flavin-containing monooxygenases or
epoxide hydrolases). CYP450s exist in several distinct iso-
enzyme families and subfamilies with different levels of amino
acid homology. Each CYP subfamily has a different, albeit
often overlapping, pattern of substrate specificities. The major
drug metabolizing CYPs with important substrates, inhibitors
and inducers are shown in Table 5.1.
CYP450 enzymes are also involved in the oxidative
biosynthesis of mediators or other biochemically important
intermediates. For example, synthase enzymes involved in the
oxidation of arachidonic acid (Chapter 26) to prostaglandins
Introduction 24
Phase I metabolism 24
Phase II metabolism (transferase reactions) 25
Enzyme induction 27
Enzyme inhibition 28
Presystemic metabolism (‘first-pass’ effect) 28
Metabolism of drugs by intestinal organisms 29
CHAPTER 5
DRUG METABOLISM
INTRODUCTION
Drug metabolism is central to biochemical pharmacology.
Knowledge of human drug metabolism has been advanced by
the wide availability of human hepatic tissue, complemented by
analytical studies of parent drugs and metabolites in plasma and
urine.
The pharmacological activity of many drugs is reduced or
abolished by enzymatic processes, and drug metabolism is one
of the primary mechanisms by which drugs are inactivated.
Examples include oxidation of phenytoin and of ethanol.
However, not all metabolic processes result in inactivation, and
drug activity is sometimes increased by metabolism, as in acti-
vation of prodrugs (e.g. hydrolysis of enalapril, Chapter 28, to
its active metabolite enalaprilat). The formation of polar metabo-
lites from a non-polar drug permits efficient urinary excretion
(Chapter 6). However, some enzymatic conversions yield active
compounds with a longer half-life than the parent drug, causing
delayed effects of the long-lasting metabolite as it accumulates
more slowly to its steady state (e.g. diazepamhas a half-life of
20–50 hours, whereas its pharmacologically active metabolite
desmethyldiazepam has a plasma half-life of approximately
100 hours, Chapter 18).
Itis convenient to divide drug metabolism into two phases
(phasesI and II: Figure 5.1), which often, but not always, occur
sequentially.Phase Ireactions involve a metabolic modification
of the drug (commonly oxidation, reduction or hydrolysis).
Productsof phase I reactions may be either pharmacologically
active or inactive. Phase II reactions are synthetic conjugation
reactions. Phase II metabolites have increased polarity com-
paredto the parent drugs and are more readily excreted in the
urine(or, less often, in the bile), and they are usually – but not
always pharmacologically inactive. Molecules or groups
involvedin phase II reactions include acetate, glucuronic acid,
glutamine, glycine and sulphate, which may combine with
reactivegroups introduced during phase I metabolism (‘func-
tionalization’). For example, phenytoin is initially oxidized
to 4-hydroxyphenytoin which is then glucuronidated to
(dopamine, noradrenaline and adrenaline), tyramine, phenyl-
ephrine and tryptophan derivatives (5-hydroxytryptamine
and tryptamine). Oxidation of purines by xanthine oxidase
(e.g. 6-mercaptopurine is inactivated to 6-thiouric acid) is
non-microsomal.
REDUCTION
This includes, for example, enzymic reduction of double
bonds, e.g. methadone,naloxone.
HYDROLYSIS
Esterases catalyse hydrolytic conversions of many drugs.
Examples include the cleavage of suxamethonium by plasma
cholinesterase, an enzyme that exhibits pharmacogenetic varia-
tion (Chapter 14), as well as hydrolysis of aspirin(acetylsalicylic
acid) to salicylate, and the hydrolysis of enalaprilto enalaprilat.
PHASE II METABOLISM (TRANSFERASE
REACTIONS)
AMINO ACID REACTIONS
Glycine and glutamine are the amino acids chiefly involved in
conjugation reactions in humans. Glycine forms conjugates
with nicotinic acid and salicylate, whilst glutamine forms con-
jugates with p-aminosalicylate. Hepatocellular damage depletes
the intracellular pool of these amino acids, thus restricting this
pathway. Amino acid conjugation is reduced in neonates
(Chapter 10).
ACETYLATION
Acetate derived from acetyl coenzyme Aconjugates with several
drugs, including isoniazid,hydralazine and procainamide (see
Chapter 14 for pharmacogenetics of acetylation). Acetylating
activity resides in the cytosol and occurs in leucocytes, gastro-
intestinal epithelium and the liver (in reticulo-endothelial rather
than parenchymal cells).
GLUCURONIDATION
Conjugation reactions between glucuronic acid and carboxyl
groups are involved in the metabolism of bilirubin, salicylates
and thromboxanes are CYP450 enzymes with distinct
specificities.
REDUCTION
Reduction requires reduced NADP-cytochrome-c reductase or
reduced NAD-cytochrome b5 reductase.
HYDROLYSIS
Pethidine(meperidine) is de-esterified to meperidinic acid by
hepatic membrane-bound esterase activity.
NON-ENDOPLASMIC RETICULUM DRUG
METABOLISM
OXIDATION
Oxidation of ethanol to acetaldehyde and of chloral to
trichlorethanol is catalysed by a cytosolic enzyme (alcohol
dehydrogenase) whose substrates also include vitamin A.
Monoamine oxidase (MAO) is a membrane-bound mitochon-
drial enzyme that oxidatively deaminates primary amines to
aldehydes (which are further oxidized to carboxylic acids) or
ketones. Monoamine oxidase is found in liver, kidney, intestine
and nervous tissue, and its substrates include catecholamines
PHASE II METABOLISM(TRANSFERASE REACTIONS) 25
DRUG
Phase I
(predominantly
CYP450)
Phase II
(transferase
reactions)
– Oxidation
– Reduction
– Hydrolysis
– Acetylation
– Methylation
– Glucuronidation
– Sulphation
– Mercaptopuric
acid formation
– Glutathione
conjugation
Renal (or biliary)
excretion
(a)
O
OO
NH
H
N
HO
O
OO
NH
H
N
O
OH
OH
OH
CO
2
H
O
O
O
O
NH
H
N
Phase I Phase II
Phenobarbital p-Hydroxy
phenobarbital
p-Hydroxy phenobarbital
glucuronide
(b)
Figure 5.1:(a) Phases I and II of drug metabolism. (b) A specific example of phases I and II of drug metabolism, in the case of
phenobarbital.
26 DRUG METABOLISM
Table 5.1:CYP450 isoenzymes most commonly involved in drug metabolism with representative drug substrates and their
specific inhibitors and inducers
Enzyme Substrate Inhibitor Inducer
CYP1A2 Caffeine Amiodarone Insulin
Clozapine Cimetidine Cruciferous vegetables
Theophylline Fluoroquinolones Nafcillin
Warfarin (R) Fluvoxamine Omeprazole
CYP2C9
a
Celecoxib
Losartan Amiodarone Barbiturates
NSAIDs Fluconazole Rifampicin
Sulphonylureas Fluoxetine/Fluvoxamine
Phenytoin Lansoprazole
Warfarin (S) Sulfamethoxazole
Ticlopidine
CYP2C19
a
Diazepam Fluoxetine Carbamazepine
Moclobamide Ketoconazole Prednisone
Omeprazole Rifampicin
Pantoprazole
Proguanil
CYP2D6
a
Codeine (opioids) Amiodarone Dexamethasone
Dextromethorphan Celecoxib Rifampicin
Haloperidol Cimetidine
Metoprolol Ecstasy (MDMA)
Nortriptyline Fluoxetine
Pravastatin Quinidine
Propafenone
CYP2E1 Chlormezanone Diethyldithio-carbamate Ethanol
Paracetamol Isoniazid
Theophylline
CYP3A4 Alprazolam Amiodarone Barbiturates
Atorvastatin Diltiazem Carbamazepine
Ciclosporin Erythromycin (and other Efavirenz
macrolides) Glucocorticosteroids
(and other steroids)
Hydrocortisone Gestodene Nevirapine
Lidocaine Grapefruit juice Phenytoin
Lovastatin Fluvoxamine Pioglitazone
Fluconazole/Itraconazole St John’s wort
Midazolam Ketoconazole
Nifedipine (many CCBs) Nefazodone
Tamoxifen Nelfinavir/Ritonavir
Tacrolimus Verapamil
Vincristine Voriconazole
a
Known genetic polymorphisms (Chapter 14).
Approximate percentage of clinically used drugs metabolized by each CYP isoenzyme: CYP3A4, 50%; CYP2D6, 20%; CYP2C,
20%; CYP1A2, 2%; CYP2E1, 2%; other CYPs, 6%.
and lorazepam. Some patients inherit a deficiency of glu-
curonide formation that presents clinically as a non-
haemolytic jaundice due to excess unconjugated bilirubin
(Crigler–Najjar syndrome). Drugs that are normally conju-
gated via this pathway aggravate jaundice in such patients.
O-Glucuronides formed by reaction with a hydroxyl group
result in an ether glucuronide. This occurs with drugs such as
paracetamoland morphine.
METHYLATION
Methylation proceeds by a pathway involving S-adenosyl
methionine as methyl donor to drugs with free amino,
hydroxyl or thiol groups. Catechol O-methyltransferase is an
example of such a methylating enzyme, and is of physiologi-
cal as well as pharmacological importance. It is present in
the cytosol, and catalyses the transfer of a methyl group to
catecholamines, inactivating noradrenaline, dopamine and
adrenaline. Phenylethanolamine N-methyltransferase is also
important in catecholamine metabolism. It methylates the
terminal – NH
2
residue of noradrenaline to form adrenaline in
the adrenal medulla. It also acts on exogenous amines, includ-
ing phenylethanolamine and phenylephrine. It is induced by
corticosteroids, and its high activity in the adrenal medulla
reflects the anatomical arrangement of the blood supply to the
medulla which comes from the adrenal cortex and conse-
quently contains very high concentrations of corticosteroids.
SULPHATION
Cytosolic sulphotransferase enzymes catalyse the sulphation of
hydroxyl and amine groups by transferring the sulphuryl
group from 3-phosphoadenosine 5-phosphosulphate (PAPS)
to the xenobiotic. Under physiological conditions, sulphotrans-
ferases generate heparin and chondroitin sulphate. In addition,
they produce ethereal sulphates from several oestrogens,
androgens, from 3-hydroxycoumarin (a phase I metabolite of
warfarin) and paracetamol. There are a number of sulphotrans-
ferases in the hepatocyte, with different specificities.
MERCAPTURIC ACID FORMATION
Mercapturic acid formation is via reaction with the cysteine
residue in the tripeptide Cys-Glu-Gly, i.e. glutathione. It is
very important in paracetamol overdose (Chapter 54), when
the usual sulphation and glucuronidation pathways of paraceta-
mol metabolism are overwhelmed, with resulting production
of a highly toxic metabolite (N-acetyl-benzoquinone imine,
NABQI). NABQI is normally detoxified by conjugation with
reduced glutathione. The availability of glutathione is critical
in determining the clinical outcome. Patients who have
ingested large amounts of paracetamol are therefore treated
ENZYME INDUCTION 27
with thiol donors such as N-acetyl cysteine or methionine to
increase the endogenous supply of reduced glutathione.
GLUTATHIONE CONJUGATES
Naphthalene and some sulphonamides also form conjugates
with glutathione. One endogenous function of glutathione
conjugation is formation of a sulphidopeptide leukotriene,
leukotriene (LT) C4. This is formed by conjugation of glu-
tathione with LTA4, analogous to a phase II reaction. LTA4 is
an epoxide which is synthesized from arachidonic acid by a
‘phase I’-type oxidation reaction catalysed by the 5-lipoxyge-
nase enzyme. LTC4, together with its dipeptide product LTD4,
comprise the activity once known as ‘slow-reacting substance
of anaphylaxis’ (SRS-A), and these leukotrienes play a role as
bronchoconstrictor mediators in anaphylaxis and in asthma
(see Chapters 12 and 33).
ENZYME INDUCTION
Enzyme induction (Figure 5.2, Table 5.1) is a process by
which enzyme activity is enhanced, usually because of increased
enzyme synthesis (or, less often, reduced enzyme degrada-
tion). The increase in enzyme synthesis is often caused by
xenobiotics binding to nuclear receptors (e.g. pregnane X
receptor, constitutive androstane receptor, aryl hydrocarbon
receptor), which then act as positive transcription factors for
certain CYP450s.
There is marked inter-individual variability in the degree
of induction produced by a given agent, part of which is
genetically determined. Exogenous inducing agents include
not only drugs, but also halogenated insecticides (particularly
dichloro-diphenyl-trichloroethane (DDT) and gamma-benzene
hexachloride), herbicides, polycyclic aromatic hydrocarbons,
dyes, food preservatives, nicotine, ethanol and hyperforin in
St John’s wort. Apractical consequence of enzyme induction is
that, when two or more drugs are given simultaneously, then
if one drug is an inducing agent it can accelerate the metabo-
lism of the other drug and may lead to therapeutic failure
(Chapter 13).
Inducer
(slow –
1–2 weeks)
synthesis
( or degradation)
of CYP450 isoenzyme(s)
Metabolism
(t
½
)
of target drug
Plasma concentration
of target drug
Effect of target
drug
Figure 5.2:Enzyme induction.
28 DRUG METABOLISM
TESTS FOR INDUCTION OF DRUG-
METABOLIZING ENZYMES
The activity of hepatic drug-metabolizing enzymes can be
assessed by measuring the clearance or metabolite ratios of
probe drug substrates, e.g. midazolam for CYP3A4, dex-
tromethorphan for CYP2D6, but this is seldom if ever indi-
cated clinically. The
14
C-erythromycin breath test or the urinary
molar ratio of 6-beta-hydroxycortisol/cortisol have also been
used to assess CYP3A4 activity. It is unlikely that a single probe
drug study will be definitive, since the mixed function oxidase
(CYP450) system is so complex that at any one time the activity
of some enzymes may be increased and that of others reduced.
Induction of drug metabolism represents variable expression
of a constant genetic constitution. It is important in drug elim-
ination and also in several other biological processes, including
adaptation to extra-uterine life. Neonates fail to form glu-
curonide conjugates because of immaturity of hepatic uridyl
glucuronyl transferases with clinically important conse-
quences, e.g. grey baby syndrome with chloramphenicol
(Chapter 10).
ENZYME INHIBITION
Allopurinol, methotrexate, angiotensin converting enzyme
inhibitors, non-steroidal anti-inflammatory drugs and many
others, exert their therapeutic effects by enzyme inhibition
(Figure 5.3). Quite apart from such direct actions, inhibition of
drug-metabolizing enzymes by a concurrently administered
drug (Table 5.1) can lead to drug accumulation and toxicity.
For example, cimetidine, an antagonist at the histamine
H
2
-receptor, also inhibits drug metabolism via the CYP450
system and potentiates the actions of unrelated CYP450
metabolized drugs, such as warfarin and theophylline (see
Chapters 13, 30 and 33). Other potent CYP3A4 inhibitors
include the azoles (e.g. fluconazole, voriconazole) and HIV
protease inhibitors (e.g. ritonavir).
The specificity of enzyme inhibition is sometimes incom-
plete. For example, warfarin and phenytoin compete with
one another for metabolism, and co-administration results in
elevation of plasma steady-state concentrations of both drugs.
Metronidazole is a non-competitive inhibitor of microsomal
enzymes and inhibits phenytoin,warfarin and sulphonylurea
(e.g.glyburide) metabolism.
PRESYSTEMIC METABOLISM (‘FIRST-PASS’
EFFECT)
The metabolism of some drugs is markedly dependent on the
route of administration. Following oral administration, drugs
gain access to the systemic circulation via the portal vein, so the
entire absorbed dose is exposed first to the intestinal mucosa
and then to the liver, before gaining access to the rest of the
body. Aconsiderably smaller fraction of the absorbed dose goes
through gut and liver in subsequent passes because of distribu-
tion to other tissues and drug elimination by other routes.
If a drug is subject to a high hepatic clearance (i.e. it is rap-
idly metabolized by the liver), a substantial fraction will be
extracted from the portal blood and metabolized before it
reaches the systemic circulation. This, in combination with
intestinal mucosal metabolism, is known as presystemic or
‘first-pass’ metabolism (Figure 5.4).
The route of administration and presystemic metabolism
markedly influence the pattern of drug metabolism. For exam-
ple, when salbutamol is given to asthmatic subjects, the ratio
of unchanged drug to metabolite in the urine is 2:1 after intra-
venous administration, but 1:2 after an oral dose. Propranolol
undergoes substantial hepatic presystemic metabolism, and
small doses given orally are completely metabolized before
they reach the systematic circulation. After intravenous admin-
istration, the area under the plasma concentration–time curve
is proportional to the dose administered and passes through
the origin (Figure 5.5). After oral administration the relation-
ship, although linear, does not pass through the origin and
there is a threshold dose below which measurable concentra-
tions of propranolol are not detectable in systemic venous
plasma. The usual dose of drugs with substantial presystemic
metabolism differs very markedly if the drug is given by
the oral or by the systemic route (one must never estimate or
guess the i.v. dose of a drug from its usual oral dose for this
reason!) In patients with portocaval anastomoses bypassing
the liver, hepatic presystemic metabolism is bypassed, so
very small drug doses are needed compared to the usual
oral dose.
Presystemic metabolism is not limited to the liver, since the
gastro-intestinal mucosa contains many drug-metabolizing
enzymes (e.g. CYP3A4, dopa-decarboxylase, catechol-
O-methyl transferase (COMT)) which can metabolize drugs, e.g.
ciclosporin, felodipine, levodopa, salbutamol, before they
enter hepatic portal blood. Pronounced first-pass metabolism by
either the gastro-intestinal mucosa (e.g. felodipine,salbutamol,
levodopa) or liver (e.g. felodipine, glyceryl trinitrate, mor-
phine, naloxone, verapamil) necessitates high oral doses by
comparison with the intravenous route. Alternative routes of
drug delivery (e.g. buccal, rectal, sublingual, transdermal) partly
or completely bypass presystemic elimination (Chapter 4).
Drugs undergoing extensive presystemic metabolism usu-
ally exhibit pronounced inter-individual variability in drug dis-
position. This results in highly variable responses to therapy,
Inhibitor Direct inhibition
of CYP450
isoenzyme(s)
Metabolism
(t
½
)
of target drug
Rapid
Plasma concentration
of target drug
Effect
Toxicity
of target drug
Figure 5.3:Enzyme inhibition.
METABOLISMOF DRUGS BY INTE STINAL ORGANISMS 29
and is one of the major difficulties in their clinical use.
Variability in first-pass metabolism results from:
1. Genetic variations – for example, the bioavailability of
hydralazineis about double in slow compared to fast
acetylators. Presystemic hydroxylation of metoprololand
encainidealso depends on genetic polymorphisms
(CYP2D6, Chapter 14).
2. Induction or inhibition of drug-metabolizing enzymes.
3. Food increases liver blood flow and can increase the
bioavailability of drugs, such as propranolol,metoprolol
andhydralazine, by increasing hepatic blood flow and
exceeding the threshold for complete hepatic extraction.
4. Drugs that increase liver blood flow have similar effects to
food – for example, hydralazineincreases propranolol
bioavailability by approximately one-third, whereas drugs
that reduce liver blood flow (e.g. -adrenoceptor
antagonists) reduce it.
5. Non-linear first-pass kinetics are common (e.g. aspirin,
hydralazine,propranolol): increasing the dose
disproportionately increases bioavailability.
6. Liver disease increases the bioavailability of some drugs
with extensive first-pass extraction (e.g. diltiazem,
ciclosporin,morphine).
METABOLISM OF DRUGS BY INTESTINAL
ORGANISMS
This is important for drugs undergoing significant enterohep-
atic circulation. For example, in the case of estradiol, which is
excreted in bile as a glucuronide conjugate, bacteria-derived
enzymes cleave the glucuronide so that free drug is available
for reabsorption in the terminal ileum. Asmall proportion of
the dose (approximately 7%) is excreted in the faeces under
normal circumstances; this increases if gastro-intestinal dis-
ease or concurrent antibiotic therapy alter the intestinal flora.
Orally
administered
drug
Intestinal
mucosal
metabolism
Portal
vein
Hepatic
metabolism
Systemic
circulation
First-pass
metabolism
Parenterally
administered
drug
Figure 5.4:Presystemic (‘first-pass’) metabolism.
i.v.
Oral
0
0
500
1000
40 80 120 160
Area (ng/ml h)
Dose (mg)
T
Figure 5.5:Area under blood concentration–time curve after oral
() and intravenous () administration of propranolol to humans
in various doses. T is the apparent threshold for propranolol
following oral administration. (Redrawn from Shand DG, Rangno
RE.Pharmacology 1972; 7: 159, with permission of
S Karger AG, Basle.)
Key points
Drug metabolism involves two phases: phase I often
followed sequentially by phase II.
Phase I metabolism introduces a reactive group into a
molecule, usually by oxidation, by a microsomal system
present in the liver.
The CYP450 enzymes are a superfamily of
haemoproteins. They have distinct isoenzyme forms
and are critical for phase I reactions.
Products of phase I metabolism may be
pharmacologically active, as well as being chemically
reactive, and can be hepatotoxic.
Phase II reactions involve conjugation (e.g. acetylation,
glucuronidation, sulphation, methylation).
Products of phase II metabolism are polar and can be
efficiently excreted by the kidneys. Unlike the products
of phase I metabolism, they are nearly always
pharmacologically inactive.
The CYP450 enzymes involved in phase I metabolism can
be induced by several drugs and nutraceuticals (e.g.
glucocorticosteroids, rifampicin, carbamazepine, St John’s
wort) or inhibited by drugs (e.g. cimetidine, azoles, HIV
protease inhibitors, quinolones, metronidazole) and
dietary constituents (e.g. grapefruit/grapefruit juice).
Induction or inhibition of the CYP450 system are
important causes of drug–drug interactions (see
Chapter 13).
30 DRUG METABOLISM
Case history
A 46-year-old woman is brought to the hospital Accident
and Emergency Department by her sister, having swal-
lowed an unknown number of paracetamol tablets washed
down with vodka six hours previously, following an argu-
ment with her partner. She is an alcoholic and has been
taking St John’s wort for several weeks. Apart from signs
of intoxication, examination was unremarkable. Plasma
paracetamol concentration was 662μmol/L (100 mg/L).
Following discussion with the resident medical officer/
Poisons Information Service, it was decided to administer
N-acetylcysteine.
Comment
In paracetamol overdose, the usual pathway of elimination
is overwhelmed and a highly toxic product (N-acetyl benzo-
quinone imine, known as NABQI) is formed by CYP1A2, 2E1
and CYP3A4 metabolism. A plasma paracetamol concentra-
tion of 100mg/L six hours after ingestion would not usually
require antidote treatment, but this woman is an alcoholic
and is taking St John’s wort and her hepatic drug-
metabolizing enzymes (CYP1A2, CYP3A4 and probably
others) will have been induced, so the paracetamol concen-
tration threshold for antidote treatment is lowered (see
Chapter 54). N-Acetylcysteine is the specific antidote, as it
increases reduced glutathione which conjugates NABQI
within hepatocytes.
FURTHER READING AND WEB MATERIAL
Boobis AR, Edwards RJ, Adams DA, Davies DS. Dissecting the func-
tion of P450. British Journal of Clinical Pharmacology1996; 42: 81–9.
Coon MJ. Cytochrome P450: nature’s most versatile biological cata-
lyst.Annual Review of Pharmacology and Toxicology 2005; 45: 1–25.
Lin JH, Lu AY. Interindividual variability in inhibition and induction
of cytochrome P450 enzymes. Annual Review of Pharmacology and
Toxicology2001; 41: 535–67.
Nelson DR, Zeldin DC, Hoffman SM, Maltais LJ, Wain HM, Nebert
DW. Comparison of cytochrome P450 (CYP) genes from the
mouse and human genomes, including nomenclature recommen-
dations for genes, pseudogenes and alternative-splice variants.
Pharmacogenetics2004; 14: 1–18.
Website for CYP450 substrates, inhibitors and inducers:
www.medicine.iupui.edu/flockhart/table, accessed April 2007.
Introduction 31
Glomerular filtration 31
Proximal tubular secretion 31
Passive distal tubular reabsorption 32
Active tubular reabsorption 33
CHAPTER 6
RENAL EXCRETION OF DRUGS
INTRODUCTION
The kidneys are involved in the elimination of virtually every
drug or drug metabolite (Figure 6.1). The contribution of renal
excretion to total body clearance of any particular drug is
determined by its lipid solubility (and hence its polarity).
Elimination of non-polar drugs depends on metabolism
(Chapter 5) to more polar metabolites, which are then excreted
in the urine. Polar substances are eliminated efficiently by the
kidneys, because they are not freely diffusible across the tubu-
lar membrane and so remain in the urine, even though there is
a concentration gradient favouring reabsorption from tubular
to interstitial fluid. Renal elimination is influenced by several
processes that alter the drug concentration in tubular fluid.
Depending on which of these predominates, the renal clear-
ance of a drug may be either an important or a trivial compo-
nent in its overall elimination.
GLOMERULAR FILTRATION
Glomerular filtrate contains concentrations of low-molecular-
weight solutes similar to plasma. In contrast, molecules with a
molecular weight of 66000 (including plasma proteins and
drug–protein complexes) do not pass through the glomerulus.
Accordingly, only free drug passes into the filtrate. Renal
impairment (Chapter 7) predictably reduces the elimination of
drugs that depend on glomerular filtration for their clearance
(e.g.digoxin). Drugs that are highly bound to albumin or α-1
acid glycoprotein in plasma are not efficiently filtered.
PROXIMAL TUBULAR SECRETION
There are independent mechanisms for active secretion of
organic anions and organic cations (OAT and OCT) into the
proximal tubule. These are relatively non-specific in their
structural requirements, and share some of the characteristics
of transport systems in the intestine. OAT excretes drugs, such
asprobenecid and penicillin. Para-aminohippuric acid (PAH)
is excreted so efficiently that it is completely extracted from
Free drug enters
glomerular filtrate
Active secretion
Can be affected by other
drugs: main site for
interactions in the
kidney
Passive reabsorption
of lipid-soluble,
unionized drug
Ionized, lipid-insoluble drug
into urine
Collecting duct
Distal tubule
Loop of Henle
Proximal tubule
1
2
3
Figure 6.1:Urinary elimination of drugs and metabolites by
glomerular filtration and/or tubular secretion and reabsorption.
32 RENALEXCRETION OF DRUGS
the renal plasma in a single pass through the kidney (i.e. dur-
ing intravenous infusion of PAH its concentration in renal
venous blood is zero). Clearance of PAH is therefore limited
by the rate at which it is delivered to the kidney, i.e. renal
plasma flow, so PAH clearance provides a non-invasive meas-
ure of renal plasma flow.
OCT contributes to the elimination of basic drugs (e.g.
cimetidine, amphetamines).
Each mechanism is characterized by a maximal rate of
transport for a given drug, so the process is theoretically sat-
urable, although this maximum is rarely reached in practice.
Because secretion of free drug occurs up a concentration gra-
dient from peritubular fluid into the lumen, the equilibrium
between unbound and bound drug in plasma can be dis-
turbed, with bound drug dissociating from protein-binding
sites. Tubular secretion can therefore eliminate drugs effi-
ciently even if they are highly protein bound. Competition
occurs between drugs transported via these systems. e.g.
probenecid competitively inhibits the tubular secretion of
methotrexate.
PASSIVE DISTAL TUBULAR REABSORPTION
The renal tubule behaves like a lipid barrier separating the
high drug concentration in the tubular lumen and the lower
concentration in the interstitial fluid and plasma. Reabsorption
of drug down its concentration gradient occurs by passive dif-
fusion. For highly lipid-soluble drugs, reabsorption is so effec-
tive that renal clearance is virtually zero. Conversely, polar
substances, such as mannitol, are too water soluble to be
absorbed, and are eliminated virtually without reabsorption.
Tubular reabsorption is influenced by urine flow rate.
Diuresis increases the renal clearance of drugs that are pas-
sively reabsorbed, since the concentration gradient is reduced
(Figure 6.2). Diuresis may be induced deliberately in order to
increase drug elimination during treatment of overdose
(Chapter 54).
Reabsorption of drugs that are weak acids (AH) or bases
(B) depends upon the pH of the tubular fluid, because this
determines the fraction of acid or base in the charged, polar
form and the fraction in the uncharged lipid-soluble form. For
acidic drugs, the more alkaline the urine, the greater the renal
clearance, and vice versa for basic drugs, since:
and
.
Thus high pH favours A
, the charged form of the weak
acid which remains in the tubular fluid and is excreted in the
urine, while low pH favours BH
, the charged form of the
base (Figure 6.3). This is utilized in treating overdose with
aspirin (a weak acid) by alkalinization of the urine, thereby
accelerating urinary elimination of salicylate (Chapter 54).
The extent to which urinary pH affects renal excretion of
weak acids and bases depends quantitatively upon the pK
a
of
the drug. The critical range of pK
a
values for pH-dependent
excretion is about 3.0–6.5 for acids and 7.5–10.5 for bases.
Urinary pH may also influence the fraction of the total dose
which is excreted unchanged. About 57% of a dose of amphet-
amine is excreted unchanged (i.e. as parent drug, rather than
as a metabolite) in acid urine (pH 4.5–5.6), compared to about
7% in subjects with alkaline urine (pH 7.1–8.0). Administration
of amphetamines with sodium bicarbonate has been used illic-
itly by athletes to enhance the pharmacological effects of the
drug on performance, as well as to make its detection by uri-
nary screening tests more difficult.
BH BH

AH A H

Low urine
flow rate
High urine
flow rate
D
D
in urine
D
D in urine
Indicates passive reabsorption in distal tubule
Figure 6.2:Effect of diuresis (urine flow rate) on renal clearance
of a drug (D) passively reabsorbed in the distal tubule.
Low pH High pH
BH
A
in urine
Indicates passive reabsorption in distal tubule
AH
A
BH
B
AH
A
BH
B
A
BH
in urine
Figure 6.3: Effects of urine pH on renal clearance of a weak acid
(AH) and a weak base (B).
ACTIVE TUBULAR REABSORPTION
This is of minor importance for most therapeutic drugs. Uric
acid is reabsorbed by an active transport system which is
inhibited by uricosuric drugs, such as probenecid and
sulfinpyrazone. Lithium also undergoes active tubular reab-
sorption (hitching a ride on the proximal sodium ion transport
mechanism).
FURTHER READING
Carmichael DJS. Chapter 19.2 Handling of drugs in kidney disease.
In: AMADavison, J Stewart Cameron, J-P Grunfeld, C Ponticelli,
C Van Ypersele, E Ritz and C Winearls (eds). Oxford textbook of clin-
ical nephrology, 3rd edn. Oxford: Oxford University Press, 2005:
2599–618.
Eraly SA, Bush KT, Sampogna RV, Bhatnagar V, Nigam SK. The mole-
cular pharmacology of organic anion transporters: from DNAto
FDA?Molecular Pharmacology 2004; 65: 479–87.
Koepsell H. Polyspecific organic cation transporters: their functions
and interactions with drugs. Trends in Pharmacological Sciences
2004;25: 375–81.
van Montfoort JE, Hagenbuch B, Groothuis GMM, Koepsell H,
Meier PJ, Meijer DKF. Drug uptake systems in liver and kidney.
Current Drug Metabolism2003; 4: 185–211.
ACTIVE TUBULAR REABSORPTION 33
Key points
The kidney cannot excrete non-polar substances
efficiently, since these diffuse back into blood as the
urine is concentrated. Consequently, the kidney
excretes polar drugs and/or the polar metabolites of
non-polar compounds.
Renal impairment reduces the elimination of drugs that
depend on glomerular filtration, so the dose of drugs,
such as digoxin, must be reduced, or the dose interval
(e.g. between doses of aminoglycoside) must be
increased, to avoid toxicity.
There are specific secretory mechanisms for organic
acids and organic bases in the proximal tubules which
lead to the efficient clearance of weak acids, such as
penicillin, and weak bases, such as cimetidine.
Competition for these carriers can cause drug
interactions, although less commonly than induction or
inhibition of cytochrome P450.
Passive reabsorption limits the efficiency with which the
kidney eliminates drugs. W
eak acids are best eliminated
in an alkaline urine (which favours the charged form,
A
), whereas weak bases are best eliminated in an acid
urine (which favours the charged form, BH
).
The urine may be deliberately alkalinized by infusing
sodium bicarbonate intravenously in the management
of overdose with weak acids such as aspirin (see
Chapter 54, to increase tubular elimination of
salicylate.
Lithium ions are actively reabsorbed in the proximal
tubule by the same system that normally reabsorbs
sodium, so salt depletion (which causes increased
proximal tubular sodium ion reabsorption) causes
lithium toxicity unless the dose of lithium is reduced.
Case history
A house officer (HO) sees a 53-year-old woman in the
Accident and Emergency Department with a six-hour his-
tory of fevers, chills, loin pain and dysuria. She looks very ill,
with a temperature of 39.5°C, blood pressure of 80/60mmHg
and right loin tenderness. The white blood cell count is
raised at 15000/μL, and there are numerous white cells and
rod-shaped organisms in the urine. Serum creatinine is nor-
mal at 90μmol/L. The HO wants to start treatment with
aminoglycoside antibiotic pending the availability of a bed
on the intensive care unit. Despite the normal creatinine
level, he is concerned that the dose may need to be adjusted
and calls the resident medical officer for advice.
Comment
The HO is right to be concerned. The patient is hypotensive
and will be perfusing her kidneys poorly. Serum creatinine
may be normal in rapid onset acute renal failure. It is impor-
tant to obtain an adequate peak concentration to combat
her presumed Gram-negative septicaemia. It would there-
fore be appropriate to start treatment with the normal
loading dose. This will achieve the usual peak concentra-
tion (since the volume of distribution will be similar to
that in a healthy person). However, the subsequent and
maintenance doses should not be given until urgent post-
administration blood concentrations have been obtained –
the dosing interval may be appropriately prolonged if
renal failure does indeed supervene causing reduced
aminoglycoside clearance.
pancreatic secretions and bile flow, can impair the absorption
of fat-soluble vitamins. Significant reductions in the absorption
of cefalexin occur in cystic fibrosis, necessitating increased
doses in such patients. Patients with small bowel resection
may absorb lipophilic drugs poorly.
CARDIAC FAILURE
Cardiac failure affects pharmacokinetics in several ways and
these are discussed below.
ABSORPTION
Absorption of some drugs (e.g. furosemide) is altered in car-
diac failure because of mucosal oedema and reduced gastro-
intestinal blood flow. Splanchnic vasoconstriction accompanies
cardiac failure as an adaptive response redistributing blood to
more vital organs.
Introduction 34
Gastro-intestinal disease 34
Cardiac failure 34
Renal disease 35
Liver disease 37
Thyroid disease 38
CHAPTER 7
EFFECTS OF DISEASE ON DRUG
DISPOSITION
INTRODUCTION
Several common disorders influence the way in which the
body handles drugs and these must be considered before pre-
scribing. Gastro-intestinal, cardiac, renal, liver and thyroid
disorders all influence drug pharmacokinetics, and individu-
alization of therapy is very important in such patients.
GASTRO-INTESTINAL DISEASE
Gastro-intestinal disease alters the absorption of orally admin-
istered drugs. This can cause therapeutic failure, so alternative
routes of administration (Chapter 4) are sometimes needed.
GASTRIC EMPTYING
Gastric emptying is an important determinant of the rate and
sometimes also the extent of drug absorption. Several patho-
logical factors alter gastric emptying (Table 7.1). However,
there is little detailed information about the effect of disease
on drug absorption, in contrast to effects of drugs that slow
gastric emptying (e.g. anti-muscarinic drugs) which delay
C
max
. Absorption of analgesics is delayed in migraine, and a
more rapid absorption can be achieved by administeringanal-
gesics with metoclopramide, which increases gastric emptying.
SMALL INTESTINAL AND PANCREATIC DISEASE
The very large absorptive surface of the small intestine pro-
vides a substantial functional reserve, so even extensive
involvement with, for example, coeliac disease may be present
without causing a clinically important reduction in drug
absorption. Crohn’s disease typically affects the terminal
ileum. Absorption of several antibiotics actually increases in
Crohn’s disease. Cystic fibrosis, because of its effects on
Table 7.1: Pathological factors influencing the rate of gastric emptying
Decreased rate Increased rate
Trauma Duodenal ulcer
Pain (including myocardial Gastroenterostomy
infarction, acute abdomen) Coeliac disease
Diabetic neuropathy Drugs, e.g. metoclopramide
Labour
Migraine
Myxoedema
Raised intracranial pressure
Intestinal obstruction
Gastric ulcer
Anti-muscarinic drugs
RENAL DISEASE 35
DISTRIBUTION
Drug distribution is altered by cardiac failure. The apparent
volume of distribution (V
d
) of, for example, quinidine and
lidocainein patients with congestive cardiac failure is markedly
reduced because of decreased tissue perfusion and altered
partition between blood and tissue components. Usual doses
can therefore result in elevated plasma concentrations, pro-
ducing toxicity.
ELIMINATION
Elimination of several drugs is diminished in heart failure.
Decreased hepatic perfusion accompanies reduced cardiac
output. Drugs such as lidocainewith a high hepatic extraction
ratio of 70% show perfusion-limited clearance, and steady-
state levels are inversely related to cardiac output (Figure 7.1).
Duringlidocaine infusion, the steady-state concentrations are
almost 50% higher in patients with cardiac failure than in healthy
volunteers. The potential for lidocainetoxicity in heart failure
is further increased by the accumulation of its polar metab-
olites, which have cardiodepressant and pro-convulsant prop-
erties. This occurs because renal blood flow and glomerular
filtration rate are reduced in heart failure.
Theophylline clearance is decreased and its half-life is
doubled in patients with cardiac failure and pulmonary oedema,
increasing the potential for accumulation and toxicity. The
metabolic capacity of the liver is reduced in heart failure both
by tissue hypoxia and by hepatocellular damage from hepatic
congestion. Liver biopsy samples from patients with heart
failure have reduced drug-metabolizing enzyme activity.
Heart failure reduces renal elimination of drugs because of
reduced glomerular filtration, predisposing to toxicity from
drugs that are primarily cleared by the kidneys, e.g. amino-
glycosides and digoxin.
RENAL DISEASE
RENAL IMPAIRMENT
Renal excretion is a major route of elimination for many drugs
(Chapter 6), and drugs and their metabolites that are excreted
predominantly by the kidneys accumulate in renal failure.
Renal disease also affects other pharmacokinetic processes
(i.e. drug absorption, distribution and metabolism) in more
subtle ways.
ABSORPTION
Gastric pH increases in chronic renal failure because urea is
cleaved, yielding ammonia which buffers acid in the stomach.
This reduces the absorption of ferrous iron and possibly also of
other drugs. Nephrotic syndrome is associated with resistance
to oral diuretics, and malabsorption of loop diuretics through
the oedematous intestine may contribute to this.
DISTRIBUTION
Renal impairment causes accumulation of several acidic sub-
stances that compete with drugs for binding sites on albumin
0
0.1
1
10
60 120 180 240
Heart
failure
Control
Lidocaine concentration (g/ml)
Time after injection (min)
0 250 500 750 100012501500 1750
Estimated hepatic blood
flow (ml/min/m
2
)
1.0
1.4
1.8
2.2
2.6
3.0
3.4
Steady-state arterial
concentration of lidocaine (g/ml)
3.2
3.0
2.8
2.6
2.4
2.2
2.0
1.8
1.6
1.4
1.2
1.51.0 2.0 2.5 3.0 3.5 4.0 4.5
Cardiac index (I/min/m
2
)
Arterial lidocaine (g/ml)
(c)
(b)
(a)
Figure 7.1:(a) Mean values (and standard deviations) of plasma
lidocaine concentrations in seven heart failure patients and
controls following a 50-mg intravenous bolus. (b) Relationship
between arterial lidocaine level and cardiac index (dotted
vertical line is lower limit of normal cardiac index, square is mean
for low cardiac index patients, triangle is mean for patients with
normal cardiac index). (c) Relationship of steady-state arterial
lidocaine level following 50-mg bolus and infusion of
40mg/kg/min (vertical line is lower limit of normal hepatic blood
flow, square is mean for patients with low hepatic blood flow,
triangle is mean for patients with normal flow). (Reproduced
from: (a) Thompson PD et al. American Heart Journal1971; 82,
417; (b,c) Stenson RE et al. Circulation1971; 43: 205. With
permission of the American Heart Association Inc.)
and other plasma proteins. This alters the pharmacokinetics of
many drugs, but is seldom clinically important. Phenytoin
is an exception, because therapy is guided by plasma concen-
tration and routine analytical methods detect total (bound
and free) drug. In renal impairment, phenytoinprotein bind-
ing is reduced by competition with accumulated molecules
normally cleared by the kidney and which bind to the same
albumin drug-binding site as phenytoin. Thus, for any meas-
ured phenytoin concentration, free (active) drug is increased
compared to a subject with normal renal function and the
same measured total concentration. The therapeutic range
therefore has to be adjusted to lower values in patients with
renal impairment, as otherwise doses will be selected that
cause toxicity.
Tissue binding of digoxin is reduced in patients with
impaired renal function, resulting in a lower volume of distri-
bution than in healthy subjects. A reduced loading dose of
digoxinis therefore appropriate in such patients, although the
effect of reduced glomerular filtration on digoxinclearance is
even more important, necessitating a reduced maintenance
dose, as described below.
The blood–brain barrier is more permeable to drugs in
uraemia. This can result in increased access of drugs to the
central nervous system, an effect that is believed to contribute
to the increased incidence of confusion caused by cimetidine,
ranitidineand famotidine in patients with renal failure.
METABOLISM
Metabolism of several drugs is reduced in renal failure. These
include drugs that undergo phase I metabolism by CYP3A4.
Drugs that are mainly metabolized by phase II drug metabol-
ism are less affected, although conversion of sulindac to its
active sulphide metabolite is impaired in renal failure, as is the
hepatic conjugation of metoclopramidewith glucuronide and
sulphate.
RENAL EXCRETION
Glomerular filtration and tubular secretion of drugs usually
fall in step with one another in patients with renal impair-
ment. Drug excretion is directly related to glomerular filtra-
tion rate (GFR). Some estimate of GFR (eGFR) is therefore
essential when deciding on an appropriate dose regimen.
Serum creatinine concentration adjusted for age permits cal-
culation of an estimate of GFR per 1.73m
2
body surface area.
This is now provided by most chemical pathology labora-
tories, and is useful in many situations. Alternatively, Figure 7.2
shows a nomogram given plasma creatinine, age, sex and
body weight and is useful when a patient is markedly over- or
underweight. The main limitation of such estimates is that
they are misleading if GFR is changing rapidly as in acute
renal failure. (Imagine that a patient with normal serum creati-
nine undergoes bilateral nephrectomy: an hour later, his serum
creatinine would still be normal, but his GFR would be zero.
Creatinine would rise gradually over the next few days as it con-
tinued to be produced in his body but was not cleared.) Anor-
mal creatinine level therefore does not mean that usual doses
can be assumed to be safe in a patient who is acutely unwell.
36 EFFECTSOF DISEASE ON DRUG DISPOSITION
eGFR is used to adjust the dose regimen in patients with
some degree of chronic renal impairment for drugs with a low
therapeutic index that are eliminated mainly by renal excre-
tion. Dose adjustment must be considered for drugs for which
there is 50% elimination by renal excretion. The British
National Formulary tabulates drugs to be avoided or used
with caution in patients with renal failure. Common examples
are shown in Table 7.2.
Clearance
(ml/min)
Weight
(kg)
Serum
creatinine
(mg/100 ml)
R
Age
(years)
5.0
4.0
3.0
2.0
1.7
1.3
0.9
1.0
0.8
0.7
0.6
0.5
0.4
1.5
1.2
95
95
30
40
50
60
70
80
90
100
110
120
150
130
110
100
90
80
70
60
50
40
30
20
10
85
85
75
75
65
65
55
55
45
45
35
35
25
25
Figure 7.2:Nomogram for rapid evaluation of endogenous
creatinine clearance – with a ruler joining weight to age. Keep
ruler at crossing point on R, then move the right-hand side of the
ruler to the appropriate serum creatinine value and read off
clearance from the left-hand scale. To convert serum creatinine in
mol/L to mg/100mL, as is used on this scale, simply divide by
88.4. (Reproduced with permission from Siersbaek-Nielson K
et al. Lancet1971; 1: 1133. © The Lancet Ltd.)
Table 7.2:Examples of drugs to be used with particular caution or avoided
in renal failure
Angiotensin-converting enzyme Angiotensin receptor
inhibitors
a
blockers
a
Aldosterone antagonists Aminoglycosides
Amphotericin Atenolol
Ciprofloxacin Cytotoxics
Digoxin Lithium
Low molecular weight heparin Metformin
NSAIDs Methotrexate
a
ACEI and ARB must be used with caution, but can slow progressive renal
impairment (see Chapter 28).
Detailed recommendations on dosage reduction can be
found in textbooks of nephrology. These are useful for getting
treatment under way but, although precise, such recommenda-
tions are inevitably based only on the effects of reduced renal
function on drug elimination in ‘average’ populations.
Individual variation is substantial, and therapeutic monitoring
of efficacy, toxicity and sometimes of drug concentrations is
essential in patients with impaired renal function.
There are two ways of reducing the total dose to compensate
for impaired renal function. Either each dose can be reduced, or
the interval between each dose can be lengthened. The latter
method is useful when a drug must achieve some threshold con-
centration to produce its desired effect, but does not need to
remain at this level throughout the dose interval. This is the case
with aminoglycoside antibiotics. Therapy with these drugs
is appropriately monitored by measuring ‘peak’ concentrations
(in blood sampled at a fixed brief interval after dosing, sufficient
to permit at least partial tissue distribution), which indicate
whether the dose is large enough to achieve a therapeutic
plasma concentration, and ’trough’ concentrations immediately
before the next dose (see Chapter 8). If the peak concentration is
satisfactory but the trough concentration is higher than desired
(i.e. toxicity is present or imminent), the dose is not reduced but
the interval between doses is extended. This type of therapeutic
drug monitoring is modified to a single time point (after dosing
and beyond the distribution phase) when extended interval dos-
ing of aminoglycosides is used to treat patients (Chapter 43).
RENAL HAEMODYNAMICS
Patients with mild renal impairment depend on vasodilator
prostaglandin biosynthesis to preserve renal blood flow and
GFR. The same is true of patients with heart failure, nephrotic
syndrome, cirrhosis or ascites. Such patients develop acute
reversible renal impairment, often accompanied by salt and
water retention and hypertension if treated with non-steroidal
anti-inflammatory drugs (NSAIDs, see Chapter 26), because
these inhibit cyclo-oxygenase and hence the synthesis of
vasodilator prostaglandins, notably prostaglandin I
2
(prosta-
cyclin) and prostaglandin E
2
. Sulindac is a partial exception
because it inhibits cyclo-oxygenase less in kidneys than in
other tissues, although this specificity is incomplete and dose
dependent.
Angiotensin converting enzyme inhibitors (e.g. ramipril) can
also cause reversible renal failure due to altered renal haemody-
namics. This occurs predictably in patients with bilateral renal
artery stenosis (or with renal artery stenosis involving a single
functioning kidney). The explanation is that in such patients
GFR is preserved in the face of the fixed proximal obstruction by
angiotensin-II-mediated efferent arteriolar vasoconstriction.
Inhibition of angiotensin converting enzyme disables this home-
ostatic mechanism and precipitates renal failure.
NEPHROTIC SYNDROME
Plasma albumin in patients with nephrotic syndrome is low,
resulting in increased fluctuations of free drug concentration
LIVER DISEASE 37
following each dose. This could cause adverse effects, although
in practice this is seldom clinically important. The high albumin
concentration in tubular fluid contributes to the resistance to
diuretics that accompanies nephrotic syndrome. This is because
both loop diuretics and thiazides act on ion-transport processes
in the luminal membranes of tubular cells (see Chapter 36).
Protein binding of such diuretics within the tubular lumen
therefore reduces the concentration of free (active) drug in
tubular fluid in contact with the ion transporters on which
they act.
PRESCRIBING FOR PATIENTS WITH RENAL
DISEASE
1. Consider the possibility of renal impairment before drugs
are prescribed and use available data to estimate GFR.
2. Check how drugs are eliminated before prescribing them.
If renal elimination accounts for more than 50% of total
elimination, then dose reduction will probably be
necessary after the first dose, i.e. for maintenance
doses.
3. Monitor therapeutic and adverse effects and, where
appropriate, plasma drug concentrations.
4. If possible avoid potentially nephrotoxic drugs (e.g.
aminoglycosides, NSAIDs); if such drugs are essential use
them with great care.
Once a potential renal problem necessitating dose modifica-
tion has been identified, there are a number of accepted refer-
ence sources that provide guidance for dose adjustment.
