Ross & Wilson Pharmacology 2025
Ross & Wilson Pharmacology 2025
ALGrawany
Table of Contents
Cover image
Title page
Copyright
Preface
Acknowledgements
Dedication
Administration
Drug interactions
Introduction to Pharmacodynamics
Principal Targets of Drug Action
Drug Dependence
Introduction
Histamine
Glutamate
Antipsychotic drugs
Anaesthetics
Anticonvulsants
Introduction
ALGrawany
Opioids
Introduction
Cardiac Arrhythmias
Haemostasis
Introduction
Antiemetics
11. Antimicrobial Drugs
Antibacterial Drugs
AntiViral Drugs
Antifungal Drugs
Antiparasitical Drugs
Introduction
Drugs that Interfere with One or More Stages of the Cell Cycle
Hormone-Responsive Cancers
Immunotherapy
Index
ALGrawany
Copyright
© 2025, Elsevier Limited. All rights are reserved, including those for text and data
mining, AI training, and similar technologies.
Publisher’s note: Elsevier takes a neutral position with respect to territorial disputes or
jurisdictional claims in its published content, including in maps and institutional
affiliations.
The right of Allison Grant to be identified as author of this work has been asserted by
her in accordance with the Copyright, Designs and Patents Act 1988.
This book and the individual contributions contained in it are protected under copyright
by the Publisher (other than as may be noted herein).
Notices
Practitioners and researchers must always rely on their own experience and
knowledge in evaluating and using any information, methods, compounds or
experiments described herein. Because of rapid advances in the medical sciences, in
particular, independent verification of diagnoses and drug dosages should be made. To
the fullest extent of the law, no responsibility is assumed by Elsevier, authors, editors
or contributors for any injury and/or damage to persons or property as a matter of
products liability, negligence or otherwise, or from any use or operation of any
methods, products, instructions, or ideas contained in the material herein.
ISBN: 978-0-7020-8098-2
Printed in India
ALGrawany
Preface
Records dating from ancient civilisations in Sumeria, Egypt, India, and China show the
origins of medicine and mankind’s attempts to understand and treat disease. Healing
practices in antiquity often relied on the intervention of gods, the wearing of talismans
to ward off disease, a strong belief in magic, and the use of ceremonial rituals to increase
the patient’s chance of recovery; however, the concept that external substances
swallowed, applied topically, or inhaled could influence outcomes was also very familiar
to the earliest physicians. These early medicines were derived from the natural world
and included plant, animal, and mineral extracts; most were useless, and many were
actively harmful.
Fast forward to the 21st century, and the use of drugs to prevent, cure, or manage
diseases has become a cornerstone of therapeutics. Very few human diseases or
disorders are managed without the use of drugs, and all healthcare professionals require
a thorough, evidence-based understanding of clinical pharmacology to prescribe safely,
to monitor the effectiveness of treatment, and to competently advise and inform patients
from a solid, evidence-based knowledge base. This text has been written with this role in
mind. It goes beyond the simple listing of drugs, their actions, and side-effects and
explains the science underpinning the mechanisms of action of drugs, but without being
bogged down with excessive detail. Understanding what a drug does to a living system
and how it exerts its biological effects explains the rationale behind its use in the disease
for which it is being used and the reason why it may have apparently unconnected side-
effects. Understanding how the human body responds to a drug introduced into its
living tissues is an essential prerequisite for safe prescribing and when monitoring
response to treatment. It ensures that the prescriber chooses the most appropriate
medicine and drug formulation and, as far as possible, anticipates problems arising
from inter-individual drug responses, interactions, and issues.
The ‘focus boxes’ used in several chapters are designed to encapsulate and present key
content, emphasising important concepts and topic areas. ‘Key points’ boxes and ‘key
definitions’ boxes have also been used to help navigation and to emphasise significant
terms.
The pharmacology described here is anchored firmly in its scientific basis, but with a
clear clinical slant. This textbook should be a useful resource for students and
practitioners in health-related professions and would also be an appropriate resource
for students in a range of life science programmes with an element of pharmacological
content. Together with its companion book, Ross and Wilson’s Pathophysiology, this
text is designed to provide a fundamental but thorough grounding in the areas of disease
and therapeutics. Both books have been written for you, the student; I hope this
textbook meets your needs and is of direct and measurable benefit to your studies and
your practice. I am happy to receive feedback, suggestions, and comments; academic
resources are always improved by collaboration and discussion!
Allison Grant
March 2024
ALGrawany
Acknowledgements
Without the (sometimes unseen but always appreciated) efforts of a range of people, this
textbook would never have made it all the way from a few undefined ideas rattling
around in my head to the finished product you now hold in your hands. I think of it as
‘my book’, because the time and effort required to prepare it has extended over 3 years,
greedily consuming evenings and weekends and at times crowding out everything else
going on in my life, though I am very conscious of and grateful for the support of family
and friends and the huge role played by the team at Elsevier, because it would never
have happened without them.
I thank Pauline Graham, formerly of Elsevier, who suggested and initiated the project
in the first place. My gratitude is also due to Anne Waugh, who knows what I owe to her
over the 20-plus years that we have been collaborators, co-authors, and friends. Thank
you, Anne, for your time in reviewing some of this content and for everything else you’ve
contributed to this project.
I thank all those at Elsevier who have had any part to play in the realisation of this
project. I have no direct contact with the experts who deal with page layout, typesetting,
or any of the other stages in converting my digital files, figure lists, and so on to the
finished pages, but I thank you very much. In particular, I owe a huge debt of gratitude
to Fariha Nadeem, who has been a constant, steady, and rock-solid support. I am also
very pleased to thank Marie Dean, who managed to translate my coloured scrawls and
scribbled instructions into the book’s clear and colourful artwork.
I’ve been in higher education for a good few years now and taught thousands of
students, and it is for you all that academics like me pick up our pens and write
academic textbooks. Thank you for the inspiration. Stay curious, stay interested, keep
reading, take nothing at face value, always look for the evidence base, always think ‘but
why…???’, keep asking questions, don’t expect me (or my academic colleagues) to have
all the answers, and be prepared to go out and find the answers for yourselves. We are
giving you the foundation, but you are the next generation and will be standing on our
shoulders.
And finally, to my much-loved circle of family and friends, thank you for everything.
None of you helped me write any of this, so you can’t be blamed for any errors, but none
of this would have happened without you.
Allison Grant
March 2024
ALGrawany
Dedication
For my sun, moon, and stars, Seona, Struan, and Will: the adventures go on!!
1: The Story of Pharmacology
The history of human drug use from its earliest origins, for which we only have
archaeological evidence, through the scientific revolution of the 18th and 19th centuries
up to today’s position as one of the key pillars of modern medicine, is one of the most
interesting stories in science. One current definition of pharmacology from the British
Pharmacological Society is ‘the science of drugs and their effects on living systems’. A
‘drug’ can be defined as any chemical substance, natural or synthetic, that affects some
aspect of the biology of a living system. This broad definition therefore encompasses not
just the clinically useful drugs that are the subject of textbooks like this one aimed at
students and practitioners in medical and healthcare professions, but also food
additives, environmental pollutants, dyes and other chemicals, and a wide range of
substances derived from the natural world, which includes some of the most toxic
chemicals known. The conventional use of the word ‘drug’ implies a substance used to
diagnose, treat, or prevent a medical condition as well as substances used recreationally,
and the term ‘clinical pharmacology’ came into use in the 1950s to refer to all aspects of
research, policy, safety, teaching, and use of drugs in humans.
ALGrawany
the use of a drug is a scrap of Egyptian papyrus dating from 3500 BC describing alcohol
production, but archaeological evidence from even earlier periods indicates that
intoxicants were in use well before then.
The earliest recorded drugs were psychotropic agents used for recreational, ritual, and
medicinal purposes, and include alcohol, opium, and cannabis. Alcohol was being
fermented at least 10,000 years ago. Opium, the juice obtained from the seed head of
the opium poppy (Fig. 1.1), was cultivated, distributed, and sold widely throughout
large areas of the Middle East, Europe, and beyond. Its analgesic, euphoric, and sedative
properties were well understood, as was its lethality: it was used to ease the passage into
death and occasionally as a means to dispose of enemies. Archaeologists have found
little opium flasks in graves and other archaeological sites, in the shape of the opium
poppy seed head (Fig. 1.2), and analysis has shown that they contained opium and
sometimes other psychoactive substances. Hemp has been in use for production of
fibres for clothing and rope for thousands of years. The use of cannabis, derived from
hemp, also dates back to pre-history and was well-known in multiple cultures including
ancient China, the Middle East, Greece, and Rome.
FIG 1.1 The seed head of the opium poppy.Once the petals have
fallen off, exposing the seed head, opium is collected from vertical
cuts made in the seed head. From Garg A (2010) Implant dentistry,
2nd ed, Fig. 10.2. Philadelphia: Mosby.
ALGrawany
ALGrawany
FIG 1.2 Clay vessel from Crete in the shape of the opium poppy
seed head. From Karageorghis V, Kanta A, Stampolides NC, et al.
(2014). Kypriaka in Crete: from the bronze age to the end of the
archaic period. Zurich: A.G. Leventis Foundation.
The plant kingdom was mankind’s main source for medicinal and recreational drugs
for thousands of years, and there is evidence that in some cultures, manufacturing took
place on a large scale. In the late 2000s, a large kitchen dating from 2000 BC was found
in Ebla in Syria, close to the city palace. There was no evidence of food remains, but
there were traces of a range of plants used in medicine, including opium, chamomile
(used as an anti-inflammatory), and heliotrope (used to treat infections), indicating a
small-scale but well-established and well-organised drug preparation industry. In the
era before written records, knowledge of useful plants and how to prepare extracts must
have been part of a people’s oral tradition. Development of traditional medicines over
the centuries, with none of today’s understanding of chemistry, physiology, genetics, or
molecular biology, was largely a process governed by careful observation and trial and
error. That is not to dismiss natural remedies as ineffective or without an evidence base:
the use of opium as an analgesic and sedative in ancient cultures is proof of that. Many
substances found in the natural world are highly potent: extracts of foxglove (Digitalis
purpurea, Fig. 1.3) were used for centuries to treat dropsy. Dropsy is an obsolete term
for oedema, which usually collects in the lower limb. Although it can be due to several
conditions, one of the most common is heart failure, and the use of foxglove extract,
which contains digitalis, improved heart function and relieved the oedema. Digitalis is
still used today to treat heart failure and some arrhythmias.
FIG 1.3 Digitalis purpurea (foxglove) From Renneberg R (2023)
ALGrawany
Biotechnology for beginners, 3rd ed, Fig. 7.30. Philadelphia:
Academic Press.
The scientific revolution of the 16th to 19th centuries brought rapid development
across the biological, physical, and chemical sciences, driven by the change in thinking
that is the scientific method––the process by which a researcher draws factual and
substantiated conclusions after careful experimentation and rigorous testing of a
hypothesis. Key to the emergence of pharmacology as a separate speciality were the
advances made in chemistry, particularly organic chemistry, and physiology. As the
early chemists learned how to extract, separate, synthesise, purify, and concentrate
substances of interest, it became possible to test accurately measured drug quantities in
biological systems. The first drug to be isolated from its crude extract was morphine,
purified from opium by Friedrich Setürner in 1804 (Fig. 1.4). As physiological
knowledge advanced, so did the understanding of what drugs were doing to the body
and how they acted, and drug use in medicine began very slowly to move away from
traditional, often arbitrary, frequently useless, and potentially lethal practices towards a
more evidence-based approach to therapeutics grounded in observation, measurement,
analysis, and pooled knowledge.
The development of analytical chemistry directly and indirectly produced whole new
classes of drugs, many of which remain in widespread use. For example, the synthetic
dye industry emerged in the mid-1850s and spawned a range of clinically important
drug groups. The first dye to be used medicinally was methylene blue, the same dye
used by Paul Ehrlich to demonstrate the presence of the blood–brain barrier (p. 12). In
1891, he was trying to stain the protozoa Plasmodium, which causes malaria, with
methylene blue to study it under the microscope. He observed that the dye inhibited the
activity of the protozoa and reasoned that it might be a useful agent to treat malaria in
humans; as a result, methylene blue was used for this purpose until the early 20th
century, when newer drugs superseded it. Phenothiazine antipsychotics such as
chlorpromazine, antihistamines such as promethazine, tricyclic antidepressants
such as amitriptyline, thiazide diuretics such as bendrofluazide, and sulphonamide
antibacterials such as sulphamethoxazole are all derived from the synthetic dye
industry.
FIG 1.4 Friedrich Sertürner From Göttmann F (1999) Paderborn -
Geschichte der Stadt in ihrer Region. Band 2. Die frühe Neuzeit:
gesellschaftliche Stabilität und politischer Wandel. Berlin:
Schöningh.
In the first half of the 20th century, a plethora of new drugs appeared on the scene.
Generally, they were found by accident; drugs developed as a side-line of the dye
industry for example, or Alexander Fleming’s chance observation that Penicillium
moulds inhibited bacterial growth in his test plates, leading to the isolation of
penicillin (Fig. 1.5). More targeted drug development became possible as research
into the molecular basis of drug action rapidly advanced. Increasing knowledge of
receptor science, enzyme structure, and molecular and cell biology expanded our
understanding of how drugs interact with specific molecules to produce their biological
effects.
Biologics
In the last 40 years or so, biotechnology has generated a wide range of new therapeutics
ALGrawany
called biologics. Biologics are drugs whose active substance is made by a living
organism, including genetically engineered bacteria, and include antibodies, hormones,
immunomodulators, vaccines, cytokines, blood products, and growth factors.
Pharmacogenomics
Advances in genetics have shone a bright light onto the relationship between our genes
and disease, and on an individual’s response to drugs. This new field,
pharmacogenomics, is an important element in personalised medicine. It has been
estimated that between 30% and 60% of drug treatments are ineffective because of
genetically determined responses to the medication. Identifying important genetic
variations between individuals allows optimal therapeutic choices to be made based on
predicted drug responses and likely side-effects. This principle is illustrated in Fig. 1.6.
The group of patients shown in Fig. 1.6 all have the same disease, and all are being
treated with the same dose of the same drug. Within this group, the patients depicted in
green are predicted to respond well. The patients depicted in blue are predicted to
respond poorly, if at all, and for these patients a different dose or a different drug would
be a better choice. The patients depicted in red are predicted to experience significant
toxicity, and this drug should not be used in these patients.
ALGrawany
candidates can be tested to see if they inhibit the enzyme. Of these initial screens,
compounds with the most promising profiles are taken forward into more
comprehensive studies. These are called lead compounds. Most screened compounds do
not make it to this stage for one or more reasons; perhaps their molecular weight is too
high, they demonstrate toxicity in cell lines or animal studies, they may not be potent
enough, or they fail to show expected activity in animal studies.
Pre-clinical development
The small number of candidate compounds, typically 10–20, that make it through the
earlier stages are taken to pre-clinical development. This involves toxicity screening and
pharmacokinetic assessment (absorption, distribution, metabolism, and excretion) in
computer modelling, cell lines, and animal models. The test compounds will also
undergo chemical assessment looking at formulation options and any limitations to
large-scale synthesis. Drug candidates that perform satisfactorily in preclinical trials can
go forward to clinical trials in humans.
Clinical trials
There are four main stages, and the candidate drug can be stopped at any stage if
thought appropriate. In phase 1 studies, the candidate drug is given in small doses to a
small group, usually 20 to 100, of healthy volunteers. The volunteers are monitored
carefully for any adverse effects. In phase 2 studies, the drug is given to larger groups
(100–500) of patients suffering from the condition(s) for which the candidate drug is
thought to be potentially useful. Most drugs that fail at clinical trial do so during phase 2
studies, because evidence emerges of unacceptable adverse effects or toxicity, or the
drug turns out to be inadequately potent. Phase 3 trials are much larger, double-blind,
randomised clinical trials in which the candidate drug is tested at different doses in
groups of thousands of patients against current treatments and/or placebo. Assuming
the phase 3 trials show the drug to be effective, non-toxic, and not associated with
unacceptable side-effects, the company can apply to the relevant national regulatory
board for approval to bring it to market. Phase 4 trials are the processes by which the
performance of drugs on the market are monitored and rare and/or chronic adverse
ALGrawany
effects picked up. They also include any special trials required to allow the drug to be
licensed in specific groups of patients, e.g. children.
Drug names
A drug may have three names: its chemical name, its generic name, and its proprietary
name.
The full chemical name is often long, unwieldy, and rarely appropriate for use in
clinical settings. The recommendation to take two tablets of N-(4-hydroxyphenyl)
acetamide for a headache would not mean much to most people; recommending two
paracetamol tablets on the other hand is a much clearer and more familiar
instruction.
The generic (non-proprietary) name of the active drug is the name approved by an
expert regulatory body and used internationally. Paracetamol is the generic name for
this drug assigned by the World Health Organisation and is the name used outside the
US; it is called acetaminophen in the US because that is the generic name given to it
by American regulatory bodies.
Proprietary names are assigned by a manufacturer to their own formulation of a drug.
Paracetamol can be sold under the brand names Tylenol, Panadol, Metacin, Calpol,
Crocin, Hedex, and others. It is important to realise that there is not always
bioequivalence between two brands of the same dose of the same drug, meaning that
depending on the formulation, preparations from different manufacturers may give
different rates and extent of absorption. Plasma levels 1 hour after taking 500 mg of
product A from one manufacturer may be very different to plasma levels 1 hour after
taking 500 mg of exactly the same drug but as proprietary product B from another
company. Changing from one brand to another can cause issues with drugs for which
even slight changes in plasma levels can cause adverse effects, e.g. anticoagulants and
anticonvulsants.
References
1. Dollery C.T. Clinical pharmacology-the first 75 years and a view of the future. Br J
Clin Pharmacol. 2006;61(6):650–665.
2. Wainwright M. Dyes in the development of drugs and pharmaceuticals. Dyes
Pigm. 2008;76(3):582–589.
Online resources
Adams J.U. Pharmacogenomics and personalized medicine Available
at:. https://www.nature.com/scitable/topicpage/pharmacogenomics-
and-personalized-medicine-643/, 2008.
Genomics Education Programme. What is pharmacogenomics? Available
at:. https://www.genomicseducation.hee.nhs.uk/blog/what-is-
pharmacogenomics/#tab-id-4, 2018.
Matyszak P. Happy plants and laughing weeds: how people of the ancient world
used – and abused – drugs Available
at:. https://www.historyextra.com/period/ancient-history/ancient-
drug-use-history-how-what-for-opium-hemp/, 2019.
Torjesen I. Drug development: the journey of a medicine from lab to shelf Available
at:. https://pharmaceutical-journal.com/article/feature/drug-
development-the-journey-of-a-medicine-from-lab-to-shelf, 2015.
ALGrawany
2: Pharmacokinetics:
How the Body Affects the Drug
Renal Excretion
Half-Life and Steady State
Half-Life
Steady State and the Therapeutic Dose Range
Therapeutic Drug Monitoring
Administration
Drug Formulations
Oral Administration
Topical Administration
Intravenous Administration
Intramuscular Administration
Subcutaneous/Intradermal Administration
Inhalation
Intrathecal and Epidural Administration
Administration into Body Cavities
Drug Interactions
Pharmacokinetic Interactions
Absorption
Distribution
Excretion
Pharmacodynamic Interactions
Beneficial Interactions
Unwanted/Dangerous Interactions
Pharmacokinetic Special Situations
Older Age
Babies and Children
Pregnancy
Genetic Variability
ALGrawany
Pharmacokinetics is the umbrella term which describes how the tissues and organs of
the body deal with biologically active drugs, sometimes simply put as ‘what the body
does to the drug’. The body is very good at dealing with foreign or unwanted substances,
and an understanding of the physical and chemical principles governing these processes
is an important part of clinical pharmacology. For example, pharmacokinetic data
informs dosing regimens, predicts plasma levels (important to ensure that dose and
dose frequency are appropriate), and guides a prescriber when inter-individual
variability is likely to affect drug response. Discussion of the four key pharmacokinetic
processes––absorption, distribution, metabolism, and excretion, often referred to by the
mnemonic ADME––forms the core of this chapter, along with an explanation of
relevant key concepts such as half-life and steady state.
FIG. 2.1 The cell membrane. From Waugh A and Grant A (2020)
Ross & Wilson anatomy and physiology in health and illness, 14th ed,
Fig. 3.2A. Oxford: Elsevier.
ALGrawany
concentration gradient to ensure that drug diffusion and absorption continues.
However, not all drugs travel equally well across plasma membranes. The main factors
determining the efficiency of drug transfer through membranes are the lipid (fat)
solubility, molecular weight (MW), and electrical charge (degree of ionisation) of the
drug molecules.
Fat Solubility
The plasma membrane is essentially a lipid sheet, and to transfer rapidly from one side
to the other, drug molecules must be fat-soluble. The more fat-soluble the drug, the
faster and more efficiently it can transfer. Water-soluble drugs are repelled by the
phospholipid bilayer and transfer slowly, and so travel less well through body tissues.
Most drugs used in clinical practice are highly fat-soluble.
Molecular Weight
The plasma membrane is thin but densely packed, and larger molecules travel through it
less easily than smaller molecules. Water molecules, despite being insoluble in fat, are so
small (MW 18) that they travel freely through plasma membranes. To facilitate efficient
movement around the body, drug molecules are generally small, with MWs less than
600, and drugs with a MW of 80,000 or more transfer very slowly, if at all.
Degree of Ionisation
The degree to which a drug molecule is ionised (electrically charged) affects its ability to
transfer across cell membranes because it affects its water solubility and therefore its
ability to pass through the lipid-rich cell membrane. The more highly ionised (charged)
the drug is, the more water-soluble it tends to be, and it is repelled by membrane lipids,
preventing its transfer. The degree of ionisation is affected by pH, as will be explored in
more detail later; this is an important concept because there are significant differences
in pH across different areas of the body.
Ion Channels
These channels allow ions to travel across the membrane to the other side (Fig. 2.2B).
They are found in all cell membranes, but are particularly important in nerve and
muscle cells, which rely on the movement of ions to generate and propagate electrical
currents.
Facilitated Diffusion
As in simple diffusion, in facilitated diffusion drug molecules move passively down their
concentration gradient, but at a faster rate because they are facilitated by a special
carrier molecule in the membrane (Fig. 2.2C). Like simple diffusion, this is also a
passive movement because no energy is required. The tissues richest in these carrier
molecules are the intestines, kidney, and liver, the cells of which require to actively
import a range of substances: the intestines for absorption, the kidneys for excretion,
and the liver for metabolism. Carrier mechanisms allow relatively water-soluble drugs to
transfer across plasma membranes, for example, the secretion of penicillin and
diuretics into the filtrate in renal tubules. Other examples include vitamin B12
absorption in the intestine. These carrier mechanisms can represent a rate-limiting step
in the transfer of drugs from one side of the membrane to the other. Once carrier
mechanisms are fully saturated and operating at their maximal capacity, increasing drug
concentrations does not increase transfer.
ALGrawany
Active Transport
Active transport mechanisms use energy in the form of ATP to move a molecule from
one side of the membrane to another and are useful when the drug is poorly lipid-
soluble and diffusion rates are low (Fig. 2.2D). They also allow a drug to be moved
against its concentration gradient, i.e. from low to high. Active transport mechanisms
are frequently involved in the transfer of cytotoxic drugs including 5-fluorouracil, as
well as iron salts, levodopa, and propylthiouracil. A point worth noting regarding
active transport mechanisms is that they have a maximum work rate, and so there is an
upper limit on the speed of drug transfer: the mechanism can become saturated at
higher drug concentrations.
FIG. 2.3 The pH scale. From Waugh A and Grant A (2020) Ross &
Wilson anatomy and physiology in health and illness, 14th ed, Fig.
3.2A. Oxford: Elsevier.
ALGrawany
FIG. 2.4 The effect of pH on the ability of drugs to cross biological
membranes.A. The absorption of basic drugs is inhibited in the
stomach because the drug molecules attach H+ ions and become
ionised, but it is facilitated in the small intestine, where they lose the
H+ ions and revert to their uncharged form. B. The absorption of
acid drugs is facilitated in the acidic environment of the stomach,
where the molecules remain un-ionised, but it is slower in the
alkaline environment of the small intestine, because they give up H+
ions and acquire a net negative charge.
ALGrawany
they ionise and are trapped.
Absorption
Absorption is the process by which drug molecules travel from their site of
administration across biological membranes into the bloodstream. The term
bioavailability refers to the percentage of administered drug that reaches the systemic
circulation. Drugs given intravenously therefore have 100% bioavailability, but the
figure is often much lower for drugs given orally, due to drug degradation in the
intestine, first-pass metabolism (see below), and/or incomplete absorption. As
described above, the properties of a drug molecule that promote absorption are small
size, high fat solubility, and little overall electrical charge (an un-ionised state). Some
drug molecules are too large, too water-soluble, and/or too highly charged to transfer
across cell membranes, but this can be clinically useful. For example, nystatin is too
toxic to be used systemically, but because it is not absorbed from the gut, it can be given
orally to clear fungal infections of the gastrointestinal (GI) tract.
Most absorption of an orally administered drug takes place in the small intestine, and
to get there it must pass through the stomach. The gastric environment is designed to be
hostile to protect the body against potentially dangerous ingested substances: gastric
fluids are strongly acidic, which denatures peptides and proteins, and contain the
proteolytic enzyme pepsin. Some drugs are completely destroyed here, and others
significantly are degraded. Enteric-coated formulations can protect the drug until it
reaches the intestine.
The rate of gastric emptying is a major determinant of the speed at which a swallowed
drug reaches the duodenum, and therefore factors that reduce gastric motility delay
drug absorption. Gastric emptying is generally fastest when a drug is taken on an empty
stomach with a glass of cold water. Gastric emptying is slowed in pain, in shock, in
neuropathies (including diabetic neuropathy), and when there are foodstuffs in the
stomach, especially fatty foods. Some drugs, including opioids and drugs with
antimuscarinic activity, reduce gastric motility. Some foodstuffs can bind certain
drugs, reducing their absorption; for example, tetracyclines should not be taken with
dairy products because they bind the calcium present in these food items, forming an
insoluble complex which cannot be absorbed.
Conditions that reduce blood flow through the GI tract, like heart failure, can also
slow absorption.
Distribution
Distribution is the transfer of a drug from the bloodstream into body tissues and fluids.
Drugs do not distribute evenly throughout body tissues, and the main principles
governing this are explained below. For example, water-soluble drugs tend to remain in
the plasma, whereas very fat-soluble drugs are rapidly taken up and concentrated in
body fat stores. Drug delivery is usually proportional to the blood flow: the higher the
blood flow, the greater the drug delivery to the area. The kidneys, central nervous
system (CNS), and liver receive a high proportion of cardiac output relative to their
weight, and therefore can be exposed to particularly high levels of any circulating drug.
ALGrawany
and fluids, and given enough time, the drug will equilibrate between these
compartments (Fig. 2.7). For example, circulating plasma levels of a drug which enters
the cerebrospinal fluid (CSF) will equilibrate between the blood and CSF, and, provided
plasma levels remain constant, CSF levels will also remain constant. However, if plasma
levels fall because dosing is stopped, drug levels in the CSF will also fall as drug
redistributes from the CSF into the blood to maintain equilibrium. This two-way
movement of drug is indicated in Fig. 2.7 by the two-way arrows.
Body fat (adipose) tissue is an important compartment when considering drug
distribution because fat-soluble drugs are taken up and stored here. The percentage
contribution to total bodyweight from fat varies considerably and can make a significant
difference to drug dosages and plasma half-lives.
Drug levels in the body are usually measured as plasma concentrations because it is
easy and convenient to take blood samples; however, it must be remembered that
probably only a small fraction of the drug is likely to be found in the blood because most
of it will have distributed out of the plasma and into the tissues. Assuming the drug
molecules are capable of efficient transfer across biological membranes, they are likely
to access all body tissues and fluids, although this is influenced by how much of the drug
is bound to plasma proteins and how much is free (see below). Because of this extensive
distribution, very little of the administered dose reaches its target tissue.
Volume of Distribution
The volume of distribution (Vd) is an important concept in clinical pharmacokinetics
because it gives an indication of how much of the total body drug has been retained in
the plasma and how much has distributed into other body tissues and fluids. This is
important when deciding what dose of drug to use to achieve a desired plasma level, and
it determines the half-life (see below) of the drug.
To calculate the Vd, plasma levels are measured following administration of a known
quantity of drug. In a hypothetical example, 100 mg of drug A is given intravenously.
After the drug has been allowed to distribute out of the blood and into the tissues, the
drug plasma concentration is measured. In our hypothetical case, the plasma level of
drug A is 1 mg/L. Vd is calculated by dividing the total amount of drug in the body (100
mg) by plasma levels of that drug (1 mg/L), giving a Vd of 100 L. This is the total volume
of fluid that would be needed for our 100 mg of drug to achieve a homogenous plasma
concentration of 1 mg/L. Clearly, the body does not possess 100 L of plasma, and so
most of the drug must have left the plasma and entered other body compartments (Fig.
2.8A). Compare this to an intravenous (IV) 100 mg dose of hypothetical drug B. If the
plasma concentration following administration of 100 mg of drug B is 20 mg/L, the Vd is
100 mg/20 mg/L, or 5 L. This shows that a much higher proportion of the drug has
ALGrawany
remained in the plasma and much less has distributed into peripheral tissues, and in
this example, much lower doses of drug B are likely to be needed to reach a desired
plasma level than drug A (Fig. 2.8B).
Therefore, the more of an administered dose of a drug that escapes from the plasma
into other compartments, including body fat stores and general body tissues, the higher
the Vd. Because the drug must be in the plasma to be cleared from the body, whether by
metabolism or renal excretion, if most of the drug is elsewhere, then clearance is going
to be slow and the plasma half-life (see below) long. High Vd values are therefore
associated with long plasma half-lives, and drug A in the above example will likely have
a much longer half-life than drug B.
FIG. 2.8 Relationship between volume of distribution, plasma
levels, and half-life.A. When most drug leaves the circulation, plasma
levels are low and the volume of distribution is high. B. When most
ALGrawany
drug stays in the circulation, plasma levels are high and the volume
of distribution is small.
Sanctuary compartments
Drug distribution can be limited by membranes specially adapted to reduce their
permeability. The placenta, for example, reduces the transfer of unwanted substances
from the mother’s bloodstream into the fetal circulation. The blood vessels supplying the
testis are also less permeable than standard capillaries, to protect the sperm-producing
stem cells from exposure to potentially toxic substances in the circulation. The largest
sanctuary compartment comprises most of the brain and spinal cord and is protected by
the blood–brain barrier.
ALGrawany
Plasma protein binding
Blood plasma contains a range of large-MW proteins, including albumins, globulins, and
the clotting protein fibrinogen. Most plasma proteins are synthesised in the liver.
Almost all drugs bind loosely and reversibly to plasma proteins, mainly albumin. This
binding is not specific like drug-receptor binding; instead, it is a non-specific
electrostatic attraction between negatively and positively charged groups on the drug
and protein molecules. Bound drug (the ‘bound fraction’) cannot leave the bloodstream
because the drug–protein complex is too large; this means that it does not get into the
liver cells for metabolism and does not pass through the glomerular filter in the kidney.
Bound drug is therefore protected from metabolism and renal excretion. Because it
cannot leave the circulation, bound drug is also unable to reach its target tissue, and so,
although it is chemically and biologically active, it is unable to produce its
pharmacological effect. On the other hand, unbound drug, the so-called ‘free fraction’, is
subject to metabolism and excretion and is free to leave the bloodstream and enter
tissues (Fig. 2.10). The pharmacological effect of the drug is therefore exerted by the
free fraction, and depending on the degree to which a drug is plasma protein-bound, the
free fraction might be only a small proportion of the total drug present in the
bloodstream.
FIG. 2.10 Free and bound drug in the bloodstream.Unlike the free
fraction, drug molecules bound to plasma proteins are unable to
leave the circulation. Modified from Lilley LL, Collins S, and Snyder
J (2023) Pharmacology and the nursing process, 10th ed, Fig. 2.4. St.
Louis: Mosby.
For nearly all drugs at therapeutic concentrations, when administered regularly, the
free and bound drug fractions reach an equilibrium, and the proportion of drug
molecules bound to plasma proteins remains constant. This means that as plasma levels
of the free fraction fall, drug molecules dissociate from their binding sites to maintain
this equilibrium. For example, for a hypothetical drug that is 50% bound and 50% free,
ALGrawany
half the drug molecules are plasma protein-bound and half are free in the plasma. As the
free drug is cleared from the plasma by metabolism or excretion, drug molecules
dissociate from their plasma proteins to maintain the 50:50 ratio (Fig. 2.11). For this
reason, bound drug can be considered a ‘reservoir’, like drugs stored in body fat: present
in the bloodstream, not pharmacologically active, but which can replenish and maintain
plasma levels even after the drug is withdrawn.
Metabolism
Metabolism refers to any enzymatic alteration to the structure of the parent drug. Most
body tissues can metabolise drugs to some degree, but the liver and kidneys are the
main organs of metabolism, and liver and/or renal impairment can significantly reduce
an individual’s capacity to metabolise drugs. Conditions such as heart failure that reduce
blood flow to the liver and kidneys can reduce delivery of the drug to these organs and
reduce the rates of metabolism and clearance. Thyroid disease can also affect drug
metabolism because thyroxine is an important regulator of metabolism.
Hypothyroidism can reduce drug metabolism, whereas hyperthyroidism accelerates it,
and drug doses may need to be adjusted accordingly in these conditions.
The ability of metabolising enzymes to keep up with rising plasma drug levels is not
infinite; this is explored in more detail in the section headed ‘First-order and second-
order kinetics’ below.
Liver cells (hepatocytes) are packed with metabolising enzymes responsible for the
phase 1 and phase 2 reactions described below. Hepatic metabolism therefore relies
upon drugs being able to cross hepatocyte membranes and enter the cell. Because most
drugs used in clinical practice have the chemical properties to do this (small, fat-soluble,
and un-ionised), this is usually straightforward. Larger, highly charged, and more water-
soluble drug molecules such as penicillin, digoxin, and the aminoglycoside antibiotics
(e.g. gentamicin) cannot enter hepatocytes and so are poorly metabolised. The body
relies on renal excretion (see below) to clear such drugs.
ALGrawany
FIG. 2.12 Warfarin and aspirin competing for plasma protein-
binding sites.A. Warfarin alone. B. Introduction of aspirin can
displace bound warfarin and increase free warfarin levels, potentially
causing adverse effects. Modified from Howe T and Burton A (2021)
Pharmacology for the surgical technologist, 5th ed, Fig. 1.14. St.
Louis: Elsevier.
Because the biological activity of a drug is usually tied very closely to its precise
chemical shape and structure, metabolism often reduces or abolishes a drug’s
pharmacological action. However, the primary function of metabolism is not actually to
deactivate drugs, but to make them more water-soluble and enhance their excretion in
the urine. Any product of a metabolic step is called a metabolite.
Key Points
In terms of activity, there are three possible scenarios by which metabolism can affect
drugs:
The two types of metabolism are phase 1 and phase 2 reactions (Fig. 2.13).
Phase 1 metabolism
Phase 1 reactions increase the water solubility of the parent molecule by adding or
exposing one or more charged groups: these are usually oxidation, reduction, or
hydrolysis reactions. The metabolite is often more reactive than the parent molecule,
which prepares it for a phase 2 reaction. This can mean that the metabolite is more toxic
than the original drug: for example, cyclophosphamide, paracetamol, ethanol,
and halothane all produce highly toxic metabolites which must undergo immediate
further metabolism to render them harmless.
ALGrawany
drugs. Poor metabolisers clear drugs more slowly and are more likely to experience side-
effects, whereas rapid metabolisers clear drugs very quickly and may not respond to
normal doses.
Phase 2 metabolism
Phase 2 reactions may follow a phase 1 reaction, or the parent drug may undergo phase
2 metabolism directly. The water solubility of the drug or its metabolite is further
increased by adding a water-soluble group such as an acetyl group or glucuronate.
Adding another group to the drug is called conjugation. As with phase 1 reactions, there
is significant genetic variation in people’s ability to undergo phase 2 reactions: for
example, acetylation is the main mechanism by which several drugs, e.g. isoniazid,
hydralazine, procainamide, and sulphonamides, are metabolised. Poor
ALGrawany
acetylators metabolise these drugs significantly more slowly than fast acetylators.
Table 2.1
ALGrawany
Ciprofloxacin Spironolactone
Isoniazid St. John’s wort
Some foodstuffs including Some foodstuffs including Brussels sprouts,
grapefruit and grapefruit juice broccoli, barbecued meats
First-pass metabolism
This refers to any metabolism of an orally administered drug occurring between the
intestine and systemic circulation (Fig. 2.16). Significant drug loss can occur in the
hostile gastric environment because of the low pH and the action of the proteolytic
enzyme pepsin. The main site of absorption is the small intestine, whose walls are
packed with digestive enzymes, and the drug can be degraded here. Substances
absorbed from the stomach and most of the intestine travel in the hepatic portal vein to
the liver, the organ most strongly associated with first-pass metabolism. The liver has an
important role in detoxification of unwanted substances and regulating the levels of key
blood constituents, and so this arrangement ensures that potentially toxic substances
including drugs, or materials absorbed in excess from the intestines, pass first through
the liver before being delivered into the systemic circulation. After passing through the
liver, the blood then travels in the hepatic vein to the inferior vena cava.
FIG. 2.16 First-pass metabolism. Modified from Townshend A,
Worsfold P, and Poole C (2005) Encyclopedia of analytical science,
2nd ed, Fig. 3. Oxford: Elsevier.
For some drugs, like glyceryl trinitrate, the liver removes 100% of an absorbed
drug, so oral bioavailability is zero. Other drugs, such as morphine, are not completely
eliminated, but most of the dose is destroyed and only a small proportion of the drug
reaches the systemic circulation.
The veins draining the upper and lower parts of the GI tract do not empty into the
hepatic portal vein but deliver their blood directly into the systemic circulation. Drugs
absorbed across the membranes of the mouth and the rectum are therefore not exposed
to first-pass metabolism. For this reason, glyceryl trinitrate is given sublingually.
Drugs are often given rectally for reasons other than avoidance of first-pass metabolism,
e.g. for a local anti-inflammatory action in haemorrhoids or anal fissures. However,
avoiding first-pass metabolism can be a significant advantage when administering drugs
rectally for systemic absorption; analgesics, sedatives, and anticonvulsants may
all be given by this route to maximise the amount of drug reaching the circulation.
Excretion
Excretion is the removal of drugs or their metabolites from the body. The main route of
excretion for most drugs is via the kidney, but drugs may be excreted in any body fluid
ALGrawany
or tissue, including bile, faeces, breast milk, hair, nails, sweat, and exhaled droplets.
Faecal excretion
Orally administered drugs that are poorly absorbed remain in the GI tract and are
excreted in the faeces. In addition, some drugs conjugated in the liver are passed into
the bile, which is then secreted into the duodenum. Conjugated drugs can pass through
the intestines and be excreted in the faeces, but some can be reabsorbed via
enterohepatic recycling.
FIG. 2.17 Enterohepatic recycling.A drug may be conjugated in the
liver and be passed into the bile. Once in the colon, bacteria may
metabolise the drug–conjugate complex, releasing the lipid-soluble
drug again, which can then be reabsorbed into the circulation. From
Waller DG, Sampson A, and Hitchings A (2022) Medical
ALGrawany
pharmacology and therapeutics, 6th ed, Fig. 2.13. Oxford: Elsevier.
Enterohepatic Recycling
Conjugated drugs, which are now very water-soluble, can be passed into the bile and
from there into the intestines. They travel in this form through the tract, too water-
soluble to be reabsorbed. In the colon however, the environment changes: this region is
richly populated with a range of bacteria, producing enzymes which can split the drug
from its conjugated group, releasing the original fat-soluble drug molecule. The drug is
therefore reabsorbed across the tract wall, keeping drug levels in the bloodstream high
(Fig. 2.17). Enterohepatic recycling prolongs plasma levels of a range of drugs,
including morphine, digoxin, and some antibiotics.
Renal excretion
The main functions of the kidney include regulation of the composition of body fluids
and the excretion of unwanted substances, including drugs and their metabolites. Renal
physiology is discussed in more detail in Chapter 9, and the anatomy of the kidney and
its functional unit, the nephron, are shown in Figs. 9.1 and 9.2. Low-MW substances
are filtered under pressure out of the bloodstream into the glomerular capsule, forming
filtrate which travels through the nephron towards the collecting duct. The nephron has
three segments: the proximal convoluted tubule, the loop of the nephron (loop of
Henle), and the distal convoluted tubule. From here, urine, the final product, drains into
the renal pelvis and ultimately into the ureter towards the bladder. As the filtrate passes
through the nephron, important substances like glucose are reabsorbed from the filtrate
back into the bloodstream, and unwanted substances like ammonium ions are actively
secreted out of the bloodstream into the filtrate for excretion in the urine. The amount of
drug present in the urine therefore depends on how much is filtered at the glomerulus,
how much is reabsorbed from the filtrate, and how much is secreted into the filtrate.
Fig. 2.18 summarises these processes, although except for the glomerulus, for clarity, it
does not show the different regions of the nephron. A significant degree of metabolism
may also take place as the drug passes through the renal tissues.