These are useful approximations to get treatment under way,
but their mathematical precision is illusory, and must not lull
the inexperienced into a false sense of security – they do not
permit a full ‘course’ of treatment to be prescribed safely. The
patient must be monitored and treatment modified in the light
of individual responses. The British National Formulary has a
useful appendix which is concise, simple and accessible.
LIVER DISEASE
The liver is the main site of drug metabolism (Chapter 5). Liver
disease has major but unpredictable effects on drug handling.
Pharmacokinetic factors that are affected include absorption
and distribution, as well as the metabolism of drugs.
Attempts to correlate changes in the pharmacokinetics of
drugs with biochemical tests of liver function have been
unsuccessful (in contrast to the use of plasma creatinine in
chronic renal impairment described above). In chronic liver
disease, serum albumin is the most useful index of hepatic
drug-metabolizing activity, possibly because a low albumin
level reflects depressed synthesis of hepatic proteins, includ-
ing those involved in drug metabolism. Prothrombin time also
shows a moderate correlation with drug clearance by the liver.
However, in neither case has a continuous relationship been
38 EFFECTSOF DISEASE ON DRUG DISPOSITION
demonstrated, and such indices of hepatic function serve
mainly to distinguish the severely affected from the milder
cases. Clearances of indocyanine green, antipyrine and lido-
cainehave also been disappointing.
Currently, therefore, cautious empiricism coupled with an
awareness of an increased likelihood of adverse drug effects
and close clinical monitoring is the best way for a prescriber to
approach a patient with liver disease. Drugs should be used
only if necessary, and the risks weighed against potential bene-
fit. If possible, drugs that are eliminated by routes other than
the liver should be employed.
EFFECTS OF LIVER DISEASE ON DRUG
ABSORPTION
Absorption of drugs is altered in liver disease because of portal
hypertension, and because hypoalbuminaemia causes mucosal
oedema. Portal/systemic anastomoses allow the passage of
orally administered drug directly into the systemic circulation,
bypassing hepatic presystemic metabolism and markedly
increasing the bioavailability of drugs with high presystemic
metabolism such as propranolol, morphine, verapamil and
ciclosporin, which must therefore be started in low doses in
such patients and titrated according to effect.
DISTRIBUTION OF DRUGS IN PATIENTS WITH
LIVER DISEASE
Drug distribution is altered in liver disease. Reduced plasma
albumin reduces plasma protein binding. This is also influ-
enced by bilirubin and other endogenous substances that
accumulate in liver disease and may displace drugs from bind-
ing sites. The free fraction of tolbutamideis increased by 115%
in cirrhosis, and that of phenytoinis increased by up to 40%.
It is particularly important to appreciate this when plasma
concentrations of phenytoin are being used to monitor ther-
apy, as unless the therapeutic range is adjusted downward,
toxicity will be induced, as explained above in the section on
drug distribution in renal disease.
Reduced plasma protein binding increases the apparent V
d
if other factors remain unchanged. Increased V
d
of several
drugs (e.g. theophylline) is indeed observed in patients with
liver disease. Disease-induced alterations in clearance and V
d
often act in opposite directions with regard to their effect on
t
1/2
. Data on t
1/2
in isolation provide little information about
the extent of changes in metabolism or drug distribution
which result from liver disease.
DRUG METABOLISM IN LIVER DISEASE
CYP450-mediated phase I drug metabolism is generally
reduced in patients with very severe liver disease, but drug
metabolism is surprisingly little impaired in patients with
moderate to severe disease. There is a poor correlation
between microsomal enzyme activity from liver biopsy speci-
mens in vitro and drug clearance measurements in vivo. Even
in very severe disease, the metabolism of different drugs is not
affected to the same extent. It is therefore hazardous to extrapo-
late from knowledge of the handling of one drug to effects on
another in an individual patient with liver disease.
Prescribing for patients with liver disease
1. Weigh risks against hoped for benefit, and minimize non-
essential drug use.
2. If possible, use drugs that are eliminated by routes
other than the liver (i.e. in general, renally cleared
drugs).
3. Monitor response, including adverse effects (and
occasionally drug concentrations), and adjust therapy
accordingly.
4. Avoid sedatives and analgesics if possible: they are
common precipitants of hepatic coma.
5. Predictable hepatotoxins (e.g. cytotoxic drugs) should
only be used for the strongest of indications, and then
only with close clinical and biochemical monitoring.
6. Drugs that are known to cause idiosyncratic liver disease
(e.g.isoniazid, phenytoin, methyldopa) are not
necessarily contraindicated in stable chronic disease, as
there is no evidence of increased susceptibility. Oral
contraceptives are not advisable if there is active liver
disease or a history of jaundice of pregnancy.
7. Constipation favours bacterial production of false
neurotransmitter amines in the bowel: avoid drugs that
cause constipation (e.g. verapamil, tricyclic
antidepressants) if possible.
8. Drugs that inhibit catabolism of amines (e.g. monoamine
oxidase inhibitors) also provoke coma and should be
avoided.
9. Low plasma potassium provokes encephalopathy: avoid
drugs that cause this if possible. Potassium-sparing
drugs, such as spironolactone, are useful.
10. Avoid drugs that cause fluid overload or renal failure
(e.g. NSAID) and beware those containing sodium (e.g.
sodium-containing antacids and high-dose
carbenicillin).
11. Avoid drugs that interfere with haemostasis (e.g. aspirin,
anticoagulants and fibrinolytics) whenever possible,
because of the increased risk of bleeding (especially in the
presence of varices!).
THYROID DISEASE
Thyroid dysfunction affects drug disposition partly as a result
of effects on drug metabolism and partly via changes in renal
elimination. Existing data refer to only a few drugs, but it is
prudent to anticipate the possibility of increased sensitivity of
hypothyroid patients to many drugs when prescribing.
Information is available for the following drugs.
DIGOXIN
Myxoedematous patients are extremely sensitive to digoxin,
whereas unusually high doses are required in thyrotoxicosis.
In general, hyperthyroid patients have lower plasma digoxin
concentrations and hypothyroid patients have higher plasma
concentrations than euthyroid patients on the same dose.
There is no significant difference in half-life between these
groups, and a difference in V
d
has been postulated to explain
the alteration of plasma concentration with thyroid activity.
Changes in renal function, which occur with changes in thy-
roid status, complicate this interpretation. GFR is increased in
thyrotoxicosis and decreased in myxoedema. These changes
in renal function influence elimination, and the reduced plasma
levels of digoxincorrelate closely with the increased creatinine
clearance in thyrotoxicosis. Other factors including enhanced
biliary clearance, digoxin malabsorption due to intestinal
hurry and increased hepatic metabolism, have all been postu-
lated as factors contributing to the insensitivity of thyrotoxic
patients to cardiac glycosides.
ANTICOAGULANTS
Oral anticoagulants produce an exaggerated prolongation of
prothrombin time in hyperthyroid patients. This is due to
increased metabolic breakdown of vitamin K-dependent clot-
ting factors (Chapter 30), rather than to changes in drug phar-
macokinetics.
GLUCOCORTICOIDS
Glucocorticoids are metabolized by hepatic mixed-function
oxidases (CYP3A4) which are influenced by thyroid status.
In hyperthyroidism, there is increased cortisol production
and a reduced cortisol half-life, the converse being true in
myxoedema.
THYROXINE
The normal half-life of thyroxine(six to seven days) is reduced
to three to four days by hyperthyroidism and prolonged to
nine to ten days by hypothyroidism. This is of considerable
clinical importance when deciding on an appropriate interval
at which to increase the dose of thyroxine in patients treated
for myxoedema, especially if they have coincident ischaemic
heart disease which would be exacerbated if an excessive
steady-statethyroxine level were achieved.
ANTITHYROID DRUGS
The half-life of propylthiouracil and methimazole is pro-
longed in hypothyroidism and shortened in hyperthyroidism.
THYROID DISEASE 39
Key points
Disease profoundly influences the response to many drugs
by altering pharmacokinetics and/or pharmacodynamics.
Gastro-intestinal disease:
(a)diseases that alter gastric emptying influence
the response to oral drugs (e.g. migraine reduces
gastric emptying, limiting the effectiveness of
analgesics);
(b)
ileum/pancreas – relatively minor effects.
Heart failure:
(a)absorption of drugs (e.g. furosemide) is reduced as a
result of splanchnic hypoperfusion;
(b)elimination of drugs that are removed very
ef
ficiently by the liver (e.g. lidocaine) is reduced as a
result of reduced hepatic blood flow, predisposing
to toxicity;
(c) tissue hypoperfusion increases the risk of lactic
acidosis with metformin (cor pulmonale especially
predisposes to this because of hypoxia).
Renal disease:
(a)chronic renal failure – as well as reduced excretion,
drug absorption, distribution and metabolism may
also be altered. Estimates of creatinine clearance or
GFR based on serum creatinine concentration/
weight/age/sex/ ethnicity provide a useful index of
the need for maintenance dose adjustment in
chronic renal failure;
(b)nephrotic syndrome leads to altered drug
distribution because of altered binding to albumin
and altered therapeutic range of concentrations for
drugs that are extensively bound to albumin (e.g.
some anticonvulsants). Albumin in tubular fluid
binds diuretics and causes diuretic resistance.
Glomerular filtration rate is preserved in nephrotic
syndrome by compensatory increased prostaglandin
synthesis, so NSAIDs (see Chapter 26) can precipitate
renal failure.
Liver disease – as well as ef
fects on drug metabolism,
absorption and distribution may also be altered because
of portal systemic shunting, hypoalbuminaemia and
ascites. There is no widely measured biochemical marker
(analogous to serum creatinine in chronic renal failure)
to guide dose adjustment in liver disease, and a cautious
dose titration approach should be used.
Thyroid disease:
(a)hypothyroidism increases sensitivity to digoxin and
opioids;
(b)hyperthyroidism increases sensitivity to warfarin and
reduces sensitivity to digoxin.
These values return to normal on attainment of the euthyroid
state, probably because of altered hepatic metabolism.
OPIATES
Patients with hypothyroidism are exceptionally sensitive to
opioid analgesics, which cause profound respiratory depres-
sion in this setting. This is probably due to reduced metab-
olism and increased sensitivity.
40 EFFECTSOF DISEASE ON DRUG DISPOSITION
FURTHER READING
Carmichael DJS. Chapter 19.2 Handling of drugs in kidney disease. In:
AMA Davison, J Stewart Cameron, J-P Grunfeld, C Ponticelli,
C Van Ypersele, E Ritz and C Winearls (eds). Oxford textbook of clin-
ical nephrology, 3rd edn. Oxford: Oxford University Press, 2005:
2599–618.
Rowland M, Tozer TN. Disease. In: Clinical pharmacokinetics: concepts
and applications, 3rd edn. Baltimore: Williams and Wilkins, 1995:
248–66.
Case history
A 57-year-old alcoholic is admitted to hospital because of
gross ascites and peripheral oedema. He looks chronically
unwell, is jaundiced, and has spider naevi and gynaecomas-
tia. His liver and spleen are not palpable in the presence of
marked ascites. Serum chemistries reveal hypoalbuminuria
(20g/L), sodium 132 mmol/L, potassium 3.5mmol/L, creati-
nine 105μmol/L, and international normalized ratio (INR) is
increased at 1.8. The patient is treated with furosemide
and his fluid intake is restricted. Over the next five days he
loses 10.5kg, but you are called to see him because he has
become confused and unwell. On examination, he is drowsy
and has asterixis (‘liver flap’). His blood pressure is
100/54mmHg with a postural drop. His serum potassium is
2.6mmol/L, creatinine has increased to 138 μmol/L and the
urea concentration has increased disproportionately.
Comment
It is a mistake to try to eliminate ascites too rapidly in patients
with cirrhosis. In this case, in addition to prerenal renal fail-
ure, the patient has developed profound hypokalaemia,
which is commonly caused by furosemide in a patient with
secondary hyperaldosteronism with a poor diet. The
hypokalaemia has precipitated hepatic encephalopathy. It
would have been better to have initiated treatment with
spironolactone to inhibit his endogenous aldosterone. Low
doses of furosemide could be added to this if increasing
doses of spironolactone up to the maximum had not pro-
duced adequate fluid/weight loss. Great caution will be
needed in starting such treatment now that the patient’s
renal function has deteriorated, and serum potassium
levels must be monitored closely.
Introduction 41
Role of drug monitoring in therapeutics 41
Practical aspects 41
Drugs for which therapeutic drug monitoring is used 42
CHAPTER 8
THERAPEUTIC DRUG MONITORING
INTRODUCTION
Drug response differs greatly between individuals. This vari-
ability results from two main sources:
1. variation in absorption, distribution, metabolism or
elimination (pharmacokinetic);
2. variation at or beyond tissue receptors or other
macromolecular drug targets (pharmacodynamic).
Monitoring of drug therapy by biological response encom-
passes both kinds of variability. There must be a continuous
variable that is readily measured and is closely linked to the
desired clinical outcome. Such responses are said to be good
‘surrogate markers’. (‘Surrogate’ because what the prescriber
really wants to achieve is to reduce the risk of a clinical event,
such as a stroke, heart attack, pulmonary embolism, etc.)
For example, antihypertensive drugs are monitored by their
effect on blood pressure (Chapter 28), statins by their effect
on serum cholesterol (Chapter 27), oral anticoagulants by
their effect on the international normalized ratio (INR) (Chapter
30). Many other examples will be encountered in later chapters.
In some circumstances, however, there is no good continu-
ous variable to monitor, especially for diseases with an unpre-
dictable or fluctuating course. Measuring drug concentrations
in plasma or serum identifies only pharmacokinetic variabil-
ity, but can sometimes usefully guide dose adjustment, for
example in treating an epileptic patient with an anticonvulsant
drug. Measuring drug concentrations for use in this way is
often referred to as ‘therapeutic drug monitoring’, and is the
focus of this chapter.
ROLE OF DRUG MONITORING IN
THERAPEUTICS
Measurement of drug concentrations is sometimes a useful
complement to clinical monitoring to assist in selecting the
best drug regimen for an individual patient. Accurate and
convenient assays are necessary. Measurements of drug con-
centrations in plasma are most useful when:
1. There is a direct relationship between plasma
concentration and pharmacological or toxic effect, i.e. a
therapeutic range has been established. (Drugs that work
via active metabolites, and drugs with irreversible actions,
are unsuited to this approach. Tolerance also restricts the
usefulness of plasma concentrations.)
2. Effect cannot readily be assessed quantitatively by clinical
observation.
3. Inter-individual variability in plasma drug concentrations
from the same dose is large (e.g. phenytoin).
4. There is a low therapeutic index (e.g. if the ratio of toxic
concentration/effective concentration is2).
5. Several drugs are being given concurrently and serious
interactions are anticipated.
6. Replacement treatment (for example, of thyroxine) is to be
optimized.
7. Apparent ‘resistance’ to the action of a drug needs an
explanation, when non-compliance is suspected.
Another indication, distinct from therapeutic drug monitor-
ing, for measuring drug concentrations in plasma is in clinical
toxicology. Such measurements can guide management when
specific intervention is contemplated in treating a poisoned
patient (e.g. with paracetamolor aspirin).
PRACTICAL ASPECTS
Drug distribution and the location (tissue and cell) of the
drug’s target influence the relationship between plasma drug
concentration and effect. A constant tissue to plasma drug
concentration ratio only occurs during the terminal β-phase of
elimination. Earlier in the dose interval, the plasma concentra-
tion does not reflect the concentration in the extracellular tis-
sue space accurately. Figure 8.1 illustrates an extreme example
of this in the case of digoxin. Measurements must be made
when enough time has elapsed after a dose for distribution to
have occurred. Greater care is therefore required in the timing
and labelling of specimens for drug concentration determina-
tion than is the case for ‘routine’ chemical pathology speci-
mens. Usually during repeated dosing a sample is taken just
before the next dose to assess the ‘trough’ concentration, and a
sample may also be taken at some specified time after dosing
(depending on the drug) to determine the ‘peak’ concentration.
Given this information, the laboratory should be able to
produce useful information. Advice on the interpretation of
this information is sometimes available from a local therapeutic
drug-monitoring service, such as is provided by some clinical
pharmacology and/or clinical pharmacy departments. In gen-
eral, the cost of measuring drug concentrations is greater than
for routine clinical chemical estimations, and to use expensive
facilities to produce ‘numbers’ resulting from analysis of sam-
ples taken at random from patients described only by name or
number is meaningless and misleading, as well as being a
waste of money.
Analytical techniques of high specificity (often relying on
high-performance liquid chromatography (HPLC), or HPLC-
tandem mass spectroscopy or radioimmunoassay) avoid the
pitfalls of less specific methods which may detect related com-
pounds (e.g. drug metabolites). Even so, quality control moni-
toring of anticonvulsant analyses performed by laboratories
both in the UK and in the USA have revealed that repeated
analyses of a reference sample can produce some startlingly
different results. The most important principle for the clin-
ician is that plasma drug concentrations must always be inter-
preted in the context of the patient’s clinical state.
There are few prospective studies of the impact of thera-
peutic drug-monitoring services on the quality of patient care.
A retrospective survey conducted at the Massachusetts
General Hospital showed that before the use of digoxin
monitoring, 13.9% of all patients receiving digoxin showed
evidence of toxicity, and that this figure fell to 5.9% following
the introduction of monitoring.
DRUGS FOR WHICH THERAPEUTIC DRUG
MONITORING IS USED
Table 8.1 lists those drugs which may be monitored
therapeutically.
1. Digoxin: measuring the plasma concentration can help
optimize therapy, especially for patients in sinus rhythm
where there is no easy pharmacodynamic surrogate
marker of efficacy, and is also useful in suspected toxicity
or poor compliance.
2. Lithium: plasma concentrations are measured 12 hours
after dosing.
3. Aminoglycoside antibiotics – for gentamicin, peak
concentrations measured 30 minutes after dosing of
7–10mg/L are usually effective against sensitive
organisms, and trough levels, measured immediately
before a dose, of 1–2mg/L reduce the risk of toxicity; for
amikacin, the desirable peak concentration is 4–12mg/L,
with a trough value of 4mg/L. With extended interval
aminoglycoside dosing (a single daily dose of 5–7mg/kg),
a single drug concentration determined at a time after the
completion of the distribution phase is used to define
further dosing intervals using validated nomograms.
4. Phenytoin: it is important to be aware of its non-linear
pharmacokinetics (Chapters 3 and 22), and of the possible
effects of concurrent renal or hepatic disease or of pregnancy
on its distribution. Therapeutic drug monitoring
is also widely used for some other anticonvulsants,
such as carbamazepineand sodium valproate.
5. Methotrexate: plasma concentration is an important
predictor of toxicity, and concentrations of 5μmol/L
24 hours after a dose or 100nmol/L 48 hours after dosing
usually require folinic acid administration to prevent
severe toxicity.
6. Theophylline: has a narrow therapeutic index (Figure 8.2)
and many factors influence its clearance (Figure 8.3).
Measurement of plasma theophyllineconcentration can
help to minimize toxicity (e.g. cardiac dysrhythmias or
seizures). Atherapeutic range of 5–20 mg/Lis quoted.
(Plasma concentrations 15mg/L are, however, associated
with severe toxicity in neonates due to decreased protein
binding and accumulation of caffeine, to which
theophyllineis methylated in neonates, but not in older
children.)
7. The therapeutic ranges of plasma concentrations of
several anti-dysrhythmic drugs (e.g. lidocaine) have been
established with reasonable confidence. The therapeutic
range of plasma amiodaroneconcentrations for ventricular
dysrhythmias (1.0–2.5mg/L) is higher than that needed
42 THERAPEUTICDRUG MONITORING
4
3
2
1
0
04812162024
Distribution
phase
Elimination phase
Sampling time
Time (h)
Digoxin concentration
(nmol/L)
Digoxin
administration
Figure 8.1: Serum concentration–time course following digoxin
administration.
Table 8.1:Therapeutic range of several important drugs,
for which therapeutic drug monitoring is often used.
Drug Therapeutic range
Digoxin 0.8–2 mg/L (1–2.6 nmol/L)
Lithium 0.4–1.4mmol/L
a
Phenytoin 10–20 mg/L (40–80μmol/L)
Theophylline 5–20mg/L (28–110 μmol/L)
Ciclosporin 50–200μg/L
a
An upper limit of 1.6mmol/L has also been advocated.
for atrial dysrhythmias (0.5–1.5mg/L). The clinical utility
of predicting toxicity by measuring a metabolite (desethyl
amiodarone) is under evaluation.
8. Immunosuppressants: Ciclosporin compliance is a
particular problem in children, and deterioration in renal
function can reflect either graft rejection due to inadequate
ciclosporinconcentration or toxicity from excessive
concentrations.Sirolimus use should be monitored,
especially when used with ciclosporinor when there is
hepatic impairment or during or after treatment with
inducers or inhibitors of drug metabolism.
DRUGS FOR WHICH THERAPEUTIC DRUG M ONITORINGIS U SED 43
Life-threatening toxicity possible
Coma
Dysrythymias
Convulsions
Dependent on drug concentration
Sinus tachycardia
Excitement
Hypokalaemia
Vomiting
Partially dependent on drug concentration
Nausea
Dyspepsia
Insomnia
Headache
25
20
10
Figure 8.2:Theophylline plasma concentrations (mg/L). Note that
there is a wide variation in the incidence and severity of adverse
effects. (Adapted from Mant T, Henry J, Cochrane G. In: Henry J,
Volans G (eds). ABC of poisoning. Part 1: Drugs. London: British
Medical Journal.)
Decreased
Cirrhosis
Heart failure
Age >50 years
Neonates
Obesity
Severe renal failure
Cimetidine
Erythromycin
Ciprofloxacin
Smoking
Marijuana
Age 1–20 years
High protein diet
Phenobarbitone
Shortened half-lifeProlonged half-life
Increased
Figure 8.3:Theophylline clearance. (Adapted from Mant T, Henry
J, Cochrane G. In: Henry J, Volans G (eds). ABC of poisoning. Part
1: Drugs. London: British Medical Journal.)
Key points
Determining the plasma concentrations of drugs in
order to adjust therapy is referred to as therapeutic
drug monitoring. It has distinct but limited applications.
Therapeutic drug monitoring permits dose
individualization and is useful when there is a clear
relationship between plasma concentration and
pharmacodynamic effects.
The timing of blood samples in relation to dosing is
crucial. For aminoglycosides, samples are obtained for
measurement of peak and trough concentrations. To
guide chronic therapy (e.g. with anticonvulsants),
sufficient time must elapse after starting treatment or
changing dose for the steady state to have been
achieved, before sampling.
Drugs which may usefully be monitored in this way
include digoxin, lithium, aminoglycosides, several
anticonvulsants, methotrexate, theophylline, several
anti-dysrhythmic drugs (including amiodarone) and
ciclosporin.
Individualization of dosage using therapeutic drug
monitoring permits the effectiveness of these drugs to
be maximized, while minimizing their potential toxicity.
Case history
A 35-year-old asthmatic is admitted to hospital at 6 a.m.
because of a severe attack of asthma. She has been treated
with salbutamol and beclometasone inhalers supplemented
by a modified-release preparation of theophylline, 300mg
at night. She has clinical evidence of a severe attack and
does not improve with nebulized salbutamol and oxygen.
Treatment with intravenous aminophylline is considered.
Comment
Aminophylline is a soluble preparation of theophylline (80%)
mixed with ethylenediamine (20%), which has a role in
patients with life-threatening asthma. However, it is essential
to have rapid access to an analytical service to measure plasma
theophylline concentrations if this drug is to be used safely,
especially in this situation where the concentration of
theophylline resulting from the modified-release prepar-
ation that the patient took the night before admission must
be determined before starting treatment. Theophylline
toxicity (including seizures and potentially fatal cardiac
dysrhythmias) can result if the dose is not individualized in
relation to the plasma theophylline concentration.
44 THERAPEUTICDRUG MONITORING
Johannessen SI, Tomson T. Pharmacokinetic variability of newer
antiepileptic drugs – When is monitoring needed? Clinical
Pharmacokinetics2006; 45: 1061–75.
Kaplan B. Mycophenolic acid trough level monitoring in solid organ
transplant recipients treated with mycophenolate mofetil: associ-
ation with clinical outcome. Current Medical Research and Opinion
2006;22: 2355–64.
Mitchell PB. Therapeutic drug monitoring of psychotropic medica-
tions.British Journal of Clinical Pharmacology 2000; 49: 303–12.
Oellerich M, Armstrong VW. The role of therapeutic drug monitoring
in individualizing immunosuppressive drug therapy: Recent
developments.Therapeutic Drug Monitoring 2006; 28: 720–25.
Stamp L, Roberts R, Kennedy M, Barclay M, O’Donnell J, Chapman P.
The use of low dose methotrexate in rheumatoid arthritis – are we
entering a new era of therapeutic drug monitoring and pharma-
cogenomics?Biomedicine and Pharmacotherapy 2006; 60: 678–87.
FURTHER READING
Arns W, Cibrik DM, Walker RG et al. Therapeutic drug monitoring of
mycophenolic acid in solid organ transplant patients treated with
mycophenolate mofetil: Review of the literature. Transplantation
2006;82: 1004–12.
Aronson JK, Hardman M, Reynolds DJM. ABC of monitoring drug
therapy. London: BMJ Publications, 1993.
Bartelink IH, Rademaker CMA, Schobben AFAM et al. Guidelines on
paediatric dosing on the basis of developmental physiology and
pharmacokinetic considerations. Clinical Pharmacokinetics 2006;
45: 1077–97.
Fleming J, Chetty M. Therapeutic monitoring of valproate in psychia-
try: How far have we progressed? Clinical Neuropharmacology2006;
29: 350–60.
Herxheimer A. Clinical pharmacology and therapeutics. In: Warrell
DA (ed.). Oxford textbook of medicine, 4th edn. Oxford: Oxford
University Press, 2006.
Introduction 45
Harmful effects on the fetus 45
Recognition of teratogenic drugs 46
Pharmacokinetics in pregnancy 47
Prescribing in pregnancy 48
CHAPTER 9
DRUGS IN PREGNANCY
INTRODUCTION
The use of drugs in pregnancy is complicated by the potential for
harmful effects on the growing fetus, altered maternal physiol-
ogy and the paucity and difficulties of research in this field.
of the mother with atenololin pregnancy. Early in embryonic
development, exogenous substances accumulate in the neuro-
ectoderm. The fetal blood–brain barrier is not developed until
the second half of pregnancy, and the susceptibility of the cen-
tral nervous system (CNS) to developmental toxins may be
partly related to this. The human placenta possesses multiple
enzymes that are primarily involved with endogenous steroid
metabolism, but which may also contribute to drug metabo-
lism and clearance.
The stage of gestation influences the effects of drugs on
the fetus. It is convenient to divide pregnancy into four
stages, namely fertilization and implantation (17 days), the
organogenesis/embryonic stage (17–57 days), the fetogenic
stage and delivery.
Key points
There is potential for harmful effects on the growing
fetus.
Because of human variation, subtle effects to the fetus
may be virtually impossible to identify.
There is altered maternal physiology.
There is notable paucity of and dif
ficulties in research
in this area.
Assume all drugs are harmful until proven otherwise.
HARMFUL EFFECTS ON THE FETUS
Because experience with many drugs in pregnancy is severely
limited, it should be assumed that all drugs are potentially
harmful until sufficient data exist to indicate otherwise. ‘Social’
drugs (alcohol and cigarette smoking) are definitely damaging
and their use must be discouraged.
In the placenta, maternal blood is separated from fetal
blood by a cellular membrane (Figure 9.1). Most drugs with a
molecular weight of less than 1000 can cross the placenta. This
is usually by passive diffusion down the concentration gradi-
ent, but can involve active transport. The rate of diffusion
depends first on the concentration of free drug (i.e. non-protein
bound) on each side of the membrane, and second on the
lipid solubility of the drug, which is determined in part by
the degree of ionization. Diffusion occurs if the drug is in the
unionized state. Placental function is also modified by changes
in blood flow, and drugs which reduce placental blood flow
can reduce birth weight. This may be the mechanism which
causes the small reduction in birth weight following treatment
Maternal
blood
space
Trophoblast
Fetal
capillary
Carrier
protein
Small
(MW <1000)
drugs
Larger
(MW >1000)
drugs
DD
D+
D−
D+
D−
D+
D−
D
DDD
Passive
diffusion
Passive
diffusion
Figure 9.1:Placental transfer of drugs from mother to fetus.
Key points
A cellular membrane separates the maternal and fetal
blood.
Most drugs cross the placenta by passive diffusion.
Placental function is modified by changes in blood flow.
There are multiple placental enzymes, primarily
involved with endogenous steroid metabolism, which
may also contribute to drug metabolism.
FERTILIZATION AND IMPLANTATION
Animal studies suggest that interference with the fetus before
17 days gestation causes abortion, i.e. if pregnancy continues
the fetus is unharmed.
ORGANOGENESIS/EMBRYONIC STAGE
At this stage, the fetus is differentiating to form major organs,
and this is the critical period for teratogenesis. Teratogens cause
deviations or abnormalities in the development of the embryo
that are compatible with prenatal life and are observable post-
natally. Drugs that interfere with this process can cause gross
structural defects (e.g. thalidomidephocomelia).
Some drugs are confirmed teratogens (Table 9.1), but for
many the evidence is inconclusive. Thalidomidewas unusual
in the way in which a very small dose of the drug given on
only one or two occasions between the fourth and seventh weeks
of pregnancy predictably produced serious malformations.
FETOGENIC STAGE
In this stage, the fetus undergoes further development and
maturation. Even after organogenesis is almost complete, drugs
can still have significant adverse effects on fetal growth and
development.
ACE inhibitors and angiotensin receptor blockers cause
fetal and neonatal renal dysfunction.
Drugs used to treat maternal hyperthyroidism can cause
fetal and neonatal hypothyroidism.
Tetracyclineantibiotics inhibit growth of fetal bones and
stain teeth.
Aminoglycosides cause fetal VIIIth nerve damage.
Opioids and cocaine taken regularly during pregnancy
can lead to fetal drug dependency.
Warfarincan cause fetal intracerebral bleeding.
Indometacin, a potent inhibitor of prostaglandin
synthesis, is used under specialist supervision to assist
closure of patent ductus arteriosus in premature infants.
Some hormones can cause inappropriate virilization or
feminization.
Anticonvulsants may possibly be associated with mental
retardation.
Cytotoxic drugs can cause intrauterine growth retardation
and stillbirth.
DELIVERY
Some drugs given late in pregnancy or during delivery may
cause particular problems. Pethidine, administered as an anal-
gesic can cause fetal apnoea (which is reversed with naloxone,
see Chapter 25). Anaesthetic agents given during Caesarean
section may transiently depress neurological, respiratory and
muscular functions. Warfaringiven in late pregnancy causes a
haemostasis defect in the baby, and predisposes to cerebral
haemorrhage during delivery.
46 DRUGS IN PREGNANCY
Table 9.1:Some drugs that are teratogenic in humans.
Thalidomide Androgens
Cytotoxic agents Progestogens
Alcohol Danozol
Warfarin Diethylstilbestrol
Retinoids Radioisotopes
Most anticonvulsants Some live vaccines
Ribavarin Lithium
Key points
Fertilization and implantation, 17 days.
Organogenesis/embryonic stage, 17–57 days.
Fetogenic stage.
Delivery.
THE MALE
Although it is generally considered that sperm cells damaged by
drugs will not result in fertilization, the manufacturers of griseo-
fulvin, an antifungal agent, advise men not to father children
during or for six months after treatment. Finasteride, an anti-
androgen used in the treatment of benign prostatic hyperplasia,
is secreted in semen, and may be teratogenic to male fetuses.
RECOGNITION OF TERATOGENIC DRUGS
Major malformations that interfere with normal function
occur in 2–3% of newborn babies, and a small but unknown
fraction of these are due to drugs. Two principal problems face
those who are trying to determine whether a drug is terato-
genic when it is used to treat disease in humans:
1. Many drugs produce birth defects when given experi-
mentally in large doses to pregnant animals. This does not
necessarily mean that they are teratogenic in humans at
therapeutic doses. Indeed, the metabolism and kinetics of
drugs at high doses in other species is so different from that
in humans as to limit seriously the relevance of such studies.
2. Fetal defects are common (2–3%). Consequently, if the
incidence of drug-induced abnormalities is low, a very
large number of cases has to be observed to define a
significant increase above this background level. Effects
on the fetus may take several years to become clinically
manifest. For example, diethylstilbestrolwas widely
used in the late 1940s to prevent miscarriages and preterm
births, despite little evidence of efficacy. In 1971, an
association was reported between adenocarcinoma of the
vagina in girls in their late teens whose mothers had been
givendiethylstilbestrol during the pregnancy. Exposure
tostilbestrol in utero has also been associated with a
T-shaped uterus and other structural abnormalities of the
genital tract, and increased rates of ectopic pregnancy and
premature labour.
ABSORPTION
Gastric emptying and small intestinal motility are reduced. This
is of little consequence unless rapid drug action is required.
Vomiting associated with pregnancy may make oral drug
administration impractical.
DISTRIBUTION
During pregnancy, the blood volume increases by one-third,
with expansion in plasma volume (from 2.5 to 4L at term) being
disproportionate to expansion in red cell mass, so that haemat-
ocrit falls. There is also an increase in body water due to a larger
extravascular volume and changes in the uterus and breasts.
Oedema, which at least one-third of women experience
during pregnancy, may add up to 8L to the volume of extra-
cellular water. For water-soluble drugs (which usually have a
relatively small volume of distribution), this increases the
apparent volume of distribution and, although clearance is
unaltered, their half-life is prolonged. During pregnancy, the
plasma protein concentration falls and there is increased com-
petition for binding sites due to competition by endogenous
ligands, such as increased hormone levels. These factors alter
the total amount of bound drug and the apparent volume of
distribution. However, the concentration of free drug usually
remains unaltered, because a greater volume of distribution of
free drug is accompanied by increased clearance of free drug.
Thus, in practice, these changes are rarely of pharmacological
significance. They may cause confusion in monitoring of
plasma drug levels, since this usually measures total (rather
than free) drug concentrations.
METABOLISM
Metabolism of drugs by the pregnant liver is increased, largely
due to enzyme induction, perhaps by raised hormone levels.
Liver blood flow does not change. This may lead to an increased
rate of elimination of those drugs (e.g. theophylline), for
which enzyme activity rather than liver blood flow is the main
determinant of elimination rate.
RENAL EXCRETION
Excretion of drugs via the kidney increases because renal plasma
flow almost doubles and the glomerular filtration rate increases
by two-thirds during pregnancy. This has been documented for
digoxin,lithium, ampicillin, cefalexin and gentamicin.
PHARMACOKINETICS I N PREGNANCY 47
Key points
The background incidence of serious congenital
abnormality recognized at birth is 2–3%.
Environmental and genetic factors can influence a
drug’s effect.
Maternal disease can af
fect the fetus.
Studies of large doses in pregnant animals are of
doubtful relevance.
Effects may be delayed (e.g. diethylstilbestrol).
Meticulous data collection is required for drugs
administered during pregnancy and outcome, including
long-term follow up. At present the Medicines and
Healthcare products Regulatory Agency (MHRA) requests
records of all drugs administered to a mother who
bears an abnormal fetus. More complete (but with
inherent practical difficulties) data collection by the
MHRA, the National Teratology Information Services,
the pharmaceutical industry and drug information
agencies on all prescriptions during pregnancy with
long-term follow up of of
fspring is required.
PHARMACOKINETICS I N PREGNANCY
Known differences in drug effects in pregnancy are usually
explained by altered pharmacokinetics (Figure 9.2).
Drug
Absorption
Metabolism
Excretion
Renal
blood flow
CYP450
induction
Plasma drug
concentration
↑ ↔ or
Volume of distribution
Plasma albumin
Vomiting
Gastric emptying
Small intestinal motility
Figure 9.2:Pharmacokinetic changes in pregnancy.
Key points
Known differences in drug effects can usually be explained
by altered pharmacokinetics. Increased volume of
distribution, hepatic metabolism and renal excretion all
tend to reduce drug concentration. Decreased plasma
albumin levels increase the ratio of free drug in plasma.
PRESCRIBING IN PREGNANCY
The prescription of drugs to a pregnant woman is a balance
between possible adverse drug effects on the fetus and the risk
to mother and fetus of leaving maternal disease inadequately
treated. Effects on the human fetus cannot be reliably pre-
dicted from animal studies – hence one should prescribe drugs
for which there is experience of safety over many years in
preference to new or untried drugs. The smallest effective
dose should be used. The fetus is most sensitive to adverse
drug effects during the first trimester. It has been estimated
that nearly half of all pregnancies in the UK are unplanned,
and that most women do not present to a doctor until five to
seven weeks after conception. Thus, sexually active women of
childbearing potential should be assumed to be pregnant until
it has been proved otherwise.
Delayed toxicity is a sinister problem (e.g. diethylstilbestrol)
and if the teratogenic effect of thalidomide had not produced
such an unusual congenital abnormality, namely phocomelia,
its detection might have been delayed further. If drugs (or envi-
ronmental toxins) have more subtle effects on the fetus (e.g. a
minor reduction in intelligence) or cause an increased incidence
of a common disease (e.g. atopy), these effects may never be
detected. Many publications demand careful prospective con-
trolled clinical trials, but the ethics and practicalities of such
studies often make their demands unrealistic. Amore rational
approach is for drug regulatory bodies, the pharmaceutical
industry and drug information agencies to collaborate closely
and internationally to collate all information concerning drug
use in pregnancy (whether inadvertent or planned) and associ-
ate these with outcome. This will require significant investment
of time and money, as well as considerable encouragement to
doctors and midwives to complete the endless forms.
Guidance on the use of drugs for a selection of conditions is
summarized below. If in doubt, consult the British National
Formulary, appendix 4 (which is appropriately conservative).
Information for health professionals in the UK about the
safety of drugs in pregnancy can also be obtained from the
National Teratology Information Service (Tel. 0191 232 1525).
ANTIMICROBIAL DRUGS
Antimicrobial drugs are commonly prescribed during preg-
nancy. The safest antibiotics in pregnancy are the penicillins
and cephalosporins. Trimethoprimis a theoretical teratogen
as it is a folic acid antagonist. The aminoglycosides can
cause ototoxicity. There is minimal experience in pregnancy
with the fluoroquinolones (e.g. ciprofloxacin) and they should
be avoided. Erythromycinis probably safe. Metronidazole is
a teratogen in animals, but there is no evidence of teratogenic-
ity in humans, and its benefit in serious anaerobic sepsis prob-
ably outweighs any risks. Unless there is a life-threatening
infection in the mother, antiviral agents should be avoided in
pregnancy. Falciparum malaria (Chapter 47) has an especially
high mortality rate in late pregnancy. Fortunately, the stan-
dard regimens of intravenous and oral quinine are safe in
pregnancy.
ANALGESICS
Opioids cross the placenta. This is particularly relevant in the
management of labour when the use of opioids, such as pethi-
dine, depresses the fetal respiratory centre and can inhibit
the start of normal respiration. If the mother is dependent on
opioids, the fetus can experience opioid withdrawal syn-
drome during and after delivery, which can be fatal. In
neonates, the chief withdrawal symptoms are tremor, irritabil-
ity, diarrhoea and vomiting. Chlorpromazine is commonly
used to treat this withdrawal state. Paracetamolis preferred to
aspirin when mild analgesia is required. In cases where a sys-
temic anti-inflammatory action is required (e.g. in rheumatoid
arthritis), ibuprofen is the drug of choice. Non-steroidal
anti-inflammatory drugs can cause constriction of the ductus
arteriosus. Occasionally, this may be used to therapeutic
benefit.
ANAESTHESIA
Anaesthesia in pregnancy is a very specialist area and should
only be undertaken by experienced anaesthetists. Local anaes-
thetics used for regional anaesthesia readily cross the pla-
centa. However, when used in epidural anaesthesia, the drug
remains largely confined to the epidural space. Pregnant
women are at increased risk of aspiration. Although com-
monly used, pethidine frequently causes vomiting and may
also lead to neonatal respiratory depression. Metoclopramide
should be used in preference to prochlorperazine(which has
48 DRUGS IN PREGNANCY
Key points
Prescribing in pregnancy is a balance between the risk of
adverse drug effects on the fetus and the risk of leaving
maternal disease untreated. The effects on the human
fetus are not reliably predicted by animal experiments.
However, untreated maternal disease may cause morbidity
and/or mortality to mother and/or fetus.
Therefore,
minimize prescribing;
use ‘tried and tested’ drugs whenever possible in
preference to new agents;
use the smallest effective dose;
remember that the fetus is most sensitive in the first
trimester;
consider pregnancy in all women of childbearing
potential;
discuss the potential risks of taking or withholding
therapy with the patient;
seek guidance on the use of drugs in pregnancy in the
British National Formulary, Drug Information Services,
National Teratology Information Service (NTIS);
warn the patient about the risks of smoking, alcohol,
over
-the-counter drugs and drugs of abuse.
an anti-analgesic effect when combined with pethidine), and
naloxone (an opioid antagonist) must always be available.
Respiratory depression in the newborn is not usually a prob-
lem with modern general anaesthetics currently in use in
Caesarean section. Several studies have shown an increased
incidence of spontaneous abortions in mothers who have had
general anaesthesia during pregnancy, although a causal rela-
tionship is not proven, and in most circumstances failure to
operate would have dramatically increased the risk to mother
and fetus.
ANTI-EMETICS
Nausea and vomiting are common in early pregnancy, but are
usually self-limiting, and ideally should be managed with
reassurance and non-drug strategies, such as small frequent
meals, avoiding large volumes of fluid and raising the head of
the bed. If symptoms are prolonged or severe, drug treatment
may be effective. An antihistamine, e.g. promethazine or
cyclizine may be required. If ineffective, prochlorperazine is
an alternative. Metoclopramide is considered to be safe and
efficacious in labour and before anaesthesia in late pregnancy,
but its routine use in early pregnancy cannot be recommended
because of the lack of controlled data, and the significant inci-
dence of dystonic reactions in young women.
DYSPEPSIA AND CONSTIPATION
The high incidence of dyspepsia due to gastro-oesophageal
reflux in the second and third trimesters is probably related to
the reduction in lower oesophageal sphincter pressure. Non-
drug treatment (reassurance, small frequent meals and advice
on posture) should be pursued in the first instance, particu-
larly in the first trimester. Fortunately, most cases occur later
in pregnancy when non-absorbable antacids, such as algi-
nates, should be used. In late pregnancy, metoclopromideis
particularly effective as it increases lower oesophageal sphinc-
ter pressure. H
2
-receptor blockers should not be used for non-
ulcer dyspepsia in this setting. Constipation should be managed
with dietary advice. Stimulant laxatives may be uterotonic
and should be avoided if possible.
PEPTIC ULCERATION
Antacids may relieve symptoms. Cimetidine and ranitidine
have been widely prescribed in pregnancy without obvious
damage to the fetus. There are inadequate safety data on the
use of omeprazole or other proton pump inhibitors in preg-
nancy. Sucralfate has been recommended for use in preg-
nancy in the USA, and this is rational as it is not systemically
absorbed. Misoprostol, a prostaglandin which stimulates the
uterus, is contraindicated because it causes abortion.
ANTI-EPILEPTICS
Epilepsy in pregnancy can lead to fetal and maternal morbid-
ity/mortality through convulsions, whilst all of the anticon-
vulsants used have been associated with teratogenic effects
(e.g. phenytoin is associated with cleft palate and congenital
heart disease). However, there is no doubt that the benefits of
good seizure control outweigh the drug-induced teratogenic
risk. Thorough explanation to the mother, ideally before a
planned pregnancy, is essential, and it must be emphasized
that the majority (90%) of epileptic mothers have normal
babies. (The usual risk of fetal malformation is 2–3% and in
epileptic mothers it is up to 10%.) In view of the association of
spina bifida with many anti-epileptics, e.g. sodium valproate
andcarbamazepine therapy, it is often recommended that the
standard dose of folic acid should be increased to 5mg daily.
Both of these anti-epileptics can also cause hypospadias. As
in non-pregnant epilepsy, single-drug therapy is preferable.
Plasma concentration monitoring is particularly relevant for
phenytoin, because the decrease in plasma protein binding and
the increase in hepatic metabolism may cause considerable
changes in the plasma concentration of free (active) drug. As
always, the guide to the correct dose is freedom from fits and
absence of toxicity. Owing to the changes in plasma protein
binding, it is generally recommended that the therapeutic
range is 5–15mg/L, whereas in the non-pregnant state it is
10–20mg/L. This is only a rough guide, as protein binding
varies.
The routine injection of vitamin K recommended at birth
counteracts the possible effect of some anti-epileptics on
vitamin K-dependent clotting factors.
Magnesium sulphate is the treatment of choice for the pre-
vention and control of eclamptic seizures.
PRESCRIBING IN PREGNANCY 49
Key points
Epilepsy in pregnancy can lead to increased fetal
and maternal morbidity/mortality.
All anticonvulsants are teratogens.
The benefits of good seizure control outweigh
drug-induced teratogenic risk.
Give a full explanation to the mother (preferably
before pregnancy): most epileptic mothers (90%)
have normal babies.
Advise an increase in the standard dose of folic acid up
to 12 weeks.
Make a referral to the neurologist and obstetrician.
If epilepsy is well controlled, do not change
therapy.
Monitor plasma concentrations (levels tend to fall,
and note that the bound: unbound ratio changes); the
guide to the correct dose is freedom from fits and
absence of toxicity
.
An early ultrasound scan at 12 weeks may detect gross
neural tube defects.
Detailed ultrasound scan and α-fetoprotein at 16–18
weeks should be considered.
ANTICOAGULATION
Warfarinhas been associated with nasal hypoplasia and chon-
drodysplasia when given in the first trimester, and with CNS
abnormalities after administration in later pregnancy, as well
as a high incidence of haemorrhagic complications towards
the end of pregnancy. Neonatal haemorrhage is difficult to
prevent because of the immature enzymes in fetal liver and
the low stores of vitamin K. It is not rcommended for use in
pregnancy unless there are no other options. Low molecular
weight heparin (LMWH), which does not cross the placenta,
is the anticoagulant of choice in pregnancy in preference to
unfractionated heparin. LMWH has predictable pharmacoki-
netics and is safer – unlike unfractionated heparin there has
never been a case of heparin-induced thrombocytopenia/
thrombosis (HITT) associated with it in pregnancy. Moreover
there has only been one case of osteoporotic fracture world-
wide, whereas there is a 2% risk of osteoporotic fracture with
nine months use of unfractionated heparin (see Chapter 30).
LMWHis given twice daily in pregnancy due to the increased
renal clearance of pregnancy. Women on long-term oral anti-
coagulants should be warned that these drugs are likely to
affect the fetus in early pregnancy. Self-administered subcuta-
neous LMWH must be substituted for warfarin before six
weeks’ gestation. Subcutaneous LMWH can be continued
throughout pregnancy and for the prothrombotic six weeks
post partum.Patients with prosthetic heart valves present a
special problem, and in these patients, despite the risks to the
fetus, warfarin is often given up to 36 weeks. The prothrom-
bin time/international normalized ratio (INR) should be mon-
itored closely if warfarinis used.
thiazide diuretics in women with essential hypertension, who
are already stabilized on these drugs. Modified-release prepara-
tions of nifedipine are also used for hypertension in preg-
nancy, but angiotensin-converting enzyme inhibitors and
angitensin II receptor antagonists must be avoided.
HORMONES
Progestogens, particularly synthetic ones, can masculinize the
female fetus. There is no evidence that this occurs with the small
amount of progestogen(or oestrogen) present in the oral con-
traceptive – the risk applies to large doses. Corticosteroids do
not appear to give rise to any serious problems when given via
inhalation or in short courses. Transient suppression of the
fetal hypothalamic–pituitary–adrenal axis has been reported.
Rarely, cleft palate and congenital cataract have been linked
with steroids in pregnancy, but the benefit of treatment usually
outweighs any such risk. Iodine and antithyroid drugs cross
the placenta and can cause hypothyroidism and goitre.
TRANQUILLIZERS AN D ANTIDEPRESSANTS
Benzodiazepines accumulate in the tissues and are slowly
eliminated by the neonate, resulting in prolonged hypotonia
(‘floppy baby’), subnormal temperatures (hypothermia), peri-
odic cessation of respiration and poor sucking. There is no evi-
dence that the phenothiazines, tricyclic antidepressants or
fluoxetineare teratogenic. Lithium can cause fetal goitre and
possible cardiovascular abnormalities.
NON-THERAPEUTIC DRUGS
Excessiveethanol consumption is associated with spontaneous
abortion, craniofacial abnormalities, mental retardation, con-
genital heart disease and impaired growth. Even moderate
alcohol intake may adversely affect the baby – the risk of hav-
ing an abnormal child is about 10% in mothers drinking
30–60mL ethanol per day, rising to 40% in chronic alcoholics.
Fetal alcohol syndrome describes the distinct pattern of abnor-
mal morphogenesis and central nervous system dysfunction
in children whose mothers were chronic alcoholics, and this
syndrome is a leading cause of mental retardation. After birth,
the characteristic craniofacial malformations diminish, but
microcephaly and to a lesser degree short stature persist.