Filtration
Small-MW drugs and metabolites are filtered out of the bloodstream into the filtrate,
although they do not necessarily stay there. Drugs bound to plasma proteins are not
filtered because the drug–protein complex is too large. Drug molecules too large to cross
the glomerular filter also stay in the bloodstream and do not enter the filtrate.
Reabsorption
As the blood flows from the glomerular capillaries into the peritubular capillaries
supplying the remainder of the nephron, further exchange of drugs and their
metabolites may take place. Filtered drugs that are very water-soluble remain in the
filtrate, but if they retain some lipid solubility, they may be reabsorbed across the tubule
wall and return to the bloodstream. Water-soluble drugs like digoxin and gentamicin
are poorly metabolised because they cannot diffuse into hepatocytes and so escape
metabolism and arrive in the kidney in their active forms. As the filtrate passes through
the nephron and most of the water is reabsorbed, they become progressively more
concentrated. Gentamicin causes significant renotoxicity because of this. An
additional consideration here is that the body relies on renal clearance to get rid of
drugs that are not metabolised, and so renal function must be carefully assessed when
these drugs are used. Some drugs are actively reabsorbed, piggybacking on carrier and
transport mechanisms used to reabsorb nutrients and other substances the body needs
to retain, but this is not of much clinical importance as few drugs are not reabsorbed this
way.
Urinary pH also influences drug reabsorption into the bloodstream. Normal urine has
a pH of 5–6, and so acidic drugs will tend to stay un-ionised (uncharged) and be
reabsorbed into the plasma (pH 7.4). Basic drugs will tend to ionise in the acid urine and
so be less able to diffuse out of the tubule back into the bloodstream, promoting their
excretion in the urine (see section on pH and pH partitioning above).
Secretion
Drugs too large to pass through the glomerular filter may be actively secreted out of the
bloodstream into the tubule, allowing the kidney to clear unwanted substances that
escape filtration: examples include furosemide, metformin, digoxin, many
antivirals, and penicillin. The transporters responsible for secretion are saturable,
meaning that they have a maximum work rate. If two or more drugs are competing for
the same transport mechanism and the transport maximum is reached, then the level of
both drugs in the blood may rise. This is an important source of drug interactions: for
example, secretion of digoxin is reduced when co-administered with verapamil or
amiodarone, because these drugs compete for the digoxin transporter, increasing
plasma digoxin levels. Plasma protein binding protects most drugs from secretion,
although diuretics including furosemide are actively secreted in their bound form; this
is the route by which they reach the lumen of the nephron, where they exert their
pharmacological effect (p.174 and Fig. 9.6).
Half-life
The half-life (t1/2) of a drug is the time required for its concentration to reduce by 50%.
It can be measured in any body fluid or tissue, but in practice for convenience it is
generally measured in the plasma. It is an important pharmacokinetic measurement,
although it is not applicable in every situation (see saturation kinetics below).
The relationship between half-life and plasma levels is shown in Fig. 2.20. The x-axis
shows time, and the y-axis shows drug levels, measured in percentage of the starting
drug concentration. After one half-life, the drug level has fallen by half and is now 50%
of the starting concentration. After another half-life, the drug level has fallen by another
50% and is now only 25% of starting levels. Another half-life later, the drug
concentration is now 12.5% of the original.
Bear in mind that drug half-lives, in absolute time, vary hugely. For example, the
plasma half-life of penicillin is less than half an hour, whereas the half-lives of some
antidepressants and some anticonvulsants can be days. Plasma half-lives are
affected by a range of factors. Rapid metabolism and/or excretion shorten the plasma
half-life; penicillin’s short half-life is due to exceptionally efficient renal clearance.
Conversely, poor metabolism and/or sluggish renal excretion extend half-lives. Very fat-
soluble drugs which are stored in large amounts in adipose tissue, or drugs which are
highly plasma protein-bound, also tend to have long half-lives because of these
significant drug reservoirs: when the drug is withdrawn, plasma levels can remain high
for an extended time as drug lost to elimination processes is replaced by drug leaching
from these stores.
In some cases, this direct relationship between drug levels and metabolism is lost.
This is usually because the metabolising enzymes become saturated: that is, they are
working at their maximal capacity and cannot work any harder. This is called saturation
or zero-order kinetics.
Metabolism switches from first- to zero-order kinetics with any drug if the
concentration rises high enough, but fortunately this does not happen with most drugs
within normal therapeutic drug ranges. However, for some drugs, including
phenytoin, salicylates, and sodium valproate, enzyme saturation occurs within the
therapeutic range. This presents a problem in drug management, because when the
enzyme’s maximum work rate is exceeded, additional drug accumulates in the plasma,
and plasma levels rise rapidly and steeply. When adjusting doses of drugs in this
scenario, special care is needed, because even a small dose increase can result in sudden
onset of toxicity. In addition, there is significant variability between individuals, adding
further unpredictability. Fig. 2.21 shows the effect of increasing dose on plasma levels
of phenytoin in three patients. The black line shows the shape of curve expected if
phenytoin metabolism followed first-order kinetics: there is a proportionate,
predictable, and steady rise in plasma levels as the daily dose increases. At lower doses,
all three curves show this relationship, but at plasma concentrations below therapeutic
levels, drug metabolism is maximal and additional drug accumulates, sending plasma
levels sharply and steeply upwards. Patient 1 reaches the maximum therapeutic plasma
level of 80 μM at around 200 mg daily; for patient 3, the corresponding dose is nearly
500 mg, showing significant inter-individual variation.
FIG. 2.21 Saturation kinetics. From Waller DG, Sampson A, and
Hitchings A (2022) Medical pharmacology and therapeutics, 6th ed,
Fig. 2.13. Oxford: Elsevier.
The therapeutic dose range is the interval between the upper desired plasma level,
above which toxic effects are likely, and the lowest desired plasma level, below which the
drug is unlikely to exert a therapeutic action. Steady-state plasma levels should normally
fall within this range. The dose and the dosing frequency should be such that peak levels
do not reach toxic levels and (usually) that trough levels do not fall so low that the
therapeutic action of the drug is lost, i.e. that the steady-state range remains within the
therapeutic dose range for the drug.
Administration
Choosing the appropriate route of administration is crucial to ensure optimal drug
delivery to the desired target site with minimal side-effects. Oral administration is often
the best choice, with the added benefit of often being the cheapest option, but in some
situations an alternative route may be better.
Drug formulations
The active drug is only one component of a medicine. The term given to the combination
of the active drug with other ingredients is its formulation. The formulation determines
the medicine’s physical properties and the pharmacokinetics of the drug, including its
stability, palatability, solubility, particle size, rates of absorption, and rate of release. It
should not be assumed that one manufacturer’s formulation of a drug, even across
equivalent doses, will give identical plasma levels and effects to another’s, and care
should be taken when switching formulations of certain drugs, including
anticonvulsants and digoxin.
Ingredients other than the active drug are called excipients and generally contribute
about 90% to the bulk of the final product. Typical excipients include stabilisers,
colourants, diluents, binding agents, and coatings and films used to seal tablets and
capsules. An ideal excipient is biologically inert, non-toxic, and non-allergenic, but this
ideal is rarely achieved, and excipients can cause adverse reactions to medicines. A
number have been associated with allergic responses: for example, lanolin (wool fat)
used as an emulsifier in some topical preparations, the colouring agent tartrazine, and
the antioxidants sodium metabisulphite and propyl gallate.
Different formulations of the same drug allow it to be given by different routes, to
modify its rate of release from the site of delivery or to target specific cells or tissues.
Hydrocortisone, for example, is formulated differently in preparations intended for
oral, IV, or topical use. Acid-sensitive drugs, e.g. omeprazole, may be formulated as
enteric-coated tablets or capsules to protect them against gastric juices and to release
the drug in the alkaline environment of the small intestine. Conversely, drugs irritant to
the stomach, including non-steroidal anti-inflammatory drugs, can be enteric-
coated to help protect the gastric lining. Drugs given by IM injection may be formulated
in an oil base to give a depot in the muscle, from which the drug is released over an
extended period, removing the need for frequent injections. Examples include
contraceptive preparations and antipsychotic medications. Other modified
formulations include sustained or slow-release preparations, which release their drug
slowly in the intestines and keep plasma levels high over an extended period.
Formulation technology is becoming progressively more sophisticated, producing a
number of novel drug delivery vehicles. For example, liposomes are tiny, artificially
engineered spheres made of lipid, used to deliver drugs that may be poorly absorbed or
poorly delivered into cells because of low lipid solubility or large MW (Fig. 2.23). Fat-
soluble (hydrophobic) drugs can be inserted into the lipid bilayer, and water-soluble
(hydrophilic) drugs can be enclosed within the liposome. For example, many
anticancer drugs are much more water-soluble than lipid-soluble, so their ability to
cross biological membranes is limited. Packaging the drug in liposomes, which readily
cross membranes, greatly enhances drug delivery to its target. In addition, the outer
surface of liposomes can be coated with molecules that bind specifically to target cells,
allowing the drug to be selectively delivered to the desired location. The advantage of
this, for example in cancer chemotherapy, is obvious: cytotoxic drugs cause significant
damage to healthy cells as well as malignant cells, so targeted therapy reduces general
toxicity. Liposomes modified with polyethylene glycol (PEGylated) are more stable than
standard liposomes and reduce the uptake of liposomes by macrophages in the liver and
spleen, prolonging the drug’s presence in the bloodstream.
Oral administration
Advantages of oral administration include convenience and ease. For people with
difficulty swallowing tablets or capsules, liquid formulations are often available,
although some may taste unpleasant. Oral administration is not an option for patients
who are unable or unwilling to swallow, who are actively vomiting, or whose
consciousness is lost or impaired. Additionally, acid-sensitive drugs are destroyed in the
stomach and protein-based drugs are digested or degraded here. If blood flow to the GI
tract is reduced, i.e. in acute pain, developing shock, or heart failure, oral absorption of
medication is slowed. Malabsorption and inflammatory conditions of the GI tract can
also reduce absorption.
Because it usually takes some time for plasma levels to rise, oral administration is not
ideal if a rapid response is needed. To accelerate the rise in plasma levels following oral
administration, an initial loading dose may be used: this increases the concentration
gradient between the intestinal lumen and the bloodstream and speeds up drug transfer
into the blood (Fig 2.24).
Topical administration
Topical absorption means the drug is applied directly to a body surface or membrane.
Often this is the preferred route when localised drug action is needed, e.g.
hydrocortisone cream for a skin allergy. A range of drugs, including anti-
inflammatories and antimicrobials, can be formulated for direct application to the
eye or ear canal. One important advantage of topical application for local effect is that
lower drug concentrations are required, and although some drug is likely to be absorbed
into the circulation, exposure of other body tissues to the drug is greatly reduced.
Application of drugs to the skin and internal mucosal surfaces, e.g. buccal (tucked into
the pouch between the cheek and teeth), nasal, vaginal (per vagina, PV), or rectal
membranes, may also be useful for a local effect, but sometimes the drug is intended to
be absorbed across the membrane and reach the systemic circulation. Drug absorption
across the healthy skin barrier (transdermal absorption) is generally slow, but this can
be advantageous in some circumstances, giving slow and sustained drug release;
examples of drugs that can be given in skin patches include fentanyl, nicotine, and
contraceptive hormones.
FIG. 2.24 Blood drug levels following a loading dose compared to
standard dose.Therapeutic drug levels are reached more quickly
when a loading dose is used. Modified from Taylor K and Aulton M
(2022) Aulton’s pharmaceutics, 6th ed, Fig. 22.8. Oxford: Elsevier.
The mucous membranes of the mouth and rectum have a good blood supply, and
drugs are generally absorbed reasonably well here. Rectal (per rectum, PR), sublingual
(SL, under the tongue), and buccal administration have the advantage that absorbed
drug is not subject to first-pass metabolism (see above). Rectal administration may be
an alternative route for drugs that are destroyed in or irritant to the stomach, like non-
steroidal anti-inflammatory agents. It may also be used when the oral or IV routes
are not available or potentially hazardous, for example the use of benzodiazepines to
terminate a seizure, or in severe vomiting or gastric stasis.
Intravenous administration
Drugs given directly into the bloodstream are usually injected into a vein (IV) rather
than an artery (intra-arterial, IA) because veins are thinner and easier to penetrate, tend
to run more superficially under the skin so are more accessible, and carry blood under
lower pressure so bleeding from the site is usually less likely. First-pass metabolism is
bypassed, and bioavailability is 100%. There is no risk to the drug from gastric acid
degradation, the onset of drug action is rapid, and providing venous access is available,
this route permits medication delivery when the patient is asleep or unconscious. Drugs
may be given as a one-off (bolus) dose and/or a continual infusion and stopped
immediately at any time. Potential risks include infection, bleeding, and local tissue
damage, especially if an in-situ cannula dislodges and infusion continues into local
tissues rather than the vein.
IA injection is used to target delivery of a drug to a particular organ, e.g. cytotoxic
drugs can be injected directly into a tumour’s arterial blood supply.
Intramuscular administration
The main skeletal muscles used for IM injection (Fig. 2.25) are the gluteal muscles of
the buttock, the deltoid muscle which forms the fleshy part of the shoulder, and the
vastus lateralis of the thigh. Skeletal muscle has a good blood supply and drug
absorption is usually reliable; IM administration may be suitable if the patient cannot
take medication orally or other routes are not suitable. Depot formulations given as deep
IM injection release active drug slowly over an extended period, but it must be borne in
mind that the drug cannot then be withdrawn, and adverse effects may therefore be
prolonged. Risks include infection, bleeding, and damage to adjacent structures if the
injection is too deep or not accurately placed, and of course it is likely to be painful.
Absorption from the muscle is reduced if blood flow to skeletal muscle falls, for example
in developing shock.
Subcutaneous/intradermal administration
Immediately below the epidermis of the skin lies the dermis and below that lies the
subcutaneous tissue, which is rich in poorly perfused adipose tissue (Fig. 2.25).
Subcutaneous injection therefore delivers the drug into a very fat-rich environment with
little blood flow, so drugs given this way are absorbed slowly into the bloodstream; this
is ideal when sustained blood levels are required: insulin and heparin are routinely
given by this route. Only small volumes (up to 2 mL) can be delivered by a single
subcutaneous injection, but some drugs, e.g. opioid analgesics in terminal care, can
be administered by subcutaneous infusion. In situations where skin blood flow is
reduced, such as in developing shock, absorption from subcutaneous tissue can be
significantly impaired. Potential complications include infection and abscess formation,
and in the case of repeated injections in the same site, degenerative tissue changes
(lipodystrophy).
FIG. 2.26 Sites of needle placement for intrathecal and epidural
drug administration.CSF, cerebrospinal fluid. From Waugh A and
Grant A (2020) Ross & Wilson anatomy and physiology in health
and illness, 14th ed, Fig 7.14. Oxford: Elsevier.
Inhalation
Inhalation of drugs is usually used to deliver drugs locally into the airways, but
occasionally for systemic drug administration, e.g. inhaled general anaesthetics.
Inhalation, for example, of anti-inflammatory drugs and bronchodilators in
obstructive airway disease such as asthma gives targeted delivery and allows drug doses
to be kept small. It must be borne in mind that airway tissues have a good blood supply,
and drug absorption into the systemic circulation is a real possibility, especially at
higher doses and with frequent administration. Delivery of drugs by inhalation is
discussed in more detail on p. 158.
Drug interactions
Drug interactions refer to the situation where a drug interferes with some aspect of the
pharmacokinetic or pharmacodynamic properties or behaviour of another. They are a
common cause of problems and adverse effects with drug therapy, and consideration
should always be given to interactions caused by foodstuffs, non-prescription medicines,
and herbal and natural remedies.
It must also be borne in mind that not all interactions are harmful or unwanted. For
example, beta agonists like salbutamol and anti-inflammatory steroids such as
budesonide are regularly used together in maintenance treatment of asthma. In
combination, the therapeutic effects of these two drugs synergise and enhance each
other (see also p.166 and Fig. 8.11).
Pharmacokinetic interactions occur when a drug interferes with the disposition of
another in the body, and several examples are discussed in earlier sections of this
chapter. Pharmacodynamic interactions occur when a drug interferes with the biological
activity of another, often by antagonising or acting additively to it. Pharmacodynamic
principles are the subject of Chapter 3, but pharmacodynamic interactions are
discussed here for consistency. This section does not attempt to provide a
comprehensive list of all important drug interactions because the list of potential and
confirmed interactions is vast, but rather offers a selection of examples to illustrate the
main principles. A suitable drug formulary should always be consulted for detailed
information on potentially hazardous drug combinations.
Pharmacokinetic interactions
Drugs can interfere with another’s absorption, distribution, metabolism, or excretion.
Most pharmacokinetic interactions are due to one drug interfering with another’s
metabolism, and this is discussed above.
Absorption
Dairy products, which are rich in calcium, interfere with tetracycline absorption
because the calcium complexes with the drug to form an insoluble complex. Drugs that
reduce gastric motility, e.g. opioids, and drugs with antimuscarinic properties, e.g.,
some antihistamines and antidepressants, slow the rate of absorption of other
drugs because they impair drug delivery into the small intestine. Adrenaline co-
injected with a local anaesthetic acts as a vasoconstrictor, reducing the loss of the
anaesthetic from the area in the bloodstream and prolonging its effect, and is thus useful
in longer procedures.
Distribution
Drugs may potentially compete for plasma protein-binding sites: the standard example,
aspirin and warfarin, is discussed above.
Excretion
Drugs that impair renal function or reduce renal blood flow can reduce excretion of
other drugs and increase their circulating blood levels. Carrier mechanisms in the renal
tubule that secrete particular drugs may be inhibited by competing drugs. For example,
non-steroidal anti-inflammatory drugs inhibit the secretion of methotrexate
and can increase methotrexate levels in the blood.
Pharmacodynamic interactions
Drugs can interfere with each other’s physiological activity in a range of ways because
pharmacodynamic processes are so complex. Some interactions are beneficial and
exploited clinically, and some are not. The most serious drug interactions can be lethal.
Beneficial interactions
The combined use of analgesics with different mechanisms of action, e.g. codeine and
paracetamol, is common. The analgesic action of the drugs is additive and allows the
doses of the individual drugs to be kept lower than if each was being used alone,
reducing the likelihood of adverse effects. Another example of a therapeutically
exploited interaction is the use of reversal agents when two drugs compete for the same
binding site, for example, when naloxone or another opioid antagonist is used to
reverse morphine overdose.
Unwanted/dangerous interactions
Sometimes both drugs have the same biological effect, and when used together, produce
an excessive response: for example, the risk of haemorrhage is increased when drugs
that increase bleeding times, including warfarin, heparin, and aspirin, are used
together. Another example is the use of dopamine receptor agonists such as ropinirole
to treat Parkinson’s disease and dopamine receptor antagonists such as olanzapine in
the treatment of psychiatric disorders: this drug combination is avoided as they directly
interfere with each other’s pharmacological action. Many diuretics, including
furosemide, predispose to hypokalaemia, which increases the risks of digoxin
toxicity.
Older age
Physiological processes tend to decline in older adults. Gastric emptying may be slower,
delaying drug delivery into the small intestine. Age-related reduction in liver and kidney
function may affect metabolic and excretory mechanisms in healthy older people,
although there is significant variability in the degree to which this affects drug
disposition, and patients should be carefully assessed on an individual basis. First-pass
metabolism may be reduced, increasing the amount of an orally administered drug
reaching the general circulation and necessitating a reduction in drug dose. From the
age of 50 years, the glomerular filtration rate (GFR) may be reduced by 25% and by 50%
by 75 years, reducing renal drug excretion.
Plasma protein levels are lower in older people, potentially increasing the drug-free
fraction and necessitating lower doses, especially at the start of a course of drugs or
when drugs are being administered in an acute setting. In general, the adipose–lean
body mass balance tips towards the deposition of adipose tissue, increasing the potential
for drug accumulation in body fat stores. Older people may be more sensitive to the
central effects of drugs, including sedation, confusion, amnesia, and impaired cognition.
Pregnancy
Drug use in pregnancy presents possible dangers to two people: the mother and the
baby. Fetal liver and kidney function are immature throughout pregnancy, and so the
baby is largely reliant on the mother to eliminate circulating drugs. Even in a healthy
pregnancy, physiological changes affect the pharmacokinetic profiles of drugs.
Pregnancy-associated nausea and vomiting can limit the usefulness of orally
administered drugs. Blood volume rises in pregnancy, diluting plasma proteins and
potentially increasing the free fraction. Cardiac output rises, increasing blood flow
through the kidney and liver, and potentially accelerating drug elimination. In addition,
hepatic metabolism increases because enzyme levels rise, which also speeds up drug
clearance. When therapeutic drugs must be used in pregnancy, particular care must be
taken in monitoring the health of both the mother and the baby.
Genetic variability
A significant source of inter-individual variation in metabolism is genetically
determined, discussed in more detail above. However, genetic differences can also affect
receptor populations, absorption mechanisms, and hypersensitivity responses and affect
both how the body deals with the drug as well as how the drug affects the body.
Identifying important genetic differences in pharmacokinetic and pharmacodynamic
profiles and using this information in planning and prescribing drug therapy is
becoming slowly but increasingly a part of personalised medicine. One drug may be
chosen over another because it is known that the patient will metabolise it better, is
likely to have a better clinical response to it, or is likely to have less severe adverse
effects with it. This is truly a situation of the story being written in the genes!
References
1. Fernandez E, Perez R, Hernandez A, et al. Factors and mechanisms for
pharmacokinetic differences between pediatric population and
adults. Pharmaceutics. 2011;3(1):53–72.
Online resources
Haywood A, Glass B.D. Pharmaceutical excipients – where do we begin? Aust
Prescr. 2011;34:112–114 Available at:. https://www.nps.org.au/australian-
prescriber/articles/pharmaceutical-excipients-where-do-we-begin.
Le J. Overview of pharmacokinetics. MSD Manual. 2022 Available
at:. https://www.msdmanuals.com/en-gb/professional/clinical-
pharmacology/pharmacokinetics/overview-of-pharmacokinetics.
Pharmacology Education Project, . Clinical pharmacokinetics Available
at:. https://www.pharmacologyeducation.org/clinical-
pharmacology/clinical-pharmacokinetics, 2023.
Yartsev A. Required reading: pharmacokinetics. Deranged
Physiology. 2024 Available
at:. https://derangedphysiology.com/main/cicm-primary-
exam/required-reading/pharmacokinetics.
3: Pharmacodynamics:
What the Drug Does to the Body
Introduction to Pharmacodynamics
The Levels of Drug Action
Principal Targets of Drug Action
Drug Receptors
Receptors and Their Actions
Ion Channels
Receptor Desensitisation and Drug
Tolerance
Membrane Transport Mechanisms
Enzymes
Non-Mammalian Cell Targets
Assessment of Drug Action
Dose–Response Relationships
Therapeutic Index
Adverse Drug Reactions
Mechanisms of Adverse Drug Reactions
Immunologically Mediated Adverse Drug
Reactions
Pharmacologically Mediated Adverse Drug
Reactions
Introduction to Pharmacodynamics
Pharmacodynamics is the umbrella term describing the mechanisms by which drugs
exert their effects on body tissues, sometimes simply put as ‘what the drug does to the
body’. The response of the body to a typical drug is likely to be complex. A drug given to
treat a specific condition or to relieve a specific symptom will frequently produce
additional, usually unwanted adverse effects, often affecting the function of organs or
systems that are not the desired target of the drug and often at drug doses in the desired
therapeutic range. A sound grasp of basic pharmacodynamic principles is essential to
understanding why this occurs.
Drug Receptors
Key Definitions
• Ligand:
• Agonist:
a drug which binds to a receptor and activates it. Using a ‘lock and key’ analogy, the
drug is the equivalent of a key which fully engages a lock (the receptor) and can either
lock or open it.
• Antagonist:
a drug which binds to a receptor and blocks it. Using a ‘lock and key’ analogy, an
antagonist acts like a key that is a close enough match to enter a lock but not a close
enough fit to operate it. However, while this ‘false’ key occupies the lock, it prevents a
‘true’ key from being inserted, and so the lock is useless.
Table 3.1
5-HT, 5-hydroxytryptamine
FIG. 3.2 The action of an endogenous agent, an agonist, and an
antagonist at a receptor. Modified from Lilley LL, Collins S, and
Snyder J (2023) Pharmacology and the nursing process, 10th ed,
Fig. 2.7. St. Louis: Mosby.
Key Definitions
• Specificity:
• Selectivity:
the ability of a drug to differentiate between subtypes of the same receptor family:
e.g. antihistamines (H1 receptor antagonists) selectively block H1 receptors but not H2
receptors.
Receptor Specificity
Receptors are usually protein molecules folded into complex and specific three-
dimensional shapes. This means they can only bind chemicals that have a
complementary shape and ‘fit’ the receptor binding site, like a lock and a key. The ability
of a ligand such as a neurotransmitter, hormone, or drug to discriminate between
different receptor types and to bind to one and not another, even if the two are
structurally very similar, is called specificity and is essential in normal healthy body
function. For example, there are currently more than 100 known neurotransmitters in
the human nervous system, each with its own particular and precise range of roles to
play in the sophisticated and finely tuned neurological control of body physiology. It is
essential that each is only active at its own receptors and does not affect receptors for
other transmitters because this would significantly disrupt this intricate and complex
control network. In clinical pharmacology, drug specificity is very important, because
the more specific the drug is for its intended receptor, the less likely it is to cause
unwanted adverse effects through binding to other receptor types. The lower the drug–
receptor specificity, the greater the risk and incidence of adverse effects unrelated to the
drug’s intended therapeutic aim. Many drugs are not absolutely specific for their target
receptor and affect other receptor types. Fig. 3.3A shows that older (first-generation)
antihistamines, such as chlorphenamine, block H1 histamine receptors, the
mechanism by which they exert their anti-inflammatory effect. However, although the
fit is not perfect, they also bind to and block muscarinic cholinergic receptors in the
autonomic nervous system. Unwanted antimuscarinic effects (p. 51), including dry
mouth, sedation, and blurred vision, are therefore common with these drugs. There is
no interaction with, for example, dopamine receptors, because there is no molecular fit.
For example, consider the histamine receptor family. Currently four histamine
receptor subtypes have been identified, named H1, H2, H3, and H4. Although they are all
in some way involved in immunity and body defence and they all respond to histamine,
they are not distributed evenly throughout histamine-responsive tissues, and they
mediate different functions of histamine. The pharmacology of histamine and its
receptors is discussed in more detail in Chapters 6 and 10, but let us use the example
here to briefly illustrate the point that different receptor subtypes mediate different
functions of the same chemical and can be blocked by different drugs. H1 receptors are
found in blood vessels and secretory epithelia (e.g. the linings of the nose), and
histamine acting here produces the standard signs and symptoms of allergic
inflammation: an insect bite gives red, swollen, itchy skin, and hay fever gives a runny
nose and red, itchy eyes. Antihistamines like chlorphenamine and cetirizine block
H1 receptors and relieve these symptoms. H2 receptors are found in the stomach, and
histamine acting here increases gastric acid secretion, another important defence
mechanism. However, antihistamines have no effect on H2 receptors; a different class of
drugs, the H2 receptor blockers including cimetidine and famotidine, are used to
reduce gastric juice secretion in conditions associated with excess acid production.
Increasing dose can affect the specificity and selectivity of a drug. For example, the β-
receptor family all respond to adrenaline and noradrenaline, but different subtypes
are found in different tissues. The myocardium in the heart has mainly β1 receptors, and
airway smooth muscle has mainly β2 receptors. Some β-blockers, for example atenolol,
have relative selectivity for β1 cardiac receptors and bind preferentially to this subtype
over other subtypes (Fig. 3.3B). However, as atenolol doses increase, its effects on
other subtypes become more significant, including β2-mediated bronchoconstriction,
which may be a problem in people with obstructive airways disorders such as asthma.
Competitive Antagonism
This model of drug–receptor interactions explains why a reversal agent can be used to
block the action of a drug already in the system. For example, naloxone is a MOR
(Table 3.1) antagonist that can be used to reverse an opioid overdose. Because the
opioid molecules bind and dissociate from their MORs multiple times a second,
naloxone molecules can also bind while the receptor is momentarily free. This is called
competitive antagonism (Fig. 3.4), because the reversal agent competes with the opioid
for receptor occupancy, and while naloxone molecules are present in appropriate
concentrations at the receptors, they limit opioid binding and reduce opioid effects.
Cell-Surface Receptors
Drug–receptor binding on the cell membrane is only the first stage in a chain of steps
which convert the interaction of the drug with its receptor to its final biological effect. It
could be likened to speaking to a receptionist at the front door of an office block, which
is only the first step in making it to your appointment with someone on the fifth floor.
The receptionist must trigger a chain of communication within the building, perhaps by
telephoning or emailing someone to let them know you have arrived, or by asking an
assistant to take you in the lift to the correct floor. A similar chain of messages is
triggered by drug–receptor binding. Drug–receptor binding usually initiates a
conformational change in the receptor which triggers a series of biochemical
communication steps within the cell, transferring the message from the receptor to
intracellular mechanisms that will produce the final cellular response. These are called
transduction mechanisms, and there are several different types. Only G protein-coupled
receptor mechanisms are discussed here in more detail because they are particularly
important.
G Protein-Coupled Receptors
Around a third of therapeutically important cell-surface drug receptors are linked to a
special protein called a G protein, which is bound to the internal surface of the cell
membrane. They are therefore called G protein-coupled receptors (GPCRs). They are a
large and diverse receptor family, including receptors for many neurotransmitters,
inflammatory mediators, and peptide hormones. A GPCR is formed from a long protein
chain, folded seven times, and embedded in the cell membrane. One end is exposed on
the outer surface of the cell membrane and acts as the drug receptor. The internal end of
the receptor protein is directly linked to the G protein (Fig. 3.7). G proteins are
sometimes called go-between proteins because they act as intermediaries between the
cell-surface receptor and the final stage in the sequence of events transmitting the drug’s
message into the cell’s interior. The final stages in the communication chain, responsible
for generating the final effect of the drug on the cell, are called second-messenger
pathways.
Second-Messenger Pathways
The two most important second-messenger pathways are the cyclic AMP
(cAMP)/adenylate cyclase system and the phosphatidyl inositol/phospholipase C
systems. Although the biochemical cascades are different for each, they both involve the
activation of intracellular enzymes called kinases. Kinases are key enzymes in the
regulation of protein activity in the cell. They convert ATP to ADP, releasing energy-rich
phosphate groups which phosphorylate and activate or inactivate a wide range of
cellular enzymes and other proteins, including ion channels and transport proteins.
There are hundreds of different kinase subtypes in the human cell, reflecting their
central role in cellular biochemistry and metabolism.
FIG. 3.6 The main locations of drug receptors.
Nuclear Receptors
Some drugs, e.g. steroid hormones, do not bind to a cell-surface receptor. They are
highly fat-soluble, so they cross the plasma membrane and bind to their receptor, called
a steroid-binding protein, in the cytosol. The drug–receptor complex then crosses the
nuclear membrane and binds directly to a specific binding site on one or more genes
contained within the cell’s DNA. This may activate the gene, resulting in the production
(transcription) of a molecule of messenger RNA (mRNA). The mRNA molecule leaves
the nucleus and attaches to ribosomes in the cytoplasm, which produce the protein that
the gene encodes (Fig. 3.9). Alternatively, steroid binding to a gene may switch the
gene off and halt protein production. For example, anti-inflammatory glucocorticoids
such as hydrocortisone switch off genes that code for pro-inflammatory proteins and
activate genes that code for proteins that suppress inflammation. The onset of action of
drugs which affect gene expression is often slow because it takes time for the target
protein levels to either rise or fall sufficiently to make a difference to cell function.
Ion Channels
Ion channels embedded in the cell membrane open and close to control the flow of ions
such as Ca2+, sodium (Na+), and Cl− across the cell membrane. Opening the channel
allows ions to travel down their concentration gradient and either enter or leave the cell.
Because ions are electrically charged, their movement in or out of the cell regulates the
electrical excitability of the cell and allows nerve and muscle cells to generate and
propagate electrical signals. Sodium and calcium channels permit Na+ and Ca2+
respectively to flow into the cell, which depolarise (activate) nerve and muscle cells.
Potassium (K+) channels allow K+ to leave the cell, which hyperpolarises (desensitises)
an excitable cell, making a nerve, for example, less able to fire. Chloride channels allow
Cl− to enter the cell, which hyperpolarises it, making nerve cells less likely to fire.
Generally, drugs which affect ion channel function have a rapid onset of action because
they cause an immediate change in ion concentrations in the cell.
Drugs affecting ion channel function usually work in one of two ways: they may bind
to a receptor site attached to the channel and regulate the channel opening, or they may
bind directly to the channel protein and physically block the channel opening.
FIG. 3.9 The mechanism of action of steroid drugs. Modified from
Jones R and Lopez KH (2006) Human reproductive biology, 3rd ed,
Fig. 1.5. San Diego: Academic Press.
Enzymes
Enzymes are biological catalysts and speed up chemical reactions inside and outside
cells without themselves being changed: the ultimate recyclers! The body contains tens
of thousands of enzymes, each driving one or more steps in the millions of chemical
reactions taking place every second. Drugs can themselves be enzymes: for example,
streptokinase is a thrombolytic drug used to break down a blood clot blocking a
coronary or cerebral artery and restore blood flow. More commonly, drugs affecting
enzyme action are inhibitors and block the enzyme; for example, ibuprofen blocks the
enzyme cyclo-oxygenase, which produces pro-inflammatory prostaglandins. This is the
basis of ibuprofen’s anti-inflammatory action, but because prostaglandins have
additional physiological functions, inhibiting their production causes a range of other
side-effects (see Chapter 6).
Dose–Response Relationships
In general, there is a direct relationship between drug dose and the biological response it
produces: the higher the dose, the greater the effects, both therapeutic and unwanted.
This can be quantified using dose–response (DR) curves, which demonstrate key
features of drug action including the therapeutic range and the maximally effective dose.
The y-axis plots the drug response under consideration. The x-axis of the DR curve
shows drug concentrations, usually plotted as their logarithms (logs) to allow a wide
range of values to be plotted along a shorter axis, and gives a characteristic sigmoid (S)-
shaped curve with a central linear portion (Fig. 3.10).
DR curves can be constructed to show the activity of a drug in a lab-based experiment;
for example, to show how increasing drug concentrations inhibit the activity of a specific
enzyme (in which case it is more properly known as a concentration–response curve) or
to show the effect of a drug in a living system. Fig. 3.10 shows a DR curve presenting
the relationship between increasing dose of a hypothetical diuretic drug and the increase
in daily urine output. It clearly shows that as drug concentration rises, there is a steady
rise in urine output. It also shows that below the point marked with one star, the drug
shows little or no effect: this is the sub-therapeutic drug range. Above the point marked
with two stars, the diuretic effect plateaus, and increasing the dose to supra-therapeutic
levels gives no further clinical benefit. In this example, the therapeutic range
(sometimes called the therapeutic window), which is the dose range across which
maximal therapeutic benefit is likely to be obtained, is also shown. It is important that
drug concentrations in body tissues, usually measured in the plasma, fall within the
therapeutic range. If they fall below the therapeutic minimum, the drug is unlikely to
produce its desired benefits. If they exceed the therapeutic maximum, the risk of adverse
effects increases. It is important to realise that for most drugs, some adverse effects may
occur even when plasma levels fall within the therapeutic range. Whether or not this is
acceptable to the patient and the clinical team is likely to depend upon the situation.
Significant unwanted effects occurring within the therapeutic range may be tolerated if
the disease is serious or if other treatment options are limited; for example, cytotoxic
agents used in cancer can produce horrible adverse effects which may be accepted
because the treatment may be life-saving.
FIG. 3.10 Dose–response curve for a hypothetical diuretic drug,
showing the therapeutic range and the ED50.
The ED50 (the dose producing an effective therapeutic response in half of a test
population) is also shown on this graph. The ED50 is important when calculating the
therapeutic index (TI; see below).
Therapeutic Index
DR curves can be used to show the relationship between dose and likely toxicity. The TI,
in its simplest form, is a measure of the difference between the therapeutic dose of a
drug and the dose likely to cause a particular adverse effect. It is calculated as a ratio of
the toxic dose to the effective dose. The closer the toxic dose is to the therapeutic dose,
the closer the ratio is to 1. A low TI therefore indicates that adverse effects are likely to
be seen at drug doses close to the therapeutic dose. Higher TI values indicate a wider
difference between therapeutic and toxic doses, which is clearly more desirable.
Fig. 3.11 demonstrates this relationship for the same hypothetical diuretic drug
shown in Fig. 3.10 and used to treat high blood pressure. Its standard DR curve is
shown in green. It has two known unwanted effects: renal toxicity and hair loss. We can
plot the DR curve showing the relationship between drug dose and hair loss; this is
shown in orange. We can see that the two curves are very close together, so that for most
doses across the therapeutic range, there is also likely to be hair loss. The DR curve
showing the relationship between drug levels and kidney toxicity is shown in grey. This
curve is significantly shifted to the right, showing that renal toxicity is likely only at
supra-therapeutic drug doses.
It seems obvious from simply eyeballing the graph that hair loss is a much more
common adverse effect than renal toxicity, but the TI is a quantitative value usually
calculated as a ratio. The TI is expressed as the ratio between the TD50, which is the dose
of the drug producing the adverse effect in half of a test population, and the ED50, and it
is calculated as TD50/ED50. The closer the TD50 is to the ED50, the higher the risk of the
given side-effect. If TD50 and ED50 are the same or very close, the ratio is 1 or close to 1,
and the toxic effect is pretty much guaranteed to occur at therapeutic doses. For
example, if the TD50 is 20 mg and the ED50 is 10 mg, the TI is 20/10=2, which is low and
suggests a high likelihood of the adverse effect occurring. However, if the TD50 is much
higher than the ED50, the ratio is much higher and reflects a greater safety margin with
respect to toxicity. For example, if the TD50 is 1000 mg and the ED50 is 1 mg, the TI is
1000/1=1000.
It is important to realise, however, that not all adverse effects are seen more
frequently or more severely with increasing dose. Allergic and anaphylactic drug
reactions can occur even after minimal exposure, and cough, a common and
troublesome side-effect of angiotensin-converting enzyme (ACE) inhibitors like
captopril, is not dose-dependent. In these circumstances, DR curves and TI are not
relevant.
FIG. 3.11 Dose–response curves for a hypothetical diuretic drug,
illustrating a narrow therapeutic index for hair loss and a wide
therapeutic index for renal toxicity.
Adverse Drug Reactions
Adverse drug reactions (ADRs) are common, often troublesome, and occasionally
catastrophic. Management of unwanted effects caused by one drug by prescribing
additional drugs is an important cause of polypharmacy, which in turn greatly increases
the risks of further side-effects and interactions. For example, Ca2+-channel blockers
like verapamil can cause acid reflux, which can be troublesome enough to require the
addition of a proton-pump inhibitor like omeprazole to the prescription. ADRs are a
common reason for people to consult their healthcare provider and are the cause of a
significant proportion of hospital admissions. Risk factors for ADRs include renal and
liver insufficiency, female sex, serious illness and/or immunocompromise, extremes of
age, and polypharmacy.
Reproductive Function
The use of both prescription and non-prescription drugs in pregnancy presents potential
risks to the developing baby. Most drugs, being fat-soluble, cross the placenta and many
do so in significant concentrations. The decision to prescribe new drugs or to continue
with pre-existing prescriptions in pregnancy or in women wishing to become pregnant
must be carefully weighed up in terms of the risk-benefit ratio to both mother and child.
In the first 3 months, while the baby’s cells and tissues are differentiating and the organ
systems are being laid down, drugs may cause fetal abnormalities; drugs that do this are
called teratogens. Significant teratogens include ethanol, many anticonvulsants
including phenytoin and valproate, cytotoxic drugs, warfarin, ACE inhibitors,
methotrexate, and retinoids. In the second two trimesters, when organogenesis is
completed, ADRs on the fetus are often similar to those seen post-natally: for example,
anticoagulants increase the risk of bleeding, oestrogens can feminise male fetuses,
and ACE inhibitors can interfere with fetal kidney function. Ethanol can cause
significant developmental retardation at all stages of pregnancy, including low
birthweight and facial and cardiac malformations.
Quantitative and evidence-based data on the effect of drugs on mother and baby
during pregnancy and in breastfeeding women is often limited, because for obvious
reasons it is not ethical to include pregnant women in clinical trials. In general, the use
of drugs in pregnancy should be avoided if possible, especially in the first trimester. If
drug treatment is unavoidable, the lowest possible doses of long-standing drugs that are
believed to be safe should be used.
Some drugs can interfere with sperm production and ideally should be avoided in men
trying to father a child. These include ethanol, opioids, anabolic steroids, and
cytotoxic agents.
Drug Dependence
Substance dependence occurs when an individual is unable to function without regular
intake of a substance, experiences distress when deprived of it, and continues to use the
substance despite associated problems. It is frequently associated with recreational
drugs, including ethanol, but can also arise with drugs used therapeutically, including
opioid analgesics, antidepressants, and benzodiazepines.
It is characteristic of human behaviour that we like to repeat pleasurable and
rewarding experiences. The evolution of powerful reward pathways, seen across the
animal kingdom and not just in humans, ensures the continuation of the species because
it reinforces essential survival behaviours such as eating, sexual and reproductive
activity, and parent–child bonding. The key nerve pathways in the brain responsible for
regulating reward-related behaviour are found in the mesolimbic system, and the main
neurotransmitter involved in promoting rewarding behaviours is dopamine. Although
other brain centres are also involved, the key areas associated with reward are the
ventral tegmental area (VTA) in the midbrain and the nucleus accumbens (NA), located
anteriorly to the VTA (Fig. 3.14A). The VTA projects dopamine-releasing neurones to
the NA, which is the main centre for reward-orientated learning and behaviour.