Cigarette smoking is associated with spontaneous abortion,
premature delivery, small babies, increased perinatal mortal-
ity and a higher incidence of sudden infant death syndrome
(cot death). Cocaine causes vasoconstriction of placental ves-
sels. There is a high incidence of low birth weight, congenital
abnormalities and, in particular, delayed neurological and
behavioural development.
50 DRUGS IN PREGNANCY
Key points
Pregnancy is associated with a hypercoagulable state.
Warfarin has been associated with nasal hypoplasia and
chondrodysplasia in the first trimester, and with CNS
abnormalities in late pregnancy, as well as haemorrhagic
complications.
Heparin does not cross the placenta. Low molecular
weight heparins (LMWH
) are preferable as they have
better and more predictable pharmacokinetics, are
safer with no evidence of heparin-induced thrombo-
cytopenia and thrombosis (HITT) and osteoporotic
fracture is very rare.
Refer to the guidelines of the Royal College of
Obstetricians for thromboprophylaxis and management
of established venous thromboembolism in pregnancy.
CARDIOVASCULAR DRUGS
Hypertension in pregnancy (see Chapter 28) can normally be
managed with either methyldopa which has the most exten-
sive safety record in pregnancy, or labetalol. Parenteral
hydralazine is useful for lowering blood pressure in pre-
eclampsia. Diuretics should not be started to treat hypertension
in pregnancy, although some American authorities continue
FURTHER READING
Anon. Antiepileptics, pregnancy and the child. Drugs and Therapeutics
Bulletin2005; 43 no 2.
Koren G. Medication, safety in pregnancy and breastfeeding: the evidence-
based A–Z clinicians pocket guide. Maidenhead: McGraw-Hill, 2006.
Rubin PC. Prescribing in pregnancy, 3rd edn. London: Blackwell, BMJ
Books, 2000.
McElhatton PR. General principles of drug use in pregnancy.
Pharmaceutical Journal2003; 270: 305–7.
FURTHER INFORMATION FOR HEALTH
PROFESSIONALS
National Teratology Information Service
Regional Drug and Therapeutics Centre
Wolfson Unit
Clarement Place
Newcastle upon Tyne
NE1 4LP
Tel. 0191 232 1525
PRESCRIBING IN PREGRANCY 51
Case history
A 20-year-old female medical student attended her GP
requesting a course of Septrin® (co-trimoxazole) for cysti-
tis. She tells her GP that her last menstrual bleed was about
six weeks earlier. She did not think she was at risk of preg-
nancy as her periods had been irregular since stopping the
oral contraceptive one year previously due to fears about
thrombosis, and her boyfriend used a condom. Physical exam-
ination, which did not include a vaginal examination, was
normal. Urinalysis was 1positive for blood and a trace of
protein.
Question
Why should the GP not prescribe co-trimoxazole for this
patient?
Answer
Until proven otherwise, it should be assumed that this
woman is pregnant. Co-trimoxazole (a combination of sul-
famethoxazole and trimethoprim) has been superseded by
trimethoprim alone as a useful drug in lower urinary tract
infection (UTI). The sulfamethoxazole does not add signifi-
cant antibacterial advantage in lower UTI, but does have
sulphonamide-associated side effects, including the rare
but life-threatening Stevens–Johnson syndrome. Both sul-
famethoxazole and trimethoprim inhibit folate synthesis
and are theoretical teratogens. If pregnancy is confirmed
(urinary frequency is an early symptom of pregnancy in
some women, due to a progesterone effect) and if the
patient has a lower UTI confirmed by pyuria and bacteria
on microscopy whilst awaiting culture and sensitivity results,
amoxicillin is the treatment of choice. Alternatives include
an oral cephalosporin or nitrofurantoin. Note that lower
urinary tract infection in pregnancy can rapidly progress to
acute pyelonephritis.
Introduction 52
Pharmacokinetics 52
Pharmacodynamics 53
Breast-feeding 53
Practical aspects of prescribing 54
Research 54
CHAPTER 10
DRUGS IN INFANTS AND
CHILDREN
INTRODUCTION
Children cannot be regarded as miniature adults in terms
of drug response, due to differences in body constitution,
drug absorption and elimination, and sensitivity to adverse
reactions. Informed consent is problematic and commercial
interest has been limited by the small size of the market,
so clinical trials in children have lagged behind those in
adults. Regulatory agencies in the USAand Europe now rec-
ognize this problem and are attempting to address it, for
example, by introducing exclusivity legislation designed to
attract commercial interest. Traditionally, paediatricians have
used drugs ‘off-label’ (i.e. for unlicensed indications), often
gaining experience in age groups close to those for which a
product is licensed and then extending this to younger chil-
dren. That this empirical approach has worked (at least to
some extent) is testament to the biological fact that while
not just ‘miniature adults’ children do share the same drug
targets (e.g. receptors, enzymes), cellular transduction mecha-
nisms and physiological processes with their parents. Drug
responses are thus usually qualitatively similar in children
and adults, although there are important exceptions, including
some central nervous system (CNS) responses and immuno-
logical responses to ciclosporin. Furthermore, some adverse
effects occur only during certain stages of development, for
example, retrolental fibroplasia induced by excess oxygen
in the premature neonate and staining of teeth by
tetracycline which occurs only in developing enamel. The
processes of drug elimination are, however, immature at
birth so quantitative differences (e.g. in dose) are important.
Establishing optimal doses for drugs prescribed for children is
thus an extremely important clinical challenge. Current
regimes have been arrived at empirically, but guidelines are
evolving for paediatric dosing in clinical trials and in future
greater use may be made of pharmacokinetic/pharmacody-
namic modelling in children, so hopefully this Cinderella
of therapeutics will soon be making her (belated) entry to
the ball.
PHARMACOKINETICS
ABSORPTION
Gastro-intestinal absorption is slower in infancy, but absorption
from intramuscular injection is faster. The rate of gastric empty-
ing is very variable during the neonatal period and may be
delayed by disease, such as respiratory distress syndrome and
congenital heart disease. To ensure that adequate blood concen-
trations reach the systemic circulation in the sick neonate, it is
common practice to use intravenous preparations. In older and
less severely ill children, oral liquid preparations are commonly
used, resulting in less accurate dosing and a more rapid rate of
absorption. This is important for drugs with adverse effects that
occur predictably at high plasma concentration, and which show
lack of efficacy if trough concentration is low (e.g. carbamazepine
andtheophylline). Infant skin is thin and percutaneous absorp-
tion can cause systemic toxicity if topical preparations (e.g. of
potent corticosteroids) are applied too extensively.
DISTRIBUTION
Body fat content is relatively low in children, whereas water
content is greater, leading to a lower volume of distribution of
fat-soluble drugs (e.g. diazepam) in infants. Plasma protein
binding of drugs is reduced in neonates due to a lower plasma
albumin concentration and altered binding properties. The
risk of kernicterus caused by displacement of bilirubin from
albumin by sulphonamides (see Chapter 12) is well recog-
nized. The blood–brain barrier is more permeable in neonates
and young children, leading to an increased risk of CNS
adverse effects.
BREAST-FEEDING 53
METABOLISM
At birth, the hepatic microsomal enzyme system (see Chapter
5) is relatively immature (particularly in the preterm infant),
but after the first four weeks it matures rapidly.
Chloramphenicol can produce ‘grey baby syndrome’ in
neonates due to high plasma levels secondary to inefficient
elimination. Conversely, hepatic drug metabolism can be
increased once enzyme activity has matured in older infants
and children, because the ratio of the weight of the liver to
body weight is up to 50% higher than in adults. Drugs admin-
istered to the mother can induce neonatal enzyme activity
(e.g. barbiturates). Phenobarbitone metabolism is faster in
children than in adults because of greater induction of hepatic
enzyme activity.
PHARMACODYNAMICS
Documented evidence of differences in receptor sensitivity in
children is lacking, and the apparently paradoxical effects
of some drugs (e.g. hyperkinesia with phenobarbitone, sedation
of hyperactive children with amphetamine) are as yet
unexplained. Augmented responses to warfarinin prepubertal
patients occur at similar plasma concentrations as in adults,
implying a pharmacodynamic mechanism. Ciclosporin added
in vitro to cultured monocytes (hence there is no opportunity for
a pharmacokinetic effect) has greater effects in cells isolated from
infants, providing another example of an age-related pharmaco-
dynamic difference.
BREAST-FEEDING
Breast-feeding can lead to toxicity in the infant if the drug
enters the milk in pharmacological quantities. The milk con-
centration of some drugs (e.g. iodides) may exceed the mater-
nal plasma concentration, but the total dose delivered to the
baby is usually very small. However, drugs in breast milk may
cause hypersensitivity reactions even in very low doses.
Virtually all drugs that reach the maternal systemic circulation
will enter breast milk, especially lipid-soluble unionized low-
molecular-weight drugs. Milk is weakly acidic, so drugs that
are weak bases are concentrated in breast milk by trapping of
the charged form of the drug (compare with renal elimination;
see Chapter 6). However, the resulting dose administered to
the fetus in breast milk is seldom clinically appreciable,
although some drugs are contraindicated (Table 10.2), and
breast-feeding should cease during treatment if there is no
safer alternative. Appendix 5 of the British National
Formulary provides very helpful practical advice.
Key points
Prevalence of chronic illness in children requiring drug
therapy:
one in eight children have asthma;
one in 250 children have epilepsy;
one in 750 children have diabetes mellitus.
Key points
At birth, renal and hepatic function are less efficient than
in adulthood. Drug effects may be prolonged and
accumulation may occur. These factors are exaggerated in
the premature infant.
EXCRETION
All renal mechanisms (filtration, secretion and reabsorption)
are reduced in neonates, and renal excretion of drugs is rela-
tively reduced in the newborn. Glomerular filtration rate
(GFR) increases rapidly during the first four weeks of life, with
consequent changes in the rate of drug elimination (Table 10.1).
Table 10.1: Changes in rate of drug elimination with development
Stage of development Plasma half-life of gentamicin
Premature infant
48 hours old 18 hours
5–22 days old 6 hours
Normal infant
1–4 weeks old 3 hours
Adult 2 hours
Table 10.2:Some drugs to be avoided during breast-feeding
Amiodarone
Aspirin
Benzodiazepines
Chloramphenicol
Ciclosporin
Ciprofloxacin
Cocaine
Combined oral contraceptives
Cytotoxics
Ergotamine
Octreotide
Stimulant laxatives
Sulphonylureas
Thiazide diuretics
Vitamin A/retinoid analogues (e.g. etretinate)
54 DRUGS IN INFANTS AND CHILDREN
The infant should be monitored if β-adrenoceptor antago-
nists, carbimazole, corticosteroids or lithium are prescribed
to the mother. β-Adrenoceptor antagonists rarely cause signif-
icant bradycardia in the suckling infant. Carbimazoleshould
be prescribed at its lowest effective dose to reduce the risk
of hypothyroidism in the neonate/infant. In high doses,
corticosteroids can affect the infant’s adrenal function and
lithium may cause intoxication. There is a theoretical risk of
Reye’s syndrome if aspirinis prescribed to the breast-feeding
mother. Warfarin is not contraindicated during breast-feed-
ing. Bromocriptine suppresses lactation and large doses of
diuretics may do likewise. Metronidazole gives milk an
unpleasant taste.
PRACTICAL ASPECTS OF PRESCRIBING
COMPLIANCE AND ROUTE OF AD MINISTRATION
Sick neonates will usually require intravenous drug adminis-
tration. Accurate dosage and attention to fluid balance are
essential. Sophisticated syringe pumps with awareness of
‘dead space’ associated with the apparatus are necessary.
Children under the age of five years may have diffi-
culty in swallowing even small tablets, and hence oral
preparations which taste pleasant are often necessary to
improve compliance. Liquid preparations are given by means
of a graduated syringe. However, chronic use of sucrose-
containing elixirs encourages tooth cavities and gingivitis.
Moreover, the dyes and colourings used may induce hyper-
sensitivity.
Pressurized aerosols (e.g. salbutamolinhaler, see Chapter
33) are usually only practicable in children over the age of ten
years, as co-ordinated deep inspiration is required unless a
device such as a spacer is used. Spacers can be combined with
a face mask from early infancy. Likewise, nebulizers may be
used to enhance local therapeutic effect and reduce systemic
toxicity.
Only in unusual circumstances, i.e. extensive areas of
application (especially to inflamed or broken skin), or in
infants, does systemic absorption of drugs (e.g. steroids,
neomycin) become significant following topical application
to the skin.
Intramuscular injection should only be used when
absolutely necessary. Intravenous therapy is less painful, but
skill is required to cannulate infants’ veins (and a confident
colleague to keep the target still!). Children find intravenous
infusions uncomfortable and restrictive. Rectal administration
(see Chapter 4) is a convenient alternative (e.g. metronidazole
to treat anaerobic infections). Rectal diazepamis particularly
valuable in the treatment of status epilepticus when intra-
venous access is often difficult. Rectal diazepammay also be
administered by parents. Rectal administration should also be
considered if the child is vomiting.
Paramount to ensuring compliance is full communication
with the child’s parents and teachers. This should include
information not only on how to administer the drug, but
also on why it is being prescribed, for how long the treat-
ment should continue and whether any adverse effects are
likely.
Case history
A two-year-old epileptic child is seen in the Accident and
Emergency Department. He has been fitting for at least 15
minutes. The casualty officer is unable to cannulate a vein
to administer intravenous diazepam. The more experi-
enced medical staff are dealing with emergencies else-
where in the hospital.
Question
Name two drugs, and their route of administration,
with which the casualty officer may terminate the convul-
sions.
Answer
Rectal diazepam solution.
Rectal or intramuscular paraldehyde.
DOSAGE
Even after adjustment of dose according to surface area,
calculation of the correct dose must consider the relatively
large volume of distribution of polar drugs in the first four
months of life, the immature microsomal enzymes and
reduced renal function. The British National Formulary and
specialist paediatric textbooks and formularies provide
appropriate guidelines and must be consulted by physicians
who are not familiar with prescribing to infants and
children.
ADVERSE EFFECTS
With a few notable exceptions, drugs in children generally
have a similar adverse effect profile to those in adults. Of par-
ticular significance is the potential of chronic corticosteroid
use, including high-dose inhaled corticosteroids, to inhibit
growth. Aspirinis avoided in children under 16 years (except
in specific indications, such as Kawasaki syndrome) due to an
association with Reye’s syndrome, a rare but often fatal illness
of unknown aetiology consisting of hepatic necrosis and
encephalopathy, often in the aftermath of a viral illness.
Tetracyclines are deposited in growing bone and teeth, caus-
ing staining and occasionally dental hypoplasia, and should
not be given to children. Fluoroquinolone antibacterial
drugs may damage growing cartilage. Dystonias with meto-
clopramide occur more frequently in children and young
adults than in older adults. Valproate hepatotoxicity is
increased in young children with learning difficulties receiv-
ing multiple anticonvulsants. Some adverse effects cause
RESEARCH 55
lifelong effects as a result of toxicity occurring at a sensitive
point in development (a ‘critical window’) during fetal
or neonatal life (‘programming’) as with thalidomide/
phocomelia or hypothyroid drugs/congenital hypothy-
roidism
RESEARCH
Research in paediatric clinical pharmacology is limited. Not
only is there concern about the potential for adverse effects
of new drugs on those who are growing and developing
mentally, but there are also considerable ethical problems
encountered in research involving individuals who are too
young to give informed consent. New drugs are often given to
children for the first time only when no alternative is available
or when unacceptable side effects have been encountered in a
particular individual with established drugs. Pharmaceutical
companies seldom seek to license their products for use in
children. When drugs are prescribed to children that are
not licensed for use in this age group, it is important to
make careful records of both efficacy and possible adverse
effects. Prescribers take sole responsibility for prescribing
unlicensed preparations (e.g. formulated to appeal to chil-
dren) or for prescribing licensed preparations outside the
licensed age range. Parents should be informed and their con-
sent obtained.
FURTHER READING
Baber N, Pritchard D. Dose estimation in children. British Journal of
Clinical Pharmacology2003; 56: 489–93.
British National Formulary for Children2007. www.bnfc.org
Kearns GL, Abdel-Rahmen SM. Developmental pharmacology – drug
disposition, action and therapy in infants and children. New
England Journal of Medicine2003; 349: 1157–67.
Paediatric Special Issue. British Journal of Clinical Pharmacology2005; 59 (6).
Paediatric formulary, 7th edn. London: Guy’s, St Thomas’, King’s
College and Lewisham Hospitals, revised 2005.
Case history
A 14-year-old boy with a history of exercise-induced
asthma, for which he uses salbutamol as necessary (on aver-
age two puffs twice daily and before exercise) is seen by his
GP because of malaise and nocturnal cough. On examina-
tion, he has a mild fever (38°C), bilateral swollen cervical
lymph nodes and bilateral wheeze. Ampicillin is prescribed
for a respiratory tract infection. The next day the boy
develops a widespread maculopapular rash.
Question 1
What is the cause of the rash?
Question 2
What is the likely cause of the nocturnal cough and how
may this be treated?
Answer 1
Ampicillin rash in infectious mononucleosis (glandular fever).
Answer 2
Poorly controlled asthma. Regular inhaled glucocorticos-
teroid or cromoglicate.
Introduction 56
Pharmacokinetic changes 56
Pharmacodynamic changes 57
Compliance in the elderly 58
Effect of drugs on some major organ systems in
the elderly 58
Practical aspects of prescribing for the elderly 60
Research 60
CHAPTER 11
DRUGS IN THE ELDERLY
INTRODUCTION
The proportion of elderly people in the population is increas-
ing steadily in economically developed countries. The elderly
are subject to a variety of complaints, many of which are
chronic and incapacitating, and so they receive a great deal of
drug treatment. There is a growing evidence base for the use of
drugs in elderly patients, with important implications for pre-
scribing of many important classes of drugs, including statins,
β-adrenoceptor antagonists, thrombolytics, ACE inhibitors,
angiotensin receptor blockers, vitamin D and bisphosphonates
(see reviews by Mangoni and Jackson, 2006). Adverse drug
reactions and drug interactions become more common with
increasing age. In one study, 11.8% of patients aged 41–50
years experienced adverse reactions to drugs, but this
increased to 25% in patients over 80 years of age. There are
several reasons for this.
1. Elderly people take more drugs. In one survey in general
practice, 87% of patients over 75 years of age were on
regular drug therapy, with 34% taking three to four
different drugs daily. The most commonly prescribed
drugs were diuretics (34% of patients), analgesics (27%),
tranquillizers and antidepressants (24%), hypnotics (22%)
and digoxin (20%). All of these are associated with a high
incidence of important adverse effects.
2. Drug elimination becomes less efficient with increasing
age, leading to drug accumulation during chronic dosing.
3. Homeostatic mechanisms become less effective with
advancing age, so individuals are less able to compensate
for adverse effects, such as unsteadiness or postural
hypotension.
4. The central nervous system becomes more sensitive to the
actions of sedative drugs.
5. Increasing age produces changes in the immune response
that can cause an increased liability to allergic reactions.
6. Impaired cognition combined with relatively complex
dose regimens may lead to inadvertent overdose.
PHARMACOKINETIC CHANGES
ABSORPTION
Absorption of carbohydrates and of several nutrients, includ-
ing iron, calcium and thiamine, is reduced in elderly people.
Lipid-soluble drugs are absorbed by simple diffusion down
the concentration gradient (Chapter 3), and this is not
impaired by age. Intestinal blood flow is reduced by up to 50%
in the elderly. However, age per se does not affect drug
absorption to a large extent (Figure 11.1).
Drug
Absorption
Metabolism
Excretion
Renal blood flow
GFR
Concentration of
fat-soluble drugs
Concentration of
water-soluble drugs
Weight
Lean body mass
Fat
Gastric motility
Intestinal blood flow
Hepatic blood flow
Figure 11.1:Pharmacokinetic changes with age.
PHARMACODYNAMIC CHANGES 57
DISTRIBUTION
Ageing is associated with loss of lean body mass, and with an
increased ratio of fat to muscle and body water. This enlarges
the volume of distribution of fat-soluble drugs, such as
diazepam and lidocaine, whereas the distribution of polar
drugs such as digoxinis reduced compared to younger adults.
Changes in plasma proteins also occur with ageing, especially
if associated with chronic disease and malnutrition, with a fall
in albumin and a rise in gamma-globulin concentrations.
HEPATIC METABOLISM
There is a decrease in the hepatic clearance of some but not all
drugs with advancing age. Aprolonged plasma half-life (Figure
11.2), can be the result either of reduced clearance or of increased
apparent volume of distribution. Ageing reduces metabolism of
some drugs (e.g. benzodiazepines) as evidenced by reduced
hepatic clearance. The reduced clearance of benzodiazepines
has important clinical consequences, as does the long half-life of
several active metabolites (Chapter 18). Slow accumulation may
lead to adverse effects whose onset may occur days or weeks
after initiating therapy. Consequently, confusion or memory
impairment may be falsely attributed to ageing rather than to
adverse drug effects.
RENAL EXCRETION
The most important cause of drug accumulation in the elderly
is declining renal function. Many healthy elderly individuals
have a glomerular filtration rate (GFR) 50mL/min. Although
glomerular filtration rate declines with age, this is not necessar-
ily reflected by serum creatinine, which can remain within the
range defined as ‘normal’ for a younger adult population
despite a marked decline in renal function. This is related to the
lower endogenous production of creatinine in the elderly sec-
ondary to their reduced muscle mass. Under-recognition of
renal impairment in the elderly is lessened by the routine
reporting by many laboratories of an estimated GFR (eGFR)
based on age, sex and serum creatinine concentration and
reported in units normalized to 1.73m
2
body surface area
(mL/min/1.73m
2
). When estimating doses of nephrotoxic
drugs, it is important to remember that the drug elimination
depends on the absolute GFR (in mL/min) rather than that nor-
malized to an ideal body surface area (in mL/min/1.73m
2
),
and to estimate this if necessary using a nomogram (see
Chapter 7) that incorporates height and weight, as well as age,
sex and creatinine.
Examples of drugs which may require reduced dosage in
the elderly secondary to reduced renal excretion and/or
hepatic clearance are listed in Table 11.1.
The principal age-related changes in pharmacokinetics are
summarized in Figure 11.1.Key points
120
100
80
60
40
20
01020304050
Age (years)
60 70 80 100
Diazepamt
1/2
(h)
Figure 11.2:Relationship between diazepam half-life and age in
33 normal individuals. Non-smokers,
°
; smokers,
. (Redrawn
with permission from Klotz U et al. Journal of Clinical
Investigation1975; 55: 347.)
Key points
Pharmacokinetic changes in the elderly include:
Absorption of iron, calcium and thiamine is reduced.
There is an increased volume of distribution of fat-
soluble drugs (e.g. diazepam).
There is a decreased volume of distribution of polar
drugs (e.g. digoxin).
There is reduced hepatic clearance of long half-life
benzodiazepines.
Declining renal function is the most important cause of
drug accumulation.
PHARMACODYNAMIC CHANGES
Evidence that the elderly are intrinsically more sensitive to
drugs than the young is scarce. However, the sensitivity of the
elderly to benzodiazepines as measured by psychometric tests
is increased, and their effects last longer than in the young. It is
common clinical experience that benzodiazepines given to the
elderly at hypnotic doses used for the young can produce pro-
longed daytime confusion even after single doses. The inci-
dence of confusion associated with cimetidine is increased in
the elderly. Other drugs may expose physiological defects that
are a normal concomitant of ageing. Postural hypotension can
occur in healthy elderly people, and the incidence of postural
hypotension from drugs such as phenothiazines, β-adrenoceptor
Table 11.1: Examples of drugs requiring dose adjustment in the elderly
Aminoglycosides (e.g. gentamicin)
Atenolol
Cimetidine
Diazepam
Digoxin
Non-steroidal anti-inflammatory drugs
Oral hypoglycaemic agents
Warfarin
COMPLIANCE IN THE E LDERLY
Incomplete compliance is extremely common in elderly people.
This is commonly due to a failure of memory or to not under-
standing how the drug should be taken. In addition, many
patients store previously prescribed drugs in the medicine
cupboard which they take from time to time. It is therefore
essential that the drug regimen is kept as simple as possible
and explained carefully. There is scope for improved methods
of packaging to reduce over- or under-dosage. Multiple drug
regimens are confusing and increase the risk of adverse inter-
actions (see Chapter 13).
EFFECT OF DRUGS ON SOME MAJOR
ORGAN SYSTEMS IN THE ELDERLY
CENTRAL N ERVOUS SYSTEM
Cerebral function in old people is easily disturbed, resulting
in disorientation and confusion. Drugs are one of the factors
that contribute to this state; sedatives and hypnotics can easily
precipitate a loss of awareness and clouding of consciousness.
NIGHT SEDATION
The elderly do not sleep as well as the young. They sleep for a
shorter time, their sleep is more likely to be broken and they are
more easily aroused. This is quite normal, and old people should
not have the expectations of the young as far as sleep is con-
cerned. Before hypnotics are commenced, other possible factors
should be considered and treated if possible. These include:
1. pain, which may be due to such causes as arthritis;
2. constipation – the discomfort of a loaded rectum;
3. urinary frequency;
4. depression;
5. anxiety;
58 DRUGS IN THE ELDERLY
6. left ventricular failure;
7. dementia;
8. nocturnal xanthine alkaloids, e.g. caffeine in tea,
theophylline.
A little more exercise may help, and ‘catnapping’ in the day
reduced to a minimum and regularized (as in Mediterranen
cultures).
The prescription of hypnotics (see Chapter 18) should be
minimized and restricted to short-term use.
ANTIDEPRESSANTS
Although depression is common in old age and may indeed
need drug treatment, this is not without risk. Tricyclic anti-
depressants (see Chapter 20) can cause constipation, urinary
retention and glaucoma (due to their muscarinic blocking
action which is less marked in the case of lofepramine than
other drugs of this class), and also drowsiness, confusion, pos-
tural hypotension and cardiac dysrhythmias. Tricyclic antide-
pressants can produce worthwhile remissions of depression
but should be started at very low dosage.
Selective 5-hydroxytryptamine reuptake inhibitors (e.g.
fluoxetine) are as effective as the tricyclics and have a distinct
side-effect profile (see chapter 20). They are generally well
tolerated by the elderly, although hyponatraemia has been
reported more frequently than with other antidepressants.
ANTI-PARKINSONIAN DRUGS
The anticholinergic group of anti-parkinsonian drugs (e.g.
trihexyphenidyl,orphenadrine) commonly cause side effects
in the elderly. Urinary retention is common in men. Glaucoma
may be precipitated or aggravated and confusion may occur
with quite small doses. Levodopacombined with a peripheral
dopa decarboxylase inhibitor such as carbidopa can be effec-
tive, but it is particularly important to start with a small dose,
which can be increased gradually as needed. In patients with
dementia, the use of antimuscarinics, levodopaor amantidine
may produce adverse cerebral stimulation and/or hallucin-
ations, leading to decompensation of cerebral functioning,
with excitement and inability to cope.
CARDIOVASCULAR SYSTEM
HYPERTENSION
There is excellent evidence that treating hypertension in the
elderly reduces both morbidity and mortality. The agents used
(starting with a C or D drug) are described in Chapter 28. It is
important to start with a low dose and monitor carefully.
Some adverse effects (e.g. hyponatraemia from diuretics) are
much more common in the elderly, who are also much more
likely to suffer severe consequences, such as falls/fractures
from common effects like postural hypotension. Alpha-
blockers in particular should be used as little as possible.
Methyldopa might be expected to be problematic in this age
group but was in fact surprisingly well tolerated when used as
add-on therapy in a trial by the European Working Party on
Hypertension in the Elderly (EWPHE).
Key points
Pharmacodynamic changes in the elderly include:
increased sensitivity to central nervous system (CNS)
effects (e.g. benzodiazepines, cimetidine);
increased incidence of postural hypotension (e.g.
phenothiazines, beta-blockers, tricyclic antidepressants,
diuretics);
reduced clotting factor synthesis, reduced warfarin for
anticoagulation;
increased toxicity from NSAIDs;
increased incidence of allergic reactions to drugs.
antagonists, tricyclic antidepressants and diuretics is increased
in elderly patients. The QT interval is longer in the elderly,
which may predispose to drug-induced ventricular tachy-
dysrhythmias. Clotting factor synthesis by the liver is reduced
in the elderly, and old people often require lower warfarin
doses for effective anticoagulation than younger adults.
DIGOXIN
Digoxintoxicity is common in the elderly because of decreased
renal elimination and reduced apparent volume of distribution.
Confusion, nausea and vomiting, altered vision and an acute
abdominal syndrome resembling mesenteric artery obstruction
are all more common features of digoxintoxicity in the elderly
than in the young. Hypokalaemia due to decreased potassium
intake (potassium-rich foods are often expensive), faulty homeo-
static mechanisms resulting in increased renal loss and the con-
comitant use of diuretics is more common in the elderly, and is
a contributory factor in some patients. Digoxin is sometimes
prescribed when there is no indication for it (e.g. for an irregu-
lar pulse which is due to multiple ectopic beats rather than
atrial fibrillation). At other times, the indications for initiation of
treatment are correct but the situation is never reviewed. In one
series of geriatric patients on digoxin, the drug was withdrawn
in 78% of cases without detrimental effects.
DIURETICS
Diuretics are more likely to cause adverse effects (e.g. postural
hypotension, glucose intolerance and electrolyte disturbances)
in elderly patients. Too vigorous a diuresis may result in urin-
ary retention in an old man with an enlarged prostate, and
necessitate bladder catheterization with its attendant risks.
Brisk diuresis in patients with mental impairment or reduced
mobility can result in incontinence. For many patients, a thia-
zide diuretic, such as bendroflumethiazide, is adequate. Loop
diuretics, such as furosemide, should be used in acute heart
failure or in the lowest effective dose for maintenance treatment
of chronic heart failure. Clinically important hypokalaemia is
uncommon with low doses of diuretics, but plasma potassium
should be checked after starting treatment. If clinically important
hypokalaemia develops, a thiazide plus potassium-retaining
diuretic (amilorideor triamterene) can be considered, but there
is a risk of hyperkalaemia due to renal impairment, especially
if an ACE inhibitor and/or angiotensin receptor antagonist
and aldosterone antagonist are given together with the diuretic
for hypertension or heart failure. Thiazide-induced gout and
glucose intolerance are important side effects.
ISCHAEMIC HEART DISEASE
This is covered in Chapter 29.
ANGIOTENSIN CONVERTING ENZYME INHIBITORS
(ACEI) AND ANGIOTENSIN RECEPTOR BLOCKERS (ARB)
These drugs plays an important part in the treatment of chronic
heart failure, as well as hypertension (see Chapters 28 and 31),
and are effective and usually well tolerated in the elderly.
However, hypotension, hyperkalaemia and renal failure are
more common in this age group. The possibility of atheroma-
tous renal artery stenosis should be borne in mind and serum
creatinine levels checked before and after starting treatment.
Potassium-retaining diuretics should be co-administered only
with extreme caution, because of the reduced GFR and plasma
potassium levels monitored. Despite differences in their phar-
macology, ACEI and ARB appear similar in efficacy, but ARB
do not cause the dry cough that is common with ACEI. The
EFFECT OF DRUGS ON SOME MAJOR O RGAN SYSTEMS I N THE ELDERLY 59
question of whether co-administration of ACEI with ARB has
much to add remains controversial; in elderly patients with
reduced GFR, the safety of such combined therapy is an impor-
tant consideration.
ORAL HYPOGLYCAEMIC AGENTS
Diabetes is common in the elderly and many patients are
treated with oral hypoglycaemic drugs (see Chapter 37). It is
best for elderly patients to be managed with diet if at all possi-
ble. In obese elderly diabetics who remain symptomatic on
diet, metformin should be considered, but coexisting renal,
heart or lung disease may preclude its use. Short-acting
sulphonylureas (e.g. gliclazide) are preferred to longer-acting
drugs because of the risk of hypoglycaemia: chlorpropamide
(half-life 36 hours) can cause prolonged hypoglycaemia and is
specifically contraindicated in this age group, glibenclamide
should also be avoided. Insulinmay be needed, but impaired
visual and cognitive skills must be considered on an individual
basis, and the potential need for dose reduction with advanc-
ing age and progressive renal impairment taken into account.
ANTIBIOTICS
The decline in renal function must be borne in mind when an
antibiotic that is renally excreted is prescribed, especially if it is
nephrotoxic (e.g. an aminoglycoside or tetracycline). Appendix 3
of the British National Formulary is an invaluable practical guide.
Over-prescription of antibiotics is a threat to all age groups,
but especially in the elderly. Broad-spectrum drugs including
cephalosporins and other beta-lactams, and fluoroquinones are
common precursors of Clostridium difficile infection which has a
high mortality rate in the elderly. Amoxicillinis the most com-
mon cause of drug rash in the elderly. Flucloxacillin induced
cholestatic jaundice and hepatitis is more common in the elderly.
Case history
An 80-year-old retired publican was referred with ‘congest-
ive cardiac failure and acute retention of urine’. His wife
said his symptoms of ankle swelling and breathlessness had
gradually increased over a period of six months despite the
GP doubling the water tablet (co-amilozide) which he was
taking for high blood pressure. Over the previous week he
had become mildly confused and restless at night, for
which the GP had prescribed chlorpromazine. His other
medication included ketoprofen for osteoarthritis and fre-
quent magnesium trisilicate mixture for indigestion. He
had been getting up nearly ten times most nights for a year
to pass urine. During the day, he frequently passed small
amounts of urine. Over the previous 24 hours, he had been
unable to pass urine. His wife thought most of his problems
were due to the fact that he drank two pints of beer each
day since his retirement seven years previously.
On physical examination he was clinically anaemic, but
not cyanosed. Findings were consistent with congestive
cardiac failure. His bladder was palpable up to his umbili-
cus. Rectal examination revealed an enlarged, symmetrical
prostate and black tarry faeces. Fundoscopy revealed a
grade II hypertensive retinopathy.
60 DRUGS IN THE ELDERLY
4.Use the fewest possible number of drugs the patient needs.
5. Consider the potential for drug interactions and
co-morbidity on drug response.
6. Drugs should seldom be used to treat symptoms without
first discovering the cause of the symptoms (i.e. first
diagnosis, then treatment).
7. Drugs should not be withheld because of old age, but it
should be remembered that there is no cure for old age
either.
8. A drug should not be continued if it is no longer necessary.
9. Do not use a drug if the symptoms it causes are worse
than those it is intended to relieve.
10. It is seldom sensible to treat the side effects of one drug
by prescribing another.
In the elderly, it is often important to pay attention to mat-
ters such as the formulation of the drug to be used – many old
people tolerate elixirs and liquid medicines better than tablets
or capsules. Supervision of drug taking may be necessary, as
an elderly person with a serious physical or mental disability
cannot be expected to comply with any but the simplest drug
regimen. Containers require especially clear labelling, and
should be easy to open – child-proof containers are often also
grandparent-proof!
RESEARCH
Despite their disproportionate consumption of medicines, the
elderly are often under-represented in clinical trials. This may
result in the data being extrapolated to an elderly population
inappropriately, or the exclusion of elderly patients from new
treatments from which they might benefit. It is essential that,
both during a drug’s development and after it has been licensed,
subgroup analysis of elderly populations is carefully examined
both for efficacy and for predisposition to adverse effects.
Initial laboratory results revealed that the patient had
acute on chronic renal failure, dangerously high potassium
levels (7.6mmol/L) and anaemia (Hb 7.4 g/dL). Emergency
treatment included calcium chloride, dextrose and insulin,
urinary catheterization, furosemide and haemodialysis.
Gastroscopy revealed a bleeding gastric ulcer. The patient
was discharged two weeks later, when he was symptomat-
ically well. His discharge medication consisted of regular
doxazosin and ranitidine, and paracetamol as required.
Question
Describe how each of this patient’s drugs prescribed before
admission may have contributed to his clinical condition.
Answer
Co-amilozide – hyperkalaemia: amiloride, exacerbation of
prostatic symptoms: thiazide
Chlorpromazine – urinary retention
Ketoprofen – gastric ulcer, antagonism of thiazide diuretic,
salt retention, possibly interstitial nephritis
Magnesium trisilicate mixture – additional sodium load
(6mmol Na
/10mL).
Comment
Iatrogenic disease due to multiple drug therapy is common
in the elderly. The use of amiloride in renal impairment
leads to hyperkalaemia. This patient’s confusion and rest-
lessness were most probably related to his renal failure.
Chlorpromazine may mask some of the symptoms/signs and
delay treatment of the reversible organic disease. The anal-
gesic of choice in osteoarthritis is paracetamol, due to its
much better tolerance than NSAID. The sodium content of
some antacids can adversely affect cardiac and renal failure.
NON-STEROIDAL ANTI-INFL AMMATORY DRUGS
The elderly are particularly susceptible to non-steroidal anti-
inflammatory drug (NSAID)-induced peptic ulceration, gastro-
intestinal irritation and fluid retention. An NSAID is frequently
prescribed inappropriately for osteoarthritis before physical
and functional interventions and oral paracetamolhave been
adequately utilized. If an NSAID is required as adjunctive
therapy, the lowest effective dose should be used. Ibuprofen
is probably the NSAID of choice in terms of minimizing gas-
tro-intestinal side effects. Aproton pump inhibitor should be
considered as prophylaxis against upper gastro-intestinal
complications in those most at risk.
PRACTICAL ASPECTS OF PRESCRIBING
FOR THE ELDERLY
Improper prescription of drugs is a common cause of morbid-
ity in elderly people. Common-sense rules for prescribing do
not apply only to the elderly, but are especially important in
this vulnerable group.
1. Take a full drug history (see Chapter 1), which should
include any adverse reactions and use of over-the-counter
drugs.
2. Know the pharmacological action of the drug employed.
3. Use the lowest effective dose.
Case history
A previously mentally alert and well-orientated 90-year-old
woman became acutely confused two nights after hospital
admission for bronchial asthma which, on the basis of peak
flow and blood gases, had responded well to inhaled salbu-
tamol and oral prednisolone. Her other medication was
cimetidine (for dyspepsia), digoxin (for an isolated episode
of atrial fibrillation two years earlier) and nitrazepam (for
night sedation).
Question
Which drugs may be related to the acute confusion?
Answer
Prednisolone, cimetidine, digoxin and nitrazepam.
Comment
If an H
2
-antagonist is necessary, ranitidine is preferred in the
elderly. It is likely that the patient no longer requires digoxin
(which accumulates in the elderly). Benzodiazepines should
not be used for sedation in elderly (or young) asthmatics.
They may also accumulate in the elderly. The elderly tend to
be more sensitive to adverse drug effects on the central ner-
vous system (CNS).
RESEARCH 61
FURTHER READING
Dhesi JK, Allain TJ, Mangoni AA, Jackson SHD. The implications of a
growing evidence base for drug use in elderly patients. Part 4.
Vitamin D and bisphosphonates for fractures and osteoporosis.
British Journal of Clinical Pharmacology2006; 61: 520–8.
Hanratty CG, McGlinchey P, Johnston GD, Passmore AP. Differential
pharmacokinetics of digoxin in elderly patients. Drugs and Aging
2000;17: 353–62.
Mangoni AA, Jackson SHD. The implications of a growing evidence
base for drug use in elderly patients. Part 1. Statins for primary
and secondary cardiovascular prevention. British Journal of Clinical
Pharmacology2006; 61: 494–501.
Mangoni AA, Jackson SHD. The implications of a growing evidence
base for drug use in elderly patients. Part 2. ACE inhibitors and
angiotensin receptor blockers in heart failure and high cardiovas-
cular risk patients. British Journal of Clinical Pharmacology2006; 61:
502–12.
Mangoni AA, Jackson SHD. The implications of a growing evidence
base for drug use in elderly patients. Part 3. β-adrenoceptor block-
ers in heart failure and thrombolytics in acute myocardial infarc-
tion.British Journal of Clinical Pharmacology 2006; 61: 513–20.
Sproule BA, Hardy BG, Shulman KI. Differential pharmacokinetics in
elderly patients. Drugs and Aging2000; 16: 165–77.
Introduction 62
Identification of the drug at fault 63
Adverse drug reaction monitoring/surveillance
(pharmacovigilance) 63
Allergic adverse drug reactions 66
Prevention of allergic drug reactions 67
Examples of allergic and other adverse
drug reactions 68
CHAPTER 12
ADVERSE DRUG REACTIONS
INTRODUCTION
Adverse drug reactions are unwanted effects caused by nor-
mal therapeutic doses. Drugs are great mimics of disease,
and adverse drug reactions present with diverse clinical
signs and symptoms. The classification proposed by Rawlins
and Thompson (1977) divides reactions into type Aand type B
(Table 12.1).
Type Areactions, which constitute approximately 80% of
adverse drug reactions, are usually a consequence of the drug’s
primary pharmacological effect (e.g. bleeding from warfarin)
or a low therapeutic index (e.g. nausea from digoxin), and they
are therefore predictable. They are dose-related and usually
mild, although they may be serious or even fatal (e.g. intracra-
nial bleeding from warfarin). Such reactions are usually due to
inappropriate dosage, especially when drug elimination is
impaired. The term ‘side effects’ is often applied to minor type
Areactions.
Type B (‘idiosyncratic’) reactions are not predictable from
the drug’s main pharmacological action, are not dose-related
and are severe, with a considerable mortality. The underlying
pathophysiology of type B reactions is poorly if at all under-
stood, and often has a genetic or immunological basis. Type B
reactions occur infrequently (1:1000–1:10000 treated subjects
being typical).
Adverse drug reactions due to specific drug–drug inter-
actions are considered in Chapter 13. Three further minor cat-
egories of adverse drug reaction have been proposed:
1. type C – continuous reactions due to long-term drug use
(e.g. neuroleptic-related tardive dyskinesia or analgesic
nephropathy);
2. type D – delayed reactions (e.g. alkylating agents
leading to carcinogenesis, or retinoid-associated
teratogenesis);
3. type E end-of-use reactions, such as adrenocortical
insufficiency following withdrawal of glucocorticosteroids,
or withdrawal syndromes following discontinuation of
treatment with benzodiazepines or β-adrenoceptor
antagonists.
In the UK there are between 30000 and 40000 medicinal
products available directly or on prescription. Surveys sug-
gest that approximately 80% of adults take some kind of medi-
cation during any two-week period. Exposure to drugs in the
population is thus substantial, and the incidence of adverse
reactions must be viewed in this context. Type Areactions are
reported to be responsible for 2–3% of consultations in general
practice. In a recent prospective analysis of 18820 hospital
admissions by Pirmohamed et al. (2004), 1225 were related to
an adverse drug reaction (prevalence 6.8%), with the adverse
drug reaction leading directly to admission in 80% of cases.
Median bed stay was eight days, accounting for 4% of hospi-
tal bed capacity. The projected annual cost to the NHS is £466
million. Overall fatality was 0.15%. Most reactions were either
definitely or probably avoidable. Adverse drug reactions are
most frequent and severe in the elderly, in neonates, women,
patients with hepatic or renal impairment, and individuals
with a history of previous adverse drug reactions. Such reac-
tions often occur early in therapy (during the first one to ten
days). Drugs most commonly implicated include low-dose
aspirin(antiplatelet agents), diuretics, warfarin and NSAIDs.
A systematic review by Howard et al. (2006) of preventable
adverse drug reactions which caused hospitalization, impli-
cated the same major drug classes.
Table 12.1: Some examples of type A and type B reactions.
Drug Type A Type B
Chlorpromazine Sedation Cholestatic jaundice
Naproxen Gastro-intestinal Agranulocytosis
haemorrhage
Phenytoin Ataxia Hepatitis,
lymphadenopathy
Thiazides Hypokalaemia Thrombocytopenia
Quinine Tinnitus Thrombocytopenia
Warfarin Bleeding Breast necrosis
ADVERSEDRUG R EACTION MONITORING/SURVEILLANCE(P HARMACOVIGILANCE) 63
Factors involved in the aetiology of adverse drug reactions
can be classified as shown in Table 12.2.
IDENTIFICATION OF THE DRUG AT FAULT
It is often difficult to decide whether a clinical event is drug
related, and even when this is probable, it may be difficult to
determine which drug is responsible, as patients are often tak-
ing multiple drugs. One or more of several possible approaches
may be appropriate.
1. A careful drug history is essential. The following
considerations should be made to assess causality of the
effect to the drug: did the clinical event and the time-
course of its development fit with the duration of suspected
drug treatment and known adverse drug effects? Did the
adverse effect reverse upon drug withdrawal and, upon
rechallenge with the drug, reappear? Were other possible
causes reasonably excluded? Apatient’s drug history may
not always be conclusive because, although allergy to a
drug implies previous exposure, the antigen may have
occurred in foods (e.g. antibiotics are often fed to livestock
and drug residues remain in the flesh), in drug mixtures
or in some casual manner.
2. Provocation testing. This involves giving a very small
amount of the suspected drug and seeing whether a
reaction ensues, e.g. skin testing, where a drug is applied
as a patch, or is pricked or scratched into the skin or
injected intradermally. Unfortunately, prick and scratch
testing is less useful for assessing the systemic reaction to
drugs than it is for the more usual atopic antigens (e.g.
pollens), and both false-positive and false-negative results
can occur. Patch testing is safe, and is useful for the
diagnosis of contact sensitivity, but does not reflect
systemic reactions and may itself cause allergy. Provocation
tests should only be undertaken under expert guidance,
after obtaining informed consent, and with resuscitation
facilities available.
3. Serological testing and lymphocytes testing. Serological
testing is rarely helpful, circulating antibodies to the drug
do not mean that they are necessarily the cause of the
symptoms. The demonstration of transformation occurring
when the patient’s lymphocytes are exposed to a drug
ex vivo suggests that the patient’s T-lymphocytes are
sensitized to the drug. In this type of reaction, the hapten
itself will often provoke lymphocyte transformation, as
well as the conjugate.
4. The best approach in patients on multiple drug therapy is
to stop all potentially causal drugs and reintroduce them
one by one until the drug at fault is discovered. This should
only be done if the reaction is not serious, or if the drug
is essential and no chemically unrelated alternative is
available. All drug allergies should be recorded in the case
notes and the patient informed of the risks involved in
taking the drug again.
Key points
Type A reaction – an extension of the pharmacology of
the drug, dose related, and accounts for most adverse
reactions (e.g. β-adrenoreceptor antagonist-induced
bradycardia or AV block).
Type B reaction – idiosyncratic reaction to the drug, not
dose related, rare but severe (e.g. chloramphenicol-
induced aplastic anaemia).
Other types of drug reaction (much rarer):
type C reaction – continuous reactions due to long-
term use: analgesic nephropathy;
type D reaction – delayed reactions of
carcinogenesis or teratogenesis;
type E reaction – drug withdrawal reactions (e.g.
benzodiazepines).
ADVERSE DRUG REACTION MONITORING/
SURVEILLANCE (PHARMACOVIGILAN CE)
The evaluation of drug safety is complex, and there are many
methods for monitoring adverse drug reactions. Each of these
has its own advantages and shortcomings, and no single
Table 12.2:Factors involved in adverse drug reactions.
Intrinsic Extrinsic
Patient factors
Age – neonatal, infant and elderly Environment – sun
Sex – hormonal environment Xenobiotics (e.g. drugs,
Genetic abnormalities (e.g. herbicides)
enzyme or receptor Malnutrition
polymorphisms)
Previous adverse drug reactions,
allergy, atopy
Presence of organ dysfunction –
disease
Personality and habits –
adherence (compliance),
alcoholic, drug addict,
nicotine
Prescriber factors
Incorrect drug or drug combination
Incorrect route of administration
Incorrect dose
Incorrect duration of therapy
Drug factors
Drug–drug interactions (see
Chapter 13)
Pharmaceutical – batch problems,
shelf-life, incorrect dispensing
64 ADVERSEDRUG REACTIONS
system can offer the 100% accuracy that current public opinion
expects. The ideal method would identify adverse drug reactions
with a high degree of sensitivity and specificity and respond
rapidly. It would detect rare but severe adverse drug reactions,
but would not be overwhelmed by common ones, the incidence
of which it would quantify together with predisposing factors.
Continued surveillance is mandatory after a new drug has
been marketed, as it is inevitable that the preliminary testing
of medicines in humans during drug development, although
excluding many ill effects, cannot identify uncommon adverse
effects. A variety of early detection systems have been intro-
duced to identify adverse drug reactions as swiftly as possible.
PHASE I/II/III TRIALS
Early (phase I/II) trials (Chapter 15) are important for assess-
ing the tolerability and dose–response relationship of new
therapeutic agents. However, these studies are, by design,
very insensitive at detecting adverse reactions because they
are performed on relatively few subjects (perhaps 200–300).