Dopamine, acting on D1 receptors in the NA, reinforces activities found to be positive,
beneficial, or enjoyable and increases the likelihood that the behaviour will be repeated.
A range of drugs via a direct or indirect action on the VTA increase dopamine levels in
the NA and so increases the likelihood that the drug will be taken again (Fig. 3.14B).
Repeated use of the drug produces sustained elevated dopamine levels in reward
pathways and brain structures, which over time leads to neuroadaptive changes, which
may be irreversible. The reward pathways become tolerant of consistently elevated
dopamine levels and cannot function without them. When drug levels fall, and
dopamine levels fall as a consequence, unpleasant withdrawal effects motivate the
individual to repeat their drug-taking.
FIG. 3.14 The reward pathway.A. The ventral tegmental area and
the nucleus accumbens. B. The action of drugs that cause
dependence on dopamine levels in the nucleus accumbens. (A)
Modified from National Institute on Drug Abuse. The Neurobiology
of Drug Addiction. Available at
http://www.nida.nih.gov/pubs/teaching/Teaching2/Teaching.html,
and (B) from Waller DG, Sampson A, and Hitchings A (2022)
Medical pharmacology and therapeutics, 6th ed, Fig. 54.1. Oxford:
Elsevier.
Withdrawal Syndromes
Withdrawal syndromes are collections of signs and symptoms seen when a drug is
discontinued, and they can be severe: for example, delirium tremens following alcohol
withdrawal can necessitate intensive care support, and seizures following
benzodiazepine withdrawal can be life-threatening.
References
1. Pasternak A.L, Ward K.M, Luzum J.A, et al. Germline genetic variants with
implications for disease risk and therapeutic outcomes. Physiol.
Genomics. 2017;49(10):567–581.
Online Resources
Farinde A. Overview of Pharmacodynamics. 2023 Available
at:. https://www.msdmanuals.com/en-gb/professional/clinical-
pharmacology/pharmacodynamics/overview-of-pharmacodynamics.
Pharmacology Education Project, . Clinical Pharmacodynamics. 2024 Available
at:. https://www.pharmacologyeducation.org/clinical-
pharmacology/clinical-pharmacodynamics.
Yartsev A. Required Reading: Pharmacodynamics. 2024 Available
at:. https://derangedphysiology.com/main/cicm-primary-
exam/required-reading/pharmacodynamics.
4: Drugs and Neurological Function
CHAPTE R OU TLINE
Introduction
Nerve Conduction and Synaptic Transmission
The neurotransmitters of the nervous system
Causes of Seizures
Types of Seizure
Pharmacological Management of Seizures
Anticonvulsants that Block Sodium Channels
Anticonvulsants that Enhance Gamma-
Aminobutyric Acid
Anticonvulsants that Block Calcium
Channels
Anticonvulsants that Reduce Glutamate
Activity
F OCUS ON
Introduction
The nervous system controls multiple aspects of body function, and because of this,
drugs that affect neurological processes often have significant and widespread effects.
Nerve conduction and synaptic transmission
Neurones are specialised cells that generate and conduct electrical impulses (action
potentials), allowing them to communicate with each other and to control activity in
muscles, glands, and other organs. A typical nerve cell has a cell body, which has
multiple fine extensions called dendrites and contains the nucleus, as well as an
extended filament called an axon, which conducts the action potential (Fig. 4.1A). The
axon terminates in a sheaf of tiny extensions called axon terminals, each of which comes
into very close association (but not direct contact) with the dendrites of another
neurone, or with a target cell of peripheral tissue, e.g. smooth muscle or a gland. Some
nerve axons have a myelin sheath, which increases the speed of nerve conduction.
The Synapse
A nerve cell does not physically make contact with other nerve or target cells: the tiny
gap between an axon terminal and the dendrite of another neurone, or the axon
terminal and a target cell, is called the synaptic cleft and the site of connection is called
the synapse. The nerve conducting an action potential towards the synapse is called the
pre-synaptic neurone, and the cell receiving the action potential is called the post-
synaptic cell (Fig. 4.1B).
FIG 4.1 A. Nerve cell structure. B. General structure of a
synapse. Modified from Waugh A and Grant A (2018) Ross & Wilson
anatomy and physiology in health and illness, 13th ed, Figs 7.2 and
7.7. Oxford: Elsevier.
Without physical contact between pre- and post-synaptic membranes, the electrical
action potential cannot be directly transmitted from cell to cell. Instead, the pre-
synaptic axon terminal synthesises a chemical called a neurotransmitter, which is
released in response to the arrival of an action potential. Neurotransmitter molecules
diffuse across the narrow synaptic cleft and bind to receptors on the post-synaptic
membrane. This produces a response in the post-synaptic cell, which may be activated
or inhibited depending on the neurotransmitter and the nature of the post-synaptic
receptors. Inhibitory neurotransmission is an essential component of neurological
control and an important target for clinically important drugs. Neurotransmitter
synthesis, release, and action on its receptors are very important drug targets.
Re-uptake Pumps
Specialised pumps and carriers in the pre-synaptic nerve cell membrane are in
continuous operation, constantly removing the transmitter from the synapse and taking
it back up into the pre-synaptic nerve (Fig. 4.2). Sometimes the transmitter is then
simply repackaged into vesicles, allowing it to be reused––an efficient recycling
mechanism! Sometimes the pumps are found on the post-synaptic membranes, in which
case the transmitter is then broken down by enzymes in the post-synaptic cell. Drugs
that block these pumps increase neurotransmitter levels in the synapse and prolong its
action. Key examples include tricyclic antidepressants and serotonin-specific re-
uptake inhibitors (SSRIs).
FIG 4.2 Removal of a neurotransmitter from the synapse by re-
uptake pumps. Modified from Burchum J and Rosenthal L (2022)
Lehne’s pharmacology for nursing care, 11th ed, Fig. 35.1. St. Louis:
Saunders.
Degradative enzymes
Enzymes in the synapse or in either the pre- or post-synaptic cells destroy the
transmitter. Important examples of drugs that inhibit these enzymes, therefore
increasing transmitter levels in the synapse and prolonging its activity, include anti-
cholinesterase drugs like neostigmine, which prolong the action of acetylcholine
(ACh), and monoamine oxidase (MAO) inhibitors like phenelzine and selegiline,
which prolong the actions of noradrenaline (NA) and related monoamine
neurotransmitters.
The membrane potential is now back to where it was before activation, at about
-90mV, but the distribution of Na+ and K+ ions is reversed, because they are now on
opposite sides of the membrane from where they started. Na+ is rapidly driven out of the
cell and K+ pulled back in by the continuous action of the Na+/K+ pump, restoring the
original excitability of the nerve. Interfering with ion movements across the nerve cell
membrane eliminates the action potential and prevents nerve conduction. Local
anaesthetics block the Na+ channels in nerve cell membranes, preventing Na+ flow
and silencing the nerve.
Autonomic pathways from the CNS to body tissues always contain two nerves (Fig.
4.4B). There are therefore two synapses per pathway, both potential targets for drug
action. The first nerve, called the pre-ganglionic nerve, leaves the CNS and terminates in
a ganglion (a collection of cell bodies), where it synapses with the cell body of the second
nerve in the pathway, the post-ganglionic nerve. The ganglia of sympathetic pathways lie
very close to the vertebral column, so the pre-ganglionic nerve is very short, and a long
post-ganglionic nerve is needed to reach the target organ. However, in the PSNS, the
pre-ganglionic nerve is very long, requiring only a short post-ganglionic nerve to reach
the target tissues. The neurotransmitter at sympathetic post-ganglionic nerve endings is
noradrenaline (NA), and acetylcholine (ACh) is the transmitter at both sympathetic and
parasympathetic ganglia and at parasympathetic nerve endings.
The receptors on which acetylcholine and noradrenaline act are not all identical but
are subdivided into subtypes, found in different tissues (see also Table 3.1).
Understanding the difference between the subtypes of autonomic receptors is important
because some drugs act at one subtype and not others, which has a direct impact on
their clinical action.
Serotonin
Serotonin (5-hydroxytryptamine, 5-HT), has multiple physiological functions, including
in the gastrointestinal tract and as an important platelet activator in clotting. The role of
serotonin in these tissues is discussed in more detail in the relevant chapters. It is
widespread in the CNS; it regulates mood and arousal, eating behaviours, vomiting,
cognitive function, sleep, sensory pathways including pain, and body temperature; and
it is hallucinogenic.
Serotonin Receptors
There are seven types of serotonin receptor, named 5-HT1-7, and each type has multiple
subtypes (see Table 3.1). All seven types are found in the brain, and the development of
drugs that act selectively at particular subtypes has yielded drugs with specific clinical
usefulness. For example, 5-HT3 receptors are found in the area postrema in the medulla,
which controls vomiting. 5-HT release by these nerves stimulates vomiting.
Ondansetron is a selective 5-HT3 antagonist and a potent anti-emetic. Methysergide
is a 5-HT2 antagonist used to treat migraine. 5-HT2C receptors are found extensively in
brain regions associated with mood and emotional responses, and stimulation of these
receptors reduces dopamine levels and is associated with depressive states. Certain
atypical antidepressants, e.g. trazodone, block these receptors.
Gamma-Aminobutyric Acid
GABA is the brain’s main inhibitory neurotransmitter. It is widespread in the brain and
controls a wide range of pathways and neurotransmitter activity. Drugs that increase
GABA levels, e.g. benzodiazepines (BDZs), sedate, cause sleep, and reduce anxiety,
aggression, and activity. They are also used to reduce the excessive, abnormal electrical
discharge that causes seizures.
Dopamine
Dopamine is a neurotransmitter in both the PNS and CNS. It is an intermediate in the
biosynthetic pathway for NA and adrenaline, so it too is a catecholamine (Fig. 4.12).
Tyrosine is converted to dihydroxyphenylalanine (DOPA, specifically, L-DOPA or
levodopa) by the enzyme tyrosine hydroxylase. Levodopa is converted to dopamine by
DOPA decarboxylase. Dopamine in turn is converted to NA by dopamine β-hydroxylase,
and noradrenaline to adrenaline by phenylethanolamine N-methyltransferase. Seventy-
five percent of the brain’s dopamine content is found in the nigrostriatal pathway,
concerned with voluntary muscle control. Dopamine is also found in pathways
concerned with behaviour, reward, motivation, and addiction; with vomiting; and with
endocrine control. Key drugs in the treatment of Parkinson’s disease and schizophrenia
work by changing dopamine levels in the brain. Several dopamine antagonists, e.g.
domperidone and metoclopramide, are used as anti-emetics.
Dopamine Receptors
There are two groups of dopamine receptors: D1 and D2. Dopamine, like many other
neurotransmitters, has both inhibitory and excitatory actions depending on the nerve
pathway involved. For example, it activates reward pathways
Table 4.1
• Cardiovascular system: The PSNS slows the heart rate, so antimuscarinic drugs
cause tachycardia. Blood vessels in the skin dilate, causing flushing.
• Gastrointestinal system: The PSNS promotes digestive activity, so
antimuscarinic activity reduces gastrointestinal motility, with delayed gastric
emptying, vomiting, gastro-oesophageal reflux, and constipation. The
production of digestive secretions, including saliva, is reduced, and dry mouth
is a very common complaint. Reduced salivary production increases the risk of
dental caries.
• Respiratory system: The PSNS contracts bronchial smooth muscle and causes
bronchoconstriction. It also promotes the production of respiratory secretions,
so antimuscarinic activity gives rise to bronchodilation and reduces respiratory
secretions.
• Secretory activity: Sweat and tear production are reduced, resulting in dry skin
and dry eyes.
• Smooth muscle: The PSNS promotes bladder contractility, and drugs with anti-
muscarinic properties cause urinary retention and overflow incontinence.
• Central effects: ACh is widespread in the brain, and its activity is reduced by
drugs with anti-muscarinic activity that cross the blood–brain barrier. This
leads to sedation, confusion and cognitive impairment, hallucinations, and
memory problems.
• Visual problems: The PSNS controls the ciliary muscle of the eye, which controls
focussing of the lens, and constricts the circular muscle of the iris, constricting
the pupil. Antimuscarinic side-effects therefore include blurred vision and
widely dilated pupils. A consequence of these actions may be raised intraocular
pressure and the development of glaucoma. When the circular muscle of the iris
and the ciliary body are constricted by parasympathetic activity, the iris is
stretched and thinned. This opens the canal of Schlemm in the iris–corneal
angle, allowing aqueous humour to drain from the interior of the eye and
keeping intraocular pressure within normal limits (Fig. 4.11A). Antimuscarinic
agents dilate the pupil by allowing the iris to relax and bulge; this can obstruct
the canal of Schlemm, block aqueous humour drainage, and predispose to
closed-angle glaucoma (Fig. 4.11B). In some people, there is little space
already between the cornea and the iris, and any drug with antimuscarinic
effects is absolutely contraindicated because it can cause a rapid rise in
intraocular pressure with permanent damage to the optic nerve.
Histamine
Histamine’s best-known activity is as an inflammatory mediator in allergy, but it is also
an important neurotransmitter in the CNS, with a role in sleep–wake rhythms, appetite,
thermoregulation, and vomiting pathways. This is the reason why many antihistamines
are sedative and explains why antihistamines are useful anti-emetics.
Histamine Receptors
There are four main types of histamine receptors: H1–H4. Each type has a characteristic
distribution in body tissues, relating directly to the different physiological roles of
histamine. This is summarised in Table 4.2, and each is further discussed in relevant
chapters. H1, H2, and H3 receptors are all found in the brain and mediate the effects
triggered by histamine released by histaminergic nerves. Some clinically important
antagonists at the different subtypes of histamine receptors are also listed in Table 4.2.
Note that the term ‘antihistamine’ is reserved for drugs that block H1 receptors.
Antihistamines (p. 122) are in widespread use as anti-allergy medications, sedatives, and
anti-emetics. H2 receptor blockers (p. 187) are used to reduce gastric acid levels.
Table 4.2
Glutamate
Glutamate is an amino acid in widespread use in the CNS as an excitatory
neurotransmitter.
There are several types of glutamate receptors, of which the N-methyl-D-aspartate
(NMDA) receptor is the best studied. Ketamine is an antagonist at this receptor.
Treatment of Depression
Pharmacological management of depression is usually only part of a treatment package
and is not necessarily the most effective option; up to 80% of people suffering from
depression do not respond to standard antidepressants, emphasising the complex
pathophysiology of this condition. Drug treatment of depression is generally most
effective when combined with non-pharmacological approaches, including cognitive
behavioural therapy and counselling. Mild to moderate depression generally responds
poorly to standard antidepressant drugs, and all current antidepressant drugs show a
time lag between beginning of treatment and the onset of a therapeutic effect. Most
antidepressants increase the activity or concentrations of monoamine
neurotransmitters, mainly norardenaline and 5-HT, in central synapses, and their
mechanisms of action are summarised in Fig. 4.16. In general, SSRIs are first-line
treatment. Tricyclic antidepressants and monoamine oxidase inhibitors are used much
less frequently, but may be useful in some situations, and both classes of drugs are
finding a place in the treatment of other conditions, e.g. amitriptyline is used in
neuropathic pain, and selegiline is used in Parkinson’s disease.
The biology of depression
Depressive illness is associated with changes in brain structure, metabolism, and
neurotransmitter release, but there is as yet no overarching understanding of the
underlying pathophysiology. There is strong evidence that depletion of key excitatory
monoamine transmitters, mainly 5-HT and NA, as well as dopamine, can lead to
depressive states. This is called the monoamine theory of depression and dates from the
mid-1960s, but not all research in the area supports it; some supporting and
contradictory evidence is summarised in Table 4.3. In addition to the monoamines,
links have been made between depression and abnormal glutamate neurotransmission,
elevated plasma cortisol levels, neuroinflammatory conditions, and reduced
neurogenesis and nerve–nerve communication, particularly in the hippocampus.
Table 4.3
Pharmacokinetics
TCAs are all given orally, are well absorbed, and are metabolised mainly in the liver.
They leave the bloodstream and bind to body tissues, which extends their half-life and
increases the risk of accumulation and toxicity. Long half-lives are characteristic of these
drugs. The half-life of amitriptyline is 24 hours, which lengthens even more in older
people and those with liver impairment.
Adverse Effects
TCAs have multiple adverse effects and should always be used with great care. They
sedate, particularly in elderly people. They interact with numerous other drug groups
and can cause life-threatening respiratory depression when used with alcohol. They
block muscarinic receptors, producing the characteristic range of antimuscarinic side-
effects: dry mouth, constipation, urinary retention, tachycardia, etc. They cause cardiac
arrhythmias and seizures, both common causes of death in TCA toxicity, because they
block Na+ channels in myocardial and nerve cell membranes, interfering with the
generation and conduction of action potentials. There are characteristic
electrocardiography (ECG) changes, including prolongation of the PR interval and wide
QRS complexes. Excitation, agitation, confusion, delirium, and coma are all seen,
especially in toxicity.
Pharmacokinetics
SSRIs are taken orally, are well absorbed, and are metabolised in the liver. Many have
long half-lives; for example, fluoxetine, sertraline, and citalopram have half-lives
ranging from 23–75 hours. Even longer half-lives are seen in reduced liver function.
Fluoxetine metabolism produces an active metabolite that has a half-life in the order
of 6 days. The likelihood of accumulation and toxicity should therefore always be borne
in mind with this group of drugs.
Adverse Effects
Largely because noradrenaline levels are relatively unaffected, SSRIs have a different
spectrum of adverse effects and interactions to the TCAs. They are generally less toxic in
overdose, less sedative, and do not cause the antimuscarinic side-effects of the TCAs.
However, they commonly cause nausea and vomiting, and they can reduce libido and
prevent orgasm, which are distressing side-effects in sexually active people. Serotonin
syndrome is a rare but potentially life-threatening adverse effect associated with mania,
confusion, hyperthermia, tachycardia, and muscle tremor and rigidity. Suicidal thoughts
and behaviour have also been reported, especially in younger people. Hyponatraemia
can occur, possibly due to reduced antidiuretic hormone secretion, which increases
blood volume and dilutes plasma sodium.
Pharmacokinetics
MAOIs are given orally, are well absorbed, and are metabolised in the liver. Most bind
irreversibly to MAO, permanently deactivating it. Because it may take 2 or 3 weeks to
restore the depleted MAO levels to normal, the effects of these drugs are long lasting and
not easily reversed in overdose. An exception is moclobemide, which inhibits MAO
reversibly and has a half-life of 2 hours.
Adverse Effects
MAOIs have multiple adverse effects, mainly relating to their inhibition of NA, 5-HT,
and dopamine breakdown. Counterintuitively for drugs that increase the levels of
sympathetic neurotransmitters, a common side-effect is postural hypotension. This is
thought to be due to increased levels of tyramine, which would normally be destroyed by
MAO being taken up into sympathetic nerve endings and converted to an inactive ‘false’
transmitter which when released does not have sympathetic activity. There may be
tremor, excitement, agitation, convulsions, and antimuscarinic side-effects.
FIG 4.17 The two forms of monoamine oxidase and their main
substrates.MAO, Monoamine oxidase. From Waller DG, Sampson A,
and Hitchings A (2022) Medical pharmacology and therapeutics, 6th
ed, Fig. 22.3. Oxford: Elsevier.
Mirtazapine
This drug blocks several receptor types, including 5-HT2C and H1 receptors (Fig. 4.16).
Its main mechanism of action is thought to be by blocking pre-synaptic ⍺2 receptors. As
explained earlier (Fig. 4.7), these receptors are part of the synaptic self-regulation
system, and when they are strongly stimulated by rising transmitter levels, they shut
down transmitter release at that synapse. Blocking them shuts down this feedback
control and increases synaptic concentrations of NA and 5-HT. Mirtazapine is sedative,
because of its antihistamine action, and can cause increased appetite and weight gain.
Trazodone
Trazodone blocks 5-HT2C receptors (Fig. 4.16), which indirectly leads to increased NA
and dopamine activity in the brain, thought to underpin its antidepressant activity. It is
sedative and may be used to treat insomnia. Unlike the TCAs and SSRIs, it has little
antimuscarinic activity, and so may be useful in patients for whom antimuscarinic
effects may be especially troublesome, e.g. in chronic constipation or benign prostatic
hypertrophy.