This is illustrated by the failure to detect the serious toxicity
of several drugs (e.g. benoxaprofen, cerivastatin, felbamate,
dexfenfluramineand fenfluramine, rofecoxib, temofloxacin,
troglitazone) before marketing. However, phase III clinical
trials can establish the incidence of common adverse reactions
and relate this to therapeutic benefit. Analysis of the reasons
given for dropping out of phase III trials is particularly valu-
able in establishing whether common events, such as
headache, constipation, lethargy or male sexual dysfunction
are truly drug related. The Medical Research Council Mild
Hypertension Study unexpectedly identified impotence as
more commonly associated with thiazide diuretics than with
placebo or β-adrenoceptor antagonist therapy. Table 12.3
illustrates how difficult it is to detect adverse drug reactions
with 95% confidence, even when there is no background inci-
dence and the diagnostic accuracy is 100%. This ‘easiest-case’
scenario approximates to the actual situation with thalido-
mide teratogenicity: spontaneous phocomelia is almost
unknown, and the condition is almost unmistakable. It is
sobering to consider that an estimated 10000 malformed
babies were born world-wide before thalidomide was with-
drawn. Regulatory authorities may act after three or more
documented events.
The problem of adverse drug reaction recognition is much
greater if the reaction resembles spontaneous disease in the
population, such that physicians are unlikely to attribute the
reaction to drug exposure: the numbers of patients that must
then be exposed to enable such reactions to be detected are
greater than those quoted in Table 12.3, probably by several
orders of magnitude.
YELLOW CARD SCHEME AND POST-MARKETING
(PHASE IV) SURVEILLANCE
Untoward effects that have not been detected in clinical trials
become apparent when the drug is used on a wider scale. Case
reports, which may stimulate further reports, remain the most
sensitive means of detecting rare but serious and unusual
adverse effects. In the UK, a Register of Adverse Reactions
was started in 1964. Currently, the Medicines and Healthcare
products Regulatory Agency (MHRA) operates a system of
spontaneous reporting on prepaid yellow postcards. Doctors,
dentists, pharmacists, nurse practitioners and (most recently)
patients are encouraged to report adverse events whether actu-
allyor potentially causally drug-related. Analogous schemes are
employed in other countries. The yellow card scheme consists
of three stages:
1. data collection;
2. analysis;
3. feedback.
Such surveillance methods are useful, but under-reporting is a
major limitation. Probably fewer than 10% of appropriate
adverse reactions are reported. This may be due partly to con-
fusion about what events to report, partly to difficulty in rec-
ognizing the possible relationship of a drug to an adverse
event – especially when the patient has been taking several
drugs, and partly to ignorance or laziness on the part of poten-
tial reporters. Afurther problem is that, as explained above, if
a drug increases the incidence of a common disorder (e.g.
ischaemic heart disease), the change in incidence must be very
large to be detectable. This is compounded when there is a
delay between starting the drug and occurrence of the event
(e.g. cardiovascular thrombotic events including myocardial
infarction following initiation of rofecoxib therapy). Doctors
are inefficient at detecting such adverse reactions to drugs,
and those reactions that are reported are in general the obvi-
ous or previously described and well-known ones. Initiatives
are in progress to attempt to improve this situation by involve-
ment of trained clinical pharmacologists and pharmacists in
and outside hospitals.
The Committee on Safety of Medicines (CSM), now part of
MHRA, introduced a system of high vigilance for newly mar-
keted drugs. For its first two years on the general market, any
newly marketed drug has a black triangle on its data sheet and
against its entry in the British National Formulary. This con-
veys to prescribers that any unexpected event should be
reported by the yellow card system. The pharmaceutical com-
pany is also responsible for obtaining accurate reports on all
patients treated up to an agreed number. This scheme was
successful in the case of benoxaprofen, an anti-inflammatory
Table 12.3:Numbers of subjects that would need to be exposed in order to
detect adverse drug reactions
Expected frequency Approximate number of patients
of the adverse effect required to be exposed
For one event For three events
1 in 100 300 650
1 in 1000 3000 6500
1 in 10000 30000 65000
ADVERSEDRUG R EACTION MONITORING/SURVEILLANCE(P HARMACOVIGILANCE) 65
analgesic. Following its release, there were spontaneous reports
to the CSM of photosensitivity and onycholysis. Further reports
appeared in the elderly, in whom its half-life is prolonged, of
cholestatic jaundice and hepatorenal failure, which was fatal
in eight cases. Benoxaprofen was subsequently taken off the
market when 3500 adverse drug reaction reports were received
with 61 fatalities. The yellow card/black triangle scheme was
also instrumental in the early identification of urticaria and
cough as adverse effects of angiotensin-converting enzyme
inhibitors. Although potentially the population under study
by this system consists of all the patients using a drug, in fact
under-reporting yields a population that is not uniformly
sampled. Such data can be unrepresentative and difficult to
work with statistically, contributing to the paucity of accurate
incidence data for adverse drug reactions.
Systems such as the yellow card scheme (e.g. FDAMedWatch
in the USA) are relatively inexpensive and easy to manage,
and facilitate ongoing monitoring of all drugs, all consumers
and all types of adverse reaction. Reports from the drug regu-
latory bodies of 22 countries are collated by the World Health
Organization (WHO) Unit of Drug Evaluation and Monitoring
in Geneva. Rapid access to reports from other countries should
be of great value in detecting rare adverse reactions, although
the same reservations apply to this register as apply to
national systems. In addition, this database could reveal geo-
graphical differences in the pattern of untoward drug effects.
CASE–CONTROL STUDIES
Avery large number of patients have to be monitored to detect
a rare type B adverse effect. An alternative approach is to iden-
tify patients with a disorder which it is postulated could be
caused by an adverse reaction to a drug, and to compare the fre-
quency of exposure to possible aetiological agents with a con-
trol group. Aprior suspicion (hypothesis) must exist to prompt
the setting up of such a study – examples are the possible con-
nection between irradiation or environmental pollution and
certain malignancies, especially where they are observed in
clusters. Artefacts can occur as a result of unrecognized bias
from faulty selection of patients and controls, and the approach
remains controversial among epidemiologists, public health
physicians and statisticians. Despite this, there is really no prac-
ticable alternative for investigating a biologically plausible
hypothesis relating to a disease which is so uncommon that it
is unlikely to be represented even in large trial or cohort popu-
lations. This methodology has had notable successes: the associ-
ation of stilboestrolwith vaginal adenocarcinoma, gatifloxacin
with hypo- and hyperglycaemia, and salmeterol or fenoterol
use with increased fatality in asthmatics.
INTENSIVE MONITORING
Several hospital-based intensive monitoring programmes are
currently in progress. The Aberdeen–Dundee system abstracts
data from some 70000 hospital admissions each year, storing
these on a computer file before analysis. The Boston
Collaborative Drug Surveillance Program (BCDSP), involving
selected hospitals in several countries, is even more compre-
hensive. In the BCDSP, all patients admitted to specially desig-
nated general wards are included in the analysis. Specially
trained personnel obtain the following information from hos-
pital patients and records:
1. background information (i.e. age, weight, height, etc.);
2. medical history;
3. drug exposure;
4. side effects;
5. outcome of treatment and changes in laboratory tests
during hospital admission.
A unique feature of comprehensive drug-monitoring sys-
tems lies in their potential to follow up and investigate adverse
reactions suggested by less sophisticated detection systems, or
by isolated case reports in medical journals. Furthermore, the
frequency of side effects can be determined more cheaply than
by a specially mounted trial to investigate a simple adverse
effect. Thus, for example, the risk of developing a rash with
ampicillin was found to be around 7% both by clinical trial
and by the BCDSP, which can quantify such associations
almost automatically from data on its files. New adverse reac-
tions or drug interactions are sought by multiple correlation
analysis. Thus, when an unexpected relationship arises, such
as the 20% incidence of gastro-intestinal bleeding in severely
ill patients treated with ethacrynic acid compared to 4.3%
among similar patients treated with other diuretics, this can-
not be attributed to bias arising from awareness of the hypoth-
esis during data collection, since the data were collected
before the hypothesis was proposed. Conversely, there is a
possibility of chance associations arising from multiple com-
parisons (‘type I’ statistical error), and such associations must
be reviewed critically before accepting a causal relationship. It
is possible to identify predisposing risk factors. In the associ-
ation between ethacrynic acid and gastro-intestinal bleeding,
these were female sex, a high blood urea concentration, previ-
ous heparin administration and intravenous administration of
the drug. An important aspect of this type of approach is that
lack of clinically important associations can also be investi-
gated. Thus, no significant association between aspirin and
renal disease was found, whereas long-term aspirinconsump-
tion is associated with a decreased incidence of myocardial
infarction, an association which has been shown to be of thera-
peutic importance in randomized clinical trials (Chapter 29).
There are plans to extend intensive drug monitoring to cover
other areas of medical practice.
However, in terms of new but uncommon adverse reac-
tions, the numbers of patients undergoing intensive monitor-
ing while taking a particular drug will inevitably be too small
for the effect to be detectable. Such monitoring can therefore
only provide information about relatively common, early reac-
tions to drugs used under hospital conditions. Patients are not
in hospital long enough for detection of delayed effects, which
are among the reactions least likely to be recognized as such
even by an astute clinician.
66 ADVERSEDRUG REACTIONS
MONITORING FROM NATIONAL STATISTICS
Agreat deal of information is available from death certificates,
hospital discharge diagnoses and similar records. From these
data, it may be possible to detect a change in disease trends
and relate this to drug therapy. Perhaps the best-known example
of this is the increased death rate in young asthmatics noted in
the mid-1960s, which was associated with overuse of bron-
chodilator inhalers containing non-specific β-adrenoceptor
agonists (e.g. adrenalineand/or isoprenaline). Although rel-
atively inexpensive, the shortcomings of this method are obvi-
ous, particularly in diseases with an appreciable mortality,
since large numbers of patients must suffer before the change is
detectable. Data interpretation is particularly difficult when
hospital discharges are used as a source of information, since
discharge diagnosis is often provisional or incomplete, and
may be revised during follow up.
However, they can combine with high molecular weight enti-
ties, usually proteins, to form an antigenic hapten conjugate.
The factors that determine the development of allergy to a
drug are not fully understood. Some drugs (e.g. penicillin)
are more likely to cause allergic reactions than others, and
type I (immediate anaphylactic) reactions are more common
in patients with a history of atopy. Acorrelation between aller-
gic reactions involving immunoglobulin E (IgE) and human
leukocyte antigen (HLA) serotypes has been reported, so genetic
factors may also be important. There is some evidence that
drug allergies are more common in older people, in women
and in those with a previous history of drug reaction. However,
this may merely represent increased frequencies of drug expo-
sure in these patient groups.
TYPES OF ALLERGY
Drugs cause a variety of allergic responses (Figure 12.1) and
sometimes a single drug can be responsible for more than one
type of allergic response.
TYPE I REACTIONS
Type I reactions are due to the production of reaginic (IgE)
antibodies to an antigen (e.g. penicillins and cephalosporins).
The antigen binds to surface bound IgE on mast cells causing
degranulation and release of histamine, eicosanoids and
cytokines. It commonly occurs in response to a foreign serum
orpenicillin, but may also occur with streptomycin and some
local anaesthetics. With penicillin, it is believed that the peni-
cilloyl moiety of the penicillinmolecule is responsible for the
production of antibodies. Treatment of anaphylactic shock is
detailed in Chapter 50.
TYPE II REACTIONS
These are due to antibodies of class IgG and IgM which, on
contact with antibodies on the surface of cells, bind comple-
ment, causing cell lysis (e.g. penicillin, cephalosporins,
methyldopaor quinine) causing, for example, Coombs’ posi-
tive haemolytic anaemia.
TYPE III IMMUN E COMPLEX ARTHUS REACTIONS
Circulating immune complexes can produce several clinical
allergic states, including serum sickness and immune complex
glomerulonephritis, and a syndrome resembling systemic lupus
erythematosus. The onset of serum sickness is delayed for sev-
eral days until features develop such as fever, urticaria,
arthropathy, lymphadenopathy, proteinuria and eosinophilia.
Recovery takes a few days. Examples of causative agents
include serum, penicillin, sulfamethoxazole/trimethopri m,
streptomycin and propylthiouracil. Amiodarone lung and
hydralazine-induced systemic lupus syndrome are also pos-
sibly mediated by immune complex-related mechanisms,
although these reactions are less well understood.
TYPE IV DELAYED HYPERSENSITIVITY REACTIONS
Type IV reactions are delayed hypersensitivity reactions, the
classical example of which is contact dermatitis (e.g. to topical
Key points
Rare (and often severe) adverse drug events may not be
detected in early drug development but only defined in
the first few years post marketing (phase IV of drug
development).
Be aware of and participate in the MHRA yellow card
system for reporting suspected adverse drug reactions.
Use of any recently marketed drug, which is identified
with a black triangle on its data sheet or in the British
National Formulary, indicates the need to
be particularly
suspicious about adverse drug reactions and to report
any suspected adverse drug reaction via the yellow card
system.
Constant vigilance by physicians for drug-induced
disease, particularly for new drugs, but also for more
established agents, is needed.
FEEDBACK
There is no point in collecting vast amounts of data on
adverse reactions unless they are analysed and conclusions
reported back to prescribing doctors. In addition to articles in
the medical journals and media, the Current Problems in
Pharmacovigilance series deals with important and recently
identified adverse drug reactions. If an acute and serious
problem is recognized, doctors will usually receive notifica-
tion from the MHRA/Commission on Human Medicines,
and often from the pharmaceutical company marketing the
product.
ALLERGIC ADVERSE DRUG REACTIONS
Immune mechanisms are involved in a number of adverse
effects caused by drugs (see below and Chapter 50). The
development of allergy implies previous exposure to the drug
or to some closely related substance. Most drugs are of low
molecular weight (300–500Da) and thus are not antigenic.
PREVENTIONOF ALLERGIC DRUG REACTIONS 67
antibiotics, such as penicillin or neomycin). The mechanism
here is that the drug applied to the skin forms an antigenic
conjugate with dermal proteins, stimulating formation of sen-
sitized T-lymphocytes in the regional lymph nodes, with a
resultant rash if the drug is applied again. Drug photosensitiv-
ity is due to a photochemical combination between the drug
(e.g. amiodarone, chlorpromazine, ciprofloxacin, tetracyc-
lines) and dermal protein. Delayed sensitivity can also result
from the systemic administration of drugs.
vitamin supplements and alternative remedies) is essential.
Ahistory of atopy, although not excluding the use of
drugs, should make one wary.
2. Drugs given orally are less likely to cause severe allergic
reactions than those given by injection.
3. Desensitization (hyposensitization) should only be used
when continued use of the drug is essential. It involves
giving a very small dose of the drug and increasing the
dose at regular intervals, sometimes under cover of a
glucocorticosteroid and β
2
-adrenoceptor agonist. An
antihistamine may be added if a drug reaction occurs, and
equipment for resuscitation and therapy of anaphylactic
shock must be close at hand. It is often successful,
although the mechanism by which it is achieved is not
fully understood.
4. Prophylactic skin testing is not usually practicable, and a
negative test does not exclude the possibility of an allergic
reaction.
Key points
How to attempt to define the drug causing the adverse
drug reaction:
Attempt to define the likely causality of the effect to
the drug, thinking through the following: Did the
reaction and its time-course fit with the duration of
suspected drug treatment and known adverse drug
effects? Did the adverse effect disappear on drug
withdrawal and, if rechallenged with the drug,
reappear? W
ere other possible causes excluded?
Provocation testing with skin testing – intradermal tests
are neither very sensitive nor specific.
Test the patient’s serum for anti-drug antibodies, or test
the reaction of the patient’s lymphocytes in vitro to the
drug and/or drug metabolite if appropriate.
Consider stopping all drugs and reintroducing essential
ones sequentially.
Carefully document and highlight the adverse drug
reaction and the most likely culprit in the case notes.
Key points
Classification of immune-mediated adverse drug reactions:
Type I – urticaria or anaphylaxis due to the production
of IgE against drug bound to mast cells, leading to
massive release of mast cell mediators locally or
systemically (e.g. ampicillin skin allergy or anaphylaxis).
Type II – IgG and IgM antibodies to drug which, on
contact with antibodies on the cell surface, cause cell
lysis by complement fixation (e.g. penicillin, haemolytic
anaemia; quinidine, thrombocytopenia).
Type III – circulating immune complexes produced by
drug and antibody to drug deposit in organs, causing
drug fever, urticaria, rash, lymphadenopathy,
glomerulonephritis, often with eosinophilia (e.g.
co-trimoxazole,β
-lactams).
Type IV – delayed-type hypersensitivity due to drug
forming an antigenic conjugate with dermal proteins
and sensitized T cells reacting to drug, causing a rash
(e.g. topical antibiotics).
Central immune
apparatus
Type IV response
Type I, II and III responses
Humoral antibodies
T
Lymphocytes
Sensitized
lymphocytes
Cell
membrane
Drug or its
metabolites
Drug or its
metabolites
Drug
(large molecule)
Protein Antigen
B
Lymphocytes
Macrophages
Figure 12.1:The immune response to drugs.
PREVENTION OF ALLERGIC DRUG
REACTIONS
Although it is probably not possible to avoid all allergic drug
reactions, the following measures can decrease their incidence:
1. Taking a detailed drug history (prescription and
over-the-counter drugs, drugs of abuse, nutritional and
68 ADVERSEDRUG REACTIONS
EXAMPLES OF ALLERGIC AND OTHER
ADVERSE DRUG REACTIONS
Adverse drug reactions can be manifested in any one or mul-
tiple organ systems, and in extraordinarily diverse forms.
Specific instances are dealt with throughout this book. Some
examples to illustrate the diversity of adverse drug reactions
are given here.
RASHES
These are one of the most common manifestations of drug
reactions. A number of immune and non-immune mech-
anisms may be involved which produce many different types
of rash ranging from a mild maculopapular rash to a severe
erythema multiforme major (Stevens Johnson syndrome;
Figures 12.2 and 12.3). Commonly implicated drugs/drug
classes include beta-lactams, sulphonamides and other anti-
microbial agents; anti-seizure medications (e.g. phenytoin,
carbamazepine); NSAIDs. Some drugs may give rise to direct
tissue toxicity (e.g. DMPS, used as chelating therapy in patients
with heavy metal poisoning; Figure 12.4, see Chapter 54).
LYMPHADENOPATHY
Lymph-node enlargement can result from taking drugs (e.g.
phenytoin). The mechanism is unknown, but allergic factors
may be involved. The reaction may be confused with a lymph-
oma, and the drug history is important in patients with lym-
phadenopathy of unknown cause.
BLOOD DYSCRASIAS
Thrombocytopenia, anaemia (aplastic, iron deficiency, macro-
cytic, haemolytic) and agranulocytosis can all be caused by
drugs.
Thrombocytopenia can occur with many drugs, and in
many but not all instances the mechanism is direct suppres-
sion of the megakaryocytes rather than immune processes.
Drugs that cause thrombocytopenia include:
heparin;
gold salts;
cytotoxic agents (e.g. azathioprine/6-mercaptopurine);
quinidine;
sulphonamides;
thiazides.
Haemolytic anaemia can be caused by a number of
drugs, and sometimes immune mechanisms are responsible.
Glucose-6-phosphate dehydrogenase deficiency (Chapter 14)
Figure 12.2:Mouth ulcer as part of Stevens Johnson syndrome as
a reaction to phenytoin therapy (see Chapter 22).
Figure 12.3:Stevens Johnson syndrome following
commencement of penicillin therapy (see Chapter 43).
Figure 12.4:Mouth ulcer following DMPS treatment (see
Chapter 54).
EXAMPLES OF ALLERGIC AND OTHER ADVERSE D RUGREACTIONS 69
predisposes to non-immune haemolysis (e.g. primaquine).
Immune mechanisms include the following:
1. Combination of the drug with the red-cell membrane,
with the conjugate acting as an antigen. This has been
shown to occur with penicillin-induced haemolysis, and
may also occur with chlorpromazineand sulphonamides.
2. Alteration of the red-cell membrane by the drug so that it
becomes autoimmunogenic. This may happen with
methyldopa, and a direct positive Coombs’ test develops
in about 20% of patients who have been treated with this
drug for more than one year. Frank haemolysis occurs in
only a small proportion of cases. Similar changes can take
place with levodopa,mefenamic acid and beta-lactam
antibiotics.
3. Non-specific binding of plasma protein to red cells, and
thus causing haemolysis. This is believed to occur with
cephalosporins.
Aplastic anaemia as an isolated entity is not common, but
may occur either in isolation or as part of a general depression
of bone marrow activity (pancytopenia). Examples include
chloramphenicol and (commonly and predictably) cytotoxic
drugs.
Agranulocytosis can be caused by many drugs. Several
different mechanisms are implicated, and it is not known
whether allergy plays a part. The drugs most frequently impli-
cated include the following:
most cytotoxic drugs (Chapter 48);
antithyroid drugs (methimazole,carbimazole,
propylthiouracil; Chapter 38);
sulphonamides and sulphonylureas (e.g. tolbutamide,
glipizide; Chapter 37);
antidepressants (especially mianserin; Chapter 20) and
antipsychotics (e.g. phenothiazines, clozapine; Chapter 20);
anti-epileptic drugs (e.g. carbamazepine,felbamate;
Chapter 22).
SYSTEMIC LUP US ERYTHEMATOSUS
Several drugs (including procainamide,isoniazid, hydralazine,
chlorpromazine and anticonvulsants) produce a syndrome
that resembles systemic lupus together with a positive anti-
nuclear factor test. The development of this is closely related
to dose, and in the case of hydralazineit also depends on the
rate of acetylation, which is genetically controlled (Chapter
14). There is some evidence that the drugs act as haptens, com-
bining with DNA and forming antigens. Symptoms usually
disappear when the drug is stopped, but recovery may
be slow.
VASCULITIS
Both acute and chronic vasculitis can result from taking
drugs, and may have an allergic basis. Acute vasculitis with
purpura and renal involvement occurs with penicillins,
sulphonamides and penicillamine. Amore chronic form can
occur with phenytoin.
RENAL DYSFUNCTION
All clinical manifestations of renal disease can be caused by
drugs, and common culprits are non-steroidal anti-inflammatory
drugs and angiotensin-converting enzyme inhibitors (which
cause functional and usually reversible renal failure in suscep-
tible patients; Chapters 26 and 28). Nephrotic syndrome
results from several drugs (e.g. penicillamine, high-dose cap-
topril, gold salts) which cause various immune-mediated
glomerular injuries. Interstitial nephritis can be caused by sev-
eral drugs, including non-steroidal anti-inflammatory drugs
and penicillins, especially meticillin. Cisplatin, aminoglyco-
sides, amphotericin, radiocontrast media and vancomycin
cause direct tubular toxicity. Many drugs cause electrolyte or
acid-base disturbances via their predictable direct or indirect
effects on renal electrolyte excretion (e.g. hypokalaemia and
hypomagnesaemia from loop diuretics, hyperkalaemia from
potassium-sparing diuretics, converting enzyme inhibitors
and angiotensin II receptor antagonists, proximal renal
tubular acidosis from carbonic anhydrase inhibitors), and
some cause unpredictable toxic effects on acid-base balance
(e.g. distal renal tubular acidosis from amphotericin).
Obstructive uropathy can be caused by uric acid crystals con-
sequent upon initiation of chemotherapy in patients with
haematological malignancy, and – rarely – poorly soluble
drugs, such as sulphonamides, methotrexateor indinavir, can
cause crystalluria.
OTHER REACTIONS
Fever is a common manifestation of drug allergy, and
should be remembered in patients with fever of unknown
cause.
Liver damage (hepatitis with or without obstructive fea-
tures) as a side effect of drugs is important. It may be insidi-
ous, leading slowly to end-stage cirrhosis (e.g. during chronic
treatment with methotrexate) or acute and fulminant (as in
some cases of isoniazid, halothane or phenytoin hepatitis).
Chlorpromazine or erythromycin may cause liver involve-
ment characterized by raised alkaline phosphatase and biliru-
bin (‘obstructive’ pattern). Gallstones (and mechanical
obstruction) can be caused by fibrates and other lipid-lowering
drugs (Chapter 27), and by octreotide, a somatostatin ana-
logue used to treat a variety of enteropancreatic tumours,
including carcinoid syndrome and VIPomas (vasoactive intes-
tinal polypeptide) (see Chapter 42). Immune mechanisms are
implicated in some forms of hepatic injury by drugs, but are
seldom solely responsible.
70 ADVERSEDRUG REACTIONS
FURTHER READING AND WEB MATERIAL
Davies DM, Ferner RE de Glanville H. Textbook of adverse drug reac-
tions, 5th edn. Oxford: Oxford Medical Publications, 1998.
Dukes MNG, Aronson JA: 2000: Meylers’s side-effects of drugs, vol. 14.
Amsterdam: Elsevier (see also companion volumes Side-effects of
drugs annuals, 2003, published annually since 1977).
FDAMedwatch website. www.fda.gov/medwatch
Gruchalla RS, Pirmohamed M. Antibiotic allergy. New England Journal
of Medicine2006; 354: 601–609 (practical clinical approach).
Howard RL, Avery AJ, Slavenburg S et al. Which drugs cause prevent-
able admissions to hospital? Asystematic review. British Journal of
Clinical Pharmacology2006; 63: 136–47.
MHRAand the Committee on Safety of Medicines and the Medicine
Control Agency. Current problems in pharmacovigilance. London:
Committee on Safety of Medicines and the Medicine Control
Agency. (Students are advised to monitor this publication for
ongoing and future adverse reactions.)
MHRA Current problems in pharmacovigilance website.
www.mhra.gov.uk/home/idcplg?IdcServiceSS_GET_PAGE&
nodeId368.
Pirmohamed M, James S, Meakin S et al. Adverse drug reactions as
cause of admission to hospital: prospective analysis of 18.820
patients.British Medical Journal 2004; 329: 15–19.
Rawlins MD, Thompson JW. Pathogenesis of adverse drug reactions,
2nd edn. Oxford: Oxford University Press, 1977.
Case history
A 73-year-old man develops severe shoulder pain and is
diagnosed as having a frozen shoulder, for which he is pre-
scribed physiotherapy and given naproxen, 250mg three
times a day, by his family practitioner. The practitioner knows
him well and checks that he has normal renal function for
his age. When he attends for review about two weeks later,
he is complaining of tiredness and reduced urine frequency.
Over the past few days he noted painful but non-swollen
joints and a maculopapular rash on his trunk and limbs. He
is afebrile and apart from the rash there are no other
abnormal physical signs. Laboratory studies show a normal
full blood count; an absolute eosinophil count raised at
490/mm
3
. His serum creatinine was 110μmol/L at baseline
and is now 350μmol/L with a urea of 22.5mmol/L; elec-
trolytes and liver function tests are normal. Urinalysis
shows 2protein, urine microscopy contains 100 leuko-
cytes/hpf with 24% eosinophils.
Question 1
If this is an adverse drug reaction, what type of reaction is
it and what is the diagnosis?
Question 2
What is the best management plan and should this patient
ever receive naproxen again?
Answer 1
The patient has developed an acute interstitial nephritis,
probably secondary to the recent introduction of naproxen
treatment. This is a well-recognized syndrome, with the
clinical features that the patient displays in this case. It can
be associated with many NSAIDs (both non selective NSAIDs
and COX-2 inhibitors), particularly in the elderly. This is a
type B adverse drug reaction whose pathophysiology is
probably a combination of type III and type IV hypersensi-
tivity reactions.
Answer 2
Discontinuation of the offending agent is vital and this
is sometimes sufficient to produce a return to baseline
values of renal function and the disappearance of systemic
symptoms of fever and the rash. Recovery may possibly be
accelerated and further renal toxicity minimized by a short
course (five to seven days) of high-dose oral corticosteroids,
while monitoring renal function. The offending agent
should not be used again in this patient unless the benefits
of using it vastly outweigh the risks associated with its use
in a serious illness.
Introduction 71
Useful interactions 72
Trivial interactions 72
Harmful interactions 73
CHAPTER 13
DRUG INTERACTIONS
INTRODUCTION
Drug interaction is the modification of the action of one drug
by another. There are three kinds of mechanism:
1. pharmaceutical;
2. pharmacodynamic;
3. pharmacokinetic.
Pharmaceutical interactions occur by chemical reaction or
physical interaction when drugs are mixed. Pharmacodynamic
interactions occur when different drugs each infuence the same
physiological function (e.g. drugs that influence state of alert-
ness or blood pressure); the result of adding a second such
drug during treatment with another may be to increase the
effect of the first (e.g. alcohol increases sleepiness caused by
benzodiazepines). Conversely, for drugs with opposing
actions, the result may be to reduce the effect of the first (e.g.
indometacinincreases blood pressure in hypertensive patients
treated with an antihypertensive drug such as losartan).
Pharmacokinetic interactions occur when one drug affects the
pharmocokinetics of another (e.g. by reducing its elimin-ation
from the body or by inhibiting its metabolism). These mecha-
nisms are discussed more fully below in the section on adverse
interactions grouped by mechanism. A drug interaction can
result from one or a combination of these mechanisms.
Drug interaction is important because, whereas judicious
use of more than one drug at a time can greatly benefit
patients, adverse interactions are not uncommon, and may be
catastrophic, yet are often avoidable. Multiple drug use
(‘polypharmacy’) is extremely common, so the potential for
drug interaction is enormous. One study showed that on
average 14 drugs were prescribed to medical in-patients
per admission (one patient received 36 different drugs). The
problem is likely to get worse, for several reasons.
1. Many drugs are not curative, but rather ameliorate chronic
conditions (e.g. arthritis). The populations of western
countries are ageing, and elderly individuals not
uncommonly have several co-morbid conditions.
2. It is all too easy to enter an iatrogenic spiral in which a
drug results in an adverse effect that is countered by the
introduction of another drug, and so on. Prescribers
should heed the moral of the nursery rhyme about the old
lady who swallowed a fly! Hospital admission provides
an opportunity to review all medications that any patient
is receiving, to ensure that the overall regimen is rational.
Out-patients also often receive several prescribed drugs, plus
proprietary over-the-counter medicines, ‘alternative’ remedies
60
50
40
30
Percentage of patients with
adverse drug reactions
Mortality
rate (%)
Average hospital
stay (days)
20
10
10
6
2
0
04
1–10 1611–15
8121620
Number of drugs administered(a)
(b)
(c)
Number of drugs administered
30
20
10
1–10 1611–15
Number of drugs administered
Figure 13.1:Relationship of number of drugs administered to
(a) adverse drug reactions, (b) mortality rate and (c) average
duration of hospital stay. (Redrawn by permission of the British
Medical Journal from Smith JW et al. Annals of Internal Medicine
1966;65: 631.)
(see Chapter 17) and ‘lifestyle’ drugs taken for social reasons.
The greater the number of drugs taken, the more likely things
are to go wrong (Figure 13.1).
Drug interactions can be useful, of no consequence, or
harmful.
USEFUL INTERACTIONS
INCREASED EFFECT
Drugs can be used in combination to enhance their effective-
ness. Disease is often caused by complex processes, and drugs
that influence different components of the disease mechanism
may have additive effects (e.g. an antiplatelet drug with a fibri-
nolytic in treating myocardial infarction, Chapter 29). Other
examples include the use of a β
2
agonist with a glucocorticoid in
the treatment of asthma (to cause bronchodilation and suppress
inflammation, respectively; Chapter 33).
Combinations of antimicrobial drugs are used to prevent
the selection of drug-resistant organisms. Tuberculosis is the
best example of a disease whose successful treatment requires
this approach (Chapter 44). Drug resistance via synthesis of a
microbial enzyme that degrades antibiotic (e.g. penicillinase-
producing staphylococci) can be countered by using a combi-
nation of the antibiotic with an inhibitor of the enzyme:
co-amoxiclavis a combination of clavulanic acid, an inhibitor
of penicillinase, with amoxicillin.
Increased efficacy can result from pharmacokinetic
interaction. Imipenem (Chapter 43) is partly inactivated by a
dipeptidase in the kidney. This is overcome by administering
imipenem in combination with cilastin, a specific renal
dipeptidase inhibitor. Another example is the use of the com-
bination of ritonavirand saquinavir in antiretroviral therapy
(Chapter 46). Saquinavir increases the systemic bioavailabil-
ity of ritonavir by inhibiting its degradation by gastro-
intestinal CYP3Aand inhibits its faecal elimination by block-
ing the P-glycoprotein that pumps it back into the intestinal
lumen.
Some combinations of drugs have a more than
additive effect (‘synergy’). Several antibacterial combinations
are synergistic, including sulfamethoxazole with trimetho-
prim (co-trimoxazole), used in the treatment of Pneumocystis
carinii (Chapter 46). Several drugs used in cancer chemother-
apy are also synergistic, e.g. cisplatin plus paclitaxel
(Chapter 48).
Therapeutic effects of drugs are often limited by the acti-
vation of a physiological control loop, particularly in the case of
cardiovascular drugs. The use of a low dose of a second drug
that interrupts this negative feedback may therefore enhance
effectiveness substantially. Examples include the combination
of an angiotensin converting enzyme inhibitor (to block the
renin-angiotensin system) with a diuretic (the effect of which
is limited by activation of the renin-angiotensin system) in
treating hypertension (Chapter 28).
MINIMIZE SIDE EFFECTS
There are many situations (e.g. hypertension) where low
doses of two drugs may be better tolerated, as well as more
effective, than larger doses of a single agent. Sometimes drugs
with similar therapeutic effects have opposing undesirable
metabolic effects, which can to some extent cancel out when
the drugs are used together. The combination of a loop
diuretic (e.g. furosemide) with a potassium-sparing diuretic
(e.g.spironolactone) provides an example.
Predictable adverse effects can sometimes be averted by
the use of drug combinations. Isoniazidneuropathy is caused
by pyridoxine deficiency, and is prevented by the prophylac-
tic use of this vitamin. The combination of a peripheral dopa
decarboxylase inhibitor (e.g. carbidopa) with levodopa
permits an equivalent therapeutic effect to be achieved with a
lower dose of levodopa than is needed when it is used as a sin-
gle agent, while reducing dose-related peripheral side effects
of nausea and vomiting (Chapter 21).
BLOCK ACUTELY AN UNWANTED (TOXIC) EFFECT
Drugs can be used to block an undesired or toxic effect, as for
example when an anaesthetist uses a cholinesterase inhibitor
to reverse neuromuscular blockade, or when antidotes such
as naloxone are used to treat opioid overdose (Chapter 54).
Uses of vitamin K or of fresh plasma to reverse the effect of
warfarin(Chapter 30) are other important examples.
TRIVIAL INTERACTIONS
Many interactions are based on in vitro experiments, the
results of which cannot be extrapolated uncritically to the clin-
ical situation. Many such potential interactions are of no prac-
tical consequence. This is especially true of drugs with
shallow dose–response curves and of interactions that depend
on competition for tissue binding to sites that are not directly
involved in drug action but which influence drug distribution
(e.g. to albumin in blood).
SHALLOW DOSE–RESPONSE CURVES
Interactions are only likely to be clinically important when
there is a steep dose–response curve and a narrow therapeutic
window between minimum effective dose and minimum
toxic dose of one or both interacting drugs (Figure 13.2). This
is often not the case. For example, penicillin, when used in
most clinical situations, is so non-toxic that the usual dose is
more than adequate for therapeutic efficacy, yet far below that
which would cause dose-related toxicity. Consequently, a second
drug that interacts with penicillin is unlikely to cause either
toxicity or loss of efficacy.
72 DRUG INTERACTIONS
PLASMA AND TISSUE BINDING SITE
INTERACTIONS
One large group of potential drug interactions that are seldom
clinically important consists of drugs that displace one
another from binding sites on plasma albumin or α-1 acid glyco-
protein (AAG) or within tissues. This is a common occurrence
and can readily be demonstrated in plasma or solutions of
albumin/AAG in vitro. However, the simple expectation that
the displacing drug will increase the effects of the displaced
drug by increasing its free (unbound) concentration is seldom
evident in clinical practice. This is because drug clearance
(renal or metabolic) also depends directly on the concentra-
tion of free drug. Consider a patient receiving a regular main-
tenance dose of a drug. When a second displacing drug is
commenced, the free concentration of the first drug rises only
transiently before increased renal or hepatic elimination
reduces total (bound plus free) drug, and restores the free con-
centration to that which prevailed before the second drug was
started. Consequently, any increased effect of the displaced
drug is transient, and is seldom important in practice. It must,
however, be taken into account if therapy is being guided by
measurements of plasma drug concentrations, as most such
determinations are of total (bound plus free) rather than just
free concentration (Chapter 8).
An exception, where a transient increase in free concentra-
tion of a circulating substance (albeit not a drug) can have dev-
astating consequences, is provided by bilirubin in premature
babies whose ability to metabolize bile pigments is limited.
Unconjugated bilirubin is bound by plasma albumin, and inju-
dicious treatment with drugs, such as sulphonamides, that
displace it from these binding sites permits diffusion of free
bilirubin across the immature blood–brain barrier, consequent
staining of and damage to basal ganglia (‘kernicterus’) and
subsequent choreoathetosis in the child.
Instances where clinically important consequences do
occur on introducing a drug that displaces another from
tissue binding sites are in fact often due to additional actions
of the second drug on elimination of the first. For instance,
quinidine displaces digoxin from tissue binding sites, and
can cause digoxintoxicity, but only because it simultaneously
reduces the renal clearance of digoxin by a separate mech-
anism. Phenylbutazone (an NSAID currently reserved for
ankylosing spondylitis unresponsive to other drugs, Chapter
26) displaces warfarin from binding sites on albumin, and
causes excessive anticoagulation, but only because it also
inhibits the metabolism of the active isomer of warfarin
(S-warfarin), causing this to accumulate at the expense of the
inactive isomer. Indometacin(another NSAID) also displaces
warfarin from binding sites on albumin, but does not inhibit
its metabolism and does not further prolong prothrombin
time in patients treated with warfarin, although it can cause
bleeding by causing peptic ulceration and interfering with
platelet function.
HARMFUL INTERACTIONS
It is impossible to memorize reliably the many clinically
important drug interactions, and prescribers should use suit-
able references (e.g. the British National Formulary) to check
for potentially harmful interactions. There are certain drugs
with steep dose–response curves and serious dose-related tox-
icities for which drug interactions are especially liable to cause
HARMFULINTERACTIONS 73
Response
Dose
Therapeutic range Toxic range
Steep dose–response curve
Narrow therapeutic index
Adverse effect likely Adverse effect unlikely
Shallow dose–response curve
Wide therapeutic index
Therapeutic range Toxic range
Dose
Response
Figure 13.2:Drug dose–response curves illustrating likelihood of adverse effect if an interaction increases its blood level.
harm (Figure 13.2), and where special caution is required with
concurrent therapy. These include:
warfarin and other anticoagulants;
anticonvulsants;
cytotoxic drugs;
drugs for HIV/AIDS;
immunosuppressants;
digoxin and other anti-dysrhythmic drugs;
oral hypoglycaemic agents;
xanthine alkaloids (e.g. theophylline);
monoamine oxidase inhibitors.
The frequency and consequences of an adverse interaction
when two drugs are used together are seldom known pre-
cisely. Every individual has a peculiar set of characteristics
that determine their response to therapy.
RISK OF ADVERSE DRUG INTERACTIONS
In the Boston Collaborative Drug Surveillance Program, 234 of
3600 (about 7%) adverse drug reactions in acute-care hospitals
were identified as being due to drug interactions. In a smaller
study in a chronic-care setting, the prevalence of adverse
interactions was much higher (22%), probably because of
the more frequent use of multiple drugs in elderly patients
with multiple pathologies. The same problems exist for the
detection of adverse drug interactions as for adverse drug
reactions (Chapter 12). The frequency of such interactions will
be underestimated by attribution of poor therapeutic outcome
to an underlying disease. For example, graft rejection follow-
ing renal transplantation is not uncommon. Historically, it
took several years for nephrologists to appreciate that epilep-
tic patients suffered much greater rejection rates than did non-
epileptic subjects. These adverse events proved to be due to an
interaction between anticonvulsant medication and immuno-
suppressant cortico-steroid therapy, which was rendered inef-
fective because of increased drug metabolism. In future, a
better understanding of the potential mechanisms of such
interactions should lead to their prediction and prevention by
study in early-phase drug evaluation.
SEVERITY OF ADVERSE DRUG INTERACTIONS
Adverse drug interactions are diverse, including unwanted
pregnancy (from failure of the contraceptive pill due to con-
comitant medication), hypertensive stroke (from hypertensive
crisis in patients on monoamine oxidase inhibitors), gastro-
intestinal or cerebral haemorrhage (in patients receiving war-
farin), cardiac arrhythmias (e.g. secondary to interactions
leading to electrolyte disturbance or prolongation of the QTc)
and blood dyscrasias (e.g. from interactions between allopuri-
nol and azathioprine). Adverse interactions can be severe. In
one study, nine of 27 fatal drug reactions were caused by drug
interactions.
ADVERSE INTERACTIONS GROUPED BY
MECHANISM
PHARMACEUTICAL INTERACTIONS
Inactivation can occur when drugs (e.g. heparinwith gentam-
icin) are mixed. Examples are listed in Table 13.1. Drugs may
also interact in the lumen of the gut (e.g. tetracycline with
iron, and colestyraminewith digoxin).
PHARMACODYNAMIC INTERACTIONS
These are common. Most have a simple mechanism consisting
of summation or opposition of the effects of drugs with,
respectively, similar or opposing actions. Since this type of
interaction depends broadly on the effect of a drug, rather
than on its specific chemical structure, such interactions are
non-specific. Drowsiness caused by an H
1
-blocking antihista-
mine and by alcohol provides an example. It occurs to a
greater or lesser degree with all H
1
-blockers irrespective of the
chemical structure of the particular drug used. Patients must
be warned of the dangers of consuming alcohol concurrently
when such antihistamines are prescribed, especially if they
drive or operate machinery. Non-steroidal anti-inflammatory
agents and antihypertensive drugs provide another clinically
important example. Antihypertensive drugs are rendered less
effective by concurrent use of non-steroidal anti-inflammatory
drugs, irrespective of the chemical group to which they
belong, because of inhibition of biosynthesis of vasodilator
prostaglandins in the kidney (Chapter 26).
74 DRUG INTERACTIONS
Key points
Drug interactions may be clinically useful, trivial or
adverse.
Useful interactions include those that enable efficacy to
be maximized, such as the addition of an angiotensin
converting enzyme inhibitor to a thiazide diuretic in a
patient with hypertension inadequately controlled on
diuretic alone (see Chapter 28). They may also enable
toxic effects to be minimized, as in the use of
pyridoxine to prevent neuropathy in malnourished
patients treated with isoniazid for tuberculosis, and
may prevent the emergence of resistant organisms
(e.g. multi-drug regimens for treating tuberculosis, see
Chapter 44).
Many interactions that occur in vitro (e.g. competition
for albumin) are unimportant in vivo because
displacement of drug from binding sites leads to
increased elimination by metabolism or excretion and
hence to a new steady state where the total
concentration of displaced drug in plasma is reduced,
but the concentration of active, free (unbound) drug is
the same as before the interacting drug was
introduced. Interactions involving drugs with a wide
safety margin (e.g. penicillin) are also seldom clinically
important.
Adverse drug interactions are not uncommon, and can
have profound consequences, including death from
hyperkalaemia and other causes of cardiac dysrhythmia,
unwanted pregnancy, transplanted organ rejection, etc.
PHARMACOKINETIC INTERACTIONS
Absorption
In addition to direct interaction within the gut lumen (see
above), drugs that influence gastric emptying (e.g. metoclo-
pramide,propantheline) can alter the rate or completeness of
absorption of a second drug, particularly if this has low
bioavailability. Drugs can interfere with the enterohepatic
recirculation of other drugs. Failure of oral contraception can
result from concurrent use of antibiotics, due to this mech-
anism. Many different antibiotics have been implicated.
Phenytoin reduces the effectiveness of ciclosporin partly by
reducing its absorption.
Distribution
As explained above, interactions that involve only mutual
competition for inert protein- or tissue-binding sites seldom, if
ever, give rise to clinically important effects. Examples of com-
plex interactions where competition for binding sites occurs in
conjunction with reduced clearance are mentioned below.
Metabolism
Decreased efficacy can result from enzyme induction by a
second agent (Table 13.3). Historically, barbiturates were clin-
ically the most important enzyme inducers, but with the
decline in their use, other anticonvulsants, notably carba-
mazepine and the antituberculous drug rifampicin, are now
the most common cause of such interactions. These necessitate
special care in concurrent therapy with warfarin,phenytoin,
oral contraceptives, glucocorticoids or immunosuppressants
(e.g.ciclosporin, sirolimus).
Drugs with negative inotropic effects can precipitate heart
failure, especially when used in combination. Thus, beta-
blockers and verapamil may precipitate heart failure if used
sequentially intravenously in patients with supraventricular
tachycardia.
Warfarininterferes with haemostasis by inhibiting the coagu-
lation cascade, whereas aspirin influences haemostasis by
inhibiting platelet function. Aspirinalso predisposes to gastric
bleeding by direct irritation and by inhibition of prostaglandin
E
2
biosynthesis in the gastric mucosa. There is therefore the
potential for serious adverse interaction between them.
Important interactions can occur between drugs acting at a
common receptor. These interactions are generally useful
when used deliberately, for example, the use of naloxone to
reverse opiate intoxication.
One potentially important type of pharmacodynamic drug
interaction involves the interruption of physiological control
loops. This was mentioned above as a desirable means of
increasing efficacy. However, in some situations such control
mechanisms are vital. The use of β-blocking drugs in patients
with insulin-requiring diabetes is such a case, as these patients
may depend on sensations initiated by activation of β-receptors
to warn them of insulin-induced hypoglycaemia.
Alterations in fluid and electrolyte balance represent an
important source of pharmacodynamic drug interactions (see
Table 13.2). Combined use of diuretics with actions at different
parts of the nephron (e.g. metolazone and furosemide) is
valuable in the treatment of resistant oedema, but without
close monitoring of plasma urea levels, such combinations
readily cause excessive intravascular fluid depletion and pre-
renal renal failure (Chapter 36). Thiazide and loop diuretics
commonly cause mild hypokalaemia, which is usually of no
consequence. However, the binding of digoxin to plasma
membrane Na
/K
adenosine triphosphatase (Na
/K
ATPase), and hence its toxicity, is increased when the extracel-
lular potassium concentration is low. Concurrent use of such
diuretics therefore increases the risk of digoxin toxicity.
β
2
-Agonists, such as salbutamol, also reduce the plasma
potassium concentration, especially when used intravenously.
Conversely, potassium-sparing diuretics may cause hyper-
kalaemia if combined with potassium supplements and/or
angiotensin converting enzyme inhibitors (which reduce cir-
culating aldosterone), especially in patients with renal impair-
ment. Hyperkalaemia is one of the most common causes of
fatal adverse drug reactions.
HARMFULINTERACTIONS 75
Table 13.1: Interactions outside the body
Mixture Result
Thiopentone and suxamethonium Precipitation
Diazepam and infusion fluids Precipitation
Phenytoin and infusion fluids Precipitation
Heparin and hydrocortisone Inactivation of heparin
Gentamicin and hydrocortisone Inactivation of gentamicin
Penicillin and hydrocortisone Inactivation of penicillin
Table 13.2:Interactions secondary to drug-induced alterations of fluid and
electrolyte balance
Primary drug Interacting drug Result of
effect interaction
Digoxin Diuretic-induced Digoxin toxicity
hypokalaemia
Lidocaine Diuretic-induced Antagonism of anti-
hypokalaemia dysrhythmic effects
Diuretics NSAID-induced salt Antagonism of
and water retention diuretic effects
Lithium Diuretic-induced Raised plasma lithium
reduction in lithium
clearance
Angiotensin Potassium chloride Hyperkalaemia
converting and/ or potassium-
enzyme inhibitor retaining diuretic-
induced
hyperkalaemia
NSAID, non-steroidal anti-inflammatory drug.
Withdrawal of an inducing agent during continued admin-
istration of a second drug can result in a slow decline in
enzyme activity, with emergence of delayed toxicity from the
second drug due to what is no longer an appropriate dose.
For example, a patient receiving warfarinmay be admitted to
hospital for an intercurrent event and receive treatment with
an enzyme inducer. During the hospital stay, the dose of
warfarin therefore has to be increased in order to maintain
measurements of international normalized ratio (INR) within
the therapeutic range. The intercurrent problem is resolved,
the inducing drug discontinued and the patient discharged
while taking the larger dose of warfarin. If the INR is
not checked frequently, bleeding may result from an
excessive effect of warfarin days or weeks after discharge
from hospital, as the effect of the enzyme inducer gradually
wears off.
Inhibition of drug metabolism also produces adverse
effects (Table 13.4). The time-course is often more rapid than
for enzyme induction, since it depends merely on the attain-
ment of a sufficiently high concentration of the inhibiting
drug at the metabolic site. Xanthine oxidase is responsible for
inactivation of 6-mercaptopurine, itself a metabolite of aza-
thioprine. Allopurinol markedly potentiates these drugs
by inhibiting xanthine oxidase. Xanthine alkaloids (e.g.
theophylline) are not inactivated by xanthine oxidase, but
rather by a form of CYP450. Theophyllinehas serious (some-
times fatal) dose-related toxicities, and clinically important
interactions occur with inhibitors of the CYP450 system,
notably several antibiotics, including ciprofloxacin and clar-
ithromycin. Severe exacerbations in asthmatic patients
are often precipitated by chest infections, so an awareness of
these interactions before commencing antibiotic treatment is
essential.
Hepatic CYP450 inhibition also accounts for clinically
important interactions with phenytoin (e.g. isoniazid) and
withwarfarin (e.g. sulphonamides). Non-selective monoamine
oxidase inhibitors (e.g. phenelzine) potentiate the action of
indirectly acting amines such as tyramine, which is present in a
wide variety of fermented products (most famously soft
cheeses: ‘cheese reaction’).
Clinically important impairment of drug metabolism may
also result indirectly from haemodynamic effects rather than
enzyme inhibition. Lidocaine is metabolized in the liver and
the hepatic extraction ratio is high. Consequently, any drug
that reduces hepatic blood flow (e.g. a negative inotrope) will
reduce hepatic clearance of lidocaineand cause it to accumu-
late. This accounts for the increased lidocaine concentration
and toxicity that is caused by β-blocking drugs.
Excretion
Many drugs share a common transport mechanism in the
proximal tubules (Chapter 6) and reduce one another’s excre-
tion by competition (Table 13.5). Probenecid reduces peni-
cillin elimination in this way. Aspirin and non-steroidal
anti-inflammatory drugs inhibit secretion of methotrexate
into urine, as well as displacing it from protein-binding
sites, and can cause methotrexate toxicity. Many diuretics
reduce sodium absorption in the loop of Henle or the distal
tubule (Chapter 36). This leads indirectly to increased proxi-
mal tubular reabsorption of monovalent cations. Increased
proximal tubular reabsorption of lithium in patients treated
with lithium salts can cause lithium accumulation and
toxicity. Digoxinexcretion is reduced by spironolactone, ver-
apamil and amiodarone, all of which can precipitate digoxin
toxicity as a consequence, although several of these inter-
actions are complex in mechanism, involving displacement
from tissue binding sites, in addition to reduced digoxin
elimination.
Changes in urinary pH alter the excretion of drugs that are
weak acids or bases, and administration of systemic alkalinizing
or acidifying agents influences reabsorption of such drugs
76 DRUG INTERACTIONS
Table 13.3:Interactions due to enzyme induction
Primary drug Inducing agent Effect of
interaction
Warfarin Barbiturates Decreased anticoagulation
Ethanol
Rifampicin
Oral contraceptives Rifampicin Pregnancy
Prednisolone/ Anticonvulsants Reduced
ciclosporin immunosuppression
(graft rejection)
Theophylline Smoking Decreased plasma
theophylline
Table 13.4:Interactions due to CYP450 or other enzyme inhibition
Primary drug Inhibiting drug Effect of
interaction
Phenytoin Isoniazid Phenytoin intoxication
Cimetidine
Chloramphenicol
Warfarin Allopurinol Haemorrhage
Metronidazole
Phenylbutazone
Co-trimoxazole
Azathioprine, 6-MP Allopurinol Bone-marrow
suppression
Theophylline Cimetidine Theophylline toxicity
Erythromycin
Cisapride Erythromycin Ventricular tachycardia
Ketoconazole
6-MP, 6-mercaptopurine.
FURTHER READING
There is a very useful website for CYP450 substrates with inhibitors
and inducers: http://medicine.iupui.edu/flockhart/
British Medical Association and Royal Pharmaceutical Society of
Great Britain. British National Formulary 54. London: Medical
Association and Royal Pharmaceutical Society of Great Britian,
2007. (Appendix 1 provides an up-to-date and succinct alphabet-
ical list of interacting drugs, highlighting interactions that are
potentially hazardous.)
Brown HS, Ito K, Galetin Aet al. Prediction of in vivo drug–drug inter-
actions from in vitro data: impact of incorporating parallel path-
ways of drug elimination and inhibitor absorption rate constant.
British Journal of Clinical Pharmacology2005; 60: 508–18.
Constable S, Ham A, Pirmohamed M. Herbal medicines and acute
medical emergency admissions to hospital. British Journal of
Clinical Pharmacology2007; 63: 247–8.
De Bruin ML, Langendijk PNJ, Koopmans RPet al. In-hospital cardiac
arrest is associated with use of non-antiarrhythmic QTc-prolonging
drugs. British Journal of Clinical Pharmacology2007; 63: 216–23.
Fugh-Berman A, Ernst E. Herb–drug interactions: Review and assess-
ment of report reliability. British Journal of Clinical Pharmacology
2001;52: 587–95.
Hurle AD, Navarro AS, Sanchez MJG. Therapeutic drug monitoring
of itraconazole and the relevance of pharmacokinetic interactions.
Clinical Microbiology and Infection2006; 12 (Suppl. 7): 97–106.
Jackson SHD, Mangoni AA, Batty GM. Optimization of drug prescrib-
ing.British Journal of Clinical Pharmacology 2004; 57: 231–6.
Karalleidde L, Henry J. Handbook of drug interactions. London: Edward
Arnold, 1998.
Mertens-Talcott SU, Zadezensky I, De Castro WV et al.
Grapefruit–drug interactions: Can interactions with drugs be
avoided?Journal of Clinical Pharmacology 2006; 46: 1390–1416.
HARMFULINTERACTIONS 77
Table 13.5:Competitive interactions for renal tubular transport
Primary drug Competing drug Effect of
interaction
Penicillin Probenecid Increased penicillin
blood level
Methotrexate Salicylates Bone marrow
suppression
Sulphonamides
Salicylate Probenecid Salicylate toxicity
Indometacin Probenecid Indometacin toxicity
Digoxin Spironolactone Increased plasma
Amiodarone digoxin
Verapamil
Key points
There are three main types of adverse interaction:
pharmaceutical;
pharmacodynamic;
pharmacokinetic.
Pharmaceutical interactions are due to in vitro
incompatibilities, and they occur outside the body (e.g.
when drugs are mixed in a bag of intravenous solution,
or in the port of an intravenous cannula).
Pharmacodynamic interactions between drugs with a
similar effect (e.g. drugs that cause drowsiness) are
common. In principle, they should be easy to anticipate,
but they can cause serious problems (e.g. if a driver fails
to account for the interaction between an
antihistamine and ethanol).
Pharmacokinetic interactions are much more difficult to
anticipate. They occur when one drug influences the
way in which another is handled by the body:
(a) absorption(e.g. broad-spectrum antibiotics
interfere with enterohepatic recirculation of
oestrogens and can cause failure of oral
contraception);
(b) distribution– competition for binding sites seldom
causes problems on its own but, if combined with
an ef
fect on elimination (e.g. amiodarone/digoxin
or NSAID/methotrexate), serious toxicity may
ensue;
(c) metabolism– many serious interactions stem from
enzyme induction or inhibition. Important
inducing agents include ethanol, rifampicin,
rifabutin, many of the older anticonvulsants,
St John’s wort, nevirapine and pioglitazone.
Common inhibitors include many antibacterial
drugs (e.g. isoniazid, macrolides, co-trimoxazole
and metronidazole), the azole antifungals,
cimetidine, allopurinol, HIV protease inhibitors;
(d) excretion(e.g. diuretics lead to increased
reabsorption of lithium, reducing its clearance
and predisposing to lithium accumulation and
toxicity).
Case history
A 64-year-old Indian male was admitted to hospital with mil-
iary tuberculosis. In the past he had had a mitral valve
replaced, and he had been on warfarin ever since. Treatment
was commenced with isoniazid, rifampicin and pyrazi-
namide, and the INR was closely monitored in anticipation of
increased warfarin requirements. He was discharged after
several weeks with the INR in the therapeutic range on a
much increased dose of warfarin. Rifampicin was subse-
quently discontinued. Two weeks later the patient was again
admitted, this time drowsy and complaining of headache
after mildly bumping his head on a locker. His pupils were
unequal and the INR was 7.0. Fresh frozen plasma was
administered and neurosurgical advice was obtained.
Comment
This patient’s warfarin requirement increased during treat-
ment with rifampicin because of enzyme induction, and
the dose of warfarin was increased to maintain anticoagu-
lation. When rifampicin was stopped, enzyme induction
gradually receded, but the dose of warfarin was not
readjusted. Consequently, the patient became over-anti-
coagulated and developed a subdural haematoma in
response to mild trauma. Replacment of clotting factors
(present in fresh frozen plasma) is the quickest way to
reverse the effect of warfarin overdose (Chapter 30).
from urine (e.g. the excretion of salicylateis increased in an alka-
line urine). Such effects are used in the management of overdose
(Chapter 54).
Neuvonen PJ, Niemi M, Backman JT. Drug interactions with lipid-
lowering drugs: mechanisms and clinical relevance. Clinical
Pharmacology and Therapeutics2006; 80: 565–81.
Perucca E. Clinically relevant drug interactions with antiepileptic
drugs. British Journal of Clinical Pharmacology2006; 61: 246–55.
Stockley I. Drug interactions, 2nd edn. Oxford: Blackwell Scientific
Publications, 1991.
Westphal JF. 2000 Macrolide-induced clinically relevant drug interac-
tions with cytochrome P-450A(CYP) 3A4: an update focused on
clarithromycin, azithromycin and dirithromycin. British Journal of
Clinical Pharmacology2000; 50: 285–95.
Whitten DL, Myers SP, Hawrelak JAet al. The effect of St John’s wort
extracts on CYP3A: a systematic review of prospective clinical
trials.British Journal of Clinical Pharmacology 2006; 62: 512–26.
78 DRUG INTERACTIONS
Introduction: ‘personalized medicine’ 79
Genetic influences on drug metabolism 79
Genetic influences on drug disposition 82
Genetic influences on drug action 82
Inherited diseases that predispose to drug toxicity 83
CHAPTER 14
PHARMACOGENETICS
INTRODUCTION: ‘PERSONALIZED MEDICINE’
Variability in drug response between individuals is due to genetic
and environmental effects on drug absorption, distribution,
metabolism or excretion (pharmacokinetics) and on target protein
(receptor) or downstream protein signalling (pharmacodynam-
ics). Several idiosyncratic adverse drug reactions (ADRs) have
been explained in terms of genetically determined variation in the
activity of enzymes involved in metabolism, or of other proteins
(e.g. variants of haemoglobin and haemolysis). The study of vari-
ation in drug responses under hereditary control is known as
pharmacogenetics. Mutation results in a change in the nucleotide
sequence of DNA. Single nucleotide polymorphisms (SNPs) are
very common. They may change the function or level of expres-
sion of the corresponding protein. (Not all single nucleotide vari-
ations change the coded protein because the genetic code is
‘redundant’ – i.e. more than one triplet of nucleotides codes for
each amino acid – so a change in one nucleotide does not always
change the amino acid coded by the triplet, leaving the structure
of the coded protein unaltered.) Balanced polymorphisms, when
a substantial fraction of a population differs from the remainder
in such a way over many generations, results when heterozygotes
experience some selective advantage. Tables 14.1 and 14.2 detail
examples of genetic influences on drug metabolism and response.
It is hoped that by defining an individual’s DNAsequence from a
blood sample, physicians will be able to select a drug that will be
effective without adverse effects. This much-hyped ‘personalized
medicine’ has one widely used clinical application currently, that
of genotyping the enzyme thiopurine methyl-transferase (which
inactivates 6-mercaptopurine (6-MP)) to guide dosing 6-MP in
children with acute lymphocytic leukaemia, but could revolu-
tionize therapeutics in the future.
Throughout this chapter, italics are used for the gene and
plain text for the protein product of the gene.
GENETIC INFLUENCES ON DRUG
METABOLISM
Abnormal sensitivity to a drug may be the result of a
genetic variation of the enzymes involved in its metabolism.
Inheritance may be autosomal recessive and such disorders
are rare, although they are important because they may have
severe consequences. However, there are also dominant pat-
terns of inheritance that lead to much more common varia-
tions within the population. Balanced polymorphisms of drug
metabolizing enzymes are common. Different ethnic popula-
tions often have a different prevalence of the various enzyme
polymorphisms.
PHASE I DRUG METABOLISM
CYP2D6
The CYP2D6 gene is found on chromosome 22 and over 50
polymorphic variants have been defined in humans. The func-
tion of this enzyme (e.g. 4-hydroxylation of debrisoquine, an
adrenergic neurone-blocking drug previously used to treat
hypertension but no longer used clinically) is deficient in about
7–10% of the UK population (Table 14.1). Hydroxylation poly-
morphisms in CYP2D6 explain an increased susceptibility to
several ADRs:
nortriptyline – headache and confusion (in poor
metabolizers);
codeine – weak (or non-existent) analgesia in poor
metabolizers (poor metabolizers convert little of it to
morphine);
phenformin – excessive incidence of lactic acidosis (in
poor metabolizers).
Several drugs (including other opioids, e.g. pethidine, mor-
phineand dextromethorphan; beta-blockers, e.g. metoprolol,
propranolol; SSRIs, e.g. fluoxetine; antipsychotics, e.g.
haloperidol) are metabolized by CYP2D6. The many geno-
typic variants yield four main phenotypes of CYP2D6 – poor
metabolizers (PM) (7–10% of a Caucasian population), inter-
mediate (IM) and extensive metabolizers (EM) (85–90% of
Caucasians) and ultra-rapid metabolizers (UM) (1–2% of
Caucasians, but up to 30% in Egyptians) due to possession of
multiple copies of the CYP2D6 gene. UM patients require
higher doses of CYP2D6 drug substrates for efficacy.
80 PHARMACOGENETICS
Table 14.1: Variations in drug metabolism/pharmacodynamics due to genetic polymorphisms
Pharmacogenetic Mechanism Inheritance Occurrence Drugs involved
variation
Phase I drug metabolism:
Defective CYP2D6 Functionally defective Autosomal recessive 7–10% Caucasians, Originally defined by
1% Saudi Arabians, reduced CYP2D6
30% Chinese debrisoquine hydroxylation;
Beta blockers: metoprolol;
TCAs: nortriptyline; SSRIs:
fluoxetine; Opioids: morphine;
Anti-dysrhythmics: encainide
Ultra-rapid metabolism:
CYP2D6 Duplication 2D6 1–2% Caucasians, Rapid metabolism of 2D6
30% Egyptians drug substrates above
Phase II drug metabolism:
Rapid-acetylator status Increased hepatic Autosomal dominant 45% Caucasians Isoniazid; hydralazine; some
N-acetyltransferase sulphonamides; phenelzine;
dapsone; procainamide
Impaired glucuronidation Reduced activity UGT1A1 7–10% Caucasians Irinotecan (CPT-11)
Abnormal pharmacodynamic responses:
Malignant hyperthermia Polymorphism in Autosomal dominant 1:20000 of Some anaesthetics, especially
with muscular rigidity ryanodine population inhalational, e.g. isoflurane,
receptors (RyR1) suxamethonium
Other:
Suxamethonium Several types of Autosomal recessive Most common Suxamethonium
sensitivity abnormal plasma form 1:2500
pseudocholinesterase
Ethanol sensitivity Relatively low rate of Usual in some ethnic Orientals Ethanol
ethanol metabolism by groups
aldehyde dehydrogenase
CYP2C9 POLYMORPHISM (TOLBUTAMIDE
POLYMORPHISM)
The CYP2C9 gene is found on chromosome 10 and six poly-
morphic variants have been defined. Pharmacogenetic vari-
ation was first described after the finding of a nine-fold range
between individuals in the rate of oxidation of a sulphony-
lurea drug, tolbutamide. CYP2C9 polymorphisms cause
reduced enzyme activity, with 1–3% of Caucasians being poor
(slow) metabolizers. Drugs metabolized by CYP2C9 are elim-
inated slowly in poor metabolizers, who are therefore suscep-
tible to dose-related ADRs. Such drugs include S-warfarin,
losartanand celecoxib, as well as the sulphonylureas.
CYP2C19POLYMORPHISM
CYP2C19 is found on chromosome 10 and four polymorphic
variants have been defined. These polymorphisms produce
reduced enzyme activity and 3–5% of Caucasians and 15–20%
of Asians have genotypes which yield a poor (slow) metabo-
lizer phenotype. Such patients require lower doses of drugs
metabolized by the CYP2C19 enzyme. These include proton
pump inhibitors (omeprazole, lansoprazole, pantoprazole)
and some anticonvulsants, e.g. phenytoin,phenobarbitone.
PHASE II DRUG METABOLISM
ACET YLAT OR S TATUS (N-ACETYLTRANSFERASE-2)
Administration of identical doses (per kilogram body weight)
of isoniazid (INH), an antituberculous drug, results in great
variation in blood concentrations. Adistribution histogram of
such concentrations shows two distinct groups (i.e. a ‘bimodal’
distribution; Figure 14.1). INH is metabolized in the liver by
rapid acetylators, particularly when the drug is not given
daily, but twice weekly. In addition, slow acetylators are more
likely to show phenytoin toxicity when this drug is given
with INH, because the latter inhibits hepatic microsomal
hydroxylation of phenytoin. Isoniazid hepatitis may be
more common among rapid acetylators, but the data are
conflicting.
Acetylator status affects other drugs (e.g. procainamide,
hydralazine) that are inactivated by acetylation. Approxi-
mately 40% of patients treated with procainamide for six
months or longer develop antinuclear antibodies. Slow acety-
lators are more likely to develop such antibodies than rapid
acetylators (Figure 14.2) and more slow acetylators develop
procainamide-induced lupus erythematosus. Similarly, lower
doses of hydralazine are needed to control hypertension in
slow acetylators (Figure 14.3) and these individuals are more
susceptible to hydralazine-induced systemic lupus erythe-
matosus (SLE).
SULPHATION
Sulphation by sulfotransferase (SULT) enzymes shows poly-
morphic variation. SULT enzymes metabolize oestrogens,
progesterones and catecholamines. The polymorphic forms
have reduced activity and contribute to the considerable
variability in metabolism of these compounds.
SUXAMETHONIUM SENSITIVITY
The usual response to a single intravenous dose of suxa-
methoniumis muscular paralysis for three to six minutes. The
effect is brief because suxamethonium is rapidly hydrolysed
by plasma pseudocholinesterase. Occasional individuals
show a much more prolonged response and may remain
GENETIC INFLUENCES ON DRUG METABOLISM 81
Table 14.2:Variations in drug response due to disease caused by genetic mutations
Pharmacogenetic Mechanism Inheritance Occurrence Drugs involved
variation
G6PD deficiency, favism, 80 distinct forms X-linked incomplete 10000 000 affected Many – including 8-
drug-induced of G6PD codominant world-wide aminoquinolines, antimicrobials
haemolytic anaemia and minor analgesics (see text)
Methaemoglobinaemia: Methaemoglobin Autosomal recessive 1:100 are heterozygotes Same drugs as for G6PD
drug-induced reductase deficiency (heterozygotes show deficiency
haemolysis some response)
Acute intermittent
porphyria: exacerbation Increased activity of Autosomal dominant Acute intermittent type Barbiturates, cloral,
induced by drugs D-amino levulinic 15:1000000 in Sweden; chloroquine, ethanol,
synthetase secondary Porphyria cutanea tarda sulphonamides, phenytoin,
to defective porphyrin 1:100 in Afrikaaners griseofulvin and many others
synthesis
0
25
20
15
10
Number of subjects
5
0
246
Plasma isoniazid concentration (μg/mL)
81012
Figure 14.1:Plasma isoniazid concentrations in 483 subjects six
hours after oral isoniazid (9.8mg/kg). Acetylator polymorphism
produces a bimodal distribution into fast and slow acetylators.
(Redrawn from Evans DAP et al. British Medical Journal1960; 2:
485, by permission of the editor.)
acetylation. Individuals who acetylate the drug more rapidly
because of a greater hepatic enzyme activity demonstrate lower
concentrations of INH in their blood following a standard dose
than do slow acetylators. Acetylator status may be measured
usingdapsone by measuring the ratio of monoacetyldapsone to
dapsonein plasma following a test dose.
Slow and rapid acetylator status are inherited in a simple
Mendelian manner. Heterozygotes, as well as homozygotes,
are rapid acetylators because rapid metabolism is autosomal
dominant. Around 55–60% of Europeans are slow acetylators
and 40–45% are rapid acetylators. The rapid acetylator pheno-
type is most common in Eskimos and Japanese (95%) and
rarest among some Mediterranean Jews (20%).
INH toxicity, in the form of peripheral neuropathy, most
commonly occurs in slow acetylators, whilst slower response
and higher risk of relapse of infection are more frequent in
Heterozygotes are unaffected carriers and represent about 4%
of the population.
GENETIC INFLUENCES ON DRUG
DISPOSITION
Several genotypic variants occur in the drug transporter pro-
teins known as ATPbinding cassette proteins (ABC proteins).
The best known is P-glycoprotein now renamed ABCB1. This
has several polymorphisms leading to altered protein expres-
sion/activity. Effects of drug transporter polymorphisms on
drug disposition depend on the individual drug and the
genetic variant, and are still incompletely understood.
GENETIC INFLUENCES ON DRUG ACTION
RECEPTOR/DRUG TARGET POLYMORPHISMS
There are many polymorphic variants in receptors, e.g. oestro-
gen receptors, β-adrenoceptors, dopamine D
2
receptors and
opioid μ receptors. Such variants produce altered receptor
expression/activity. One of the best studied is the β
2
-adreno-
ceptor polymorphism. SNPs resulting in an Arg-to-Gly amino
acid change at codon 16 yield a reduced response to salbuta-
molwith increased desensitization.
Variants in platelet glycoprotein IIb/IIIa receptors modify
the effects of eptifibatide. Genetic variation in serotonin
transporters influences the effects of antidepressants, such
as fluoxetine and clomiprimine. There is a polymorphism
of the angiotensin-converting enzyme (ACE) gene which
involves a deletion in a flanking region of DNAthat controls
the activity of the gene; suggestions that the double-deletion
genotype may be a risk factor for various disorders are
controversial.
WARFARIN SUSCEPTIBILITY
Warfarininhibits the vitamin K epoxide complex 1 (VKORC1)
(Chapter 30). Sensitivity to warfarin has been associated with
the genetically determined combination of reduced metabolism
of the S-warfarin stereoisomer by CYP2C9 *2/*3 and *3/*3
polymorphic variants and reduced activity (low amounts) of
VKORC1. This explains approximately 40% of the variability in
warfarindosing requirement. Warfarin resistance (requirement
for very high doses of warfarin) has been noted in a few pedi-
grees and may be related to poorly defined variants in CYP2C9
combined with VKORC1.
FAMILIAL HYPERCHOLESTEROLAEMIA
Familial hypercholesterolaemia (FH) is an autosomal disease
in which the ability to synthesize receptors for low-density
paralysed and require artificial ventilation for two hours or
longer. This results from the presence of an aberrant form of
plasma cholinesterase. The most common variant which
causes suxamethonium sensitivity occurs at a frequency of
around one in 2500 and is inherited as an autosomal recessive.
82 PHARMACOGENETICS
I I
I I
I I
Slow acetylators
Time to conversion (months)
Percentage of patients
with antinuclear antibodies
I I
I I
I I
9
9
9
0
0
20
40
60
80
100
2 4 6 8 10 12 77
9
9
8
Rapid acetylators
8
Figure 14.2:Development of procainamide-induced antinuclear
antibody in slow acetylators () and rapid acetylators () with
time. Number of patients shown at each point. (Redrawn with
permission from Woosley RL et al. New England Journal of
Medicine1978; 298: 1157.)
0.4
0.3
0.2
0.1
Slow
acetylators
Fast
acetylators
0
Serum concentration
dose
μg/mL
mg/kg/day
(
(
Figure 14.3:Relationship between acetylator status and dose-
normalized serum hydralazine concentration (i.e. serum
concentration corrected for variable daily dose). Serum
concentrations were measured one to two hours after oral
hydralazine doses of 25–100mg in 24 slow and 11 fast
acetylators. (Redrawn with permission from Koch-Weser J.
Medical Clinics of North America1974; 58: 1027.)
lipoprotein (LDL) is impaired. LDL receptors are needed for
hepatic uptake of LDL and individuals with FH consequently
have very high circulating concentrations of LDL, and suffer
from atheromatous disease at a young age. Homozygotes
completely lack the ability to synthesize LDL receptors and
may suffer from coronary artery disease in childhood, whereas
the much more common heterozygotes have intermediate
numbers of receptors between homozygotes and healthy indi-
viduals, and commonly suffer from coronary disease in young
adulthood. β-Hydroxy-β-methylglutaryl coenzyme A (HMG
CoA) reductase inhibitors (otherwise known as statins, an
important class of drug for lowering circulating cholesterol lev-
els) function largely by indirectly increasing the number of
hepatic LDL receptors. Such drugs are especially valuable for
treating heterozygotes with FH, because they restore hepatic
LDLreceptors towards normal in such individuals by increas-
ing their synthesis. In contrast, they are relatively ineffective in
homozygotes because such individuals entirely lack the genetic
material needed for LDL-receptor synthesis.
INHERITED DISEASES THAT PREDISPOSE
TO DRUG TOXICITY
GLUCOSE-6-PHOSPHATE DEHYDROGENASE
DEFICIENCY
Glucose-6-phosphatase dehydrogenase (G6PD) catalyses the
formation of reduced nicotinamide adenine dinucleotide phos-
phate (NADPH), which maintains glutathione in its reduced
form (Figure 14.4). The gene for G6PD is located on the
X-chromosome, so deficiency of this enzyme is inherited in a
sex-linked manner. G6PD deficiency is common, especially in
Mediterranean peoples, those of African or Indian descent and
in East Asia. Reduced enzyme activity results in methaemoglo-
binaemia and haemolysis when red cells are exposed to oxidiz-
ing agents (e.g. as a result of ingestion of broad beans (Vicia
faba), naphthalene or one of several drugs). There are over 80
distinct variants of G6PD, but not all of them produce haem-
olysis. The lower the activity of the enzyme, the more severe is
the clinical disease. The following drugs can produce haemol-
ysis in such patients:
1. analgesics – aspirin;
2. antimalarials – primaquine, quinacrine, quinine;
3. antibacterials – sulphonamides, sulphones,
nitrofurantoin, fluoroquinolones: ciprofloxacin
4. miscellaneous – quinidine, probenecid.
Patients with G6PD deficiency treated with an 8-aminoquino-
line (e.g. primaquine) should spend at least the first few
days in hospital under supervision. If acute severe haemolysis
occurs, primaquine may have to be withdrawn and blood
transfusion may be needed. Hydrocortisone is given intra-
venously and the urine is alkalinized to reduce the likelihood
of deposition of acid haematin in the renal tubules. The
high incidence of this condition in some areas is attributed
to a balanced polymorphism. It is postulated that the selec-
tive advantage conferred on heterozygotes is due to a protec-
tive effect of partial enzyme deficiency against falciparum
malaria.
METHAEMOGLOBINAEMIA
Several xenobiotics oxidize haemoglobin to methaemoglobin,
including nitrates, nitrites, chlorates, sulphonamides, sul-
phones, nitrobenzenes, nitrotoluenes, anilines and topical local
anesthetics. In certain haemoglobin variants (e.g. HbM, HbH),
the oxidized (methaemoglobin) form is not readily converted
back into reduced, functional haemoglobin. Exposure to the
above substances causes methaemoglobinaemia in individuals
with these haemoglobin variants. Similarly, nitrites, chlorates,
dapsoneand primaquine can cause cyanosis in patients with a
deficiency of NADH-methaemoglobin reductase.
MALIGNANT HYPERTHERMIA
This is a rare but potentially fatal complication of general
anaesthesia (Chapter 24). The causative agent is usually an
inhalational anaesthetic (e.g. halothane, isoflurane) and/or
suxamethonium. Sufferers exhibit a rapid rise in temperature,
muscular rigidity, tachycardia, increased respiratory rate,
sweating, cyanosis and metabolic acidosis. There are several
forms, one of the more common ones (characterized by
halothane-induced rigidity) being inherited as a Mendelian
dominant. The underlying abnormality is a variant in the
ryanodine R1 receptor (Ry1R) responsible for controlling
intracellular calcium flux from the sarcolemma. The preva-
lence is approximately 1:20000. Individuals can be genotyped
INHERITED DISEAS ES THAT PREDISPOSE TO DRUG TOXICITY 83
Figure 14.4:Physiological role of glucose-6-phosphate
dehydrogenase.
for Ry1R or undergo muscle biopsy to assess their predisposi-
tion to this condition. Muscle from affected individuals is
abnormally sensitive to caffeine in vitro, responding with a
strong contraction to low concentrations. (Pharmacological
doses of caffeine release calcium from intracellular stores
and cause contraction even in normal muscle at sufficiently
high concentration.) Affected muscle responds similarly to
halothaneor suxamethonium.
ACUTE PORPHYRIAS
This group of diseases includes acute intermittent porphyria,
variegate porphyria and hereditary coproporphyria. In each
of these varieties, acute illness is precipitated by drugs
because of inherited enzyme deficiencies in the pathway of
haem biosynthesis (Figure 14.5). Drugs do not precipitate
acute attacks in porphyria cutanea tarda, a non-acute por-
phyria, although this condition is aggravated by alcohol,
oestrogens, iron and polychlorinated aromatic compounds.
Drug-induced exacerbations of acute porphyria (neuro-
logical, psychiatric, cardiovascular and gastro-intestinal dis-
turbances that are occasionally fatal) are accompanied by
increased urinary excretion of 5-aminolevulinic acid (ALA)
and porphobilinogen. An extraordinarily wide array of drugs
can cause such exacerbations. Most of the drugs that have
been incriminated are enzyme inducers that raise hepatic ALA
synthetase levels. These drugs include phenytoin, sulphonyl-
ureas, ethanol, griseofulvin, sulphonamides, sex hormones,
methyldopa, imipramine, theophylline, rifampicin and
pyrazinamide. Often a single dose of one drug of this type can
precipitate an acute episode, but in some patients repeated
doses are necessary to provoke a reaction.
Specialist advice is essential. Avery useful list of drugs that
are unsafe to use in patients with porphyrias is included in the
British National Formulary.
GILBERT’S DISEASE
This is a benign chronic form of primarily unconjugated hyper-
bilirubinaemia caused by an inherited reduced activity/lack of
the hepatic conjugating enzyme uridine phosphoglucuronyl
transferase (UGT1A1). Oestrogens impair bilirubin uptake and
aggravate jaundice in patients with this condition, as does pro-
tracted fasting. The active metabolite of irinotecan is glu-
curonidated by UGT1A1, so irinotecantoxicity is increased in
Gilbert’s disease.
84 PHARMACOGENETICS
Figure 14.5:Porphyrin metabolism, showing sites of enzyme
deficiency.
Glycine succinyl CoA
-aminolevulinic acid (ALA)
Porphobilinogen (PBG)
Hydroxymethylbilane
Uroporphyrinogen (UPG) III
Coproporphyrinogen (CPG) III
Uroporphyrin
Coproporphyrin
UPG decarboxylase
UPG III synthetase
PBG
deaminase
CPG
oxidase
PPG
oxidase
Ferrochelatase
Protoporphyrinogen (PPG)
Protoporphyrin IX
Haem
ALA synthetase
CO
2
Deficient in
acute intermittent
porphyria
Deficient in
hereditary
coproporphyria
Deficient in
variegate
porphyria
Case history
A 26-year-old Caucasian woman has a three-month history
of intermittent bloody diarrhoea and is diagnosed with
ulcerative colitis. She is initially started on oral prednisolone
30mg/day and sulfasalazine 1 g four times a day with little
improvement in her colitic symptoms. Her gastroenterolo-
gist, despite attempting to control her disease with increas-
ing doses of her initial therapy, reverts to starting low-dose
azathioprine at 25mg three times a day and stopping her
sulfasalazine. Two weeks later, on review, her symptoms of
colitis have improved, but she has ulcers on her oropharynx
with a sore mouth. Her Hb is 9.8g/dL and absolute neu-
trophil count is 250/mm
3
and platelet count 85000.
Question
What is the most likely cause of this clinical situation?
Answer
The patient has haematopoietic toxicity due to azathioprine
(a prodrug of 6-MP). 6-MP is inactivated by the enzyme
thiopurine methyltransferase (TPMT). In Caucasians 0.3%
(one in 300) of patients are genetically deficient in this
enzyme because of polymorphisms in the gene (*3/*4 is
most common) and 11% of Caucasians who have a het-
erozygous genotype have low levels of the enzyme. Patients
with absent or low TPMT expression are at a higher risk of
bone marrow suppression. In this patient, the azathioprine
should be stopped and her TPMT genotype defined. Once
her bone marrow has recovered (with or without
haematopoietic growth factors), she could be restarted on
very low doses (e.g 6.25–12mg azathioprine daily).
INHERITED DISEAS ES THAT PREDISPOSE TO DRUG TOXICITY 85
Key points
Genetic differences contribute substantially to
individual (pharmacokinetic and pharmacodynamic)
variability (20–50%) in drug response.
Mendelian traits that influence drug metabolism
include:
(a) deficient thiopurine methyltransferase (TPMT)
which inactivates 6-MP (excess haematopoietic
suppression);
(b) deficient CYP2D6 activity which hydroxylates
several drug classes, including opioids, β
-blockers,
tricyclic antidepressants and SSRIs;
(c) deficient CYP2C9 activity which hydroxylates several
drugs including sulphonylureas, S-warfarin,
losartan;
(d) acetylator status (NAT-2), a polymorphism that
affects acetylation of drugs, including isoniazid,
hydralazine and dapsone;
(e) pseudocholinesterase deficiency; this leads to
prolonged apnoea after suxamethonium, which is
normally inactivated by this enzyme.
Several inherited diseases predispose to drug toxicity:
(a) glucose-6-phosphate dehydrogenase deficiency
predisposes to haemolysis following many drugs,
including primaquine;
(b) malignant hyperthermia is a Mendelian dominant
af
fecting the ryanodine receptor in striated muscle,
leading to potentially fatal attacks of hyperthermia
and muscle spasm after treatment with
suxamethonium and/or inhalational anaesthetics;
(c) acute porphyrias, attacks of which are particularly
triggered by enzyme-inducing agents, as well as
drugs, e.g. sulphonamides, rifampicin and anti-
seizure medications.
FURTHER READING
Evans DA, McLeod HL, Pritchard S, Tariq M, Mobarek A. Inter-ethnic
variability in human drug responses. Drug Metabolism and
Disposition2001; 29: 606–10.
Evans WE, McLeod HL. Drug therapy: pharmacogenomics – drug
disposition, drug targets, and side effects. New England Journal of
Medicine2003; 348: 538–49.
Wang L, Weinshilboum R. Thiopurine S-methyltransferase pharmaco-
genetics: insights, challenges and future directions. Oncogene2006;
25: 1629–38.
Weinshilboum R. Inheritance and drug response. New England Journal
of Medicine2003; 348: 529–37.
Weinshilboum R, Wang L. Pharmacogenomics: bench to bedside.
Nature Reviews. Drug Discovery2004; 3: 739–48.
Wilkinson GR. Drug therapy: drug metabolism and variability among
patients in drug response. New England Journal of Medicine 2005;
352: 2211–21.
HISTORY
Many years before Christ, humans discovered that certain
plants influence the course of disease. Primitive tribes used
extracts containing active drugs such as opium, ephedrine,
cascara,cocaine, ipecacuanha and digitalis. These were prob-
ably often combined with strong psychosomatic therapies and
the fact that potentially beneficial agents survived the era of
magic and superstition says a great deal about the powers of
observation of those early ‘researchers’.
Many useless and sometimes deleterious treatments also
persisted through the centuries, but the desperate situation of
the sick and their faith in medicine delayed recognition of the
harmful effects of drugs. Any deterioration following drug
administration was usually attributed to disease progression,
rather than to adverse drug effects. There were notable excep-
tions to this faith in medicine and some physicians had a short
life expectancy as a consequence!
Over the last 100 years, there has been an almost exponential
growth in the number of drugs introduced into medicine.
Properly controlled clinical trials, which are the cornerstone of
new drug development and for which the well-organized vac-
cine trials of the Medical Research Council (MRC) must take
much credit, only became widespread after the Second World
War. Some conditions did not require clinical trials (e.g. the early
use of penicillinin conditions with a predictable natural history
and high fatality rate). (Florey is credited with the remark that
‘if you make a real discovery, you don’t need to call in the
statisticians’.) Ethical considerations relating to the use of a ‘non-
treatment’ group in early trials were sometimes rendered irrele-
vant by logistic factors such as the lack of availability of drugs.
It was not until the 1960s that the appalling potential of
drug-induced disease was realized world-wide. Thalidomide
was first marketed in West Germany in 1956 as a sedative/
hypnotic, as well as a treatment for morning sickness. The
drug was successfully launched in various countries, includ-
ing the UK in 1958, and was generally accepted as a safe and
effective compound, and indeed its advertising slogan was
History 86
UK regulatory system 86
The process of drug development 86
Preclinical studies 87
Clinical trials 87
Clinical drug development 89
Generic drugs 90
Ethics committees 91
Globalization 91
CHAPTER 15
INTRODUCTION OF NEW DRUGS
AND CLINICAL TRIALS
‘the safe hypnotic’. However, in 1961, it became clear that its
use in early pregnancy was causally related to rare congenital
abnormality, phocomelia, in which the long bones fail to
develop. At least 600 such babies were born in England and
more than 10000 afflicted babies were born world-wide.
The thalidomide tragedy stunned the medical profession, the
pharmaceutical industry and the general public. In 1963, the
Minister of Health of the UK established a Committee on
the Safety of Drugs, since it was clear that some control over
the introduction and marketing of drugs was necessary. These
attempts at regulation culminated in the Medicines Act (1968).
UK REGULATORY SYSTEM
The UK comes under European Community (EC) legislation
regarding the control of human medicines, which is based upon
safety, quality and efficacy. The UK Medicines and Healthcare
products Regulation Agency (MHRA) or the European Agency
for the Evaluation of Medicinal Products (EMEA) must approve
any new medicine before it can be marketed in the UK. All UK
clinical trials involving a medicinal product must be approved
by the MHRA. The MHRA is assisted by expert advisory
groupsthrough the Commission on Human Medicines (CHM)
to assess new medicines during their development and licens-
ing. The MHRAis also responsible for the quality and safety
monitoring of medicines after licensing. Product labels,
patient leaflets, prescribing information and advertising are
subject to review by the MHRA. In the UK, there is also exten-
sive ‘self-regulation’ of the pharmaceutical industry through
the Association of the British Pharmaceutical Industry (ABPI).
The National Institute for Health and Clinical Excellence
(NICE) is independent of the MHRA.
THE PROCESS OF DRUG DEVELOPMENT
Drug development is a highly regulated process which
should be performed under internationally recognized codes
CLINICAL TRIALS 87
CLINICAL TRIALS
Physicians read clinical papers, review articles and pharma-
ceutical advertisements describing clinical trial results. Despite
peer review, the incompetent or unscrupulous author can con-
ceal deficiencies in design and possibly publish misleading
data. The major medical journals are well refereed, although
supplements to many medical journals are less rigorously
reviewed for scientific value. An understanding of the essen-
tial elements of clinical trial design enables a more informed
interpretation of published data.
Assessment of a new treatment by clinical impression is
not adequate. Diseases may resolve or relapse spontaneously,
coincidental factors may confound interpretation, and the
power of placebo and enthusiastic investigators are a major
influence on subjective response. In order to minimize these
factors and eliminate bias, any new treatment should be rigor-
ously assessed by carefully designed, controlled clinical trials.
All physicians involved in clinical trials must follow the
guidelines of the Declaration of Helsinki and subsequent
amendments.
OBJECTIVES
The first step in clinical trial design is to determine the ques-
tions to be addressed. Primary and achievable objectives must
be defined. The question may be straightforward. For example,
does treatment Aprolong survival in comparison with treat-
ment B following diagnosis of small-cell carcinoma of the
lung? Survival is a clear and objective end-point. Less easily
measured end-points such as quality of life must also be
assessed as objectively as possible. Prespecified subgroups of
patients may be identified and differences in response deter-
mined. For example, treatment A may be found to be most
effective in those patients with limited disease at diagnosis,
whereas treatment B may be most effective in those with
widespread disease at diagnosis. Any physician conducting a
clinical trial must not forget that the ultimate objective of all
studies is to benefit patients. The patients’ welfare must be of
paramount importance.
RANDOMIZATION
Patients who agree to enter such a study must be randomized
so that there is an equal likelihood of receiving treatment Aor B.
If treatment is not truly randomized, then bias will occur. For
example, the investigator might consider treatment B to be
less well tolerated and thus decide to treat particularly frail
patients with treatment A. Multicentre studies are often neces-
sary in order to recruit adequate numbers of patients, and it is
essential to ensure that the treatments are fairly compared. If
treatment Ais confined to one centre/hospital and treatment
B to another, many factors may affect the outcome of the
study due to differences between the centres, such as interval
of practice, namely Good Manufacturing Practice (GMP),
Good Laboratory Practice (GLP) and Good Clinical Practice
(GCP). Good Clinical Practice is an international ethical and
scientific quality standard for designing, conducting, record-
ing and reporting trials that involve the participation of
human subjects. The stages of drug development are outlined
in Figure 15.1.
DRUG DISCOVERY, DESIGN AND SYNTHESIS
Whilst random screening and serendipity remain important
in the discovery of new drugs, new knowledge of the role of
receptors, enzymes, ion channels and carrier molecules in
both normal physiological processes and disease now permits
a more focused approach to drug design. Using advances in
combinatorial chemistry, biotechnology, genomics, high out-
put screening and computer-aided drug design, new drugs
can now be identified more rationally.
PRECLINICAL STUDIES
New chemical entities are tested in animals to investigate their
pharmacology, toxicology, pharmacokinetics and potential
efficacy in order to select drugs of potential value in humans.
Although there is considerable controversy concerning the
value of some studies performed in animals, human drug devel-
opment has an excellent safety record, and there is under-
standable reluctance on the part of the regulatory authorities
to reduce requirements. At present, the European guidelines
require that the effects of the drug should be assessed in two
mammalian species (one non-rodent) after two weeks of dos-
ing before a single dose is administered to a human. In addition,
safety pharmacology and mutagenicity tests will have been
assessed. Additional and longer duration studies are conducted
before product licence approval. The timing, specific tests and
duration of studies may relate to the proposed human usage
in both the clinical trials and eventual indications.
Proof of principle
Proof of concept
Cost is approximately £500 million, 60% of which is spent in
clinical trials. Time from discovery to registration approximatel
y
10–13 years
• Discovery
• Screening
• Preclinical testing
• Phase I
(usually healthy volunteers)
• Phase IIa
• Phase IIb
• Phase III
(1000–5000 patients)
* Registration
• Phase IV
Early (exploratory)
development
Late (confirmatory)
development
Figure 15.1:Stages of drug development.
88 INTRODUCTIONOF NEW DRUGS AND CLINICAL TRIALS
between diagnosis and treatment, individual differences in
determining entry criteria, facilities for treatment of complica-
tions, differing attitude to pain control, ease of transport, etc.
INCLUSION AND EXCLUSION CRITERIA
For any study, inclusion and exclusion criteria must be defined.
It is essential to maximize safety and minimize confounding
factors, whilst also ensuring that the criteria are not so strict
that the findings will be applicable only to an unrepresenta-
tive subset of the patient population encountered in usual
practice. The definition of a healthy elderly subject is problem-
atic. Over the age of 65 years, it is ‘normal’ (in the sense that it
is common) to have a reduced creatinine clearance, to be on
some concomitant medication and to have a history of allergy.
If these are exclusion criteria, a trial will address a ‘superfit’
elderly population and not a normal population.
DOUBLE-BLIN D DESIGN
A‘double-blind’ design is often desirable to eliminate psycho-
logical factors such as enthusiasm for the ‘new’ remedy. This
is not always possible. For example, if in the comparison of
treatment A and treatment B described above, treatment A
consists of regular intravenous infusions whilst treatment B
consists of oral medication, the ‘blind’ is broken. As ‘survival’
duration is ‘hard’ objective data, this should not be influenced
markedly, whereas softer end-points, such as the state of well-
being, are more easily confounded. In trials where these are
especially important, it may be appropriate to use more elabor-
ate strategies to permit blinding, such as the use of a ‘double
dummy’ where there is a placebo for both dosage forms. In
this case patients are randomized to active tablets plus placebo
infusion or to active infusion plus placebo tablets.
WITHDRAWALS
The number of patients who are withdrawn from each treat-
ment and the reason for withdrawal (subjective, objective or
logistic) must be taken into account. For example, if in an anti-
hypertensive study comparing two treatments administered
for three months only the data from those who completed three
months of therapy with treatment X or Yare analysed, this may
suggest that both treatments were equally effective. However,
if 50% of the patients on treatment X withdrew after one week
because of lack of efficacy, that conclusion is erroneous. Again,
if patients are withdrawn after randomization but before dos-
ing, this can lead to unrecognized bias if more patients in one
group die before treatment is started than in the other group,
leading to one group containing a higher proportion of fitter
‘survivors’. Conversely, if patients are withdrawn after ran-
domization but before dosing, adverse events cannot be attrib-
uted to the drug. Hence both an ‘intention-to-treat’ analysis
and a ‘treatment-received’ analysis should be presented.
PLACEBO
If a placebo control is ethical and practical, this simplifies
interpretation of trial data and enables efficacy to be deter-
mined more easily (and with much smaller numbers of sub-
jects) than if an effective active comparator is current standard
treatment (and hence ethically essential). It is well recognized
that placebo treatment can have marked effects (e.g. lowering
of blood pressure). This is partly due to patient familiarization
with study procedures, whose effect can be minimized by a
placebo ‘run-in’ phase.
TRIAL DESIGN
There is no one perfect design for comparing treatments.
Studies should be prospective, randomized, double-blind and
placebo-controlled whenever possible. Parallel-group studies
are those in which patients are randomized to receive different
treatments. Although tempting, the use of historical data as a
control is often misleading and should only be employed in
exceptional circumstances. Usually one of the treatments is
the standard, established treatment of choice, i.e. the control,
whilst the other is an alternative – often a new treatment which
is a potential advance. In chronic stable diseases, a crossover
design in which each subject acts as his or her own control can
be employed. Intra-individual variability in response is usu-
ally much less than inter-individual variability. The treatment
sequence must be evenly balanced to avoid order effects and
there must be adequate ‘washout’ to prevent a carry-over
effect from the first treatment. This design is theoretically
more ‘economical’ in subject numbers, but is often not appli-
cable in practice.
STATISTICS
It is important to discuss the design and sample size of any
clinical trial with a statistician at the planning phase.
Research papers often quote Pvalues as a measure of whether
or not an observed difference is ‘significant’. Conventionally,
the null hypothesis is often rejected if P0.05 (i.e. a difference
of the magnitude observed would be expected to occur by
chance in less than one in 20 trials – so-called type I error, see
Figure 15.2). This is of limited value, as a clinically important
difference may be missed if the sample size is too small (type II
error, see Figure 15.2). To place reliance on a negative result,
the statistical power of the study should be at least 0.8 and
preferably 0.9 (i.e. a true difference of the magnitude pre-
specified would be missed in 20% or 10% of such trials,
respectively). It is possible to calculate the number of patients
required to establish a given difference between treatments at
a specified level of statistical confidence. For a continuous
variable, one needs an estimate of the mean and standard
deviation which one would expect in the control group. This is
usually available from historical data, but a pilot study may be
necessary. The degree of uncertainty surrounding observed
CLINICAL DRUG DEVELOPMENT 89
differences should be reported as confidence intervals (usu-
ally 95% confidence intervals). Such intervals will diminish as
the sample size is increased. Confidence intervals reflect the
effects of sampling variability on the precision of a procedure,
and it is important to quote them when a ‘non-significant’
result is obtained, and when comparing different estimates of
effectiveness (e.g. drug A in one trial may have performed
twice as well as placebo, whereas drug B in another trial may
have performed only 1.5 times as well as placebo; whether
drug Ais probably superior to drug B will be apparent from
inspection of the two sets of confidence intervals).
If many parameters are analysed, some apparently ‘signifi-
cant’ differences will be identified by chance. For example, if
100 parameters are analysed in a comparison of two treat-
ments, one would expect to see a ‘significant’ difference in
approximately five of those parameters. It is therefore very
important to prespecify the primary trial end-point and sec-
ondary end-points that will be analysed. Statistical corrections
can be applied to allow for the number of comparisons made.
One must also consider the clinical importance of any statistic-
ally significant result. For example, a drug may cause a statis-
tically significant decrease in blood pressure in a study, but if
it is only 0.2mmHg it is not of any clinical relevance.
CLINICAL DRUG DEVELOPM ENT
For most new drugs, the development process – following a
satisfactory preclinical safety evaluation – proceeds through
four distinct phases. These are summarized below. Figure 15.3
illustrates the overall decision-making process for determin-
ing whether or not a new therapy will be clinically useful.