Agomelatine
Disruption of sleep patterns and poor sleep quality are frequently seen in depression
and can significantly reduce quality of life and cause slow recovery. The suprachiasmatic
nucleus (SCN) in the hypothalamus is the main driver of the circadian rhythms directing
the body’s internal 24-hour body clock. The SCN is linked to the pineal gland, which
releases melatonin, and at night, the SCN stimulates melatonin release from the pineal
gland, promoting sleep. SCN function is disrupted in depressive disorders, interrupting
the sleep–wake cycle, and agomelatine, a melatonin analogue, is used to regulate sleep
patterns in depression (and other conditions featuring sleep disorders). It promotes
sleep by activating the body’s natural melatonin receptors. Agomelatine also antagonises
5-HT2C receptors (see also trazodone), which may contribute to its usefulness in
depression. It is generally well tolerated, although adverse effects include insomnia,
gastrointestinal disturbances, increased sweating, headache, drowsiness, and dizziness.
Future Developments
Ketamine, introduced in the 1960s as an anaesthetic, has a history of illegal and
recreational use. In the early 2000s, researchers trialled the closely related agent
esketamine in severely depressed patients resistant to standard treatments, and not
only did the drug relieve the signs and symptoms of depression, it did so rapidly,
sometimes within hours of administration. This contrasts markedly with conventional
treatments, all of which require weeks for achievement of full therapeutic effect. It is
believed to act by increasing levels of the excitatory transmitter glutamate in the brain.
It is not currently licensed for use in the UK because of safety concerns but research is
ongoing in the search for analogues with fewer side-effects and less potential for
addiction and abuse.
Anxiety Disorders
Anxiety disorders are common and include generalised anxiety disorder, phobic states,
and panic disorder. They may co-exist with depression, and there is often a genetic
component. The main neurotransmitters in the brain thought to be important in anxiety
disorders include GABA, noradrenaline, serotonin, and dopamine, and structural
changes in areas of the brain involved in emotional and anxiety responses, including the
amygdala, hippocampus, and other limbic system structures, have been identified. The
main anxiolytic drugs include the benzodiazepines, but other agents including the
SSRIs (increasingly the first choice in treating anxiety disorders), beta-blockers,
certain anticonvulsant drugs like gabapentin, and some atypical (second-generation)
antipsychotics, e.g. olanzapine, have all proved useful in anxiety disorder
management.
Non-benzodiazepine anxiolytics
BDZ-mediated sedation can be a significant disadvantage in treating anxiety disorders
because daytime sleepiness interferes with normal daily living. In addition, the risk of
dependence is high. The search for non-sedative agents with anxiolytic activity has led
researchers to investigate medications acting at a range of other neurotransmitters,
including glutamate.
Buspirone
Buspirone was initially developed as an antipsychotic agent, but its activity in this area
was disappointing, and its anxiolytic activity was explored instead. It is most useful in
generalised anxiety states and is usually tolerated better than BDZs because it is not
sedative and does not interfere with motor control. Buspirone is an agonist at 5-HT
receptors, although how this translates into its anxiolytic effect is uncertain. There is a
delay of 2–3 weeks between starting treatment and improvement of symptoms, so it is
believed that whatever its mode of action, buspirone must trigger some form of
adaptation in the brain, particularly in the amygdala, which takes time to develop. It
also activates a range of other receptors, including dopamine and noradrenaline
receptors, but the relevance of this to its clinical action is unknown.
Pharmacokinetics
Buspirone is given orally and is well absorbed, with a half-life of 2–4 hours. Much of an
oral dose is destroyed by the liver in first-pass metabolism, and its bioavailability is
therefore low. Because of this, it is important to consider hepatic function when
prescribing. Reduced liver function can increase the proportion of an oral dose reaching
the bloodstream several fold, causing potentially toxic plasma levels.
Adverse Effects
Buspirone commonly causes dizziness, reported by over 10% of patients. Other side-
effects include nausea, diarrhoea, headache, confusion, drowsiness or excitement, mood
changes, and nervousness. It is not associated with tolerance or dependence and does
not interfere with sexual desire or performance, unlike SSRIs.
Hypnotics
Hypnotics, drugs that induce sleep, are used to treat insomnia. Insomnia is a persistent
inability to fall asleep or stay asleep despite appropriate time and opportunity, leading
to daytime fatigue, irritability, poor concentration and memory, and impairment of
normal daily function, and it is experienced by nearly everybody at some point in their
lives. Insomnia is frequently short-term, e.g. related to jet lag or the stress of an
upcoming examination. More prolonged periods of insomnia may follow bereavement
or other personal stressors. Chronic insomnia may be caused by psychiatric or physical
illness, e.g. in chronic obstructive pulmonary disease, dyspnoea can interrupt sleep.
Pain, acute or chronic, and snoring can also cause poor sleep. Chronic insomnia is
associated with increased rates of depression and poor quality of life and requires
careful investigation. The initial approach to treatment should attempt to identify the
cause of the insomnia through taking of sleep history, a general medical and psychiatric
history, a review of the patient’s current social and personal situation, and a review of all
medications, including caffeine, tobacco, and alcohol use. Following good sleep
hygiene practices, e.g. avoiding stimulants and screen use prior to bedtime, following a
regular bedtime routine, and ensuring a comfortable and quiet sleep environment, often
improve or resolve sleep problems. Hypnotics may provide a temporary improvement in
sleep times and sleep quality, but they disrupt the normal sleep cycle (described below)
and can reduce the length of time spent in deep sleep or in rapid-eye movement (REM)
sleep.
Benzodiazepines
BDZs, discussed in detail in the Focus Box below, all have hypnotic activity. Their action
in enhancing GABA activity promotes the onset of sleep, and they are widely used in
sleep complaints. However, they predispose to dependence; reduce the length of time
spend in deep sleep; cause hangover effects the next day because of their long half-lives;
increase the risk of accident, injury, and falls, especially in elderly people; and cause
cognitive and memory impairment.
Anxiety
Acute panic attacks
Insomnia
Prevention of seizures
Emergency treatment of status epilepticus/febrile convulsions/seizures
caused by poisoning
Adjunct to anaesthesia/conscious sedation
Muscle relaxant: useful in reducing physical tension in anxiety, when used as
an adjunct to anaesthesia, and in conditions featuring muscle spasm
Alcohol withdrawal
Adverse Effects
BDZs cause sedation, impaired memory, confusion, and cognitive impairment,
especially in elderly people. They cause skeletal muscle weakness and ataxia, and
impair co-ordination and fine motor control, increasing the risk of accidents, falls
(especially in elderly people), and injury, e.g. from operating machinery. They
potentiate the effects of other CNS depressants, including alcohol and opioids, and
in combination with these agents can cause life-threatening respiratory depression.
Tolerance develops rapidly, requiring steady increase in dosing when used in the
medium to long term. Dependence is common in medium- to long-term treatment,
and it is recommended that treatment should not exceed 4 weeks. Both physiological
and psychological dependence develop, with well-defined withdrawal syndromes. The
likelihood of dependence increases with the duration of treatment, with higher doses,
and with shorter-acting agents such as lorazepam. Withdrawal symptoms include
anxiety, insomnia, irritability, depression, and sometimes convulsions.
Benzodiazepine Toxicity
BDZs are safer in overdose than older anxiolytic and hypnotic agents like the
barbiturates, and if taken alone in excessive doses generally produce extended sleep
from which the person eventually wakes naturally. However, if overdose reversal is
required, for example if other CNS depressants such as opioids or alcohol have also
been taken, the BDZ antagonist flumazenil is given. Flumazenil binds to and blocks
the BDZ receptor, but its half-life is only about 1 hour, considerably shorter than that
of the BDZs it is being used to reverse, so is usually given by infusion rather than as a
one-off dose.
Pharmacokinetics
These drugs are given orally, are well absorbed, and are metabolised in the liver to
inactive metabolites. One advantage over the BDZs is their shorter half-life, usually no
longer than 5 hours, reducing the incidence of accumulation and hangover effects in the
morning.
Adverse Effects
The Z-drugs cause dependence, cognitive impairment, and hangover effects despite
their relatively short half-life.
Table 4.4
Mood-stabilising drugs
The main mood-stabilising agent is lithium, but the range of drugs used for this
purpose is expanding and includes certain anticonvulsant drugs, e.g. lamotrigine and
sodium valproate, and some atypical antipsychotics, e.g. risperidone and
olanzapine. Mood stabilisers prevent mood swings in bipolar disorder.
Bipolar Disorder
This condition has a strong hereditable component and usually manifests in young
adulthood. It is characterised by fluctuating and alternating episodes of depression,
episodes of mania (elevated mood), and periods of euthymia (normal mood). The
depressive component is usually more marked than the manic condition and is treated
with standard antidepressant therapy. The manic episodes feature euphoria, reckless
and impulsive behaviour, loss of inhibition and increased libido, and even psychotic
symptoms such as delusions and hallucinations. The risk of suicide is significantly
increased in bipolar disorder. Lithium or sodium valproate (p. 75) are the mainstays
of treatment, with antidepressants, antipsychotics, and anticonvulsants used as
supplementary agents.
Lithium
Lithium is usually effective in regulating mood and preventing relapse, but it has
significant side-effects and toxicity, and controlling plasma levels is challenging. It is
given as lithium carbonate, which dissociates to release lithium ion (Li+), the active
agent. The mechanism by which lithium stabilises mood is not known, although it has
been shown to have multiple effects on enzyme activity, gene expression, and nerve
excitability in the brain, and accumulates inside body cells because it passes through
Na+ channels.
Pharmacokinetics
Lithium is given orally and is excreted by the kidney. Because it is not metabolised,
clearance of lithium from the body depends entirely on renal function, so care must be
taken in individuals with any degree of renal compromise. Lithium has a long half-life,
partly because so much of it collects within cells via Na+ channel uptake. Initiating
lithium treatment must therefore be done gradually, with regular measurements of
plasma levels, because it takes 2 weeks or more to achieve steady state, and reversing
toxic levels is slow because excretion is slow. The therapeutic plasma range is narrow,
only 0.4–1 mmol/L, making safe lithium management even harder.
Adverse Effects
Lithium’s side-effects are common and significant and contribute to the declining use of
this drug despite its well-documented efficacy in stabilising mood. It causes nausea in
up to 20% of patients, diarrhoea in up to 10% of patients, and vomiting. Up to 70% of
patients experience excessive thirst (polydipsia) and production of large volumes of
dilute urine (polyuria). Polyuria is due to lithium’s interference with the kidney’s ability
to respond to anti-diuretic hormone, which acts on the distal tubules and collecting
ducts, increasing water reabsorption and concentrating the urine. Lithium blocks this
action, leading to the production of large quantities of dilute urine, and triggering thirst.
Tremor is seen in about 25% of patients and can be severe or inconvenient enough to
require additional management, usually with a β-blocker. Weight gain is common and
frequently a reason for the patient failing to comply with treatment or wishing to
discontinue. The reasons for lithium-induced weight gain are not known, but it is
thought that the drug interferes in some way with fundamental control mechanisms in
the brain that regulate bodyweight, metabolism, or appetite. Lithium accumulates in the
thyroid gland and can impair thyroid hormone synthesis and release, causing goitre and
hypothyroidism.
Antipsychotic drugs
Psychotic illness is characterised by a loss of contact with reality. It features
hallucinations (experiencing events that are not happening), delusions (false and
abnormal beliefs), and emotional blunting and disordered thinking, and the person has
no insight into their condition. Although psychotic episodes may be caused by a range of
situations, e.g. alcohol withdrawal, recreational drug use (e.g. cocaine,
amphetamines, cannabis), childbirth (post-puerperal psychosis), and some
therapeutic drugs (e.g. vigabatrin), it may manifest itself as part of a chronic
psychiatric illness, notably schizophrenia. The term ‘antipsychotic agent’ is generally
taken to mean a drug used to treat schizophrenia.
Schizophrenia
This is a common and serious psychiatric condition, affecting 20 million people
worldwide, and is more common in men than women. It generally manifests early in life,
has a strong hereditable component, and increases the risk of premature death, partly
because of higher suicide rates. The signs and symptoms of schizophrenia are classified
into four groups: positive symptoms, negative symptoms, cognitive symptoms, and
mood symptoms, shown in Box 4.2. Positive symptoms are ‘add-ons’ to normal
behaviour, whereas negative symptoms are elements of behaviour and emotional
responses that are missing or depressed compared to normal. Individuals whose
schizophrenia displays predominantly positive symptoms tend to respond better to drug
treatment than those whose illness features mainly negative symptoms.
Neurophysiology of Schizophrenia
The underlying neurophysiology is not fully understood, but in schizophrenia there are
abnormalities in brain structure and volume and neurotransmitter activity. The most
clearly implicated neurotransmitter is dopamine, and all known antipsychotic drugs
block dopamine receptors, particularly D2 receptors, emphasising the role of dopamine
in the aetiology of schizophrenia. Dopamine is released at nerve endings in four main
pathways in the brain, three of which are shown in Fig. 4.19. The nigrostriatal pathway
is essential to skeletal muscle control, and its degeneration is the cause of Parkinson’s
disease. The fourth important pathway releasing dopamine, not shown here, is the
tuberoinfundibular pathway, which controls prolactin release and therefore breast
growth and lactation. Release of dopamine here inhibits prolactin release (see Fig. 5.2).
Antipsychotic drugs
These are generally considered under two main headings: the older, typical
antipsychotics, including chlorpromazine, haloperidol, and flupentixol, and the
newer atypical antipsychotics, sometimes also called second-generation antipsychotics,
including clozapine and risperidone. The distinction between the two groups is not
clear-cut, but in general, typical antipsychotics are fairly selective antagonists for D2
receptors, whereas the atypical agents have a wider spectrum of receptor-blocking
activity, including at 5-HT and histamine receptors.
Endocrine Effects
The release of dopamine in the tuberoinfundibular pathway switches off prolactin
secretion via D2 receptors (see Fig. 5.2). Dopamine antagonists release the
hypothalamus and the posterior pituitary from its suppression, and prolactin is secreted
into the bloodstream. This can cause swelling of the breasts and lactation in both men
and women.
FIG 4.20 Torticollis: an example of an acute antipsychotic-induced
dystonia. From Wikipedia: James Heilman, MD
Anti-Emetic Activity
Dopamine is a transmitter in the chemosensitive trigger zone and stimulates vomiting.
Antipsychotic drugs including metoclopramide and domperidone are therefore
anti-emetic (see p. 194).
Chlorpromazine
This was the first drug introduced to clinical medicine specifically as an antipsychotic
and was in widespread use by the late 1950s. Structurally, it belongs to the same family
as the antihistamines, and like the antihistamines it is very sedative; this led
researchers to investigate the possibility of its being useful as a premedication before
surgery, and from there its usefulness in psychotic disorders was quickly recognised. It
is sometimes used for intractable hiccups and tics, and in nausea and vomiting in
serious illness when other anti-emetics have failed.
Pharmacokinetics
Chlorpromazine is well absorbed orally and is highly plasma protein-bound. It is heavily
metabolised in the liver and has a half-life of around 30 hours. It should not be taken
with alcohol, and it can be an irritant in the gastrointestinal tract, so taking with food is
recommended.
Haloperidol
Haloperidol is one of the most potent antipsychotic drugs and can be given orally or as a
depot preparation given as a deep intramuscular injection, giving effective plasma levels
over an extended period. This smooths out plasma levels and eliminates the possibility
of the patient forgetting to take a dose. It is sometimes used as an anti-emetic in serious
illness, and to calm and sedate in a range of conditions associated with agitated,
aggressive, or hyperactive behaviour, e.g. Tourette’s syndrome and acute psychotic
episodes.
Flupentixol
Flupentixol is absorbed slowly and incompletely after oral administration and is often
used as a depot preparation. It is highly bound to plasma proteins, and its half-life
generally falls between 19 and 39 hours. It is sometimes also used in depression.
Clozapine
Clozapine and the related agent olanzapine bind to a wide range of receptors in the
brain, and their dopamine receptor blockade is actually fairly weak, which probably
accounts for their relative lack of extrapyramidal symptoms. They bind strongly to H1
receptors and so are sedative, although less so than the typical antipsychotics. They also
bind strongly to 5-HT receptors and muscarinic receptors. Clozapine is especially useful
in patients whose psychosis has not responded to other treatments. However, it can
cause dangerous intestinal obstruction, fatal agranulocytosis, and potentially fatal
myocarditis.
Pharmacokinetics
Clozapine is rapidly and almost completely absorbed following oral administration and
is very highly plasma protein-bound. Its average half-life is about 8 hours, and it is
metabolised in the liver.
Risperidone
Risperidone is used in a range of psychotic disorders and to treat mania. It is associated
with a higher incidence of extrapyramidal side-effects than clozapine, but less than the
typical antipsychotic agents. It is well absorbed from an oral dose, is highly plasma
protein-bound, and its half-life can range from 3 to 20 hours. It is metabolised in the
liver to the active metabolite paliperidone and cleared by the kidney.
Aripiprazole
Aripiprazole is also used in bipolar disorder. It is associated with fewer side-effects than
other atypical antipsychotics, possibly because it has an unusual interaction with
dopamine receptors. It has partial agonist activity, so that even though it occupies the
receptor and prevents dopamine from attaching, it causes a minor degree of receptor
stimulation. The incidence of extrapyramidal symptoms is reduced and prolactin levels
do not increase. In addition, weight gain is not usually a problem. It also has few
antimuscarinic side-effects.
Pharmacokinetics
Aripiprazole is well absorbed orally, although if taken with a fatty meal, absorption is
delayed. It is almost 100% plasma protein-bound and has a very long half-life of 75
hours. It is metabolised in the liver to an active product which has an even longer half-
life. This extended half-life means that aripiprazole can be useful in maintenance
treatment.
Anaesthetics
‘Anaesthesia’ means ‘absence of sensation’. General anaesthetics (GAs) depress
nerve function in the CNS and produce controlled unconsciousness associated with
analgesia and amnesia. Local anaesthetics (LAs) interrupt the conduction of action
potentials in peripheral sensory nerves and cause loss of sensation in the tissues served
by these nerves; clinically, eliminating pain signals is the primary aim, but temperature
and pressure sensation is also lost or impaired. In high enough concentrations, LAs
block conduction in all electrically excitable cells, and therefore also silence motor
nerves, central nerves, and muscle cells.
General anaesthetics
Before the introduction of the first successful inhalational anaesthetic, ether, used in
1846 for a dental extraction, surgical procedures had to be fast and crude, and the
unfortunate patient physically restrained. Many patients died of shock from the pain or
from infection. Alcohol and opioids could be given beforehand, but doses that
achieved adequate sedation were in themselves life-threatening. The horrors and
dangers of pre-anaesthetic surgery were such that people tolerated intensely painful or
debilitating conditions rather than present themselves for treatment. The introduction
of general anaesthesia transformed surgery, giving surgeons time to perform
increasingly complex and precise procedures. Early anaesthetics included ether,
hazardous because of its high flammability, and chloroform, which was associated
with a high death rate because of cardiovascular and hepatic toxicity. Both have been
superseded with newer, safer agents, and both inhalational and intravenous drugs are
available.
Chemically speaking, GAs are a diverse group of substances. The inert gas xenon and
nitrous oxide (N2O), both simple structures, induce anaesthesia. Other agents have
much more complex molecular structures. However, they do have one important
property in common: they are all highly lipid-soluble and on administration are rapidly
and extensively taken up into tissues, including the CNS. Anaesthetic potency therefore
increases with lipid solubility.
Table 4.5
Table 4.6
Inhalational anaesthetics
These are low-molecular-weight gases or volatile liquids (i.e. liquids that release gas).
They are inhaled into the alveoli of the lungs, from where they rapidly diffuse into the
bloodstream. Because they are so lipid-soluble, they dissolve poorly in the water-based
medium of the blood and are rapidly taken up into the CNS because of its rich blood
supply and relatively high lipid content.
The MAC decreases with age and is reduced in certain situations, e.g. anaemia,
hypoxia, pregnancy, and alcoholic intoxication. It is increased in chronic alcohol use and
in people with (naturally) red hair.
Pharmacokinetics
Achieving MAC depends on the concentration of inhaled anaesthetic, the rate and depth
of respiration, and the presence of certain pathologies, e.g. emphysema. The higher the
anaesthetic concentration and the deeper and faster the respiratory effort of the patient,
the faster the MAC is reached. In emphysema, total lung volume is increased, although
the number of functional alveoli is reduced, so the anaesthetic concentration in
functional alveoli is diluted and it takes longer to achieve MAC. With continued
administration, the drug equilibrates between the alveoli, the bloodstream, and the
tissues, including the CNS (Fig. 4.21A). Blood flow to an organ is an important
determinant of equilibration; well-perfused tissues receive higher quantities of drug and
equilibrate quickly, whereas tissues with a lower blood supply, e.g. adipose tissue,
equilibrate more slowly. Termination of action (reversal) of an inhaled anaesthetic relies
almost entirely on pulmonary excretion. On withdrawal of the anaesthetic, its alveolar
concentration immediately falls, meaning that it is no longer in equilibrium between
alveolar air and the bloodstream, and the drug transfers down its concentration gradient
into the alveoli and is excreted in the breath (Fig. 4.21B). As blood levels fall, the drug
transfers from tissues into the bloodstream, reducing levels in the CNS and initiating
recovery. The ‘hangover’ effect seen with some inhalational anaesthetics is due to the
accumulation of the drug in body fat stores. Because adipose tissue has a poor blood
supply, anaesthetics are taken up relatively slowly despite their high fat solubility, but
for the same reason, they are released slowly after reversal and can lead to drowsiness
and impaired motor function for up to 24 hours. In recovery, the patient passes through
the four stages of anaesthesia (Table 4.6) in reverse, including the excitation stage. An
ideal inhaled anaesthetic therefore reverses very quickly, minimising time spent in this
potentially dangerous period. Factors that depress respiration, e.g. opioid
administration, slow down recovery.
Modified Ethers
Examples: desflurane, isoflurane, sevoflurane
These are simple molecules based on the chemical structure of ether. They target
multiple channels and molecular sites in the CNS, inhibiting synaptic transmission and
reducing nerve activity. Sevoflurane and desflurane give faster induction and
recovery than isoflurane. They all suppress respiration by reducing the sensitivity of
the respiratory centres in the brainstem to carbon dioxide, an important stimulant to the
inherent rate and rhythm of breathing. They relax smooth muscle, including in blood
vessel walls, causing vasodilation and hypotension. This reduces perfusion of key organs
including the heart and the liver. In addition, they may directly depress myocardial
contractility, worsening any hypotension. They relax the uterus, which can slow labour
and increase the risk of haemorrhage, because the contracting uterus normally
compresses blood vessels that are damaged as the placenta separates during the third
stage of labour. They induce postoperative nausea and vomiting because they stimulate
the chemosensitive trigger zone of the medulla oblongata, and they may irritate the
respiratory tract, causing coughing and laryngospasm. Sevoflurane is the least irritant
in this group.
FIG 4.21 Equilibration of inhaled anaesthetic between lungs,
tissues, and blood.A. Induction and maintenance. 1: The anaesthetic
equilibrates between alveolar air and arterial blood. Provided the
minimum alveolar concentration (MAC) has been reached, this
means that blood levels are high enough to achieve anaesthesia. 2:
Well-perfused tissues, e.g. the central nervous system (CNS), liver,
and other organs, rapidly equilibrate. Provided arterial drug levels
remain high enough, anaesthesia is induced and maintained. 3:
Poorly perfused tissues, e.g. adipose tissue, also equilibrate but more
slowly. B. Reversal. 1: When administration stops, alveolar
anaesthetic concentration rapidly drops. The drug diffuses rapidly
out of the bloodstream into the alveoli and is excreted in the breath.
2: As blood levels fall, anaesthetic diffuses rapidly down its
concentration gradient from the tissues, including the CNS, into the
blood. Well-perfused tissues clear the drug very quickly. 3:
Anaesthetic levels fall more slowly in poorly perfused tissues, which
may still contain measurable levels of anaesthetic for hours after
administration has stopped.
Nitrous Oxide
N2O induces anaesthesia rapidly and is pleasant to use because it is odourless and non-
irritant. Its mechanism of action has not been clearly explained, but it is known to
interact with a range of neurotransmitter receptors in the brain, including inhibition of
the excitatory NMDA receptor family. It also releases endogenous opioids in the
brainstem, activating endogenous pain-modulating pathways in the spinal cord, and
presumably contributing at least in part to its excellent analgesic activity. It is less
cardiodepressant and less emetic than the modified ethers, but it does not achieve high
enough alveolar concentrations to give full anaesthesia on its own. It is often used with
other inhaled anaesthetics, which allows the concentration of the second agent to be
reduced, and it is used as an analgesic in labour and other situations where short-term
or emergency pain relief may be needed, e.g. after fracture or burns.
Intravenous anaesthetics
Examples: thiopental, propofol, etomidate, ketamine
These may be used as a bolus to induce anaesthesia or by infusion to maintain
anaesthesia in shorter procedures.
Pharmacokinetics
These lipid-soluble drugs enter the CNS within seconds following intravenous injection,
giving fast onset of anaesthesia. If given as a single bolus, anaesthesia is short-lived,
because although the drug is initially taken up into the CNS due to its rich blood supply,
it then steadily distributes throughout body tissues and CNS levels quickly fall. Reversal
therefore depends upon drug distribution. If the anaesthetic is infused over a period of
time, it equilibrates throughout body tissues, and reversal becomes dependent upon
metabolism.
Thiopental
Thiopental is a barbiturate, a class of drugs largely abandoned in clinical medicine
because of dangerous side-effects. It is very lipid-soluble and gives a very fast induction,
but it has significant depressant activity on cardiovascular and respiratory function.
Thiopental induces anaesthesia by enhancing GABA activity.
Propofol
Propofol potentiates the inhibitory action of GABA at GABAA receptors. It is mainly
used as an induction agent and in lower doses for conscious sedation. It depresses
cardiovascular and respiratory function and can cause pain on injection because it has
very low solubility in water and is formulated as an oil-in-water suspension. However, it
causes little nausea or vomiting, reverses rapidly with little hangover, and its amnesiac
action is useful for patients undergoing short procedures with conscious sedation.
Etomidate
Etomidate potentiates the inhibitory action of GABA at GABAA receptors, but it is not
chemically related either to the benzodiazepines or the barbiturates. It is very rapid
acting and has a short plasma half-life of 6–10 minutes, because it is rapidly degraded
by plasma and liver esterases to inactive products. It causes less cardiovascular
depression than propofol, but it can be painful on injection and causes postoperative
nausea and vomiting. It is not used for maintenance of anaesthesia because it causes
adrenal suppression. Adrenal steroids, e.g. cortisol, are essential for an effective stress
response, essential to supporting physiological function in illness and trauma, including
surgery. Adrenal suppression therefore increases risk, especially in severely ill patients.
Ketamine
Ketamine affects the function of a range of receptors in the brain, including blockade of
excitatory NMDA receptors. Its analgesic properties are probably due to activation of
internal opioid pain modulation mechanisms. It gives slow induction and slow recovery,
and causes hangover effects and postoperative nausea and vomiting. It induces a state
called ‘dissociative anaesthesia’, in which the patient appears awake, with eye opening,
good muscle tone, and preserved laryngeal reflexes, but with excellent analgesia. It
stimulates the sympathetic nervous system, supporting cardiovascular and respiratory
function, and has a significant psychotropic effect––people report hallucinations, vivid
dreams, and detachment from reality.
Table 4.7
Neuronal Factors
Small-diameter neurones are more susceptible to LA action than larger ones. This is
fortunate, because pain neurones are small-diameter free nerve endings, whereas
other sensory and motor nerves are larger and require higher drug concentrations
before their conductance is impaired. Additionally, a myelin sheath presents an extra
barrier to LA penetration, so that non-myelinated nerves are more susceptible than
myelinated ones. This too is clinically advantageous, because sensory pain fibres are
non-myelinated. In addition, the duration of the action potential affects tissue
susceptibility. Pain neurones and cardiac muscle cells both have longer than average
action potentials, meaning their Na+ channels stay open for longer. This is beneficial
when abolishing pain signals and explains why LAs can significantly depress heart
function if they achieve high enough levels in the bloodstream. Sensory modalities are
lost in a predictable order as LA concentration rises, reflecting the sensitivity of
sensory and motor nerves to LA: pain sensation is lost first, followed by temperature,
then touch, then deep pressure, and finally motor function.
Use-dependence
Neurones with high rates of firing, e.g. pain fibres, are highly susceptible to LA action
because their Na+ channels spend more time in the open state. This is clearly
clinically useful. High rates of firing are also characteristic of other electrically active
tissues, including ectopic foci in the heart. This is the reason why LAs, e.g. lidocaine,
are used to treat some arrhythmias.
Pharmacodynamics
LAs are lipid-soluble, allowing them to diffuse across nerve cell membranes, and the
more lipid-soluble they are, the more potent they are. The degree to which they bind
to tissue proteins determines their duration of action; strong protein binding helps
the drug to be retained at its site of administration and prolongs its action. Cocaine
and tetracaine have a very short plasma half-life (less than 3 minutes), because they
are metabolised by plasma enzymes, but the amides, e.g. lidocaine and
benzocaine, are metabolised in the liver and have plasma half-lives ranging from 1
to 3 hours. The key properties of some important LAs are given in Table 4.7.
Side-Effects
There may be a localised allergic reaction, but the most dangerous side-effects of LAs
occur when they escape into the circulation in high enough concentrations to inhibit
excitable tissues elsewhere. They reduce myocardial contractility and can interfere
with the heart’s internal pacemaker activity and impulse conduction. This
cardiodepressant activity along with their hypotensive action can lead to
cardiovascular collapse and death. Cocaine is an exception to the general pattern of
hypotension and cardiac depression because it causes vasoconstriction, stimulates the
heart, and increases blood pressure. Local anaesthetic toxicity in the CNS shows a
biphasic pattern, with initial excitation followed by depression. Excitation might seem
counterintuitive, since these drugs inhibit neural function, but it occurs because the
first nerves to be blocked are inhibitory neurones that suppress excitatory pathways.
With their inhibitory neurones silenced by the anaesthetic, overactivity of these
excitatory pathways leads to a spectrum of effects ranging from mild excitement to
convulsions. With increasing doses, a generalised depression of CNS function follows,
including shutting down of basic life-support mechanisms in the cardiovascular and
respiratory centres in the brainstem, coma, and death.
Anticonvulsants
A seizure is caused by a sudden and uncontrolled episode of electrical disturbance in the
brain, originating in a group of hyperexcitable neurones called the epileptogenic focus.
Epilepsy, sometimes called seizure disorder, is a chronic condition characterised by
recurrent, unprovoked seizures, but not all seizures are associated with epilepsy. The
aim of anticonvulsant therapy is, whenever possible, to prevent seizures from occurring,
or at least to reduce their frequency and severity as much as possible.
Causes of seizures
A wide range of diseases and disorders, not necessarily neuropathological in origin, can
trigger seizures. The most common cause of seizures is epilepsy, but one-off seizures
may be caused by infection, hyponatraemia (e.g. in poorly monitored diuretic therapy),
high fever (especially in young children), hypoglycaemia, recreational drug use (e.g.
cocaine), or during alcohol withdrawal. Traditionally, epilepsy has been considered to
be caused by an imbalance between excitatory and inhibitory nerve activity, permitting
episodic dominance of excitatory neurotransmission. A range of brain abnormalities
have been associated with epilepsy, including neurodegeneration, inflammation, and
structural and functional changes in excitatory and inhibitory neurotransmitters and
their receptors and associated ion channels. Epilepsy can be caused by congenital brain
malformations, cerebral hypoxia, scarring following brain injury, a tumour,
haemorrhage, or stroke. In other people, epilepsy can arise with no obvious underlying
cause. Some forms of epilepsy have a strong hereditable component, and there is an
increasing list of ‘epilepsy genes’, an important area of research in the aetiology of
epilepsy.
Types of seizure
Seizures are classified according to the location of the epileptogenic focus in the brain,
their duration, and the characteristic features of the event, which is determined by
which part(s) of the brain are affected. For example, if the abnormal neuronal discharge
occurs in the motor cortex, the seizure will involve involuntary activity in skeletal
muscle.
Focal Seizures
If the abnormal electrical discharge remains localised within one cerebral hemisphere, it
causes a focal seizure. The patient may remain aware throughout the seizure and have
full memory afterwards (focal aware seizure) or may lose awareness with limited recall
(focal impaired awareness seizure). There may be a range of involuntary activities called
automatisms, including hand rubbing and lip smacking. Drugs with a range of modes of
action, including Na+-channel blockers, e.g. carbamazepine and phenytoin, drugs
that increase the activity of GABA, e.g. the benzodiazepines and vigabatrin, and
drugs that reduce the activity of glutamate, e.g. lamotrigine, are effective in treating
focal seizures.
Generalised Seizures
In generalised seizures, the abnormal electrical activity occurs in both cerebral
hemispheres, and may either evolve from a focal seizure or begin as a generalised
seizure. Consciousness is almost always lost. Two important forms of seizure in this
category are tonic-clonic convulsions and absence seizures.
Tonic-Clonic Seizures
In tonic-clonic seizures (formerly called grand mal), there is an initial sudden and
widespread stiffening of skeletal muscle, causing respiratory arrest and often
incontinence. This is followed by the clonic phase, in which skeletal muscles alternately
contract and relax, causing violent jerking of the limbs. Afterwards, the patient is
generally sleepy, confused, irritable, and disoriented. Drugs that block Na+ channels in
nerve axons, e.g. phenytoin, or that increase the levels of GABA, e.g. vigabatrin, are
effective in controlling tonic-clonic seizures.
Absence Seizures
In absence seizures, (previously called petit mal epilepsy) which are usually seen in
children, there are few motor symptoms. The child abruptly stops whatever they were
doing, including talking, and appears to be daydreaming, with no awareness of their
surroundings. Return of full awareness is also abrupt, and the child is not aware that the
seizure has occurred. The aetiology of absence seizures is not understood, but they seem
to be associated with a particular pattern of electrical discharge in a neuronal circuit
between the thalamus and the cortex. Abnormalities in Ca2+ channels and in GABA
activity have both been demonstrated in this circuit, reflected in the agents used to treat
absence seizures. The most effective drugs include sodium valproate and
ethosuximide, which block the abnormal Ca2+ channels. Agents that increase GABA
activity, e.g. vigabatrin, can make absence seizures worse, as can Na+-channel blockers
such as phenytoin.
Status Epilepticus
This is either a single extended seizure or a continuous series of seizures with no
recovery between them persisting for over half an hour. It carries a mortality of up to
15%, and there is high risk of permanent hypoxic brain damage following recovery. If the
seizure involves convulsions, skeletal muscle damage (rhabdomyolysis) can release
significant quantities of myoglobin into the bloodstream, which may cause acute kidney
injury. Benzodiazepines e.g. diazepam and midazolam, intravenous anticonvulsants,
e.g. phenytoin, and general anaesthetics, e.g. propofol or thiopental, are used to
terminate status epilepticus.
Phenytoin
Phenytoin is used in all types of epilepsy except absence seizures.
Pharmacokinetics
Phenytoin is completely absorbed after oral dosing, and is 90% bound to plasma
proteins, giving a substantial reservoir of drug in the bloodstream. Its metabolism is
complex because it induces a range of liver enzymes, including those that catalyse its
own breakdown. This means that in the initial stages of treatment, phenytoin has a long
half-life (36 hours) which falls to 12–18 hours with continued use. It increases the
breakdown of a range of other drugs, including anticoagulants, an important cause of
interactions.
Phenytoin’s metabolism is not straightforward. As plasma levels rise, its metabolism
shifts from linear to saturation kinetics (p. 19 and Fig. 2.21). Fig. 2.21 shows plasma
levels in three individuals receiving phenytoin treatment. For all individuals, the
transition between linear kinetics and saturation kinetics, i.e. the point at which the
metabolising enzymes are working at full capacity and any additional drug simply
accumulates in the bloodstream, is seen at subtherapeutic levels. In addition, this figure
demonstrates the significant variability between individuals in the ability to metabolise
phenytoin, an additional complicating factor in safe management of phenytoin
treatment. The daily dose of phenytoin required to achieve mid-therapeutic levels for
the patient whose plasma levels are represented by the red curve is three times lower
than the patient results shown by the green curve. It also reinforces the point that even
small changes in daily dose can have significant effects on plasma levels, which would
impact on seizure control and the likelihood of side-effects. Therapeutic drug
monitoring is important to ensure that plasma levels remain in the therapeutic range,
especially when adjusting drug dose or introducing other drugs.
FIG 4.24 Summary of the mechanism of action of some important
anticonvulsant drugs.BDZ, Benzodiazepine; GABA, gamma-
aminobutyric acid.
Adverse Effects
Phenytoin has a narrow therapeutic index, and a range of dose-dependent adverse
effects caused by its interference with CNS function appear at the upper end of its
therapeutic dose range. Ataxia, tremor, and vertigo are common, and in higher doses the
drug is sedative and causes confusion and reduced intellectual function. Allergy occurs
in a small number of patients and may manifest as a mild skin rash or can involve life-
threatening desquamation reactions, including Stevens–Johnson syndrome (Fig. 3.13).
Phenytoin stimulates connective tissue growth, particularly affecting the gums, and
causes acne and hirsutism, probably through a stimulation of testosterone release. It
increases the risk of fetal and facial and digital malformations when used in pregnancy.
It can also cause blood disorders because it interferes with the metabolism of folic acid,
an essential factor in blood cell synthesis.
Carbamazepine
Carbamazepine is used in most types of epilepsy except absence seizures. It is also
sometimes used in neuropathic pain, e.g. trigeminal neuralgia and diabetic neuropathy,
because it silences the pain signals generated by abnormal or injured neurones and can
also be effective in bipolar disorder.
Pharmacokinetics
Carbamazepine is well absorbed when given orally and is up to 80% plasma protein-
bound. Like phenytoin, carbamazepine induces liver metabolic enzymes, including its
own. Its half-life in the initial stages of treatment is about 36 hours, but with continued
administration and autoinduction of metabolism, this falls to 12–17 hours. It induces the
metabolism of other drugs, notably oestrogens in contraceptive preparations, leading
to rapid clearance of oestrogen and possible contraception failure. Carbamazepine
metabolites are inactive and are mainly excreted in the urine.
Adverse Effects
Carbamazepine causes a range of dose-dependent neurological side-effects related to its
depressant activity on the CNS; they include ataxia, headache, and drowsiness. There
may be hypersensitivity reactions, mainly skin rashes, but occasionally severe allergy is
seen, including Stevens–Johnson syndrome (Fig. 3.13). It can cause water retention
and oedema because it antagonises the effects of antidiuretic hormone, and the
increased fluid load dilutes blood Na+ levels and causes hyponatremia.
Benzodiazepines
All BDZs have anticonvulsant activity, but because they are sedative and induce
dependence and tolerance, their usefulness in long-term therapy is limited. The main
agents used in maintenance treatment are clobazam and clonazepam. However,
BDZs are the drugs of choice in controlling status epilepticus and are given rectally or
intravenously. Diazepam, lorazepam, and midazolam are the main agents used.
The pharmacology of the BDZs is described above.
Vigabatrin
Vigabatrin irreversibly blocks GABA transaminase, the enzyme that destroys GABA.
Reducing its enzymatic destruction elevates GABA levels in the brain and enhances its
inhibitory action. It is an example of rational drug design, having been formulated
specifically to act against this specific enzyme. It is a second-line agent used in
combination with other anticonvulsants to improve control when the primary agent is
not fully effective. Vigabatrin is highly effective in some forms of childhood epilepsy and
in most adult epilepsies, but its use is restricted because it causes permanent peripheral
vision loss in up to 40% of patients.
Pharmacokinetics
Vigabatrin is well absorbed from an oral dose, does not bind significantly to plasma
proteins, and is not metabolised. Its plasma half-life is about 10 hours, but because it
irreversibly deactivates GABA transaminase, its duration of action depends on how
quickly the brain synthesises new enzyme rather than how long the drug persists there.
This also means that plasma levels of the drug do not correlate with its therapeutic
activity and measuring them is not useful.
Adverse Effects
The most significant side-effect of vigabatrin is permanent impairment of peripheral
vision because the drug damages retinal neurones and causes them to atrophy. The risk
is greater in males than females and increases with the duration of treatment, but it has
been reported after only 4 weeks of using the drug. It is common, with up to 40% of
patients developing constriction of their visual field. Although central vision is
unaffected, restricted peripheral vision can have a range of negative effects, including
reducing one’s ability to play sports requiring awareness of players or a ball approaching
from the side, or limiting a driver’s ability to see pedestrians stepping onto the road
(Fig. 4.25). Regular vision tests are therefore essential for patients taking this drug,
especially in children who are likely to be unaware of their gradual vision loss. A wide
range of other side-effects, including dizziness, drowsiness, memory loss, and
paraesthesias, have also been reported.
Tiagabine
Tiagabine is a GABA analogue: that is, its chemical structure is very similar to GABA,
and once it enters the brain, it competes with GABA for the GABA transporter that
removes GABA from the synapse. This preserves GABA levels in the synapse and
prolongs its inhibitory activity. It is used in conjunction with other anti-epileptic drugs
to improve seizure control when a single drug is not effective enough on its own.
FIG 4.25 Reduction of peripheral visual fields by vigabatrin.A.
Normal field of view. B. Vigabatrin treatment reduces peripheral
vision.
Ethosuximide
This anticonvulsant is used mainly in absence seizures in children (see above). It is well
absorbed when taken orally and has a half-life of around 60 hours in adults, although
this is shorter in children (30 hours) because of increased hepatic metabolism.