PHASE I
The initial studies of drugs in humans usually involve healthy
male volunteers unless toxicity is predictable (e.g. cytotoxic
agents, murine monoclonal antibodies). The first dose to be
administered to humans is usually a fraction of the dose that
produced any effect in the most sensitive animal species
tested. Subjective adverse events, clinical signs, haematology,
biochemistry, urinalysis and electrocardiography are used to
assess tolerability. Depending on the preclinical data, further,
more specific evaluations may be appropriate. The studies are
placebo controlled to reduce the influence of environment and
normal variability. If the dose is well tolerated, a higher dose
will be administered either to a different subject in a parallel
design, or to the same group in an incremented crossover
design.
This process is repeated until some predefined end-point
such as a particular plasma concentration, a pharmacody-
namic effect or maximum tolerated dose is reached. Data from
the single-dose study will determine appropriate doses and
dose intervals for subsequent multiple-dose studies. If the
drug is administered by mouth, a food interaction study
should be conducted before multiple-dose studies.
The multiple-dose study provides further opportunity for
pharmacodynamic assessments, which may demonstrate a
desired pharmacological effect and are often crucial for the
selection of doses for phase II. Having established the dose
range that is well tolerated by healthy subjects, and in some
cases identified doses that produce the desired pharmacol-
ogical effect, the phase II studies are initiated.
Key points
Phase I studies:
initial exposure of humans to investigational drug;
assessment of tolerance, pharmacokinetics and
pharmacodynamics in healthy subjects or patients;
usually healthy male volunteers;
usually single site;
40–100 subjects in total.
PHASE II
Phase II studies are usually conducted in a small number of
patients by specialists in the appropriate area to explore efficacy,
tolerance and the dose–response relationship. If it is ethical and
practicable, a double-blind design is used, employing either a
placebo control or a standard reference drug therapy as con-
trol. These are the first studies in the target population, and it
is possible that drug effects, including adverse drug reactions
and pharmacokinetics, may be different to those observed in
the healthy subjects. If the exploratory phase II studies are
promising, larger phase III studies are instigated, using a
dosage regimen defined on the basis of the phase II studies.
Key points
Phase II studies:
initial assessment of tolerance in ‘target’ population;
initial assessment of efficacy;
identification of doses for phase III studies;
well controlled with a narrowly defined patient
population;
100–300 patients in total;
usually double-blind, randomized and controlled.
PHASE III
Phase III is the phase of large-scale formal clinical trials in which
the efficacy and tolerability of the new drug is established.
Too many statistical
comparisons performed
Too small sample size
(insufficient power)
False-positive result
(significant difference found
when no difference present)
False-negative result
(no significant difference found
when difference present)
TYPE II ERRORTYPE I ERROR
Figure 15.2:Different types of statistical error.
90 INTRODUCTIONOF NEW DRUGS AND CLINICAL TRIALS
Patient groups who respond more or less well may be identi-
fied, patient exposure (both numbers and duration of therapy)
is increased, and less common type B (see Chapter 12) adverse
reactions may be identified. During this period, the manufac-
turers will be setting up plant for large-scale manufacture and
undertaking further pharmaceutical studies on drug formula-
tion, bioavailability and stability. The medical advisers to the
company, in association with their pharmacological, pharma-
ceutical and legal colleagues, will begin to collate the large
amount of data necessary to make formal application to the
MHRA or EMEA for a product licence. Marketing approval
may be general or granted subject to certain limitations which
may include restriction to hospital practice only, restriction in
indications for use, or a requirement to monitor some particular
action or organ function in a specified number of patients.
Doctors are reminded (by means of a black triangle symbol
beside its entry in the British National Formulary) that this is
a recently introduced drug, and that any suspected adverse
reaction should be reported to the MHRAor Commission on
Human Medicines.
and may also help in the detection of previously unrecognized
adverse events (see Chapter 12).
Does the therapy have
unacceptable adverse effects?
Yes
Yes
Yes
Yes
No
No
No
No
Does the therapy exhibit
potentially useful clinical effects?
Are the benefits statistically significant
over existing therapy/placebos in
well-designed clinical trials?
Are these benefits of useful
magnitude?
Not usefulUseful
Figure 15.3:Flow chart for deciding
usefulness of a new therapy.
Key points
Phase III studies:
confirmation of effective doses;
expanded tolerability profile;
collection of data on a more varied patient population
with indication;
data on overall benefit/risk;
can be placebo or more usually active controls;
multicentre;
commonly 1000–5000 patients in total;
usually double-blind.
PHASE IV
Phase IV studies are prospective trials performed after mar-
keting approval (the granting of a product licence). These may
assess the drug’s clinical effectiveness in a wider population
Key points
Phase IV studies:
performed after marketing approval and related to the
approved indications;
exposure of drug to a wider population;
different formulations, dosages, duration of treatment,
drug interactions and other drug comparisons are
studied;
detection and definition of previously unknown or
inadequately quantified adverse events and related risk
factors.
POSTMARKETING SURVEILLANCE
The MHRAclosely monitors newly licensed drugs for adverse
events through the yellow card reporting system (see Chapter
12). Direct reporting by patients of adverse events was intro-
duced in 2004. SAMM (Safety Assessment of Marketed
Medicines) studies may be initiated which can involve many
thousands of patients.
GENERIC DRUGS
Once the patent life of a drug has expired, anyone may manu-
facture and sell their version of that drug. The generic drug
producer does not have to perform any of the research and
development process other than to demonstrate that their ver-
sion of the drug is ‘bioequivalent’ to the standard formulation.
The convention accepted for such ‘bioequivalence’ is gener-
ous, and the issue is the subject of current debate by biostatis-
ticians. In practice, the essential point is that clinically
untoward consequences should not ensue if one preparation
is substituted for the other.
GLOBALIZATION 91
ETHICS COMMITTEES
Protocols for all clinical trials must be reviewed and approved
by a properly constituted independent ethics committee.
Research Ethics Committees are coordinated by NRES
(National Research Ethics Services) working on behalf of the
Department of Health in the UK.
NRES maintains a UK-wide system of ethical review that
protects the safety, dignity and well-being of research partici-
pants whilst facilitating and promoting research within the
NHS.
Interestingly, studies have shown that patients taking part
in clinical trials often have better health outcomes than those
not involved in a trial.
GLOBALIZATION
In order to facilitate world-wide drug development and
encourage good standards of practice, a series of international
conferences on harmonization of requirements for registration
of pharmaceuticals for human use have been conducted.
International Conferences on Harmonisation (ICH) are lead-
ing to a globally accepted system of drug development,
hopefully without stifling research with excessive bureaucracy
FURTHER READING
Collier J (ed.). Drug and therapeutics bulletin; from trial outcomes to clin-
ical practice. London: Which? Ltd, 1996.
Griffin JP, O’Grady J (eds). The textbook of pharmaceutical medicine, 5th
edn. London: BMJ Books, 2005.
Wilkins MR (ed.). Experimental therapeutics. Section 1: Drug discovery
and development. London: Martin Dunitz, 2003.
Case history
Rather than a clinical case history, consider a chapter in the
history of drug regulation which is instructive in illustrating
the value of toxicity testing. Triparanol is a drug that
lowers the concentration of cholesterol in plasma. It was
marketed in the USA in 1959. In 1962, the Food and Drug
Administration (FDA) received a tip-off and undertook an
unannounced inspection. This revealed that toxicology
data demonstrating cataract formation in rats and dogs
had been falsified. Triparanol was withdrawn, but some of
the patients who had been taking it for a year or longer
also developed cataracts.
and without any lowering of standards. The goal is to facili-
tate the early introduction of valuable new therapies, while at
the same time maximizing patient protection.
CHAPTER 16
CELL-BASED AN D RECOMBINANT
DNA THERAPIES
Gene therapy 94
Human stem cell therapy 95
The term ‘biotechnology’ encompasses the application of
advances in our knowledge of cell and molecular biology since
the discovery of DNA to the diagnosis and treatment of dis-
ease. Recent progress in molecular genetics, cell biology and
the human genome has assisted the discovery of the mecha-
nisms and potential therapies of disease. The identification of
a nucleotide sequence that has a particular function (e.g. pro-
duction of a protein), coupled with our ability to insert that
human nucleotide sequence into a bacterial or yeast chromo-
some and to extract from those organisms large quantities of
human proteins, has presented a whole array of new opportu-
nities in medicine. (Human gene sequences have also been
inserted into mice to develop murine models of human dis-
ease.) In 1982, the first recombinant pharmaceutical product,
human recombinant insulin, was marketed. Since then, more
than 100 medicines derived via biotechnology have been
licensed for use in patients, whilst hundreds more are cur-
rently undergoing clinical trials. Successes include hormones,
coagulation factors, enzymes and monoclonal antibodies,
extending the range of useful therapeutic agents from low
molecular weight chemical entities to macromolecules. Once
discovered, some biotechnology products are manufactured
by chemical synthesis rather than by biological processes.
Examples of recombinant products are listed in Table 16.1. In
parallel with these advances, the human genome project is
establishing associations between specific genes and specific
diseases. Detailed medical histories and genetic information
are being collected and collated from large population sam-
ples. This will identify not only who is at risk of a potential dis-
ease and may thus benefit from prophylactic therapy, but also
who may be at risk of particular side effects of certain drugs.
This carries potentially momentous implications for selecting
the right drug for the individual patient – a ‘holy grail’ known
as personalized medicine. Achieving this grail is not immi-
nent. It is not just the physical presence but, more importantly,
the expression of a gene that is relevant. Often a complex inter-
action between many genes and the environment gives rise to
disease. Despite these complexities, the human genome proj-
ect linked with products of recombinant DNA technology,
including gene therapy, offers unprecedented opportunities
for the treatment of disease.
Most recombinant proteins are not orally bioavailable, due
to the efficiency of the human digestive system. However, the
ability to use bacteria to modify proteins systematically may
aid the identification of orally bioavailable peptides. Nucleic
acids for gene therapy (see below) are also inactive when
administered by mouth. Drug delivery for such molecules is
very specialized and at present consists mainly of incorporat-
ing the gene in a virus which acts as a vector, delivering the
DNAinto the host cell for incorporation into the host genome
and subsequent transcription and translation by the cellular
machinery of the host cell.
Human proteins from transgenic animals and bacteria are
used to treat diseases that are caused by the absence or
impaired function of particular proteins. Before gene cloning
permitted the synthesis of these human proteins in large
quantities, their only source was human tissues or body flu-
ids, carrying an inherent risk of viral (e.g. hepatitis B and C
and HIV) or prion infections. An example in which protein
replacement is life-saving is the treatment of Gaucher’s dis-
ease, a lysosomal storage disease, which is caused by an
inborn error of metabolism inherited as an autosomal reces-
sive trait, which results in a deficiency of glucocerebrosidase,
which in turn results in the accumulation of glucosylceramide
in the lysosomes of the reticulo-endothelial system, particu-
larly the liver, bone marrow and spleen. This may result in
hepatosplenomegaly, anaemia and pathological fractures.
Originally, a modified form of the protein, namely alglucerase,
had to be extracted from human placental tissue. The deficient
enzyme is now produced by recombinant technology.
The production of recombinant factor VIII for the treatment
of haemophilia has eliminated the risk of blood-borne viral
infection. Likewise, the use of human recombinant growth
hormone has eliminated the risk of Creutzfeldt–Jakob disease
that was associated with human growth hormone extracted
from bulked cadaver-derived human pituitaries.
Recombinant technology is used to provide deficient pro-
teins (Table 16.1) and can also be used to introduce modifica-
tions of human molecules. In the human insulin analogue,
lispro insulin, produced using recombinant technology, the
order of just two amino acids is reversed in one chain of the
insulin molecule, resulting in a shorter duration of action than
CELL-BASED AND RECOMB INANT DNA THERAPIES 93
soluble insulin, a real advance for some patients (see
Chapter 37). Other ‘designer’ insulins have longer actions or
other kinetic features that are advantageous in specific
circumstances.
In addition to producing recombinant human hormones
(see Table 16.2) and other recombinant proteins (e.g. hirudin,
the anticoagulant protein of the leech), recombinant mono-
clonal antibodies for treating human diseases have been pro-
duced originally in immortalized clones of mouse plasma
cells. Not surprisingly, the original murine antibodies induced
antibody responses in humans which in turn caused disease
or neutralizing antibodies, rendering the monoclonal antibod-
ies ineffective if used repeatedly (Table 16.3). Immunoglobulins
have been gradually humanized to reduce the risk of an
immune response on repeated treatments.
In cancer therapy, monoclonal antibodies have been devel-
oped against a tumour-associated antigen, e.g. trastuzumab
against the HER2 protein (over-expressed in certain breast
cancers in particular). Most facilitate the body’s immune sys-
tem in destroying the cancer cells or reduce the blood supply
to the tumour. Abciximab (see Chapter 30) inhibits platelet
aggregation by blocking the glycoprotein receptor that is a key
convergence point in different pathways of platelet aggrega-
tion. It is used as an adjunct to heparin and aspirin for the
prevention of ischaemic complications in high-risk patients
undergoing percutaneous coronary intervention. It is a
murine monoclonal antibody and can only be used in an indi-
vidual patient once. Most recently developed monoclonal
antibodies have been fully humanized. In comparison to most
conventional ‘small molecule’ drugs, the antibodies’ activities
are very specific and toxicity is usually directly related to the
targeted effect either through excessive effect or a ‘down-
stream’ consequence of the effect. The effects are usually very
species-specific, so extrapolation from animal studies is more
difficult. The initial doses in humans should be a fraction of
the minimum anticipated biological effect level (MABEL, see
Figure 16.1) taking into account concentration, receptor occu-
pancy, relative potency, likely dose–response curve, and
effects of excessive pharmacology rather than just the ‘no
observable adverse effect level’ (NOAEL) which is the main-
stay of first dose calculation for conventional small molecule
drugs.
Recombinant techniques have also been of value in the
development of vaccines, thereby avoiding the use of intact
virus. Suspensions of hepatitis B surface antigen prepared
from yeast cells by recombinant DNAtechniques are already
widely used to prevent hepatitis B infection in high-risk
groups in the UK. In comparison to traditional egg-based and
cell-based vaccines, DNAvaccines using plasmid DNA coding
for specific epitopes of influenza virus may be developed,
manufactured and distributed much more rapidly and effec-
tively. With the current likelihood of an influenza pandemic
caused by a new strain of virus predicted by the World Health
Organization (WHO), the ability to produce such DNA vac-
cines may save millions of lives.
Table 16.1: Recombinant proteins/enzymes licensed in the UK (examples)
Protein/enzyme Indication
Recombinant coagulation factors VIII Haemophilia
and VIIa
Imiglucerase Gaucher’s disease
Interferon alfa Hepatitis B and C, certain lymphomas and
solid tumours
Interferon beta Multiple sclerosis
Epoetin alfa and beta (recombinant human Anaemia of chronic renal failure. To increase
erythropoietin) yield of autologous blood, e.g. during
cancer chemotherapy
Drotrecogin alfa (activated) (recombinant Severe sepsis
activated protein Cwhich reduces
microvascular dysfunction)
Table 16.2:Hormones/hormone antagonists (examples)
Mode of action Indication
Somatropin Synthetic human Growth hormone
growth hormone (hGH) deficiency
Pegvisomant Genetically modified Acromegaly
hGH that blocks hGH
receptors
Follitropin Recombinant human Infertility
alfa and follicle stimulating
beta hormone
Insulin aspart, Recombinant human Diabetes (helps
glulisine and insulin analogues, glucose control in
lispro faster onset of action some patients/
situations)
94 CELL-BASEDAND RECOMB INANT DNA THERAPIES
Table 16.4:Prevalence of some genetic disorders which result from a defect
in a single gene
Disorder Estimated prevalence
Familial hypercholesterolaemia 1 in 500
Polycystic kidney disease 1 in 1250
Cystic fibrosis 1 in 2000
Huntington’s chorea 1 in 2500
Hereditary spherocytosis 1 in 5000
Duchenne muscular dystrophy 1 in 7000
Haemophilia 1 in 10000
Phenylketonuria 1 in 12000
GENE THERAPY
The increasing potential to exploit advances in genetics and
biotechnology raises the possibility of prevention by gene
therapy both of some relatively common diseases which are
currently reliant on symptomatic drug therapy, and of genetic
disorders for which there is currently no satisfactory treat-
ment, let alone cure.
Gene therapy is the deliberate insertion of genes into
human cells for therapeutic purposes. Potentially, gene ther-
apy may involve the deliberate modification of the genetic
material of either somatic or germ-line cells. Germ-line
genotherapy by the introduction of a normal gene and/or
deletion of the abnormal gene in germ cells (sperm, egg or
zygote) has the potential to correct the genetic defect in many
devastating inherited diseases and to be subsequently trans-
mitted in Mendelian fashion from one generation to the next.
The prevalence figures for inherited diseases in which a single
gene is the major factor are listed in Table 16.4. However,
germ-line gene therapy is prohibited at present because of the
unknown possible consequences and hazards, not only to the
individual but also to future generations. Thus, currently,
gene therapy only involves the introduction of genes into
human somatic cells. Whereas gene therapy research was ini-
tially mainly directed at single-gene disorders, most of the
research currently in progress is on malignant disease. Gene
therapy trials in cancer usually involve destruction of tumour
cells by the insertion of a gene that causes protein expression
that induces an immune response against those cells, or by the
introduction of ‘suicide genes’ into tumour cells.
Cystic fibrosis (CF) is the most common life-shortening
autosomal-recessive disease in Europeans. It is caused by a
mutation in the cystic fibrosis transmembrane conductance
regulator (CFTR) gene. Over 600 different CF mutations
have been recognized, although one mutation (F508) is pres-
ent on over 70% of CF chromosomes. Phase I studies using
adenoviral or liposomal vectors to deliver the normal CFTR
gene to the airway epithelium have shown that gene transfer
is feasible, but with current methods is only transient in
Table 16.3:Licensed monoclonal antibodies (examples)
Monoclonal antibody Mode of action Indication
Abciximab Inhibits glycoprotein IIb/IIIa, platelet Angioplasty
aggregation
Omalizumab Anti-IgE Prophylaxis of severe allergic asthma
Infliximab, Adalimumab Anti-TNF
α Rheumatoid arthritis, psoriatic arthritis
Basiliximab, Daclizumab Bind to IL-2R
αreceptor on T cells, prevent Prophylaxis of acute rejection in
T-cell proliferation, causing allogenic renal transplantation
immunosuppression
Bevacizumab (Avastin®) Inhibits vascular endothelial growth factor Metastatic colorectal cancer
(VEGF), hence inhibits angiogenesis
Pegaptanib and ranibizumab lnhibit VEGF Neovascular age-related macular
degeneration
100
MABEL
Therapeutic
range
Dose or exposure
Unacceptable
toxicity
80
60
Effect
40
20
0
10 100 1000 1000
0
Figure 16.1:Explanation of minimum anticipated biological
effect level (MABEL) (kindly provided by P Lloyd, Novartis, Basel,
Switzerland). Unbroken line, desired effect; dashed line,
undesired effect.
HUMAN STEM CELL TH ERAPY 95
duration and benefit. Adenoviral vectors are more efficient
than liposomes but themselves cause serious inflammatory
reactions.
Adramatic example of the potential benefit and danger of
gene therapy has been seen in the treatment of severe com-
bined immunodeficiency (SCID) secondary to adenosine
deaminase deficiency by reinfusing genetically corrected
autologous T cells into affected children. Whilst the gene ther-
apy was effective in the immunological reconstitution of the
patients, allowing a normal life including socializing with
other children rather than living in an isolation ‘bubble’, T-cell
leukaemia has developed in some patients. This probably
reflects problems with the retrovirus vector.
Asuccess in gene therapy has occurred with recipients of
allogenic bone marrow transplants with recurrent malignan-
cies. T cells from the original bone marrow donor can mediate
regression of the malignancy, but can then potentially damage
normal host tissues. Asuicide gene was introduced into the
donor T cells, rendering them susceptible to ganciclovir
before they were infused into the patients, so that they could
be eliminated after the tumours had regressed and so avoid
future damage to normal tissues.
From the above, it will be appreciated that a major problem
in gene therapy is introducing the gene into human cells. In
some applications, ‘gene-gun’ injection of ‘naked’ (i.e. not
incorporated in a vector) plasmid DNA may be sufficient.
Minute metal (e.g. gold) particles coated with DNAare ‘shot’
into tissues using gas pressure (Figure 16.2). Some DNAis rec-
ognized as foreign by a minority of cells, and this may be suf-
ficient to induce an immune response. This method underpins
DNAvaccines. The other major problem is that for most dis-
eases it is not enough simply to replace a defective protein, it
is also necessary to control the expression of the inserted gene.
It is for reasons such as these that gene therapy has been
slower in finding clinical applications than had been hoped,
but the long-term prospects remain bright.
Despite the inherent problems of gene therapy and societal
concerns as to how information from the genotyping of indi-
viduals will be used, the development of gene therapy has
dramatic potential – not only for the replacement of defective
genes in disabling diseases such as cystic fibrosis, Duchenne
muscular dystrophy and Friedreich’s ataxia, but also for the
treatment of malignant disease, and for prevention of cardio-
vascular disease and other diseases for which there is a genetic
predisposition or critical protein target.
Another gene-modulating therapy that is currently
being evaluated is the role of anti-sense oligonucleotides.
These are nucleotides (approximately 20mers in length)
whose sequence is complementary to part of the mRNAof the
gene of interest. When the anti-sense enters cells it binds to the
complementary sequence, forming a short piece of double-
stranded DNA that is then degraded by RNase enzymes,
thus inhibiting gene expression. Examples of such agents in
development or near approval include fomiversen, which
binds to cytomegalovirus (CMV) RNA(used intraocularly for
CMV infection) and anti-Bcl-2, used to enhance apoptosis in
lymphoma cells.
HUMAN STEM CELL THERAPY
The discovery of stem cells’ ability to replace damaged cells
has led to much interest in cell-based therapies. Stem cells
retain the potential to differentiate, for example into cardiac
muscle cells or pancreatic insulin-producing cells, under
particular physiological conditions.
In the UK, stem cell therapy is already established in the
treatment of certain leukaemias and has also been used suc-
cessfully in skin grafting, certain immune system and corneal
disorders. Autologous and allogenic haemopoietic stem cells
collected from bone marrow or via leukophoresis from
peripheral blood following granulocyte colony-stimulating
factor (G-CSF) stimulation (see Chapter 49) have been used
for some years in the management of certain leukaemias.
Allogenic stem cell transplantation is associated with graft-
versus-host disease, hence concomitant immunosuppressant
treatment with prophylactic anti-infective treatment includ-
ing anti-T-cell antibodies is required. Graft-versus-host
disease and opportunistic infections remain the principal
complications.
Non-myeloblastic allogenic stem cell transplantation is
being increasingly used, particularly in the elderly. This has
an additional benefit from a graft-versus-tumour effect as
immunosuppression is less severe.
Although there has been much publicity over the potential
of stem cell regenerative and reparative effects in chronic
central nervous system disorders, such as Parkinson’s disease,
Alzheimer’s disease, motor neurone disease and multiple
sclerosis, to date there is no convincing evidence of benefit
for these conditions. There is ongoing ethical debate over
the use of embryonic stem cells, which have more therapeutic
Figure 16.2:Particle-mediated epidermal delivery (PMED) of DNA
into an antigen presenting cell (APC). The DNA elutes from the
gold particle and enters the nucleus where it is transcribed into
mRNA. The mRNA is then translated using the cellular synthetic
pathways to produce the encoded protein of interest. This
intracellular foreign protein is then processed by proteasomes
into small antigenic peptides that are presented on the cell
surface by the major histocompatibility complex (MHC).
96 CELL-BASEDAND RECOMB INANT DNA THERAPIES
potential than adult stem cells for research and possible
therapy.
The Gene Therapy Advisory Committee (GTAC) is the
national research ethics committee for gene therapy clinical
research. GTAC’s definition of gene therapy is as follows: ‘The
deliberate introduction of genetic material into human
somatic cells for therapeutic, prophylactic or diagnostic pur-
poses.’ This definition, and hence the remit of GTAC, encom-
passes techniques for delivering synthetic or recombinant
nucleic acids into humans:
genetically modified biological vectors, such as viruses or
plasmids;
genetically modified stem cells;
oncolytic viruses;
nucleic acids associated with delivery vehicles;
naked nucleic acids;
antisense techniques (for example, gene silencing, gene
correction or gene modification);
genetic vaccines;
DNAor RNA technologies, such as RNA interference;
xenotransplantation of animal cells, but not solid organs.
FURTHER READING
Anon. Understanding monoclonal antibodies. Drugs and Therapeutics
Bulletin2007; 45.
Anson DS. The use of retroviral vectors for gene therapy – what are
the risks? Areview of retroviral pathogenesis and its relevance to
retroviral vector-mediated gene delivery. Genetic Vaccines and
Therapy2004; 2: 9.
Check E. Atragic setback. Nature 2002; 420: 116–18.
Guttmacher AE, Collins FS. Genomic medicine a primer. New England
Journal of Medicine2002; 347: 1512–20.
Marshall E. Gene therapy death prompts review of adenovirus vector.
Science1999; 286: 2244–5.
Nathwani AC, Davidoff AM, Linch DC. Areview of gene therapy for
haematological disorders. British Journal of Haematology2005; 128:
3–17.
Rang HP, Dale MM, Ritter JM, Flower RJ. Chapter 55
Biopharmaceuticals and gene therapy. In: Pharmacology, 6th edn.
Oxford: Elsevier, 2007.
Safer medicines. Areport from the Academy. London: The Academy of
Medical Sciences, 2005.
Walsh G. Second-generation biopharmaceuticals. European Journal of
Pharmaceutics and Biopharmaceutics2004; 58: 185–96.
Introduction 97
Garlic 97
Ginseng 98
Ginkgo biloba 99
Echinacea 99
Soy 99
Saw palmetto 100
St John’s wort 100
Glucosamine 101
Miscellaneous herbs recently found to be toxic or
meriting their withdrawal from the market 101
CHAPTER 17
ALTERNATIVE MEDICINES: HERBALS
AND N UTRACEUTICALS
INTRODUCTION
‘Alternative’ therapies (i.e. alternative to licensed products of
proven quality, safety and efficacy) span a huge range from
frank charlatanry (e.g. products based on unscientific postu-
lates, composed of diluent or of snake oil), through physical
therapies such as massage and aroma therapies which certainly
please (‘placebo’ means ‘I will please’) and do a great deal less
harm than some conventional therapies (e.g. surgery, chemo-
therapy), through to herbal medications with undoubted
pharmacological activity and the potential to cause desired or
adverse effects, albeit less predictably than the licensed prod-
ucts that have been derived from them in the past and will no
doubt be so derived in the future. Medicine takes an empirical,
evidence-based view of therapeutics and, if supported by suffi-
ciently convincing evidence, alternative therapies can enter the
mainstream of licensed products. Overall, efforts to test homeo-
pathic products have been negative (Ernst, 2002) and it has been
argued that no more resource should be wasted on testing prod-
ucts on the lunatic fringe, even when they come with royal
endorsement and (disgracefully) public funding. Here we focus
on herbal and nutraceutical products that may cause pharmaco-
logical effects.
Herbal remedies include dietary supplements (any product
other than tobacco intended for ingestion as a supplement to
the diet, including vitamins, minerals, anti-oxidants – Chapter
35 – and herbal products), phytomedicines (the use of plants or
plants components to achieve a therapeutic effect/outcome)
and botanical medicines (botanical supplements used as
medicine). The recent increase in the use of herbal remedies by
normal healthy humans, as well as patients, is likely to be mul-
tifactorial and related to: (1) patient dissatisfaction with con-
ventional medicine; (2) patient desire to take more control of
their medical treatment; and (3) philosophical/cultural bias. In
the USA, approximately one-third of the population used some
form of complementary or alternative medicine (the majority
consuming herbal products) in the past 12 months. At a clinical
therapeutic level, it is disconcerting that 15–20 million
Americans regularly take herbal remedies, while concomi-
tantly receiving modern prescription drugs, implying a signif-
icant risk for herb–drug interactions. In Scotland, some 12% of
general practitioners and 60% of general practices prescribe
homeopathic medicines! Herbal remedies are particularly
used by certain groups of patients, notably HIV and cancer
patients. The stereotypical user is a well-educated, career pro-
fessional, white female. From a therapeutic perspective, many
concerns arise from the easy and widespread availability,
lack of manufacturing or regulatory oversight, potential
adulteration and contamination of these herbal products.
Furthermore, there is often little or no rigorous clinical trial
evidence for efficacy and only anecdotes about toxicity. Many
patients who are highly attuned to potential harms of conven-
tional drugs (such as digoxin, a high quality drug derived his-
torically from extracts of dried foxglove of variable quality
and potency) fail to recognize that current herbals have as
great or greater potential toxicities, often putting their faith in
the ‘naturalness’ of the herbal product as an assurance of
safety. This chapter briefly reviews the most commonly used
herbals (on the basis of sales, Table 17.1) from a therapeutic
perspective and addresses some of the recently identified
problems caused by these agents.
GARLIC
Garlic has been used as a culinary spice and medicinal herb for
thousands of years. One active compound in garlic is allicin,
and this is produced along with many additional sulphur
compounds by the action of the enzyme allinase when fresh
garlic is crushed or chewed. Initial clinical trials suggested the
potential of garlic to lower serum cholesterol and triglyceride,
but a recent trial has shown limited to no benefit. Garlic has
been advocated to treat many conditions, ranging from many
cardiovascular diseases, e.g. atherosclerosis including periph-
eral vascular disease, hypertension, lipid disorders and sickle
cell anaemia. Garlic can alter blood coagulability by decreas-
ing platelet aggregation and increasing fibrinolysis.
Adverse effects
The adverse effects of garlic use involve gastro-intestinal
symptoms including halitosis, dyspepsia, flatulence and
heartburn. Other reported adverse effects include headache,
haematoma and contact dermatitis.
Drug interactions
Garlic inhibits many drug-metabolizing (CYP450) enzymes in
vitro, but induces CYP450s when administered chronically in
vivo (reminiscent of many anticonvulsant drugs – Chapter 22 –
as well as ethanol). Clinical studies using probe-drug cocktails
have shown that garlic has no significant effect on the activity
of CYP1A2 (caffeine), CYP2D6 (debrisoquine, dextromethor-
phan) and CYP3A4 (alprazolam,midazolam). Clinical studies
suggest that garlic significantly decreases the bioavailability of
saquinavirand ritonavir. These HIV protease inhibitors are not
only metabolized by CYP3A4, but are also substrates for P-gly-
coprotein. The clinical importance of these interactions is uncer-
tain, but potentially appreciable.
GINSENG
There are several types of ginseng (Siberian, Asian, American
and Japanese), the most common type used in herbal prepara-
tions being the Asian variety (Panax ginseng). In humans,
ginseng has been suggested to be a sedative-hypnotic, an
aphrodisiac, an antidepressant and a diuretic, and therapeutic
benefits have been claimed for many indications (see below).
Its pharmacologic properties include actions as a phytoestro-
gen, suggesting that its use, as with soy supplementation,
could be disadvantageous in women with oestrogen-sensitive
cancers (e.g. breast or endometrium). The active component
of ginseng, ginsenoside, inhibits cAMP phosphodiesterase
and monamine oxidase. These properties may partly explain
purported central nervous system (CNS) stimulant actions
of ginseng (though not sedative/hypnotic effects), potential
modulation of the immune system and increase of glycogen
storage. However, possible efficacy of ginseng in improving
physical or psychomotor performance, cognitive function,
immune function, diabetes mellitus and herpes simplex type 2
infections is not established beyond reasonable doubt.
Adverse effects
The adverse effects of ginseng are primarily CNS effects –
agitation, irritability, insomnia and headache. Others noted
include hypertension and mastalgia.
Drug interactions
In vitro evidence suggests that ginseng extracts inhibit CYP3A4
in human hepatocytes. These in vitro data are consistent with
study data during an 18-day course of ginseng where it signifi-
cantly increased the peak plasma concentration of nifedipine, a
CYP3A4 substrate, in healthy volunteers. As with other herbs
(e.g. echinacea), substantial variability in ginsenoside content
has been reported among commercially available ginseng
preparations, indicating that clinically significant effects on the
pharmacokinetics of drugs that are metabolized by CYP3A4
could be highly variable between batches.
98 ALTERNATIVEMEDICINES: HE RBALS AND N UTRACEUTICALS
Table 17.1: Most commonly used herbal products based on dollar sales
Product Plant Intended condition to Annual sales in
be used for USA ($ millions)
Garlic Allium sativum Hyperlipidaemia– 34.5
hypercholesterolaemia
Ginkgo Ginkgo biloba Dementia and claudication 33.0
Echinacea Echinacea purpurea Prevention of common cold 32.5
Soy Glycine max Symptoms of menopause 28.0
Saw palmetto Serenoa repens Prostatic hypertrophy 23.0
Ginseng Panax ginseng Fatigue 22.0
St John’s wort Hypericum perforatum Depression (mild) 15.0
Black cohosh Actaea racemosa Menopausal symptoms 12.3
Cranberry Vaccinia macrocarpon Cystitis and UTI 12.0
Valerian Valeriana officinalis Stress and sleeplessness 8.0
Milk thistle Silybum marianum Hepatitis and cirrhosis 7.5
Evening primrose Oenothera biennis Premenstrual symptoms 6.0
Bilberry Vaccinia myrtillus Diabetic retinopathy 3.5
Grape seed Vitis vinifera Allergic rhinitis 3
UTI: urinary tract infection
Acase report has suggested a possible interaction between
ginseng consumption and warfarin, but animal studies do not
support this.
GINKGO BILOBA
Originating from Chinese medicine, ginkgo (derived from the
nuts of Ginkgo biloba – a beautiful and threatened tree rather
than the western culinary stereotype of a ‘herb’) is used for a
variety of ailments and has multiple purported actions,
including antihypoxic, antioxidant, antiplatelet, free radical-
scavenging and microcirculatory properties. It has been used
in patients with asthma, brain trauma, cochlear deafness,
depression, retinitis, impotence, myocardial reperfusion and
vertigo. The evidence for efficacy in many of these conditions
is unconvincing. A recent clinical trial, in which a leading
ginkgo extract did not improve cognitive function, may have
contributed to a decline of ginkgo from the top-selling pos-
ition it had held among such products since 1995. One of the
principal components of ginkgo, ginkgolide B, is a moderately
potent antagonist of platelet-activating factor. ‘Anti-stress’
effects claimed for ginkgo products are postulated to be due to
monamine oxidase inhibition by ginkgolides.
Adverse effects
Serious or fatal side effects of gingko include spontaneous
bleeding, fatal intracerebral bleeding, seizures and anaphyl-
actic shock. Less serious side effects are nausea, vomiting, flat-
ulence, diarrhoea, headaches and pruritus.
Drug interactions
In vitro data suggest ginkgo can inhibit hepatic drug metab-
olizing enzymes. Long-term administration of ginkgo to volun-
teers(for up to 28 days) had no effect on the pharmacokinetics
ofmidazolam, a marker of CYP3A4 activity. In another study,
however, ginkgo increased the plasma concentrations of the
CYP3A4 substrate nifedipineby 53%, confirming the potential
for enzyme inhibition observed in vitro. The discrepant find-
ings for effects of ginkgo on CYP3A4 observed in this trial and
in the phenotyping studies is possibly related to the highly
variable phytochemical composition of commercially available
ginkgo extracts. The potential importance of the change in
CYP2C19 activity noted previously in a cocktail screening
approach, was verified by the observation that ginkgo signifi-
cantly reduced the metabolism of omeprazole, a CYP2C19
substrate, in Chinese patients. Collectively, these clinical data
indicate that ginkgo may interfere with the pharmacokinetics
of drugs metabolized by CYP2C19 or CYP3A4. If it does inhibit
MAO at therapeutic doses, adverse interactions with tyramine-
containing foods and possibly with selective serotonin reup-
take inhibitors (SSRI) (Chapter 20) are to be anticipated.
ECHINACEA
Echinacea is one of the most commonly used alternative medi-
cines, representing 10% of the herbal market. There are nine
species of the genus Echinacea, a member of the sunflower fam-
ily, found in North America. The most common and widespread
of these are Echinacea angustifolia,E. purpurea and E. pallida, each
of which has a long history of medicinal use. The majority of
pharmacologic studies since 1939 have been conducted on
E. purpureapreparations made from the fresh pressed juice of the
flowering plant. Many chemical compounds have been identi-
fied from Echinacea species and it is currently not possible to
attribute the pharmacological effects to any specific substance.
Constituents that have been identified include volatile oil, caffeic
acid derivatives, polysaccharides, polyines, polyenes, isobuty-
lamides and flavonoids of the quercetin and kaempferol type.
Many studies of echinacea have pointed to effects on the immune
system. Proposed mechanisms of action include increased circu-
lating granulocytes, enhanced phagocytosis, inhibition of virus
proliferation, cytokine activation, increased T-lymphocyte pro-
duction and an increase in the CD4/CD8 T-cell ratio. Echinacea
is currently most widely used in attempts to prevent the com-
mon cold and influenza symptoms, but is also used for Candida
infections, chronic respiratory infections, prostatitis and rheuma-
toid arthritis. Well-controlled studies have shown little, if any,
benefit. One recent placebo-controlled study of echinacea in the
treatment of the common cold actually suggested echinacea did
not prevent people catching a ‘cold’ and if they did get symp-
toms they lasted slightly longer in patients taking echinacea.
Adverse effects
Adverse effects of echinacea use involve rashes, including
erythema multiforme, arthralgias, allergic reactions, gastro-
intestinal disturbances including dysgeusia, dyspepsia and
diarrhoea.
Drug interactions
Some flavonoids present in echinacea extracts can either
inhibit or activate human CYPs and drug transporters,
depending on their structures, concentrations and assay con-
ditions. Midazolam, a substrate for CYP3A4 and CYP3A5,
was cleared 42% faster during an eight-day echinacea treat-
ment in 12 volunteers and there was a 23% reduction in mida-
zolamarea under the curve (AUC). The oral bioavailability of
midazolam in this study was significantly increased from 24
to 36% in the presence of echinacea, indicating that the hepatic
and intestinal availabilities were altered in opposite direc-
tions. These data suggest that echinacea is likely to interact
with other oral drugs that are substrates for CYP3A4 and that
the interaction will depend on the relative extraction of drugs
at the hepatic and intestinal sites and the route of administra-
tion. Echinacea from retail stores often does not contain the
labelled species (a similar situation affects other herbal prepa-
rations). The high variability observed in concentration of
constituents of the herb has implications for echinacea’s abil-
ity to modulate drug absorption and disposition.
SOY
The use of soy (Glycine max) and soy-derived products for the
treatment of menopause in women is growing with the fear of
SOY 99
possible side effects of traditional hormone replacement ther-
apy. The principal constituents of soy, the isoflavones genis-
tein and daidzein, are structurally similar to 17α-oestradiol
and produce weak oestrogenic effects (i.e. they are phytoestro-
gens). It is prudent to discourage soy-derived products in
patients with oestrogen-dependent tumours (e.g. breast can-
cer or endometrial cancer) because experimental data indicate
that soy can stimulate the growth of these tumours in mice.
Furthermore, as genistein can negate the inhibitory effect of
tamoxifen on breast cancer growth, women taking this agent
should especially avoid soy. Acute vasodilatation caused by
17β-oestradiol is mediated by nitric oxide, and genistein
(which is selective for the oestrogen receptor ER
β
, as well as
having quite distinct effects attributable to tyrosine kinase
inhibition) is as potent as 17β-oestradiol in this regard, raising
the possibility of beneficial vascular effects.
Adverse reactions
Adverse reactions in soy use include allergic reactions (prur-
itus, rash, anaphylaxis) and gastro-intestinal disturbances
(nausea, dyspepsia, diarrhoea).
Drug interactions
Isoflavones, such as genistein and daidzein, also inhibit oxida-
tive and conjugative metabolism in vitro and in vivo. In 20
healthy volunteers, a 14-day course of soy extract (50mg twice a
day) did not alter the ratio of the amounts of 6β-hydroxycortisol
and cortisol excreted in the urine, suggesting that soy is not an
inducer of CYP3A4 in humans. However, genistein interacts
with transporters such as P-glycoprotein (MDR-1, ABCB1),
MRP1 (ABCC1) and MRP2 (ABCC2). Given that these trans-
porters are involved in the intestinal absorption and biliary secre-
tion of many drugs, it is reasonable to suspect that soy may alter
drug absorption and/or disposition of such agents in humans.
SAW PALMETTO
Saw palmetto (Serenoa repens) is derived from a tree native to
southeastern North America, particularly Florida. The main
constituents of saw palmetto include carbohydrates, fixed oils,
steroids, flavonoids, resin, tannin and volatile oil. Saw palmetto
is used in men with the hope of ‘toning and strengthening the
reproductive system, and specifically for symptoms of prostate
enlargement’. It has oestrogenic activity and reduces plasma
testosterone concentration. In women, the principal use of saw
palmetto is to (hopefully) reduce ovarian enlargement and to
increase the size of small breasts. Although no drug interactions
with, or medical contraindications to, the use of saw palmetto
have been reported, it would be prudent to avoid concomitant
use with other hormonal therapies, especially oestrogens, and
in patients with oestrogen-dependent cancers.
Adverse effects
The adverse effects of saw palmetto involve gastro-intestinal
intolerance, nausea and diarrhoea, hepatitis and cholestasis,
gynaecomastia and impotence.
ST JOHN’S WORT
St John’s wort (Hypericum perforatum, Figure 17.1), a perennial
plant native to Europe, North America and western Asia, is
one of the most extensively studied herbal products and many
of its uses are based on observations noted in early Greek and
Roman medicine. Currently, St John’s wort is still widely used
for the treatment of mild to moderate depression and other
nervous conditions. Reported cases and trials have shown
varying results of therapy with St John’s wort for depressive
and mood disorders. Ameta-analysis of trials in 1757 patients
concluded that treatment of depression with St John’s wort
was comparable to standard, prescription antidepressants and
superior to placebo. More recently, a randomized, double-
blind, placebo-controlled trial evaluating the safety and effi-
cacy of St John’s wort in the treatment of patients with major
depressive disorders revealed that St John’s wort was no more
effective than placebo.
St John’s wort extract is a very complex mixture of over
20 constituent compounds. These include catechin-type tannins
and condensed-type proanthocyanidins, flavonoids (mostly
hyperoside, rutin, quercetin and kaempferol), biflavonoids
(e.g. biapigenin), phloroglutinol derivatives like hyperforin,
phenolic acids, volatile oils and naphthodianthrones,
100 ALTERNATIVE MEDICINES: HERBALS AN D NUTRACEUTICALS
Figure 17.1: Drawing of perforate St John’s wort (Hypericum
perforatum). (© Natural History Museum, London. Reproduced
with permission.)
including hypericin and pseudohypericin. With regard to
the putative antidepressant effects of St John’s wort, the phar-
macological activities of hypericin and hyperforin, which
inhibit synaptic 5HT and catecholamine reuptake, could
contribute.
Adverse effects
Adverse CNS effects include headaches, drowsiness, restless-
ness, serotonin syndrome (Chapter 20) if used with SSRIs or
TCAs, skin photosensitivity. Gastro-intestinal disturbances
involve abdominal pain or discomfort, and xerostomia. Drug
interactions with therapeutic failure of concomitant drugs,
e.g. HIV protease inhibitors, ciclosporin, warfarin, theo-
phylline, antidepressants, oral contraceptives and anti-cancer
agents, such as irinotecan.
Drug interactions
Many clinical trials are now reporting significant pharmacoki-
netic interactions with long-term treatment with St John’s
wort and drugs from a variety of therapeutic classes. These
studies followed a number of case reports of serious inter-
actions between St John’s wort and digoxin, theophylline,
ciclosporin, oral contraceptives, phenprocoumon, warfarin
andsertraline, thought to be secondary to enzyme induction.
The mechanism for most of the interactions observed in subse-
quent clinical trials remains unclear, although for some
agents, induction of CYP3A4 (e.g. indinavir, midazolam,
simvastatin), P-glycoprotein-ABCB1 (e.g. digoxin, fexofena-
dine), or both (e.g. ciclosporin) may explain their increased
clearance. St John’s wort produced significantly greater
increases in CYP3A4 expression in women compared to men,
unexplained by differences in body mass index. More recently,
it was shown that St John’s wort enhanced the activity of tran-
scription factors, including the pregnane X receptor to tran-
scribe the CYP3A4 and P-gp (ABCB1) genes. Other drug
metabolism enzymes induced by St John’s wort include
CYP1A2, CYP2C9 and 2C19 and possibly UGT1A1 (Chapter
13). It should be noted that studies of St John’s wort on CYP
activity in vitro suggest acute inhibition, followed by induc-
tion in the long term.
GLUCOSAMINE
Glucosamine is available as a non-prescription dietary supple-
ment and in many products is obtained from shellfish. It is one
of several naturally occurring 6-carbon amino sugars found in
the body. Amino sugars are essential building blocks for
mucopolysaccharides, mucoproteins and mucolipids. Some
commercial products contain glucosamine in combination
with chondroitin. The precise mechanism of action of glu-
cosamine is unknown. In vitro data suggest glucosamine can
stimulate cartilage cells to synthesize glycosaminoglycans and
proteoglycans. It is more likely that the cell produces smaller,
soluble subunits; assembly of these smaller, soluble subunits
outside of the cell into a soluble form of collagen has been
proposed. Solubilized collagen, or tropocollagen, is a precursor
of mature collagen fibres. Chondroitin inhibits the enzymes
that degrade cartilage.
Several clinical studies have documented the efficacy of
glucosamine in the treatment of patients with osteoarthritis:
data from double-blind studies showed glucosamine was super-
ior to placebo and to ibuprofen in patients with osteoarthritis
of the knee. Although there is a scientific basis for administering
glucosamine in combination with chrondroitin, there is cur-
rently no evidence that the combination is more effective than
glucosamine alone for osteoarthritis. Arandomized, placebo-
controlled, double-blind study evaluated the effects of glu-
cosamine on disease progression and supported the use of
glucosamine long term (three years) for slowing progression
of knee osteoarthritis.
Adverse effects
The adverse effects associated with glucosamine involve
gastro-intestinal disturbances, including dyspepsia, nausea,
constipation and diarrhoea, skin rashes and allergic reactions
in patients with known shellfish allergy.
Drug interactions
No drug interactions have been defined with the use of
glucosamine.
MISCELLANEOUS HERBS RECENTLY
FOUND TO BE TOXIC OR MERITING THEIR
WITHDRAWAL FROM THE MARKET
Warnings about the toxicity of herbal products such as
kava kava (hepatotoxicity), aristocholic acid (nephrotoxicity)
and phen phen (pulmonary hypertension) have recently
been communicated to prescribers and the public. PC-SPES,
which was used by many prostate cancer patients because
of anecdotal and uncontrolled studies of evidence of
activity in prostate cancer, was withdrawn from sale by its
suppliers after the FDA found it contained alprazolam and
phytoestrogens.
MISCELLANEOUS HERBS 101
Key points
Herbal and nutraceutical products are widely
available over the counter in many shops and are not
regulated.
The most commonly used products are garlic, ginkgo
biloba, echinacea, soy, saw palmetto, ginseng and St
John’s wort.
The ef
ficacy of such products in many cases is not
supported by rigorous clinical trials.
Patients believe herbals are safe and are unaware of
documented or potential toxicities.
Many patients take herbal products in conjunction with
prescription medications, unknowingly risking
herb–drug interactions.
When a patient develops an unusual reaction to his or
her drug therapy (either therapeutic failure or toxicity)
a careful history concerning the use of herbal products
should be obtained.
FURTHER READING AND WEB MATERIAL
Ernst E. Asystematic review of systematic reviews of homeopathy.
British Journal of Clinical Pharmacology2002; 54: 577–82.
Goggs R, Vaughan-Thomas A, Clegg PD et al. Nutraceutical therapies
for degenerative joint diseases: a critical review. Critical Reviews in
Food Science and Nutrition2005; 45: 145–64.
Linde K, Berner M, Egger M, Mulrow C. St John’s wort for depression:
meta-analysis of randomised controlled trials. British Journal of
Psychiatry2005; 186: 99–107.
Linde K, Barrett B, Wolkart K et al. Echinacea for preventing and treat-
ing the common cold. Cochrane Database of Systematic Reviews2006;
CD000530.
Reginster JY, Bruyere O, Fraikin G, Henrotin Y. Current concepts in
the therapeutic management of osteoarthritis with glucosamine.
Bulletin (Hospital for Joint Diseases (New York))2005; 63: 31–6.
Ross S, Simpson CR, McLay JS. Homoeopathic and herbal prescribing
in general practice in Scotland. British Journal of Clinical
Pharmacology2006; 62: 646–51.
Sparreboom A, Cox MC, Acahrya MR, Figg WD. Herbal remedies in
the USA: Potential adverse reactions with anti-cancer agents.
Journal of Clinical Oncology2004; 20: 2489–503.
Walker HA, Dean TS, Sanders TAB et al. The phytoestrogen genistein
produces acute nitric oxide-dependent dilation of human forearm
vasculature with similar potency to 17 beta-estradiol. Circulation
2001;103: 258–62.
Xie HG, Kim RB. St John’s wort-associated drug interactions: short-
term inhibition and long-term induction? Clinical Pharmacology and
Therapeutics2005; 78: 19–24.
Useful websites: www.nccam.nih.gov and www.fda.gov
102 ALTERNATIVE MEDICINES: HERBALS AN D NUTRACEUTICALS
Case history
A 45-year-old Caucasian female undergoes a successful liver
transplant for primary biliary cirrhosis. Following the success-
ful operation, her immunosuppressive regimen consists of
tacrolimus, mycophenolic acid and relatively low doses of
prednisolone, which are being further reduced. During the
first six months, she remains well and her trough tacrolimus
concentrations remain between 5 and 15
μg/L. This is thera-
peutic. When seen in follow up at approximately nine months
post transplant, she is not quite feeling herself generally. Her
only other symptoms noted on systematic enquiry are that
she has not been sleeping well recently and has been anxious
about driving her car. This was because four weeks ago she
was involved in a head to head collision in a road traffic acci-
dent, but neither she nor the other driver were injured.
Current clinical examination revealed some mild subcostal
tenderness, without guarding and an otherwise normal clin-
ical examination. Her liver function tests show an increased
AST and ALT (five-fold the upper limit of normal) and a
mildly elevated conjugated bilirubin. Thorough clinical and
laboratory investigation revealed no infectious cause. A liver
biopsy is compatible with hepatic rejection and a random
tacrolimus concentration is 2
μg/L. She is adamant that she is
adhering to her medication regimen.
Question 1
Could these problems all be attributed to her liver dysfunc-
tion? Is this a possible drug–drug interaction, if so which
CYP450 enzyme system is involved?
Question 2
What else might she be taking in addition to her immunosup-
pressive regimen that could lead to this clinical situation?
Answer 1
The patient’s hepatic dysfunction is most likely due to a late
rejection episode. However, if her hepatic dysfunction were
severe enough to compromise hepatic drug metabolism
this would be accompanied by evidence of hepatic biosyn-
thetic dysfunction and drugs metabolized by the liver
would accumulate to toxic concentrations, rather than be
subtherapeutic. An alternative drug interaction with an
inducer of hepatic drug metabolism could explain the clin-
ical picture, but whereas high-dose corticosteroids would
cause a 15–30% induction of hepatic CYP3A4 enzymes, the
enzymes involved in metabolism of tacrolimus, she is on a
relatively low dose of prednisolone.
Answer 2
It is possible, but should be clarified with the patient, that
she has been taking St John’s wort for anxiety and insom-
nia. The current public view of St John’s wort is that it is a
harmless, herbal therapy that can be used to help patients
with anxiety, insomnia and depression. Some of its chemi-
cal constituents act as GABA and 5-HT receptor agonists. In
addition, one of the constituents of St John’s wort (hyper-
forin) has been shown to be a potent inducer of several
CYP450 enzymes, including 3A4, and the drug efflux trans-
porter protein P-gp (ABCB1). The induction of CYP3A4/
ABCB1 by St John’s wort constituents occurs over eight to
ten days. In the case of the magnitude of the induction
caused by CYP3A and P-gp, St Johns wort is similar to that
caused by rifampicin, and induction of both proteins is
mediated via the pregnane X nuclear receptor. St John’s
wort could be the likely cause of this patient’s subthera-
peutic tacrolimus concentrations (a 3A4/ABCB1 substrate)
and could thus have led over time to this rejection episode.
Carefully enquiring about this possibility with the patient
would be mandatory in this case. Apart from rifampicin,
other drugs that induce 3A4 (but which the patient has not
been prescribed) include phenobarbitone, carbamazepine,
other rifamycins, pioglitazone, nevirapine (see Chapter 13).
PART II
THE NERVOUS SYSTEM
This page intentionally left blank
Introduction 105
Sleep difficulties and insomnia 105
Anxiety 107
CHAPTER 18
HYPNOTICS AND ANXIOLYTICS
INTRODUCTION
Hypnotics induce sleep and anxiolytics reduce anxiety. There
is considerable overlap between them. Thus, drugs that induce
sleep also reduce anxiety, and as most anxiolytic drugs are
sedative, will assist sleep when given at night. Neither hypnotics
nor anxiolytics are suitable for the long-term management of
insomnia or anxiety, due to tolerance and dependence. In this
chapter, we discuss the management – both non-pharmacological
and pharmacological – of sleep difficulties and of anxiety, and
this is summarized in Figure 18.1.
SLEEP DIFFICULTIES AND INSOMNIA
Insomnia is common. Although no general optimal sleep
duration can be defined, sleep requirements decline in old age.
The average adult requires seven to eight hours, but some func-
tion well on as little as four hours, while others perceive more
than nine hours to be necessary. Dissatisfaction with sleep report-
edly occurs in 35% of adults and is most frequent in women
aged over 65 years. Insomnia may include complaints such as
difficulty in falling or staying asleep, and waking unrefreshed.
Hypnotics are widely prescribed despite their ineffectiveness
in chronic insomnia, as well as the problems associated with
their long-term use. Persistent insomnia is a risk factor for or
precursor of mood disorders, and may be associated with an
increased incidence of daytime sleepiness predisposing to road
traffic accidents, social and work-related problems. Insomnia
lasting only a few days is commonly the result of acute stress,
acute medical illness or jet lag. Insomnia lasting longer than
three weeks is ‘chronic’.
SLEEP
Although we spend about one-third of our lives asleep, the
function of sleep is not known. Sleep consists of two alternat-
ing states, namely rapid eye movement (REM) sleep and
non-REM sleep. During REM sleep, dreaming occurs. This is
accompanied by maintenance of synaptic connections and
increased cerebral blood flow. Non-REM sleep includes sleep
of different depths, and in the deepest form the electroenceph-
alogram (EEG) shows a slow wave pattern, growth hormone
is secreted and protein synthesis occurs.
Drugs produce states that superficially resemble physio-
logical sleep, but lack the normal mixture of REM and non-
REM phases. Hypnotics usually suppress REM sleep, and when
discontinued, there is an excess of REM (rebound) which is
associated with troubled dreams punctuated by repeated wak-
enings. During this withdrawal state, falling from wakefulness
to non-REM sleep is also inhibited by feelings of tension and
anxiety. The result is that both patient and doctor are tempted
to restart medication to suppress the withdrawal phenomena,
resulting in a vicious cycle.
GENERAL PRINC IPLES OF MANAGEMENT OF
INSOMNIA
It is important to exclude causes of insomnia that require treat-
ing in their own right. These include:
pain (e.g. due to arthritis or dyspepsia);
dyspnoea (e.g. as a result of left ventricular failure,
bronchospasm or cough);
frequency of micturition;
full bladder and/or loaded colon in the elderly;
drugs (see Table 18.1);
Table 18.1: Drugs that may cause sleep disturbances
Caffeine
Nicotine
Alcohol withdrawal
Benzodiazepine withdrawal
Amphetamines
Certain antidepressants (e.g. imipramine)
Ecstasy
Drugs that can cause nightmares (e.g. cimetidine, corticosteroids,
digoxin and propranolol)
depression;
anxiety.
Much chronic insomnia is due to dependence on hypnotic
drugs. In addition, external factors such as noise, snoring part-
ner and an uncomfortable bed may be relevant.
Drug therapy is inappropriate in individuals who need little
sleep. Shortened sleep time is common in the elderly, and
patients with dementia often have a very disturbed sleep pattern.
Hypnotics should be considered if insomnia is severe and
causing intolerable distress. They should be used for short
periods (two to four weeks at most) and, if possible, taken
intermittently. On withdrawal the dose and frequency of
use should be tailed off gradually.
Benzodiazepines are currently the hypnotics of choice, but
may fail in the elderly, and alternatives such as
clomethiazolecan be helpful. There is currently no evidence
of superiority for the newer ‘non-benzodiazepine’
hypnotics that act nonetheless on benzodiazepine
receptors (see below).
Prescribing more than one hypnotic at a time is not
recommended, and there is no pharmacological rationale
for doing this.
Drugs of other types may be needed when insomnia
complicates psychiatric illness. Sleep disturbances
accompanying depressive illness usually respond to
sedative antidepressives, such as amitriptyline.
Antipsychotics, such as chlorpromazine, may help to
settle patients suffering from dementia who have
nocturnal restlessness.
Hypnotics should not be routinely given to hospital
patients or in any other situation, except where
specifically indicated and for short-term use only.
Whenever possible, non-pharmacological methods such
as relaxation techniques, meditation, cognitive therapy,
controlled breathing or mantras should be used. Some
people experience sleepiness after a warm bath and/or
sexual activity. Amilk-based drink before bed can promote
sleep, but may cause nocturia and, in the long run, weight
gain. Caffeine-containing beverages should be avoided,
106 HYPNOTICS AND ANXIOLYTICS
INSOMNIA ANXIETY
Underlying
cause?
Chronic/
long-term
cause?
Severe
and/or
disabling?
Treat underlying
cause (Physical/
Psychological)
Non-pharmacological
methods/
behavioural
therapies
First line
Benzodiazepine
Alternative (elderly)
Clomethiazole
Second line
Zopiclone, zolpidem, zaleplon
Yes
Yes
No
No
No
Yes
Underlying
cause?
Chronic/
long-term
cause?
Severe
and/or
disabling?
Treat underlying
cause (Physical/
Psychological)
Non-pharmacological
methods/
behavioural
therapies
First line
Benzodiazepine
Second line (where sedation
is to be avoided)
Buspirone
Yes
Yes
No
No
No
Yes
Figure 18.1:Decision tree/flow chart for the management of insomnia and anxiety.
and daytime sleeping should be discouraged. Increased
daytime exercise improves sleep at night.
Alcohol should be avoided because it causes rebound
restlessness and sleep disturbance after the initial sedation
has worn off. Tolerance and dependence develop rapidly.
It also causes dehydration (gueule de bois) and other
unpleasant manifestations of hangover.
SPECIAL PROBLEMS AND SPECIAL GROU PS
JET LAG
Jet lag consists of fatigue, sleep disturbances, headache and diffi-
culty in concentrating. It is due to mismatching of the body clock
(circadian dysrhythmia) against a new time environment with
its own time cues (Zeitgebers). Resetting the internal clock is has-
tened by conforming to the new time regime. Thus, one should
rest in a dark room at night, even if not tired, and eat, work and
socialize during the day. Sufferers should not allow themselves
to sleep during the day (easier said than done!). Taking hyp-
notics at night can make things worse if sleepiness is experienced
the next day. However, short-acting benzodiazepines may be
effective if taken before going to bed for two or three nights.
Melatonin is of uncertain usefulness but may help sleep
patterns, and improves daytime well-being if taken in the
evening. It is not generally available in the UK, although it is
in several other countries including the USA.
NIGHT WORK
Night work causes more serious sleep difficulties than jet lag
because hypnotics cannot be used for long periods. Moreover,
drug-induced sleep during the day precludes family and other
non-work activities. Abetter strategy is to allow the subject to
have a short, non-drug-induced sleep during the night shift.
This improves efficiency towards the end of the night shift and
reduces sleep needs during the day.
CHILDREN
The use of hypnotics in children is not recommended, except
in unusual situations (e.g. on the night before an anticipated
unpleasant procedure in hospital). Hypnotics are sometimes
used for night terrors. Children are, however, prone to experi-
ence paradoxical excitement with these drugs. Promethazine,
an antihistamine which is available without a prescription, is
often used, but is of doubtful benefit.
ELDERLY
Anxiety and insomnia are prevalent in the elderly, for a var-
iety of psychological and physical reasons. As a rule, elderly
patients are more sensitive to the action of central nervous sys-
tem (CNS) depressant drugs than younger patients, and the
pharmacokinetics of these drugs are also altered such that their
action is more prolonged with increasing age. Hypnotics
increase the risk of falls and nocturnal confusion. Even short-
acting drugs can lead to ataxia and hangover the next morn-
ing. In the treatment of insomnia, when short-term treatment
with drugs is considered necessary, short-acting hypnotics
should be used in preference to long-acting drugs but with
explanation from the outset that these will not be continued
long term. (Short-acting benzodiazepines have the greatest
abuse potential.) Insomnia occurring in the context of docu-
mented psychiatric disorders or dementia may be better
treated with low doses of antipsychotic drugs.
ANXIETY 107
Case history
A 42-year-old man with chronic depression presents to his
general practitioner with a long history of difficulty in sleep-
ing at night, associated with early morning waking. His gen-
eral practitioner had made the diagnosis of depression and
referred him some years previously for cognitive behavioural
therapy, but this had not resulted in significant improvement
of his symptoms. His difficulty in sleeping is now interfering
with his life quite significantly, so that he feels tired most
of the day and is having difficulty holding down his job as
an insurance clerk. The GP decides that he would benefit
from taking temazepam at night; he prescribes him this,
but says that he will only give it for a maximum of a month,
as he does not want his patient to become addicted.
Question 1
Is this the correct management?
Question 2
What would be a suitable alternative treatment?
Answer 1
No. Although the benzodiazepine might help in the short
term, it does not provide the patient with a long-term solu-
tion, and does not tackle the root cause of his insomnia.
Answer 2
A more appropriate treatment would be with a regular
dose of a sedating antidepressant drug, for example ami-
triptylineat night.
ANXIETY
Anxiety is fear and is usually a normal reaction. Pathological
anxiety is fear that is sufficiently severe as to be disabling. Such
a reaction may be a response to a threatening situation (e.g. hav-
ing to make a speech) or to a non-threatening event (e.g. leaving
one’s front door and going into the street). Episodes of paroxys-
mal severe anxiety associated with severe autonomic symptoms
(e.g. chest pain, dyspnoea and palpitations) are termed panic
attacks and often accompany a generalized anxiety disorder.
GENERAL PRINC IPLES AND MANAGEMENT OF
ANXIETY
Distinguish anxiety as a functional disturbance from a
manifestation of organic brain disease or somatic illness
(e.g. systemic lupus erythematosus).
Assess the severity of any accompanying depression,
which may need treatment in itself.
Most patients are best treated with cognitive therapy,
relaxation techniques and simple psychotherapy and
without drugs.
Some patients are improved by taking regular exercise.
In severely anxious patients who are given anxiolytic drugs,
these are only administered for a short period (up to two
to four weeks) because of the risk of dependence.
short-term use, as it causes intense withdrawal
phenomena and dependence.
Diazepam or midazolam i.v. before procedures such as
endoscopy, cardioversion and operations under local
anaesthesia. Early short-lived high peak blood levels are
accompanied by anterograde amnesia.
Cautions
respiratory failure;
breast-feeding;
previous addiction.
Adverse effects
drowsiness;
confusion;
paradoxical disinhibition and aggression.
Adverse effects of intravenous diazepam include:
1. Cardiovascular and respiratory depression (uncommon).
Patients with chronic lung disease, and those who have
been previously given other central depressant drugs are
at risk.
2. Local pain following i.v. injection. An emulsion of
diazepamin intralipid is less irritating to the vein.
Intra-arterial benzodiazepine can cause arterial spasm
and gangrene.
Drug dependence, tolerance and withdrawal
Benzodiazepine dependence is usually caused by large doses
taken for prolonged periods, but withdrawal states have
arisen even after limited drug exposure. Pharmacological evi-
dence of tolerance may develop within three to 14 days. The
full withdrawal picture can manifest within hours of the last
dose for the shorter-acting drugs, or may develop over up to
three weeks with the longer-duration benzodiazepines.
Withdrawal syndrome includes a cluster of features including
frank anxiety and panic attacks. Perceptual distortions (e.g.
feelings of being surrounded by cotton wool), visual and audi-
tory hallucinations, paranoia, feelings of unreality, deperson-
alization, paraesthesiae, sweating, headaches, blurring of
vision, dyspepsia and influenza-like symptoms can occur.
Depression and agoraphobia are also common. The syndrome
may persist for weeks. Withdrawal from benzodiazepines in
patients who have become dependent should be gradual. If
this proves difficult, then an equivalent dose of a long-acting
benzodiazepine should be given as a single night-time dose
instead of shorter-acting drugs. The dose should then be
reduced in small fortnightly steps. Psychological support is
important.
Drug interactions
Pharmacodynamic interactions with other centrally acting drugs
are common, whereas pharmacokinetic interactions are not.
Pharmacodynamic interactions include potentiation of the seda-
tive actions of alcohol, histamine (H
1
) antagonists and other
hypnotics.
108 HYPNOTICS AND ANXIOLYTICS
Desensitization can be useful when severe anxiety
develops in well-recognized situations (e.g. agoraphobia,
arachnophobia, etc.). Anxiolytic drugs are sometimes
given intermittently and with a flexible-dose scheme in
such situations.
Benzodiazepines are the anxiolytics normally used where
pharmacological therapy is indicated. Buspironeis as
effective as and less hypnotic than the benzodiazepines,
but has slower onset.
β-Blockers are sometimes useful in patients with
prominent symptoms, such as palpitations or tremor.
Tricyclic antidepressants may be effective in anxiety and
in preventing panic attacks.
Monoamine oxidase inhibitors (used only by specialists)
can be useful for treating anxiety with depression, phobic
anxiety, recurrent panic attacks and obsessive-compulsive
disorders.
Individual panic attacks are usually terminated by
benzodiazepines, which may have to be supplemented
with short-term treatment with phenothiazines (e.g.
chlorpromazine).
If hyperventilation is the principal ‘trigger’, advice on
controlled breathing exercises can be curative.
DRUGS USED TO TREAT SLEEP DISTURBANCES
AND ANXIETY
The distinction between hypnotics and anxiolytics is rather
arbitrary, and the same classes of drugs are used for both pur-
poses. Compounds with a short half-life tend to be used as hyp-
notics, because they cause less ‘hangover’ effects; longer half-life
drugs tend to be used as anxiolytics, since a longer duration of
action is generally desirable in this setting. Benzodiazepines
are used for the short-term alleviation of anxiety, but should
not be used long term, where antidepressants (Chapter 20) are
usually the treatment of choice.
BENZODIAZEPINES
These drugs are anxiolytic, anticonvulsant muscle relaxants
that induce sleepiness; they remain drugs of choice for the phar-
macological treatment of insomnia and anxiety. Clonazepamis
believed to be more anticonvulsant than other members of the
group at equi-sedating doses. Benzodiazepines bind to specific
binding sites in the GABA
A
receptor–chloride channel complex
in the brain, and facilitate the opening of the channel in the
presence of GABA; this increases hyperpolarization-induced
neuronal inhibition.
Examples
Diazepam – used as an anxiolytic, because of its long
half-life.
Temazepam– used as a hypnotic, because of its short
half-life.
Lorazepam – potent short half-life benzodiazepine.
Should generally be avoided for more than very
BENZODIAZEPINES VS. NEWER DRUGS
Since the advent of the newer non-benzodiazepine hypnotics
(zopiclone,zolpidem and zaleplon), there has been much dis-
cussion and a considerable amount of confusion, as to which
type of drug should be preferred. The National Institute for
Health and Clinical Excellence (NICE) has given guidance
based on evidence and experience. In essence,
1. When hypnotic drug therapy is appropriate for severe
insomnia, hypnotics should be prescribed for short
periods only.
2. There is no compelling evidence to distinguish between
zaleplon,zolpidem, zopiclone or the shorter-acting
benzodiazepine hypnotics. It is reasonable to prescribe the
drug whose cost is lowest, other things being equal. (At
present, this means that benzodiazepines are preferred.)
3. Switching from one hypnotic to another should only be
done if a patient experiences an idiosyncratic adverse effect.
4. Patients who have not benefited from one of these
hypnotic drugs should not be prescribed any of the others.
ANXIETY 109
Key points
Insomnia and anxiety are common. Most patients do not
require drug therapy.
Benzodiazepines are indicated for the short-term relief
(2–4 weeks only) of anxiety that is severe, disabling or
subjecting the individual to unacceptable levels of
distress.
The use of benzodiazepines to treat short-term ‘mild’
anxiety is inappropriate and unsuitable.
Benzodiazepines should be used to treat insomnia only
when it is severe, disabling or subjecting the individual
to extreme distress.
There is no convincing evidence to support the use of
non-benzodiazepine hypnotics and anxiolytics over
benzodiazepines.
FLUMAZENIL
Flumazenil is a benzodiazepine antagonist. It can be used to
reverse benzodiazepine sedation. It is short acting, so sedation
may return. It can cause nausea, flushing, anxiety and fits, so
is not routinely used in benzodiazepine overdose which sel-
dom causes severe adverse outcome.
OTHERS
Barbiturates are little used and dangerous in overdose.
Clomethiazole – causes conjunctival, nasal and gastric
irritation. Useful as a hypnotic in the elderly because its
short action reduces the risk of severe hangover, ataxia and
confusion the next day. It is effective in acute withdrawal
syndrome in alcoholics, but its use should be carefully
supervised and treatment limited to a maximum of nine
days. It can be given intravenously to terminate status
epilepticus. It can also be used as a sedative during
surgery under local anaesthesia.
Zopiclone, zolpidem and zaleplon – are non-
benzodiazepine hypnotics which enhance GABAactivity
by binding to the GABA–chloride channel complex at the
benzodiazepine-bindingsite. Although they lack structural
features of benzodiazepines, they also act by potentiating
GABA. Their addictive properties are probably similar to
benzodiazepines.
Buspirone – is a 5HT
1A
receptor partial agonist. Its use has
not been associated with addiction or abuse, but may be a
less potent anxiolytic than the benzodiazepines. Its
therapeutic effects take much longer to develop (two to
three weeks). It has mild antidepressant properties.
Cloral and derivatives – formerly often used in paediatric
practice. Cloral shares properties with alcohol and volatile
anaesthetics. Cloral derivatives have no advantages over
benzodiazepines, and are more likely to cause rashes and
gastric irritation.
Sedative antihistamines, e.g. promethazine, are of
doubtful benefit, and may be associated with prolonged
drowsiness, psychomotor impairment and antimuscarinic
effects.
Case history
A 67-year-old widow attended the Accident and Emergency
Department complaining of left-sided chest pain, palpita-
tions, breathlessness and dizziness. Relevant past medical his-
tory included generalized anxiety disorder following the
death of her husband three years earlier. She had been pre-
scribed lorazepam, but had stopped it three weeks previously
because she had read in a magazine that it was addictive.
When her anxiety symptoms returned she attended her GP,
who prescribed buspirone, which she had started the day
before admission.
Examination revealed no abnormality other than a regu-
lar tachycardia of 110 beats/minute, dilated pupils and sweat-
ing hands. Routine investigations, including ECG and chest
x-ray, were unremarkable.
Question 1
Assuming a panic attack is the diagnosis, what is a poten-
tial precipitant?
Question 2
Give two potential reasons for the tachycardia.
Answer 1
Benzodiazepine withdrawal.
Answer 2
1. Buspirone (note that buspirone, although anxiolytic, is
not helpful in benzodiazepine withdrawal and may
also cause tachycardia).
2. Anxiety.
3. Benzodiazepine withdrawal.
FURTHER READING
Fricchione G. Clinical practice. Generalized anxiety disorder. New
England Journal of Medicine2004; 351: 675–82.
National Institute for Clinical Excellence. 2004: Guidance on the use of
zaleplon, zolpidem and zopiclone for the short-term management
of insomnia. www.nice.org.uk/TA077guidance, 2004.
Sateia MJ, Nowell PD. Insomnia. Lancet2004; 364: 1959–73.
Stevens JC, Pollack MH. Benzodiazepines in clinical practice: consid-
eration of their long-term use and alternative agents. Journal of
Clinical Psychiatry2005; 66 (Suppl. 2), 21–7.
Schizophrenia 110
Behavioural emergencies 114
CHAPTER 19
SCHIZOPHRENIA AND
BEHAVIOURAL EMERGENCIES
SCHIZOPHRENIA
INTRODUCTION
Schizophrenia is a devastating disease that affects approxi-
mately 1% of the population. The onset is often in adolescence
or young adulthood and the disease is usually characterized by
recurrent acute episodes which may develop into chronic dis-
ease. The introduction of antipsychotic drugs such as chlorpro-
mazine revolutionized the treatment of schizophrenia so that
the majority of patients, once the acute symptoms are relieved,
can now be cared for in the community. Previously, they would
commonly be sentenced to a lifetime in institutional care.
PATHOPHYSIOLOGY
The aetiology of schizophrenia, for which there is a genetic pre-
disposition, is unknown, although several precipitating factors
are recognized (Figure 19.1). Neurodevelopmental delay has
been implicated and it has been postulated that the disease is
triggered by some life experience in individuals predisposed by
an abnormal (biochemical/anatomical) mesolimbic system.
There is heterogeneity in clinical features, course of disease
and response to therapy. The concept of an underlying
neurochemical disorder is advanced by the dopamine theory
of schizophrenia, summarized in Box 19.1. The majority of
antipsychotics block dopamine receptors in the forebrain.
5-Hydroxytryptamine is also implicated, as indicated in Box
19.2. Glutamine hypoactivity, GABAhypoactivity and α-adren-
ergic hyperactivity are also potential neurochemical targets.
About 30% of patients with schizophrenia respond inad-
equately to conventional dopamine D
2
receptor antagonists. A
high proportion of such refractory patients respond to clozap-
ine, an ‘atypical’ antipsychotic drug which binds only tran-
siently to D
2
receptors, but acts on other receptors, especially
muscarinic, 5-hydroxytryptamine receptors (5HT
2
) and D
1
,
and displays an especially high affinity for D
4
receptors. The
D
4
receptor is localized to cortical regions and may be over-
expressed in schizophrenia. Regional dopamine differences
may be involved, such as low mesocortical activity with high
mesolimbic activity. Magnetic resonance imaging (MRI) stud-
ies indicate enlargement of ventricles and loss of brain tissue,
whilst functional MRI and positron emission tomography
(PET) suggest hyperactivity in some cerebral areas, consistent
with loss of inhibitory neurone function.
012345
Odds Ratio
Winter
Urban
Place/time of birth
Infection
Prenatal
Obstetric
Influenza
Respiratory
Rubella
Poliovirus
CNS
Famine
Bereavement
Flood
Unwantedness
Maternal depression
Rhesus Incompatibility
Hypoxia
CNS damage
Low birth weight
Pre-eclampsia
Family history
678910
Figure 19.1:Predispositions to
schizophrenia. Redrawn with permission
from Sullivan PF. The genetics of
schizophrenia.PLoS Medicine 2005; 2: e212.
SCHIZOPHRENIA 111
Figure 19.2 shows a summary of putative pathways for the
development of schizophrenia.
GENERAL PRINC IPLES OF MANAGEMENT
ACUTE TREATMENT
The main principles are:
Prompt drug treatment should be instigated, usually as an
in-patient.
Oral ‘atypical antipsychotics’ should be administered, e.g.
risperidoneor olanzapine.
If the patient is very disturbed/aggressive, add
benzodiazepine, e.g. lorazepam.
Chlorpromazine may be preferred if sedation is
advantageous, e.g. in very agitated patients.
Antimuscarinic drugs, e.g. procyclidine, should be used if
acute dystonia or Parkinsonian symptoms develop.
Psychosocial support/treatment should be offered.
Behaviour usually improves quickly, but hallucinations,
delusions and affective disturbance may take weeks or
months to improve.
Once first-rank symptoms have been relieved, the patient
can usually return home and resume work on low-dose
antipsychotic treatment.
Conventional drugs, e.g. chlorpromazineor haloperidol,
are as effective in treatment of acute positive symptoms as
atypical antipsychotic drugs and are less expensive, but
adverse effects may be troublesome.
MAINTENANCE TREATMENT
Only 10–15% of patients remain in permanent remission
after stopping drug therapy following a first
schizophrenic episode.
The decision to attempt drug withdrawal should be taken
with regard to the individual patient, their views, adverse
drug effects, social support, relatives and carers.
Cognitive behavioural therapy is a treatment option.
Most patients require lifelong drug therapy, so the correct
diagnosis is essential (e.g. beware drug-induced psychosis,
as amphetamines in particular can produce acute
schizophreniform states). All antipsychotic drugs have
adverse effects. Continuing psychosocial support is critical.
Oral or intramuscular depot therapy (Box 19.3), e.g.
olanzapine(oral) or flupentixol (i.m.) should be
considered. The latter ensures compliance.
Box 19.1: Dopamine theory of schizophrenia
There is excess dopamine activity in the mesolimbic
system in schizophrenia.
Antipsychotic potency is often proportional to
D
2
-blocking potency.
Amphetamine (which increases dopamine release) can
produce acute psychosis that is indistinguishable from
acute schizophrenia (positive symptoms).
D
2
agonists (bromocriptine and apomorphine)
aggravate schizophrenia in schizophrenic patients.
There is an increase in D
2
and D
4
receptors on PET in
schizophrenic patients.
L-Dopa can cause hallucinations and acute psychotic
reactions and paranoia, but does not cause all the
features of these conditions.
There is no definite increase in brain dopamine in vivo
and post mortem.
Dopamine receptor blockade does not fully alleviate
symptoms.
Box 19.3: Intramuscular depot treatment
Esters of the active drug are formulated in oil.
There is slow absorption into the systemic circulation.
It takes several months to reach steady state.
After an acute episode, reduce the oral dose gradually
and overlap with depot treatment.
Give a test dose in case the patient is allergic to the oil
vehicle or very sensitive to extrapyramidal effects.
Rotate the injection site, e.g. flupentixol is given once
every two to four weeks (ester of active drug
formulated in an oil) or risperidone once every two
weeks.
Box 19.2: 5-Hydroxytryptamine and schizophrenia
LSD acts on 5HT receptors, causing hallucinations and
dramatic psychological effects which may mimic some
features of schizophrenia.
5HT has a modulatory effect on dopamine pathways.
Many effective antipsychotic drugs have dopamine and
5HT
2
receptor-blocking properties.
5HT
2
receptor blockade is not essential for drug efficacy.
Genetic predisposition
Obstetric complications
and other early insults
affecting CNS
Neurodevelopmental
abnormalities
Neurocognitive
impairment
Social anxiety
Isolation
Odd ideas
Frank
psychosis
Abuse of dopaminergic drugs
Social stress/isolation
Figure 19.2:Pathways for development of
schizophrenia.
112 SCHIZOPHRENIA AND B EHAVIOURALEMERGENCIES
DRUGS USED IN TREATMENT
CONVENTIONAL ANTIPSYCHOTIC DRUGS
The principal action of the conventional antipsychotic drugs
(see Table 19.1), such as chlorpromazine(a phenothiazine) and
haloperidol(a butyrophenone), is an antagonism of D
2
recep-
tors in the forebrain. The effect on D
1
receptors is variable.
Blockade of the D
2
receptors induces extrapyramidal effects.
Repeated adminstration causes an increase in D
2
-receptor sen-
sitivity due to an increase in abundance of these receptors. This
appears to underlie the tardive dyskinesias that are caused by
prolonged use of the conventional antipsychotic drugs.
The choice of drug is largely determined by the demands of
the clinical situation, in particular the degree of sedation
needed and the patient’s susceptibility to extrapyramidal tox-
icity and hypotension.
Uses
These include the following:
1. schizophrenia – antipsychotic drugs are more effective
against first-rank (positive) symptoms (hallucinations,
thought disorder, delusions, feelings of external control)
than against negative symptoms (apathy and
withdrawal);
2. other excited psychotic states, including mania and
delirium;
3. anti-emetic and anti-hiccough;
4. premedication and in neuroleptanalgesia;
5. terminal illness, including potentiating desired actions of
opioids while reducing nausea and vomiting;
6. severe agitation and panic;
7. aggressive and violent behaviour;
8. movement and mental disorders in Huntington’s disease.
Adverse effects
1. The most common adverse effects are dose-dependent
extensions of pharmacological actions:
extrapyramidal symptoms (related to tight binding to,
and receptor occupancy of, D
2
receptors) – parkinsonism
including tremor, acute dystonias, e.g. torticollis,
fixed upward gaze, tongue protrusion; akathisia
(uncontrollable restlessness with feelings of anxiety
and agitation) and tardive dyskinesia. Tardive
dyskinesia consists of persistent, repetitive, dystonic
athetoid or choreiform movements of voluntary
muscles. Usually the face and mouth are involved,
causing repetitive sucking, chewing and lip smacking.
The tongue may be injured. The movements are
usually mild, but can be severe and incapacitating.
This effect follows months or years of antipsychotic
treatment;
anticholinergic – dry mouth, nasal stuffiness,
constipation, urinary retention, blurred vision;
postural hypotension due to α-adrenergic blockade.
Gradual build up of the dose improves tolerability;
sedation (which may be desirable in agitated patients),
drowsiness and confusion. Tolerance usually develops
after several weeks on a maintenance dose. Emotional
flattening is common, but it may be difficult to
distinguish this feature from schizophrenia.
Depression may develop, particularly following
treatment of hypomania, and is again difficult to
distinguish confidently from the natural history of the
disease. Acute confusion is uncommon.
2. Jaundice occurs in 2–4% of patients taking
chlorpromazine, usually during the second to fourth
weeks of treatment. It is due to intrahepatic cholestasis
and is a hypersensitivity phenomenon associated with
eosinophilia. Substitution of another phenothiazine may
not reactivate the jaundice.
3. Ocular disorders during chronic administration include
corneal and lens opacities and pigmentary retinopathy.
This may be associated with cutaneous light sensitivity.
4. About 5% of patients develop urticarial, maculopapular
or petechial rashes. These disappear on withdrawal of
the drug and may not recur if the drug is reinstated.
Contact dermatitis and light sensitivity are common
complications. Abnormal melanin pigmentation may
develop in the skin.
5. Hyperprolactinaemia.
6. Blood dyscrasias are uncommon, but may be lethal,
particularly leukopenia and thrombocytopenia. These
usually develop in the early days or weeks of treatment.
The incidence of agranulocytosis is approximately 1 in
10000 patients receiving chlorpromazine.
7. Cardiac dysrhythmia, including torsades de pointes
(see Chapter 32) and arrest.
8. Malignant neuroleptic syndrome is rare but potentially
fatal. Its clinical features are rigidity, hyperpyrexia,
stupor or coma, and autonomic disorder. It responds to
treatment with dantrolene(a ryanodine receptor
antagonist that blocks intracellular Ca
2
mobilization).
9. Seizures, particularly in alcoholics. Pre-existing epilepsy
may be aggravated.
10. Impaired temperature control, with hypothermia in cold
weather and hyperthermia in hot weather.
Table 19.1: Conventional antipsychotic drugs
Sedation Extrapyramidal Hypotension
symptoms
Phenothiazines
Chlorpromazine   
Fluphenazine
a

Butyrophenones
Haloperidol 
Thioxanthines
Fluphenthixol
a

a
Depot preparation available.
All increase serum prolactin levels
Note: Pimozide causes a prolonged QT and cardiac arrhythmias.
The Boston Collaborative Survey indicated that adverse
reactions are most common in patients receiving high doses,
and that they usually occur soon after starting treatment.
The most common serious reactions were fits, coma, severe
hypotension, leukopenia, thrombocytopenia and cardiac
arrest.
Contraindications and cautions
These include the following:
coma due to cerebral depressants, bone marrow
depression, phaeochromocytoma, epilepsy, chronic
respiratory disease, hepatic impairment or Parkinson’s
disease;
caution is needed in the elderly, especially in hot or cold
weather;
pregnancy, lactation;
alcoholism.
Pharmacokinetics
The pharmacokinetics of conventional antipsychotic drugs
have been little studied. They have multiple metabolites and
their large apparent volumes of distribution (V
d
) (e.g. for
chlorpromazine V
d
22 L/kg) result in low plasma concen-
trations, presenting technical difficulties in estimation. Most is
known about chlorpromazine, see Box 19.4.
Drug interactions
These include the following:
alcohol and other CNS depressants – enhanced sedation;
hypotensive drugs and anaesthetics – enhanced
hypotension;
increased risk of cardiac arrhythmias with drugs that
prolong the QT interval (e.g. amiodarone,sotalol);
tricyclic antidepressants – increased antimuscarinic
actions;
metoclopramide – increased extrapyramidal effects and
akathisia;
antagonism of anti-Parkinsonian dopamine agonists (e.g.
L-dopa) (these are in any case contraindicated in
schizophrenia).
ATYPICAL ANTIPSYCHOTIC DRUGS
The term ‘atyptical antipsychotic’ is used very imprecisely.
‘Newer’ or ‘second-generation’ antipsychotics are synonymous
in some texts. In comparison to the conventional antipsychotics
where potency is closely related to D
2
receptor blockade, atyp-
ical antipsychotics bind less tightly to D
2
receptors and have
additional pharmacological activity which varies with the drug.
Efficacy against negative symptoms, as well as less extrapyra-
midal side effects, are characteristic. These may be the result of
the transient (‘hit and run’) binding to D
2
receptors.
Clozapine is the original ‘atypical’ antipsychotic and is
described below. Its use is limited to resistant patients due to
the risk of agranulocytosis. A variety of other atypical anti-
psychotic drugs are available. Features of clozapineare:
D
4
5HT
2
blockade;
D
1
D
2
blockade;
α-adrenoceptor blockade;
effective in resistant patients;
effective against negative and positive symptoms;
virtually free from extrapyramidal effects;
agranulocytosis (3%) – use is restricted to patients licensed
with a monitoring service: blood count (weekly for first
18 weeks, then every two weeks till one year, then every
four weeks);
severe postural hypotension – initiate therapy under
supervision;
sedation, dizziness, hypersalivation;
weight gain, glucose intolerance, possible intestinal
obstruction;
myocarditis and cardiomyopathy;
pulmonary embolism;
seizures.
Many newer alternatives, but none with the unique properties
of clozapine, e.g. risperidone, olanzapine, aripiprazole,
amisulpride,quetiapine and zotepine, have been introduced.
Their pharmacology, efficacy and adverse effects vary.
Although more expensive, in June 2002 NICE recommended
Box 19.4: Pharmacokinetics (chlorpromazine)
Dose regimes are largely empirical.
There is variable absorption.
There are 70 metabolites, some of which are
active.
Enterohepatic circulation is involved.
There is enormous variability in plasma concentrations
andt
1/2
.
There is a vast volume of distribution.
Brain:plasma concentration is 5:1.
Reduced doses should be prescribed in the elderly
(for both pharmacokinetic and pharmacodynamic
differences).
Case history
A 50-year-old woman whose schizophrenia is treated
with oral haloperidol is admitted to the Accident and
Emergency Department with a high fever, fluctuating level
of consciousness, muscular rigidity, pallor, tachycardia,
labile blood pressure and urinary incontinence.
Question 1
What is the likely diagnosis?
Question 2
How should this patient be managed?
Answer 1
Neuroleptic malignant syndrome.
Answer 2
1. Stop the haloperidol.
2. Initiate supportive therapy.
3. Bromocriptine (value uncertain).
4. Dantrolene (value uncertain).
SCHIZOPHRENIA 113
114 SCHIZOPHRENIA AND B EHAVIOURALEMERGENCIES
that atypical antipsychotics should be considered in newly
diagnosed schizophrenic patients and in those who have
unacceptable effects from, or inadequate response to, conven-
tional antipsychotic drugs. Risperidone blocks D
2
, D
4
and in
particular 5HT
2
receptors. Careful dose titration reduces the
risk of adverse effects, but extrapyramidal side effects are com-
mon at high doses. It is available as an intramuscular injection
for acute control of agitation and disturbed behaviour. Weight
gain and, more worryingly, an increased incidence of stroke in
elderly patients with dementia have been reported wih both
risperidone and olanzapine. Aripiprazole is a long-acting
atypical antipsychotic which is a partial agonist at D
2
recep-
tors, as well as blocking 5HT
2
. It is not associated with
extrapyramidal effects, prolactin secretion or weight gain.
treatment. The control of hypomanic and manic episodes with
chlorpromazineis often dramatic.
ACUTE PSYCHOTIC EPISODES
Patients with organic disorders may experience fluctuating
confusion, hallucinations and transient paranoid delusions.
Violent incidents sometimes complicate schizophrenic illness.
Key points
Pharmacological treatment
Receptor blockade:
–D
2
, D
4
, 5HT
2
.
Although there may be a rapid behavioural benefit, a
delay (usually of the order of weeks) in reduction of
many symptoms implies secondary effects (e.g. receptor
up/downregulation).
Conventional antipsychotics (e.g. chlorpromazine,
haloperidol, fluphenazine), act predominantly by D
2
blockade.
Atypical antipsychotics (e.g. clozapine, risperidone,
olanzapine) are less likely to cause extrapyramidal side
effects.
Key points
Adverse effects of antipsychotic drugs
Extrapyramidal motor disturbances, related to
dopamine blockade.
Endocrine distributions (e.g. gynaecomastia), related to
prolactin release secondary to dopamine blockade.
Autonomic effects, dry mouth, blurred vision,
constipation due to antimuscarinic action and postural
hypotension due to α-blockade.
Cardiac dysrhythmias, which may be related to
prolonged QT, e.g. sertindole (an atypical antipsychotic),
pimozide.
Sedation.
Impaired temperature homeostasis.
Weight gain.
Idiosyncratic reactions;
jaundice (e.g. chlorpromazine);
leukopenia and agranulocytosis (e.g. clozapine);
skin reactions;
neuroleptic malignant syndrome.
BEHAVIOURAL EMERGENCIES
MANIA
Acute attacks are managed with antipsychotics, but lithium
is a common and well-established long-term prophylactic
MANAGEMENT
Antipsychotics and benzodiazepines, either separately or
together, are effective in the treatment of patients with violent
and disturbed behaviour. Lorazepamby mouth or parenteral
injection is most frequently used to treat severely disturbed
behaviour as an in-patient.
Haloperidol can rapidly terminate violent and psychotic
behaviour, but hypotension, although uncommon, can be
severe, particularly in patients who are already critically ill.
Doses should be reduced in the elderly.
Intramuscular olanzapine or liquid risperidone are grad-
ually supplanting more conventional antipsychotics in the
acute management of psychosis.
When treating violent patients, large doses of anti-
psychotics may be sometimes needed. Consequently, extrapyra-
midal toxicity, in particular acute dystonias, develops in up to
one-third of patients. Prophylactic anti-parkinsonian drugs,
such as procyclidine, may be given, especially in patients who
are particularly prone to movement disorders.
The combination of lorazepam and haloperidol has
been successful in treating otherwise resistant delirious
behaviour.
Case history
A 60-year-old man with schizophrenia who has been
treated for 30 years with chlorpromazine develops involun-
tary (choreo-athetoid) movements of the face and tongue.
Question 1
What drug-induced movement disorder has developed?
Question 2
Will an anticholinergic drug improve the symptoms?
Question 3
Name three other drug-induced movement disorders
associated with antipsychotic drugs.
Answer 1
Tardive dyskinesia.
Answer 2
No. Anticholinergic drugs may unmask or worsen tardive
dyskinesia.
Answer 3
1. Akathisia.
2. Acute dystonias.
3. Chronic dystonias.
4. Pseudo-parkinsonism.
BEHAVIOURALEMERGENCIES 115
a vailability of flumazenilif (particularly i.v.) benzodiazepines
are used.
FURTHER READING
Anon. Which atypical antipsychotic for schizophrenia? Drugs and
Therapeutics Bulletin2004; 42: 57–60.
Freedman R. Drug therapy: schizophrenia. New England Journal of
Medicine2003; 334: 1738–49.
Oral medication, especially in liquid form, is the
preferred mode of administration, if the patient will accept
it, but intramuscular or intravenous routes may have to
be used.
Antipsychotics, such as chlorpromazineshould be avoided
in alcohol withdrawal states, in alcoholics or in those depend-
ent on benzodiazepines because of the risk of causing fits.
Ensure resuscitation facilities including those for mechan-
ical ventilation are available. Many centres insist on the
Depressive illnesses and antidepressants 116
Lithium, tryptophan and St John’s wort 121
Special groups 122
CHAPTER 20
MOOD DISORDERS
DEPRESSIVE ILLNESSES AN D
ANTIDEPRESSANTS
Many forms of depression are recognized clinically and most
respond well to drugs. From a biochemical viewpoint, there
are probably different types of depression (which do not corres-
pond predictably to clinical variants) depending on which
neurotransmitter is involved, and these may respond differ-
ently to different drugs.
PATHOPHYSIOLOGY: INSIGHTS FROM
ANTIDEPRESSANT DRUG ACTIONS
The monoamine theory of mood is mainly based on evidence
from the actions of drugs.
1. Reserpine, which depletes neuronal stores of noradrenaline
(NA) and 5-hydroxytryptamine (5HT) and α-methyltyrosine,
which inhibits NAsynthesis, cause depression.
2. Tricyclic antidepressants (TCA) of the amitriptyline type
(which raise the synaptic concentration of NAand 5HT)
are antidepressant.
3. Monoamine oxidase inhibitors (MAOIs, which increase
total brain NAand 5HT) are antidepressant.
On the basis of these actions, it was suggested that depression
could be due to a cerebral deficiency of monoamines. One dif-
ficulty with this theory is that amfetamineand cocaine, which
act like tricyclic drugs in raising the synaptic NAcontent, are
not antidepressive, although they do alter mood. Even worse,
the tricyclic antidepressants block amine reuptake from synapses
within one or two hours of administration, but take from ten
days to four weeks to alleviate depression. Such a long time-
course suggests a resetting of postsynaptic or presynaptic
receptor sensitivity.
Another theory of depression is the serotonin-only hypothesis.
This theory emphasizes the role of 5HT and downplays that of
NAin the causation of depression, and is backed by the effect-
iveness of the selective serotonin reuptake inhibitors, or SSRI
class of drugs, in the treatment of depression. However, it also
does not explain the delay in onset of the clinical effect of
antidepressant drugs, including the SSRIs, and again receptor
resetting has to be invoked. Also, many strands of evidence sug-
gest that NAdoes indeed have an important role in depression.
The permissive hypothesis of mania/depression suggests
that the control of emotional behaviour results from a balance
between NA and 5HT. According to this theory, both the
manic phase and the depressive phase of bipolar disorder are
characterized by low central 5HT function. Evidence suggests
that brain 5HT systems dampen or inhibit a range of functions
involving other neurotransmitters. Mood disorders result from
the removal of the serotonin damper. This hypothesis postu-
lates that low levels of 5HT permit abnormal levels of NAto
cause depression or mania. If 5HT cannot control NAand NA
falls to abnormally low levels, the patient becomes depressed.
On the other hand, if the level of 5HT falls and the level of
NA becomes abnormally high, the patient becomes manic.
According to this hypothesis, antidepressant drugs are effect-
ive to the degree that they restore the ability of 5HT to control
NA, thus restoring the critical balance that controls emotional
behaviour. Arecently available class of antidepressant drugs,
serotonin-noradrenaline reuptake inhibitors (SNRI), work by
selectively blocking reuptake of both NA and 5HT, thereby
increasing levels of both monoamines. The SNRIs have very
little affinity for other postsynaptic receptor sites and are there-
fore less likely to produce some of the side effects associated
with TCA.
Dysregulation of the hypothalamic–pituitary–adrenal axis
is a common biological marker of depression and the value of
antiglucocorticoid drugs is under investigation.
GENERAL PRINC IPLES OF MANAGEMENT
Depression is common, but under-diagnosed. It can be recog-
nized during routine consultations, but additional time may
be needed. Genetic and social factors are often relevant. Drug
treatment is not usually appropriate at the mild end of the
severity range. Drugs are used in more severe depression,
especially if it has melancholic (‘endogenous’) features. Even
if depression is attributable to external factors (‘exogenous’),
DEPRESSIVE ILLNESSES AND ANTIDEPRESSANTS 117
e.g. interpersonal difficulties or other life stresses (including
physical illness), antidepressant drugs may be useful. Drugs
used in the initial treatment of depression include TCAs and
related drugs, SSRIs and SNRIs. Although clinical experience
is most extensive with the TCAs, the side-effect profile of the
SSRIs is usually less troublesome, and these drugs are safer in
overdose. Therefore many psychiatrists and general practi-
tioners use SSRIs rather than TCAs as first-line treatment for
depression. SSRIs are more expensive than TCAs. The relative
side effects of the different antidepressant drugs are summar-
ized in Table 20.1.
In refractory depression, other drug treatment or electro-
convulsive therapy (ECT) are considered. Alternative drug
strategies include (1) adding lithium to a tricyclic to give a
lithium blood level of 0.6–0.8 mmol/L; (2) combining anti-
depressants; (3) augmenting with T3 (or T4), a mood stabilizer
such as lamotrigine,buspirone or estradiol; (4) MAOIs, usu-
ally prescribed only by psychiatrists; (5) MAOI plus a TCA–
but only in expert psychiatric hands; or (6) small doses of flu-
pentixol(for short-term treatment only).
Figures 20.1 and 20.2 show a treatment algorithm for man-
agement of depressive illness.
SELECTIVE SEROTONIN REUPTAKE INHIBITORS
(SSRIs)
These drugs are safer in overdose than the tricyclic group.
Selective serotonin reuptake inhibitors (SSRIs) do not stimulate
appetite and have much fewer antimuscarinic side effects than
the tricyclics and other catecholamine-uptake inhibitors. They
are also well tolerated in the elderly. Examples include fluox-
etine, fluvoxamine, paroxetine, sertraline, citalopram and
escitalopram.
Uses
These include the following:
1. in depression (they have similar efficacy to tricyclics, but
are much more expensive);
2. in chronic anxiety, and as prophylaxis for panic attacks;
3. obsessive-compulsive states;
4. bulimia nervosa;
5. seasonal affective disorder, especially if accompanied by
carbohydrate craving and weight gain;
6. possibly effective as prophylactic agents in recurrent
depression.
Adverse effects
1. The most common adverse reactions to SSRIs are nausea,
dyspepsia, diarrhoea, dry mouth, headache, insomnia and
dizziness. Sweating, erectile dysfunction and delayed
orgasm are well-recognized associations. These tend to
become less severe after one to two months of treatment.
2. They have less anticholinergic and cardiotoxic actions
than tricyclic drugs.
Table 20.1:Relative antidepressant side effects
Drug Anticholinergic Cardiac Nausea Sedation Overdose Pro-convulsant Tyramine
effects effects risk interaction
Tricyclics and related
antidepressants
Amitriptyline     
Clomipramine    
Dothiepin     
Imipramine    
Lofepramine 
Trazodone  
Selective serotonin
reuptake inhibitors
Citalopram 
Fluoxetine ?
Paroxetine ?
Sertraline ?
Monoamine oxidase
inhibitors
Phenelzine  
Moclobemide  ?
Others
Venlafaxine ? 
, little or nothing reported; , mild; , moderate; , high; ?, insufficient information available.
118 MOOD DISORDERS
3. Epilepsy can be precipitated.
4. They are usually non-sedating, but may cause insomnia
and do not usually cause orthostatic hypotension.
5. All antidepressants can cause hyponatraemia, probably
due to induction of inappropriate antidiuretic hormone
secretion, but it is reported more frequently with SSRIs
than with other antidepressants.
Contraindications
These include the following:
hepatic and renal failure;
epilepsy;
manic phase.
Drug interactions
Combinations of SSRI with lithium,tryptophan or MAOIs
may enhance efficacy, but are currently contraindicated
because they increase the severity of 5HT-related toxicity.
In the worst reactions, the life-threatening 5HT syndrome
develops. This consists of hyperthermia, restlessness, tremor,
myoclonus, hyperreflexia, coma and fits. After using MAOIs,
it is recommended that two weeks should elapse before
starting SSRIs. Avoid fluoxetinefor at least five weeks
before using MAOI because of its particularly long half-
life (about two days).
The action of warfarin is probably enhanced by
fluoxetineand paroxetine.
There is antagonism of anticonvulsants.
Fluoxetine raises blood concentrations of haloperidol.
SEROTONIN-NORADRENALINE REUPTAKE
INHIBITORS AND RELATED ANTIDEPRESSANTS
Venlafaxine: A potent 5HT and NAuptake inhibitor that
appears to be as effective as TCAs, but without anticholinergic
effects. It may have a more rapid onset of therapeutic action
than other antidepressants, but this has yet to be confirmed. It
is associated with more cardiac toxicity than the SSRIs.
Duloxetineinhibits NA and 5HT reuptake.
TRICYCLICS AND RELATED ANTIDEPRESSANTS
(TCAs)
Uses
These include the following:
1. depressive illnesses, especially major depressive episodes
and melancholic depression;
2. atypical oral and facial pain;
3. prophylaxis of panic attacks;
4. phobic anxiety;
5. obsessive–compulsive disorders;
6. imipramine has some efficacy in nocturnal enuresis.
Although these drugs share many properties, their
profiles vary in some respects, and this may alter their use in
different patients. The more sedative drugs include amitripty-
line, dosulepin and doxepin. These are more appropriate
for agitated or anxious patients than for withdrawn or apa-
thetic patients, for whom imipramine or nortriptyline,
which are less sedative, are preferred. Protriptylineis usually
stimulant.
Only 70% of depressed patients respond adequately to
TCAs. One of the factors involved may be the wide variation in
individual plasma concentrations of these drugs that is
obtained with a given dose. However, the relationship between
plasma concentration and response is not well defined. Amul-
ticentre collaborative study organized by the World Health
Organization failed to demonstrate any relationship whatso-
ever between plasma amitriptyline concentration and clinical
effect.
Diagnosis of unipolar
depression
Psychotherapy
and medication
Continue same
treatment
Advance dose
as tolerated
Go to second phase
of treatment
Evaluate response
to medication after
3–4 weeks
Evaluate response
to medication after
6–8 weeks
Significant symptoms
persist after 6 weeks
Psychotherapy
Symptoms resolving
Symptoms persist
Partial response No response
Add medication
Medication
Figure 20.1:General algorithm for the initial phase of treatment
of depression. When symptoms persist after first-line treatment,
re-evaluate the accuracy of the diagnosis, the adequacy of the dose
and the duration of treatment before moving to the second
phase of treatment. (Redrawn with permission from Aronson SC
and Ayres VE. ‘Depression: A Treatment Algorithm for the Family
Physician’,Hospital Physician Vol 36 No 7, 2000. Copyright 2000
Turner White Communications, Inc.)
DEPRESSIVE ILLNESSES AND ANTIDEPRESSANTS 119
Imipramineand amitriptyline (tertiary amines) have more
powerful anticholinergic and cardiac toxic effects than second-
ary amines (e.g. nortriptyline).
Mechanism of action
The tricyclics block uptake-1 of monoamines into cerebral (and
other) neurones. Thus, the concentration of amines in the
synaptic cleft rises. As discussed above, they may also induce a
slow adaptive decrease in pre- and/or postsynaptic amine
receptor sensitivity.
Adverse effects
Autonomic (anticholinergic)/cardiovascularDry mouth,
constipation (rarely paralytic ileus, gastroparesis),
tachycardia, paralysis of accommodation, aggravation of
narrow-angle glaucoma, retention of urine, dry skin due
to loss of sweating, and (due to α-blockade) postural
hypotension. Rarely, sudden death due to a cardiac
dysrhythmia. In overdose, a range of tachydysrhythmias
and intracardiac blocks may be produced.
Central nervous systemFine tremor and sedation, but also
(paradoxically) sometimes insomnia, decreased rapid eye
movement (REM) sleep, twitching, convulsions, dysarthria,
paraesthesia, ataxia. Increased appetite and weight gain,
particularly with the sedative tricyclics, are common. On
withdrawal of the drug, there may be gastro-intestinal
symptoms such as nausea and vomiting, headache,
giddiness, shivering and insomnia. Sometimes anxiety,
agitation and restlessness follow sudden withdrawal.
Allergic and idiosyncratic reactionsThese include bone marrow
suppression and jaundice (both rare).
HyponatraemiaHyponatraemia is an adverse effect due to
inappropriate ADH secretion, and is more common in the
elderly.
Contraindications
These include the following:
epilepsy;
recent myocardial infarction, heart block;
mania;
porphyria.
RELATED NON-TRICYCLIC ANTIDEPRESSANT DRUGS
This is a mixed group which includes 1-, 2- and 4-ring struc-
tured drugs with broadly similar properties. Characteristics of
specific drugs are summarized below.
Maprotiline– sedative, with less antimuscarinic effects, but
rashes are more common and fits are a significant risk.
Mianserin– blocks central α
2
-adrenoceptors. It is sedative,
with much fewer anticholinergic effects, but can cause
postural hypotension and blood dyscrasias, particularly in
the elderly. Full blood count must be monitored.
Assess risk factors for
treatment resistance
Assess symptom severity
Partial response to
first-line treatment
Advance dose
as tolerated
Symptoms persist after
6–8 weeks
Ensure safe maximum
tolerated dose
Trial of an alternative
medication
Nonresponders:
mild-to-moderate
symptoms, low risk
All patients:
moderate-to-severe
symptoms, or high risk
No response to
first-line treatment
Persistent symptoms
AUGMENTATION
*
Persistent symptoms
Trial of an alternative
medication
Partial responders:
mild symptoms, low risk
Figure 20.2:General algorithm for
the second phase of treatment of
depression. Augmentation*
involves the use of a combination
of medications to enhance the
efficacy of an antidepressant.
(Redrawn with permission from
Aronson SC and Ayres VE,
‘Depression: A Treatment
Algorithm for the Family
Physician’,Hospital Physician
Vol 36 No 7, 2000. Copyright
2000 Turner White
Communications, Inc.)
120 MOOD DISORDERS
Lofepramine– less sedative, and with less cardiac toxicity,
but occasionally hepatotoxic.
Mirtazapine– increases noradrenergic and serotonergic
neurotransmission via central α
2
adrenoceptors. The
increased release of 5HT stimulates 5HT
1
receptors, whilst
5HT
2
and 5HT
3
receptors are blocked. H
1
receptors are
also blocked. This combination of actions appears to be
associated with antidepressant activity, anxiolytic and
sedative effects. Reported adverse effects include increased
appetite, weight gain, drowsiness, dry mouth and (rarely)
blood dyscrasias.
Drug interactions
These include the following:
antagonism of anti-epileptics;
potentiation of sedation with alcohol and other central
depressants;
antihypertensives and diuretics increase orthostatic
hypotension;
hypertension and cardiac dysrhythmias with adrenaline,
noradrenaline and ephedrine.
MONOAMINE OXIDASE INHIBITORS (MAOIs)
These drugs were little used for many years because of their
toxicity, and particularly potentially lethal food and drug inter-
actions causing hypertensive crises. Non-selective MAOIs
should only be prescribed by specialists who are experienced
in their use. They can be effective in some forms of refractory
depression and anxiety states, for which they are generally
reserved. The introduction of moclobemide, a reversible select-
ive MAO-Ainhibitor, may lead to more widespread use of this
therapeutic class.
Tranylcypromineis the most hazardous MAOI because of
its stimulant activity. The non-selective MAOIs of choice are
phenelzineand isocarboxazid.
Uses
These include the following:
1. MAOIs can be used alone or (with close psychiatric
supervision) with a TCA, in depression which has not
responded to TCAs alone;
2. in phobic anxiety and depression with anxiety;
3. in patients with anxiety who have agoraphobia, panic
attacks or multiple somatic symptoms;
4. hypochondria and hysterical symptoms may respond well;
5. for atypical depression with biological features such as
hypersomnia, lethargy and hyperphagia.
Adverse effects
1. Common effects include orthostatic hypotension, weight
gain, sexual dysfunction, headache and aggravation of
migraine, insomnia, anticholinergic actions and oedema.
2. Rare and potentially fatal effects include hypertensive
crisis and 5HT syndrome, psychotic reactions,
hepatocellular necrosis, peripheral neuropathy and
convulsions.
3. Stopping a MAOI is more likely to produce a withdrawal
syndrome than is the case with tricyclics. The syndrome
includes agitation, restlessness, panic attacks and
insomnia.
Contraindications
These include the following:
liver failure;
cerebrovascular disease;
phaeochromocytoma;
porphyria;
epilepsy.
Drug interactions
Many important interactions occur with MAOI. Atreatment
card for patients should be carried at all times, which describes
precautions and lists some of the foods to be avoided. The
interactions are as follows:
hypertensive and hyperthermic reactions sufficient to
cause fatal subarachnoid haemorrhage, particularly with
tranylcypromine. Such serious reactions are precipitated
by amines, including indirectly acting sympathomimetic
agents such as tyramine (in cheese), dopamine (in broad
bean pods and formed from levodopa), amines formed
from any fermentation process (e.g. in yoghurt, beer,
wine),phenylephrine (including that administered as
nosedrops and in cold remedies), ephedrine,amfetamine
(all can give hypertensive reactions), other amines,
pethidine(excitement, hyperthermia), levodopa
(hypertension) and tricyclic, tetracyclic and bicyclic
antidepressants (excitement, hyperpyrexia). Buspirone
should not be used with MAOIs. Hypertensive crisis may
be treated with α-adrenoceptor blockade analogous to
medical treatment of patients with phaeochromocytoma
(see Chapter 40). Interactions of this type are much less
likely to occur with moclobemide, as its MAO inhibition
is reversible, competitive and selective for MAO-A, so that
MAO-B is free to deaminate biogenic amines;
failure to metabolize drugs that are normally oxidized,
including opioids, benzodiazepines, alcohol (reactions
with alcoholic drinks occur mainly because of their
tyramine content). These drugs will have an exaggerated
and prolonged effect;
enhanced effects of oral hypoglycaemic agents, anaesthetics,
suxamethonium, caffeine and anticholinergics (including
benzhexol and similar anti-Parkinsonian drugs);
antagonism of anti-epileptics;
enhanced hypotension with antihypertensives;
central nervous system (CNS) excitation and hypertension
withoxypertine (an antipsychotic) and tetrabenazine (used
for chorea);
increased CNS toxicity with triptans (5HT
1
agonists) and
withsibutramine.
LITHIUM, TRYPTOPHAN AND ST JOHN’S
WORT
LITHIUM
Althoughlithium is widely used in affective disorders, it has
a low toxic to therapeutic ratio, and serum concentration moni-
toring is essential. Serum is used rather than plasma because
of possible problems due to lithium heparin, which is often
used as an anticoagulant in blood sample tubes. Serum
lithium levels fluctuate between doses and serum concentra-
tions should be measured at a standard time, preferably 12
hours after the previous dose. This measurement is made fre-
quently until steady state is attained and is then made every
three months, unless some intercurrent event occurs that
could cause toxicity (e.g. desal-ination or diuretic therapy).
Use
Lithiumis effective in acute mania, but its action is slow (one
to two weeks), so antipsychotic drugs, such as haloperidol,
are preferred in this situation (see Chapter 19). Its main use
is in prophylaxis in unipolar and bipolar affective illness
LITHIUM, TRYPTOPHANAND ST J OHNS WORT 121
(therapeutic serum levels 0.4–1mmol/L). Lithium is also
used on its own or with another antidepressant in refractory
depression to terminate a depressive episode or to prevent
recurrences and aggressive or self-mutilating behaviour.
Patients should avoid major dietary changes that alter
sodium intake and maintain an adequate water intake.
Different lithiumpreparations have different bioavailabili-
ties, so the form should not be changed.
Mechanism of action
Lithium increases 5HT actions in the CNS. It acts as a 5HT
1A
agonist and is also a 5HT
2
antagonist. This may be the basis for
its antidepressant activity and may explain why it increases
the CNS toxicity of selective 5HT uptake inhibitors.
The basic biochemical activity of lithium is not known. It
has actions on two second messengers.
1. Hormone stimulation of adenylyl cyclase is inhibited,
so that hormone-stimulated cyclic adenosine
monophosphate (cAMP) production is reduced. This
probably underlies some of the adverse effects of lithium,
such as goitre and nephrogenic diabetes insipidus, since
thyroid-stimulating hormone (TSH) and antidiuretic
hormone activate adenylyl cyclase in thyroid and
collecting duct cells, respectively. The relevance of this
to its therapeutic effect is uncertain.
2. Lithium at a concentration of 1 mmol/L inhibits
hydrolysis of myoinositol phosphate in the brain, so
lithiummay reduce the cellular content of phosphatidyl
inositides, thereby altering the sensitivity of neurones
to neurotransmitters that work on receptors linked
to phospholipase C (including muscarinic and
α-adrenoceptors).
From these actions, it is clear that lithiumcan modify a wide
range of neurotransmitter effects, yet its efficacy both in mania
and in depression indicates a subtlety of action that is cur-
rently unexplained, but may be related to activation of the brain
stem raphe nuclei.
Adverse effects
1. When monitored regularly lithium is reasonably safe in
the medium term. However, adverse effects occur even in
the therapeutic range – in particular, tremor, weight gain,
oedema, polyuria, nausea and loose bowels.
2. Above the therapeutic range, tremor coarsens, diarrhoea
becomes more severe and ataxia and dysarthria appear.
Higher levels cause gross ataxia, coma, fits, cardiac
dysrhythmias and death. Serum lithiumconcentrations
greater than 1.5mmol/L may be dangerous and if greater
than 2mmol/L, are usually associated with serious
toxicity.
3. Goitre, hypothyroidism and exacerbation of psoriasis are
less common.
4. Renal tubular damage has been described in association
with prolonged use.
Key points
Antidepressant contraindications
Tricyclic antidepressants – recent myocardial infarction,
dysrhythmias, manic phase, severe liver disease.
SSRIs – manic phase.
Monamine oxidase inhibitors – acute confused state,
phaeochromocytoma.
Caution is needed – cardiac disease, epilepsy, pregnancy
and breast-feeding, elderly, hepatic and renal
impairment, thyroid disease, narrow-angle glaucoma,
urinary retention, prostatism, porphyria, psychoses,
electroconvulsive therapy (ECT) and anaesthesia.
Key points
Drug treatment of depression
Initial drug treatment is usually with SSRIs, tricyclic
antidepressants or related drugs.
The choice is usually related to the side-effect profile of
relevance to the particular patient.
Tricyclic antidepressants are more dangerous in overdose.
Tricyclic antidepressants commonly cause antimuscarinic
and cardiac effects.
Tricyclic antidepressants tend to increase appetite and
weight, whereas SSRIs more commonly reduce appetite
and weight.
SSRIs are associated with nausea, sexual dysfunction
and sleep disturbance.
There is a variable delay (between ten days and four
weeks) before therapeutic benefit is obtained.
Following remission, antidepressant therapy should be
continued for at least four to six months.
ST JOHN’S WORT
St John’s wort (Hypericum perforatum) is an unlicensed herbal
remedy in popular use for treating depression (Chapter 17).
However, it can induce drug-metabolizing enzymes, and many
important drug interactions have been identified, including
with antidepressant drugs, with which St John’s wort should
therefore not be given. The amount of active ingredient can
vary between different preparations, thus changing the prepa-
ration can alter the degree of such interactions. Importantly,
when St John’s wort is discontinued, the concentrations of
interacting drugs may increase.
SPECIAL GROUPS
THE ELDERLY
Depression is common in the elderly, in whom it tends to be
chronic and has a high rate of recurrence. Treatment with drugs
is made more difficult because of slow metabolism and sensi-
tivity to anticholinergic effects. Lower doses are therefore needed
than in younger patients.
Lack of response may indicate true refractoriness of the
depression, or sadness due to social isolation or bereavement.
The possibility of underlying disease, such as hypothyroidism
(the incidence of which increases with age), should be
considered.
Lofepramine and SSRIs cause fewer problems in patients
with prostatism or glaucoma than do the tricyclic antidepres-
sants because they have less antimuscarinic action. Dizziness
and falls due to orthostatic hypotension are less common with
nortriptyline than with imipramine. Mianserin has fewer
anticholinergic effects, but blood dyscrasias occur in about one
in 4000 patients and postural hypotension can be severe.
EPILEPSY
No currently used antidepressive is entirely safe in epilepsy,
but SSRIs are less likely to cause fits than the amitriptyline
group, mianserinor maprotiline.
Contraindications
These include the following:
renal disease;
cardiac disease;
sodium-losing states (e.g. Addison’s disease, diarrhoea,
vomiting);
myasthenia gravis;
during surgical operations;
avoid when possible during pregnancy and breast-feeding.
Pharmacokinetics
Lithium is readily absorbed after oral administration and
injectable preparations are not available. Peak serum concen-
trations occur three to five hours after dosing. The t
1/2
varies
with age because of the progressive decline in glomerular fil-
tration rate, being 18–20 hours in young adults and up to 36
hours in healthy elderly people. Sustained-release prepar-
ations are available, but in view of the long t
1/2
they are not
kinetically justified. Lithiumtakes several days to reach steady
state and the first samples for serum level monitoring should
be taken after about one week unless loading doses are given.
Lithium elimination is almost entirely renal. Like sodium,
lithiumdoes not bind to plasma protein, and it readily passes
into the glomerular filtrate; 70–80% is reabsorbed in the proxi-
mal tubules but, unlike sodium, there is no distal tubular reab-
sorption and its elimination is not directly altered by diuretics
acting on the distal tubule. However, states such as sodium
deficiency and sodium diuresis increase lithiumretention (and
cause toxicity) by stimulating proximal tubular sodium and
lithium reabsorption. An important implication of the renal
handling of lithiumis that neither loop diuretics, thiazides nor
potassium-sparing diuretics can enhance lithiumloss in a toxic
patient, but all of them do enhance its toxicity. Dialysis reduces
elevated serum lithiumconcentration effectively.
Drug interactions
Lithium concentration in the serum is increased by
diuretics and non-steroidal anti-inflammatory drugs.
Lithium toxicity is increased by concomitant
administration of haloperidol, serotonin uptake
inhibitors, calcium antagonists (e.g. diltiazem) and
anticonvulsants (phenytoinand carbamazepine) without
a change in serum concentration.
Lithium increases the incidence of extrapyramidal effects
of antipsychotics.
L-TRYPTOPHAN
Tryptophan is the amino acid precursor of 5HT. On its own
or with other antidepressants or lithium it sometimes bene-
fits refractory forms of depression. However,
L-tryptophan
should only be initiated under specialist supervision because
of its association with an eosinophilic myalgic syndrome char-
acterized by intense and incapacitating fatigue, myalgia and
eosinophilia. Arthralgia, fever, cough, dyspnoea and rash may
also develop over several weeks. A few patients develop
myocarditis.
122 MOOD DISORDERS
Case history
A 75-year-old woman with endogenous depression is
treated with amitriptyline. After three weeks, she appears
to be responding, but then seems to become increasingly
drowsy and confused. She is brought to the Accident
and Emergency Department following a series of
convulsions.
Question
What is the likely cause of her drowsiness, confusion and
convulsions?
Answer
Hyponatraemia.
Comment
Hyponatraemia (usually in the elderly) has been associated
with all types of antidepressant but most frequently with
SSRIs.
SPECIAL GROUPS 123
Case history
A 45-year-old man with agoraphobia, anxiety and depres-
sion associated with hypochondriacal features is treated
with phenelzine. He has no history of hypertension. He is
seen in the Accident and Emergency Department because
of a throbbing headache and palpitations. On examination
he is hypertensive 260/120mmHg with a heart rate of 40
beats/minute. He is noted to have nasal congestion.
Question 1
What is the likely diagnosis?
Question 2
What is the most appropriate treatment?
Answer 1
Hypertensive crisis, possibly secondary to taking a cold cure
containing an indirectly acting sympathomimetic.
Answer 2
Phentolamine, a short-acting alpha-blocker, may be given
by intravenous injection, with repeat doses titrated against
response.
FURTHER READING
Aronson SC, Ayres VD. Depression: a treatment algorithm for the
family physician. Hospital Physician2000; 44: 21–38.
Ebmeier KP, Donaghey C, Steele JD. Recent developments and current
controversies in depression. The Lancet2005; 367: 153–67.
PARKINSON’S SYNDROME AND ITS
TREATMENT
PATHOPHYSIOLOGY
James Parkinson first described the tremor, rigidity and
bradykinesia/akinesia that characterize the syndrome known
as Parkinson’s disease. Most cases of Parkinson’s disease are
caused by idiopathic degeneration of the nigrostriatal path-
way. Atherosclerotic, toxic (e.g. related to antipsychotic drug
treatment, manganese or carbon monoxide poisoning) and
post-encephalitic cases also occur. Treatment of parkinsonism
caused by antipsychotic drugs differs from treatment of the
idiopathic disease, but other aetiologies are treated similarly to
Parkinson’s syndrome and its treatment 124
Spasticity 128
Chorea 129
Drug-induced dyskinesias 129
Treatment of other movement disorders 129
Myasthenia gravis 129
Alzheimer’s disease 131
CHAPTER 21
MOVEMENT DISORDERS AND
DEGENERATIVE CNS DISEASE
Striatum
Substantia nigra
Motor
cortex
ACh
DA
GABA
Figure 21.1:Representation of relationships between cholinergic
(ACh), dopaminergic (DA) and GABA-producing neurones in the
basal ganglia.
the idiopathic disease. Parkinsonian symptoms manifest after
loss of 80% or more of the nerve cells in the substantia nigra.
The nigrostriatal projection consists of very fine nerve fibres
travelling from the substantia nigra to the corpus striatum.
This pathway is dopaminergic and inhibitory, and the motor
projections to the putamen are more affected than either those
to the cognitive areas or to the limbic and hypolimbic regions
(Figure 21.1). Other fibres terminating in the corpus striatum
include excitatory cholinergic nerves and noradrenergic and
serotoninergic fibres, and these are also affected, but to vary-
ing extents, and the overall effect is a complex imbalance
between inhibitory and excitatory influences.
Parkinsonism arises because of deficient neural transmission
at postsynaptic D
2
receptors, but it appears that stimulation of
both D
1
and D
2
is required for optimal response. D
1
receptors
activate adenylyl cyclase, which increases intracellular cyclic
adenosine monophosphate (cAMP). The antagonistic effects of
dopamine and acetylcholine within the striatum have suggested
that parkinsonism results from an imbalance between these
neurotransmitters (Figure 21.2). The therapeutic basis for treat-
ing parkinsonism is to increase dopaminergic activity or to
reduce the effects of acetylcholine. 1-Methyl-4-phenyl-1,2,5,6-
tetrahydropyridine (MPTP) has been used illicitly as a drug of
abuse and it causes severe parkinsonism. MPTPis converted by
monoamine oxidase-B (MAO-B) in neuronal mitochondria to a
toxic free-radical metabolite (MPP
), which is specifically toxic
to dopamine-producing cells. This led to the hypothesis that
idiopathic Parkinson’s disease may be due to chronically
increased free-radical damage to the cells of the substantia nigra.
However, clinical studies of anti-oxidants have so far been
disappointing.
The free-radical hypothesis has raised the worrying possibil-
ity that treatment with levodopa (see below) could accelerate
disease progression by increasing free-radical formation as the
drug is metabolized in the remaining nigro-striatal nerve fibres.
This is consistent with the clinical impression of some neurolo-
gists, but in the absence of randomized clinical trials it is
difficult to tell whether clinical deterioration is due to the natu-
ral history of the disease or is being accelerated by the therapeu-
tic agent.
PARKINSONS SYNDROME AND ITS TREATMENT 125
PRINCIPLES OF TREATMENT IN PARKINSONISM
Idiopathic Parkinson’s disease is a progressive disorder, and is
treated with drugs that relieve symptoms and if possible
slow disease progression. Treatment is usually initiated when
symptoms disrupt normal daily activities. Initial treatment is
often with a dopamine receptor agonist, e.g. bromocriptine,
particularly in younger (70) patients. Alevodopa/decarboxy-
lase inhibitor combination is commonly used in patients with
definite disability. The dose is titrated to produce optimal
results. Occasionally, amantadine or anticholinergics may be
useful as monotherapy in early disease, especially in younger
patients when tremor is the dominant symptom. In patients on
levodopa the occurrence of motor fluctuations (on–off phenom-
ena) heralds a more severe phase of the illness. Initially, such
fluctuations may be controlled by giving more frequent doses
of levodopa (or a sustained-release preparation). The addition
of either a dopamine receptor agonist (one of the non-ergot
derivatives, e.g. ropinirole) or one of the calechol-O-methyl
transferase (COMT) inhibitors (e.g. entacapone, tolcapone) to
the drug regimen may improve mobility. In addition, this usu-
ally allows dose reduction of the levodopa, while improving
‘end-of-dose’ effects and improving motor fluctuations. If on–off
phenomena are refractory, the dopamine agonist apomorphine
can terminate ‘off’ periods, but its use is complex (see below).
Selegiline, a MAO-B inhibitor, may reduce the end-of-dose
deterioration in advanced disease. Physiotherapy and psycho-
logical support are helpful. The experimental approach of
implantation of stem cells into the substantia nigra of severely
affected parkinsonian patients (perhaps with low-dose
immunosuppression) is being investigated. The potential of
stereotactic unilateral pallidotomy, for severe refractory cases of
Parkinson’s disease is being re-evaluated.
Drugs that cause parkinsonism, notably conventional
antipsychotic drugs (e.g. chlorpromazine, haloperidol) (see
Chapter 19) are withdrawn if possible, or substituted by the
newer ‘atypical’ antipsychotics (e.g. risperidoneor olanzapine),
since these have a lower incidence of extrapyramidal side effects.
Antimuscarinic drugs (e.g. trihexyphenidyl) are useful if chang-
ing the drug/reducing the dose is not therapeutically acceptable,
whereas drugs that increase dopaminergic transmission are con-
traindicated because of their effect on psychotic symptoms.
ANTI-PARKINSONIAN DRUGS
DRUGS AFFECTING THE DOPAMINERGIC SYSTEM
Dopaminergic activity can be enhanced by:
levodopa with a peripheral dopa decarboxylase inhibitor;
increasing release of endogenous dopamine;
stimulation of dopamine receptors;
inhibition of catechol-O-methyl transferase;
inhibition of monoamine oxidase type B.
LEVODOPA AND DOPA DECARBOXYLASE INHIBITORS
Use
Levodopa (unlike dopamine) can enter nerve terminals in the
basal ganglia where it undergoes decarboxylation to form
dopamine.Levodopa is used in combination with a peripheral
(extracerebral) dopa decarboxylase inhibitor (e.g. carbidopaor
benserazide). This allows a four- to five-fold reduction in levo-
dopadose and the incidence of vomiting and dysrhythmias is
reduced. However, central adverse effects (e.g. hallucinations)
are (predictably) as common as when larger doses of levodopa
are given without a dopa decarboxylase inhibitor.
Combined preparations (co-careldopa or co-beneldopa)
are appropriate for idiopathic Parkinson’s disease. (Levodopa
is contraindicated in schizophrenia and must not be used for
parkinsonism caused by antipsychotic drugs.) Combined
preparations are given three times daily starting at a low dose,
increased initially after two weeks and then reviewed at
intervals of six to eight weeks. Without dopa decarboxylase
inhibitors, 95% of levodopa is metabolized outside the brain.
In their presence, plasma levodopaconcentrations rise (Figure
21.3), excretion of dopamine and its metabolites falls, and the
availability of levodopa within the brain for conversion to
dopamine increases. The two available inhibitors are similar.
Adverse effects
These include the following:
nausea and vomiting;
postural hypotension – this usually resolves after a few
weeks, but excessive hypotension may result if
antihypertensive treatment is given concurrently;
Normal
Normal
Parkinsonism due to
excess acetylcholine
Cholinergic
system
(excitatory)
Anticholinergic
drugs
Dopaminergic
system
(inhibitory)
Parkinsonism due to
dopamine deficiency
Levodopa
Figure 21.2:Antagonistic actions of the dopaminergic
and cholinergic systems in the pathogenesis of
parkinsonian symptoms.
Key points
Parkinson’s disease
Clinical diagnosis is based on the triad of tremor,
rigidity and bradykinesia.
Parkinsonism is caused by the degeneration of
dopaminergic pathways in basal ganglia leading to
imbalance between cholinergic (stimulatory) and
dopaminergic (inhibitory) transmission.
It is induced/exacerbated by centrally acting dopamine
antagonists (e.g. haloperidol), but less so by clozapine,
risperidone or olanzapine.
126 MOVEMENT DISORDERS AND DEGEN ERATIVEC NS DISEASE
involuntary movements (dystonic reactions) – these
include akathisia (abnormal restlessness and inability to
keep still), chorea and jerking of the limbs (myoclonus).
Involuntary movements may become worse as treatment
is continued, and may necessitate drug withdrawal;
psychological disturbance, including vivid dreams,
agitation, paranoia, confusion and hallucinations;
cardiac dysrhythmias;
endocrine effects of levodopa, including stimulation of
growth hormone and suppression of prolactin.
sedation and sudden onset of sleep (avoid driving at onset
of treatment and if these symptoms recur).
Pharmacokinetics
Levodopais absorbed from the proximal small intestine and is
metabolized both by decarboxylases in the intestinal wall and
by the gut flora. Oral absorption is variable. Absorption/
bioavailability are improved by co-administration of decar-
boxylase inhibitors. Addition of a COMT inhibitor further
increases t
1/2
and AUC.
Drug interactions
Monoamine oxidase inhibitors can produce hypertension if
given concurrently with levodopa. The hypotensive actions of
other drugs are potentiated by levodopa.
INCREASED RELEASE OF E NDOGENOUS
DOPAMINE
AMANTADINE
Use
Amantadine has limited efficacy, but approximately 60% of
patients experience some benefit. Severe toxicity is rare.
Mechanism of action
Endogenous dopamine release is stimulated by amantadine,
which also inhibits reuptake of dopamine into nerve terminals.
Adverse effects
These include the following:
peripheral oedema;
gastro-intestinal upset and dry mouth;
livedo reticularis;
CNS toxicity – nightmares, insomnia, dizziness,
hallucinations, convulsions;
leukopenia (uncommon).
Pharmacokinetics
The t
1/2
of amantadine varies from 10 to 30 hours, so steady-
state concentrations are reached after four to seven days of treat-
ment. About 95% is eliminated by the kidneys and it should not
be used in patients with renal failure.
DOPAMINE RECEPTOR AGONISTS
Uses
Dopamine receptor agonists are used as initial therapy or as
adjuncts to levodopa–dopa decarboxylase inhibitor combin-
ations in patients with severe motor fluctuations (on–off
phenomena). Dopamine agonists share many of their adverse
effects with levodopa, particularly nausea due to stimulation of
dopamine receptors in the chemoreceptor trigger zone. This
brain region is unusual in that it is accessible to drugs in the sys-
temic circulation, so domperidone(a dopamine antagonist that
does not cross the blood–brain barrier) prevents this symptom
without blocking dopamine receptors in the striatum, and hence
worsening the movement disorder. Neuropsychiatric disorders
are more frequent than with levodopamonotherapy. (See also
Chapter 42 for use in pituitary disorders, and Chapter 41 for use
in suppression of lactation). Pulmonary, retroperitoneal and
pericardial fibrotic reactions have been associated with some
ergot-derived dopamine agonists. Dopamine receptor agonists
are started at a low dose that is gradually titrated upwards
depending on efficacy and tolerance. If added to levodopa, the
dose of the latter may be reduced.
Ergot derivatives include bromocriptine,lisuride, pergolide
and cabergoline. Other licensed dopamine agonists include
pramipexole,ropinirole and rotigotine.
There is great individual variation in the efficacy of
dopamine receptor agonists. The initial dose is gradually
titrated upwards depending on response and adverse effects.
Adverse effects
These are primarily due to D
2
agonist activity, although 5HT
1
and 5HT
2
effects are also relevant.
gastro-intestinal – nausea and vomiting, constipation or
diarrhoea;
central nervous system – headache, drowsiness,
confusion, psychomotor excitation, hallucination;
orthostatic hypotension (particularly in the elderly),
syncope;
cardiac dysrhythmias – bradycardia;
L
-dopa (100 mg) MK 485
L
-dopa (1000 mg)
L
-dopa (100 mg)
0
0.2
0.4
0.6
0.8
1.2
1.0
2468
Plasma dopa concentration (g/mL)
Time after
L
-dopa dose (h)
Figure 21.3:Increased plasma dopa concentrations following
combination with a peripheral dopa decarboxylase inhibitor
(MK 485) in one patient. (Redrawn with permission from Dunner DL
et al. Clinical Pharmacology and Therapeutics1971; 12: 213.)
PARKINSONSSYNDROME AND ITS TREATMENT 127
pulmonary, retroperitoneal and pericardial fibrotic
reactions have been associated with the ergot-derived
dopamine agonists (bromocriptine,cabergoline, lisuride
andpergolide).
APOMORPHINE
Apomorphine is a powerful dopamine agonist at both D
1
and
D
2
receptors, and is used in patients with refractory motor oscil-
lations (on–off phenomena). It is difficult to use, necessitating
specialist input. The problems stem from its pharmacokinetics
and from side effects of severe nausea and vomiting. The gastro-
intestinal side effects can be controlled with domperidone.
Apomorphine is started in hospital after pretreatment with
domperidonefor at least three days, and withholding other anti-
parkinsonian treatment at night to provoke an ‘off’ attack. The
subcutaneous dose is increased and when the individual dose
requirement has been established, with reintroduction of other
drugs if necessary, administration is sometimes changed from
intermittent dosing to subcutaneous infusion via a syringe pump,
with patient-activated extra boluses if needed. Apomorphine is
extensively hepatically metabolized and is given parenterally.
The mean plasma t
1/2
is approximately 30 minutes.
CATECHOL-O-METHYL TRANSFERASE INHIBITORS
Use
Tolcaponeand entacapone are used for adjunctive therapy in
patients who are already taking
L-dopa/dopa decarboxylase
inhibitor combinations with unsatisfactory control (e.g. end-of-
dose deterioration). These agents improve symptoms with less
on–off fluctuations, as well as reducing the levodopa dose
requirement by 20–30%. Adverse effects arising from increased
availability of
L-dopa centrally can be minimized by decreas-
ing the dose of levodopacombination treatment prospectively.
Because of hepatotoxicity associated with tolcapone it is
only used by specialists when entacapone is ineffective as an
adjunctive treatment.
Mechanism of action
Reversible competitive inhibition of COMT, thereby reducing
metabolism of
L-dopa and increasing its availability within
nigrostriatal nerve fibres. It is relatively specific for central ner-
vous system (CNS) COMT, with little effect on the peripheral
COMT, thus causing increased brain concentrations of
L-dopa,
while producing less of an increase in plasma concentration.
Adverse effects
These include the following:
nausea, vomiting, diarrhoea and constipation;
increased levodopa-related side effects;
neuroleptic malignant syndrome;
dizziness;
hepatitis – rare with entacapone, but potentially life-
threatening with tolcapone(liver function testing is
mandatory before and during treatment);
urine discolouration.
Pharmacokinetics
Tolcaponeis rapidly absorbed and is cleared by hepatic metab-
olism. At recommended doses it produces approximately
80–90% inhibition of central COMT.
Drug interactions
Apomorphine is metabolized by O-methylation, so inter-
action with COMT inhibitors is to be anticipated. COMT
inhibitors should not be administered with MAOIs, as block-
ade of both pathways of monoamine metabolism simultane-
ously has the potential to enhance the effects of endogen-
ous and exogenous amines and other drugs unpredictably.
MONOAMINE OXIDASE INHIBITORS – TYPE B
SELEGILINE AND RASAGILINE
Use
Initial small controlled studies in Parkinson’s disease reported
that disease progression was slowed in patients treated with
selegiline alone, delaying the need to start levodopa. Larger-
scale studies have not confirmed this conclusion. MAO type B
inhibitors, such as selegiline and rasagiline, may be used in
conjunction with levodopato reduce end-of-dose deterioration.
Mechanism of action
There are two forms of monoamine oxidase (MAO), namely
type A (substrates include 5-hydroxytryptamine and tyram-
ine) and type B (substrates include phenylethylamine). MAO-B,
is mainly localized in neuroglia. MAO-Ametabolizes endoge-
nous adrenaline, noradrenaline and 5-hydroxytryptamine,
while the physiological role of MAO-B is unclear. Both isoen-
zymes metabolize dopamine. Inhibition of MAO-B raises
brain dopamine levels without affecting other major transmit-
ter amines. Because selegiline and rasagiline selectively
inhibit MAO-B, they are much less likely to produce a hyper-
tensive reaction with cheese or other sources of tyramine than
non-selective MAOIs, such as phenelzine.
Adverse effects
Selegiline is generally well tolerated, but side effects include
the following:
agitation and involuntary movements;
confusion, insomnia and hallucinations;
nausea, dry mouth, vertigo;
peptic ulceration.
Pharmacokinetics
Oralselegiline is well absorbed (100%), but is extensively metab-
olized by the liver, first to an active metabolite, desmethylselegi-
line (which also inhibits MAO-B) and then to amphetamine and
metamphetamine. Its plasma t
1/2
is long (approximately 39h).
Drug interactions
At very high doses (six times the therapeutic dose), MAO-B
selectivity is lost and pressor responses to tyramine are
128 MOVEMENT DISORDERS AND DEGEN ERATIVEC NS DISEASE
Adverse effects
These include the following:
dry mouth, blurred vision, constipation;
precipitation of glaucoma or urinary retention – they are
therefore contraindicated in narrow angle glaucoma and
in men with prostatic hypertrophy;
cognitive impairment, confusion, excitement or psychosis,
especially in the elderly.
Pharmacokinetics
Table 21.1 lists some drugs of this type that are in common
use, together with their major pharmacokinetic properties.
SPASTICITY
Spasticity is an increase in muscle tone, for example, due to
damage to upper motor neurone pathways following stroke or
in demyelinating disease. It can be painful and disabling.
Treatment is seldom very effective. Physiotherapy, limited sur-
gical release procedures or local injection of botulinum toxin
(see below) all have a role to play. Drugs that reduce spasticity
include diazepam, baclofen, tizanidine and dantrolene, but
they have considerable limitations.
Diazepam (see Chapter 18, Hypnotics and anxiolytics)
facili-tatesγ-aminobutyric acid (GABA) action. Although spas-
ticity and flexor spasms may be diminished, sedating doses are
often needed to produce this effect.
Baclofen facilitates GABA-B receptors and also reduces
spasticity. Less sedation is produced than by equi-effective
doses of diazepam, but baclofencan cause vertigo, nausea and
hypotension. Abrupt withdrawal may precipitate hyperactiv-
ity, convulsions and autonomic dysfunction. There is specialist
interest in chronic administration of low doses of baclofen
intrathecally via implanted intrathecal cannulae in selected
patients in order to maximize efficacy without causing side
effects.
Dantrolene (a ryanodine receptor antagonist) is generally
less useful for symptoms of spasticity than baclofen because
muscle power is reduced as spasticity is relieved. It is used
intravenously to treat malignant hyperthermia and the neu-
roleptic malignant syndrome, for both of which it is uniquely
effective (see Chapter 24). Its adverse effects include:
drowsiness, vertigo, malaise, weakness and fatigue;
diarrhoea;
increased serum potassium levels.
potentiated. Hypertensive reactions to tyramine-containing
products (e.g. cheese or yeast extract) have been described,
but are rare. Amantadine and centrally active antimuscarinic
agents potenti-ate the anti-parkinsonian effects of selegiline.
Levodopa-induced postural hypotension may be potentiated.
DRUGS AFFECTING THE CHOLINERGIC SYSTEM
MUSCARINIC RECEPTOR ANTAGONISTS
Use
Muscarinic antagonists (e.g. trihexyphenidyl, benzatropine,
orphenadrine, procyclidine) are effective in the treatment of
parkinsonian tremor and – to a lesser extent – rigidity, but pro-
duce only a slight improvement in bradykinesia. They are
usually given in divided doses, which are increased every two
to five days until optimum benefit is achieved or until adverse
effects occur. Their main use is in patients with parkinsonism
caused by antipsychotic agents.
Mechanism of action
Non-selective muscarinic receptor antagonism is believed to
restore, in part, the balance between dopaminergic/cholinergic
pathways in the striatum.
Table 21.1: Common muscarinic receptor ant agonists, dosing and pharmacokinetics
Drug Route of Half-life (hours) Metabolism and Special features
administration excretion
Trihexyphenidyl Oral 3–7 Hepatic
Orphenadrine Oral 13.7–16.1 Hepatic-active Central
metabolite stimulation
Procyclidine Oral 12.6 Hepatic
Key points
Treatment of Parkinson’s disease
A combination of levodopa and a dopa-decarboxylase
inhibitor (carbidopa or benserazide) or a dopamine
agonist (e.g. ropinirole) are standard first-line therapies.
Dopamine agonists and COMT inhibitors (e.g.
entacapone) are helpful as adjuvant drugs for patients
with loss of effect at the end of the dose interval, and
to reduce ‘on–off’ motor fluctuations.
The benefit of early treatment with an MAO-B
inhibitor
, selegiline, to retard disease progression is
unproven, and it may even increase mortality.
Polypharmacy is almost inevitable in patients with
longstanding disease.
Ultimately
, disease progression requires increasing drug
doses with a regrettable but inevitable increased
incidence of side effects, especially involuntary
movements and psychosis.
Anticholinergic drugs reduce tremor, but dose-limiting
CNS side ef
fects are common, especially in the elderly.
These drugs are first-line treatment for parkinsonism
caused by indicated (essential) antipsychotic drugs.
cervical dystonia (torticollis), jaw-closing oromandibular dysto-
nia and adductor laryngeal dysphonia. Botulinum A toxin is
given by local injection into affected muscles, the injection site
being best localized by electromyography. Recently, it has also
proved successful in the treatment of achalasia. Injection of
botulinum A toxin into a muscle weakens it by irreversibly
blocking the release of acetylcholine at the neuromuscular junc-
tion. Muscles injected with botulinum A toxin atrophy and
become weak over a period of 2–20 days and recover over two
to four months as new axon terminals sprout and restore trans-
mission. Repeated injections can then be given. The best long-
term treatment plan has not yet been established. Symptoms
are seldom abolished and adjuvant conventional therapy
should be given. Adverse effects due to toxin spread causing
weakness of nearby muscles and local autonomic dysfunction
can occur. In the neck, this may cause dysphagia and aspiration
into the lungs. Electromyography has detected evidence of
systemic spread of the toxin, but generalized weakness does
not occur with standard doses. Occasionally, a flu-like reaction
with brachial neuritis has been reported, suggesting an acute
immune response to the toxin. Neutralizing antibodies to botu-
linum toxin Acause loss of efficacy in up to 10% of patients.
Botulinum B toxin does not cross-react with neutralizing anti-
bodies to botulinum toxin A, and is effective in patients with
torticollis who have botulinum toxin A-neutralizing antibodies.
The most common use of botulinum is now cosmetic.
AMYOTROPHIC LATERAL SCLEROSIS (MOTOR
NEURONE DISEAS E)
Riluzole is used to extend life or time to mechanical ventila-
tion in patients with the amyotrophic lateral sclerosis (ALS)
form of motor neurone disease (MND). It acts by inhibiting
the presynaptic release of glutamate. Side effects include nau-
sea, vomiting, dizziness, vertigo, tachycardia, paraesthesia
and liver toxicity.
MYASTHENIA GRAVIS
PATHOPHYSIOLOGY
Myasthenia gravis is a syndrome of increased fatiguability and
weakness of striated muscle, and it results from an autoimmune
process with antibodies to nicotinic acetylcholine receptors.
These interact with postsynaptic nicotinic cholinoceptors at the
neuromuscular junction. (Such antibodies may be passively
transferred via purified immunoglobulin or across the placenta
to produce a myasthenic neonate.) Antibodies vary from one
patient to another, and are often directed against receptor-pro-
tein domains distinct from the acetylcholine-binding site.
Nonetheless, they interfere with neuromuscular transmission
by reducing available receptors, by increasing receptor turnover
by activating complement and/or cross-linking adjacent recep-
tors. Endplate potentials are reduced in amplitude, and in some
fibres may be below the threshold for initiating a muscle action
CHOREA
Theγ-aminobutyric acid content in the basal ganglia is reduced
in patients with Huntington’s disease. Dopamine receptor
antagonists (e.g. haloperidol) or tetrabenazine suppress the
choreiform movements in these patients, but dopamine antag-
onists are best avoided, as they themselves may induce dyski-
nesias.Tetrabenazineis therefore preferred. It depletes neuronal
terminals of dopamine and serotonin. It can cause severe dose-
related depression. Diazepammay be a useful alternative, but
there is no effective treatment for the dementia and other mani-
festations of Huntington’s disease.
DRUG-INDUCED DYSKINESIAS
The most common drug-induced movement disorders are
‘extrapyramidal symptoms’ related to dopamine receptor
blockade.
The most frequently implicated drugs are the
‘conventional’ antipsychotics (e.g. haloperidoland
fluphenazine).Metoclopramide, an anti-emetic, also
blocks dopamine receptors and causes dystonias.
Acute dystonias can be effectively treated with parenteral
benzodiazepine (e.g. diazepam) or anticholinergic (e.g.
procyclidine).
Tardive dyskinesia may be permanent.
Extrapyramidal symptoms are less common with the newer
‘atypical’ antipsychotics (e.g. olanzapineor aripiprazole).
NON-DOPAMINE-RELATED MOVEMENT
DISORDERS
‘Cerebellar’ ataxia – ethanol, phenytoin
Tremor
β-Adrenoceptor agonists, e.g. salbutamol;
caffeine;
thyroxine;
SSRls, e.g. fluoxetine;
valproate;
withdrawal of alcohol and benzodiazepines.
vestibular toxicity – aminoglycosides;
myasthenia – aminoglycosides;
proximal myopathy – ethanol, corticosteroids;
myositis – lipid-lowering agents – statins, fibrates;
tenosynovitis – fluoroquinolones.
TREATMENT OF OTHER MOVEMENT
DISORDERS
TICS AND IDIOPATHIC DYSTONIAS
Botulinum A toxin is one of seven distinct neurotoxins pro-
duced by Clostridium botulinum and it is a glycoprotein. It is
used by neurologists to treat hemifacial spasm, blepharospasm,
MYASTHENIA GRAVIS 129