Sodium Valproate
This drug was discovered by accident in a series of experiments in 1962, during which
valproic acid was used to dissolve several experimental compounds undergoing a range
of pre-clinical tests, including seizure tests. As part of the standard experimental design,
valproic acid was used alone as a control and its anticonvulsant effect was immediately
obvious. It is now one of the most prescribed anticonvulsants worldwide, used in most
types of epilepsy including absence seizures. It is included here with drugs that block
Ca2+ channels in the brain, but this is thought to be only one of its mechanisms of
action. It also blocks Na+ channels in axon membranes, and it inhibits GABA
transaminase, the enzyme that degrades GABA, so it is likely that both action potential
inhibition and increasing GABA levels in central synapses contribute to its
anticonvulsant action. It may also increase GABA synthesis, and it suppresses the action
of pro-epileptic genes. Its full spectrum of action is clearly wide and not yet fully
understood. It is also used in bipolar disease and to prevent migraine.
Pharmacokinetics
Valproate is well absorbed after oral administration, has a plasma half-life of about 15
hours, and is metabolised in the liver.
Adverse Effects
Valproate is strongly teratogenic, causing spina bifida and other neural tube defects, and
it should not be used in sexually active women without effective contraception. It may
also cause potentially fatal hepatitis, but fortunately this is rare. It is a potent enzyme
inhibitor, reducing the metabolism and increasing the plasma levels of a range of other
drugs, including other anticonvulsants: this is an important cause of valproate-mediated
drug interactions. It has a range of other adverse effects, including hair loss (not
permanent, and the regrowth may be curly), confusion, memory loss, and tremor.
Lamotrigine
Lamotrigine blocks glutamate release from excitatory neurones, which reduces the levels
of glutamate in the synapse and reduces excitatory transmission. Lamotrigine also has
appreciable Na+-channel blockade activity. It is used in many types of seizure disorders
either as monotherapy or in combination with other anticonvulsants and in maintaining
mood stability in bipolar disorder.
Pharmacokinetics
Lamotrigine is well absorbed after an oral dose, and its bioavailability is almost 100%
because there is almost no first-pass metabolism. It is metabolised in the liver and
mainly excreted in the urine. Its half-life is long and variable between individuals,
ranging from 22 to 37 hours. It is often used with valproate, a potent enzyme inhibitor,
in which case its half-life can increase to 60 hours.
Adverse Effects
Lamotrigine can cause headache, drowsiness, nausea and vomiting, and tremor, among
others. It is sometimes associated with serious skin conditions, including Stevens–
Johnson syndrome (Fig. 3.13).
Topiramate
Topiramate was discovered by accident by researchers looking for antidiabetic agents. It
blocks glutamate receptors on the post-synaptic membrane. This reduces the excitability
of the nerve by inhibiting its ability to respond to this excitatory neurotransmitter. This
drug also blocks Na+ channels and probably Ca2+ channels, so its mechanism of action
in seizure control is probably complex. It is also used in migraine.
Pharmacokinetics
Topiramate is well absorbed orally and is not strongly bound to plasma proteins. Up to
70% is excreted unchanged by the kidney, so care must be taken in people with any
degree of renal impairment. Its half-life is usually within 19–23 hours.
Adverse Effects
This drug can cause hair loss, confusion, gastrointestinal upset, nausea, and tremor. It is
strongly associated with congenital malformations, especially cleft palate.
References
1. Atkin T, Comai S, Gobbi G. Drugs for insomnia beyond benzodiazepines:
pharmacology, clinical applications and discovery. Pharmacol.
Rev. 2018;70(2):197–245.
2. Becker D.E, Reed K.L. Essentials of local anaesthetic pharmacology. Anesth.
Prog. 2006;53(3):98–109.
3. Cipriani A, Furukawa T.A, Salanti G, et al. Comparative efficacy and acceptability
of 21 antidepressant drugs for the acute treatment of adults with major
depressive disorder: a systematic review and network meta-
analysis. Lancet. 2018;391(10128):1357–1366.
4. Egan T.D. Are opioids indispensable for general anaesthesia? Br. J.
Anaesth. 2019;122(6):e127–e135.
5. GBD 2017 Disease and Injury Incidence and Prevalence Collaborators, . Global,
regional and national incidence, prevalence and years lived with disability for 354
diseases and injuries for 195 countries and territories, 1990-2017: a systematic
analysis for the Global Burden of Disease study
2017. Lancet. 2018;392(10159):1789–1858.
6. Gitlin M. Lithium side-effects and toxicity: prevalence and management
strategies. Int. J. Bipolar. Disord. 2016;4:27.
7. Guardiola-Lemaitre B, De
Bodinat C, Delagrange P, Millan M.J, Munoz C, Mocaër E. Agomelatine:
mechanism of action and pharmacological profile in relation to antidepressant
properties. Br. J. Pharmacol. 2014;171(15):3604–3619.
8. Guzman F. The four dopamine pathways relevant to antipsychotics
pharmacology. 2019 Available
at:. https://psychopharmacologyinstitute.com/publication/the-four-
dopamine-pathways-relevant-to-antipsychotics-pharmacology-2096.
9. Jesulola E, Micalos P, Baguley I.J. Understanding the pathophysiology of
depression: from monoamines to the neurogenesis model-are we there
yet? Behav. Brain Res. 2018;341:79–90.
10. Scammell T.E, Jackson A.C, Franks N.P, Wisden W, Dauvilliers Y. Histamine:
neural circuits and new medications. Sleep. 2019;42(1):1–8.
11. Wick J.Y. The history of benzodiazepines. Consult. Pharm. 2013;28(9):538–548.
12. Wilson T.K, Tripp J. Buspirone. In: StatPearls [Internet]. Treasure Island
(FL): StatPearls Publishing; 2020 Available
from:. https://www.ncbi.nlm.nih.gov/books/NBK531477/.
5: Drugs and Endocrine Function
Introduction
F OCUS ON
Introduction
The endocrine system comprises a collection of glands and tissues dispersed around the
body and the hormones they produce and release. Many hormones are released into
local blood vessels and are carried to distant target tissues in the bloodstream. This is
called endocrine control. Other hormones act locally, in the tissues close to their site of
release, and regulate the activity and behaviour of cells in their immediate environment.
This is called paracrine control. Paracrine substances are referred to as mediators and
regulate local responses such as inflammation. They include growth factors;
inflammatory mediators including prostaglandins, histamine, and bradykinin; and
cytokines, a broad group of small peptides involved in inflammation, immunity, and the
production of blood cells. Mediators are increasingly important as drug targets in the
treatment of inflammatory, malignant, and immune disease and will be discussed, as
appropriate, in other chapters.
The main endocrine glands and tissues are shown in Fig. 5.1. Endocrine disorders
usually involve under- or over-secretion of hormones; thus, pharmacological
management generally involves hormone replacement or the use of hormone
antagonists or drugs that suppress hormone synthesis or release.
Adrenocorticotrophic Hormone
The synthetic ACTH analogue tetracosactide is used to test adrenal cortex function. It
is given by injection, and the adrenal cortex response is measured by measuring plasma
steroid levels. In adrenocortical insufficiency, the adrenal cortex cannot respond
adequately to the stimulus, and steroid levels remain low.
Growth Hormone
Growth rates are controlled by GH from the anterior pituitary, which affects all body
tissues, stimulating cell division and increasing body mass, including skeletal muscle. It
promotes cartilage cell division in the growth plates of bones, and so it is essential for
increasing height in a growing child and young person. It increases blood glucose levels
by stimulating gluconeogenesis (glucose synthesis from non-carbohydrate nutrients, e.g.
amino acids) and increases blood fatty-acid levels, ensuring that nutrient supply to
growing tissues is adequate. GH deficiency is rare and can be due to hypothalamic
failure causing a lack of GHRH, or pituitary failure causing GH deficiency. It may be
congenital, a consequence of a traumatic head injury, meningitis or other central
nervous system infection, or irradiation. In children, it retards growth and reduces
height (dwarfism), and even though GH levels fall naturally with age, this hormone
seems to be important for normal metabolism and maintenance of body mass in adults.
Excessive GH release in childhood leads to gigantism, with long limbs and unusually tall
adult stature. In adults, however, in whom the growth plates of the limb bones have
closed, growing taller is not possible, and excess GH thickens and enlarges bone and
other connective tissues, a condition called acromegaly. Coarsening of facial features is
characteristic of this disorder, with enlargement of the nose, jaw, hands, and feet.
Somatostatin
SST inhibits GH release, and therefore reduces cell division and tissue growth. It also
suppresses the secretion of a range of digestive enzymes and hormones, including
insulin, gastrin, and pancreatic enzymes, which enhance its anti-growth activity.
Somatostatin Analogues
Lanerotide and octreotide are synthetic analogues of SST given by subcutaneous or
intramuscular injection to treat conditions associated with excessive GH activity, e.g.
acromegaly. They are also used in a range of conditions featuring excessive secretion of
gastrointestinal (GI) hormones, including GI tumours. Unsurprisingly, their main
adverse effects involve the GI tract and metabolic function, and include nausea,
vomiting, abdominal pain, diarrhoea, and impaired glucose tolerance. They also
increase the risk of gallstones because they reduce the release of cholecystokinin, which
triggers gallbladder contraction. In its absence, the gallbladder empties more slowly and
less frequently, allowing bile to stagnate and increasing the risk of stone formation.
Gonadorelin
Gonadorelin is a synthetic preparation of GnRH used to test pituitary function. It is
administered by either intravenous or subcutaneous injection and the pituitary response
assessed by measuring plasma LH levels at set time intervals over the following 2 hours.
AntiDiuretic Hormone
ADH increases water re-absorption by the kidney, which increases blood volume and
blood pressure. The stimulus for ADH release is a rise in plasma osmolarity, i.e. an
increase in the plasma concentration, or a reduction in plasma volume. ADH’s target
tissues are the proximal convoluted tubule and collecting ducts of the renal nephron, the
regions of the tubule mainly responsible for water re-absorption. ADH increases the
number of water channels in the wall of the nephron, increasing water re-absorption
into the bloodstream. The plasma osmolarity therefore falls as the plasma becomes
more dilute, the blood volume rises, and a smaller, more concentrated volume of urine is
passed. ADH also causes vasoconstriction, increasing blood pressure. It also reduces
bleeding time by activating platelets and stimulating clotting. ADH itself is sometimes
used to treat diabetes insipidus, a condition characterised by inadequate ADH secretion.
Oxytocin
Oxytocin stimulates uterine contractions during childbirth and milk ejection during
lactation and is important in a range of behaviours including eating, social interactions,
and developing relationships, including establishing the parent-child bond. Synthetic
oxytocin is given by intravenous infusion to induce and speed up labour when medically
indicated and to prevent and treat post-partum haemorrhage. It can cause fluid
retention because of its ADH-like activity, which can dilute body fluids and cause
hyponatraemia.
The plasma half-life of thyroxine is 5–7 days, but T3’s half-life is 24 hours.
Functions of Calcitonin
Calcitonin reduces plasma calcium levels, opposing the effects of parathormone from
the parathyroid glands. Its influence on plasma calcium levels is, however, thought to be
relatively minor.
Hypothyroidism
Underactivity of the thyroid gland, hypothyroidism, is common. The most common
cause, Hashimoto’s disease, is an autoimmune disorder in which autoantibodies are
produced to thyroid gland TSH receptors. The autoantibodies bind to and block the
receptor, meaning that the thyroid gland cannot respond to TSH from the anterior
pituitary, and thyroid hormone release falls. Other causes include a diet low in iodine,
reversible with iodine supplements, and congenital hypothyroidism. Hypothyroidism
also often follows antithyroid treatment for hyperthyroidism. In hypothyroid states
caused by thyroid failure, plasma T4 levels are low, but TRH and TSH levels can be
significantly elevated because the hypothalamus and pituitary try to stimulate the failing
thyroid to secrete additional T4. Characteristic signs and symptoms of an underactive
thyroid are listed in Box 5.1.
Levothyroxine
This is the standard treatment for hypothyroidism. It is a synthetic drug, prepared as the
sodium salt of the natural hormone and converted to T3 in peripheral tissues.
Pharmacokinetics
Levothyroxine is given orally and moderately well absorbed. Absorption is reduced or
slowed in the presence of food and when gastric pH is higher than normal, e.g. in people
taking antacid medications. Levothyroxine has a long half-life, usually over a week in
hypothyroid patients, so it equilibrates slowly and can take several weeks to reach a
stable steady state. It is almost completely bound to T4-binding globulin, the plasma
protein responsible for transporting it in the bloodstream, which contributes to its long
half-life. It is metabolised in body tissues to T3 by removal of an iodine atom and from
there to inactive products.
Liothyronine
Synthetic T3 is available, also as a sodium salt, as the drug liothyronine. It has a much
shorter half-life than levothyroxine, usually less than 24 hours, and its onset of action is
faster. These properties mean that it is preferred to levothyroxine in acute or severe
hypothyroid states.
Adverse Effects
Excessive doses of levothyroxine or liothyronine give the same physiological effects as
the natural hormone. Cardiovascular stimulation can cause tachycardia, palpitations,
arrhythmias, and hypertension. There may be diarrhoea, weight loss, heat intolerance,
sweating, insomnia, tremor, and menstrual disturbances. Levothyroxine crosses the
placenta and can affect the fetus, so should be used very cautiously in pregnancy.
Hyperthyroidism
Overactivity of the thyroid gland, hyperthyroidism (thyrotoxicosis), is common. The
most common cause, Graves’ disease, is an autoimmune disorder in which
autoantibodies are produced to the TSH receptor on the thyroid gland. These
autoantibodies bind to and stimulate the TSH receptor, triggering thyroid hormone
production. Exophthalmos, protrusion of the eyeballs due to deposition of extracellular
matrix at the back of the eye sockets, is a characteristic of Graves’ disease. Other causes
of hyperthyroidism include toxic nodular goitre caused by a benign functional thyroid
tumour, which releases large quantities of thyroid hormone. Some drugs can also cause
hyperthyroidism; e.g. the antiarrhythmic agent amiodarone (p. 137) is structurally
similar to T4 and contains significant quantities of iodine: a 200 mg tablet (the usual
daily maintenance dose) delivers 75 mg of iodine. For reference, the daily recommended
iodine intake is only 0.14 mg/day.
Carbimazole
Like the similar agent propylthiouracil, carbimazole inhibits the enzyme thyroid
peroxidase, which catalyses the addition of iodine to tyrosine in thyroglobulin (Fig.
5.5), blocking the production of thyroid hormone. There is also evidence that these
drugs reduce levels of the anti-TSH receptor antibody that stimulates T4 production in
Graves’ disease. They are taken orally and metabolised in the liver.
Pharmacokinetics
Carbimazole is a pro-drug, converted in the bloodstream to the active agent
thiamazole. Both thiamazole and propylthiouracil are taken up and concentrated in
the thyroid gland. This means that although their plasma half-lives are relatively
short––carbimazole has a half-life of around 5 hours and propylthiouracil’s half-life is
even shorter––their effective duration of action is much longer. It can take several
weeks of treatment with carbimazole for thyroid hormone levels to fall to normal
(become euthyroid) because the half-life of T4 is so long, and there are usually
substantial T4 stores in the thyroid follicles that take some time to be used up.
Adverse Effects
The most significant adverse effect of these agents is bone marrow suppression, more
commonly with propylthiouracil than carbimazole. Although this is rare, it can cause a
rapid fall in white blood cell numbers with significant immunocompromise. Patients
must be warned to report any signs of infection, e.g. fever or sore throat. Bone marrow
function usually returns if the drug is stopped.
Radioactive Iodine
The radioactive isotope of iodine, 131I, given orally, is treated by the thyroid in exactly
the same way as the stable form: that is, it is rapidly and actively extracted by iodide
pumps in cell membranes of the thyroid cells, and built into thyroglobulin to form
mono-iodotyrosine and di-iodotyrosine. It emits radiation in the form of β particles,
which have a very short range, so that although over time they kill the thyroid gland
cells, they do not damage other local tissues. Because this agent permanently destroys
the glandular function of the thyroid, individuals invariably become hypothyroid and
need T4 replacement therapy.
Insulin
Insulin is an anabolic hormone; its overarching function in metabolism is in
conservation and storage of energy-rich nutrients. It stimulates body cells to remove
glucose, fats, and amino acids from the bloodstream to use in energy production;
promotes the storage of excess fuel molecules as glycogen or fat; and increases protein
synthesis. It is a small-molecular-weight peptide composed of 52 amino acids,
synthesised in β-cells within pancreatic islets. The main stimulus for its release is rising
blood glucose, but an increase in blood levels of fatty acids and/or amino acids has the
same effect. Insulin therefore reduces circulating blood glucose––a hypoglycaemic
effect––and levels of other nutrients. Liver, skeletal muscle, and fat are the most
insulin-responsive tissues.
Glucagon
Glucagon is synthesised by ⍺-cells in the pancreatic islets, and the main stimulus for its
release is falling blood glucose levels. Falling blood fatty-acid levels also increase
glucagon secretion. It stimulates glycogen breakdown in the liver, releasing glucose, and
breakdown of adipose and muscle tissue, releasing fatty acids and amino acids,
respectively. Its metabolic actions therefore oppose those of insulin, and it is given
subcutaneously, intravenously, or intramuscularly to increase blood glucose levels in
insulin-induced hypoglycaemia. Glucagon also has an adrenaline-like effect on the heart
and is used to improve cardiac function in shock caused by β-blocker overdose. It can
cause vomiting, so it is important to protect the airway in unconscious patients treated
with glucagon.
Diabetes Mellitus
Diabetes mellitus (DM) is a very common condition characterised by high blood glucose
levels, as well as an inability of body cells to respond appropriately to insulin (insulin
resistance) and/or an absolute insulin deficiency. Without insulin, cells cannot import
glucose no matter how much is available in the blood and are forced to rely on other
energy molecules such as fats and amino acids. This is tolerable in the short term but
unsustainable in the long term. The discovery that the pancreas is the source of the
agent that prevents diabetes was made in 1890 when the pancreas was removed from a
dog, which rapidly developed diabetes and died. It was over 30 years following that
important discovery before the first clinical use of insulin. Prior to this, type 1 diabetes
(T1D), usually presenting in childhood, invariably led to a slow, miserable, wasting
death within a year or two of diagnosis. In 1922, a team of researchers in Toronto, led by
Frederick Banting, Charles Best, and John MacLeod, treated a ward of seriously ill
diabetic children with pancreatic extracts, with dramatic and immediate success:
comatose children were restored to full consciousness, and children facing imminent
and inevitable death were, to all intents and purposes, cured. Fig. 5.6 shows the
dramatic difference in a diabetic child before and after starting treatment with insulin.
Although life expectancy in DM is shorter than average, continuing improvements in
management, including better pharmacological treatment, is slowly closing the gap.
Insulin release is also stimulated by hormones released by the GI tract during eating.
This explains the perhaps unexpected observation that consuming glucose by mouth
raises blood insulin levels faster than an intravenous glucose infusion. GI hormones that
increase insulin release are collectively called incretins and include glucagon-like
peptide 1 (GLP-1). Their blood levels rise when foodstuffs enter the GI tract and
stimulate insulin release in anticipation of rising blood sugar levels as the products of
digestion are absorbed (Fig. 5.9). Some antidiabetic drugs, e.g. liraglutide, stimulate
insulin release by mimicking the action of incretins, i.e. act as incretin agonists.
Following their release, incretins are rapidly destroyed by the enzyme dipeptidyl
peptidase (DPP-4). The gliptins, including linagliptin and vildagliptin, inhibit DPP-
4, protecting incretins from destruction and therefore increasing insulin release.
Fig. 5.10 summarises the main factors controlling blood insulin levels.
Type 2 Diabetes
This is associated with adult-age onset and obesity, and although the individual usually
continues to produce some insulin of their own (so-called relative insulin deficiency),
the cellular response to insulin is abnormal. Overeating and obesity are associated with
sustained elevated blood insulin levels, because constantly high blood nutrient levels
stimulate insulin release (Fig. 5.10). Over time, it is thought that the continual
exposure of insulin receptors on body cells to elevated insulin levels desensitises them
and disrupts the normal biochemistry of the cellular response to insulin. This is called
insulin resistance. Although type 2 diabetes (T2D) is treated with a range of drugs to
increase pancreatic release of insulin or to improve the cellular response to insulin,
many people eventually require supplementary insulin treatment too. Because these
patients usually produce at least some insulin, hypoglycaemic episodes are much less
likely than in T1D, and the main management goal is to minimise postprandial
hyperglycaemia to minimise long-term cardiovascular risk.
Insulin
Insulin cannot be given orally because it is a small peptide and is rapidly digested in the
stomach. It is generally given subcutaneously in maintenance treatment but can be
given intravenously or intramuscularly if a faster effect is needed. Its plasma half-life is
short––about 10 minutes––and it is metabolised in the liver to inactive products which
are excreted in the urine. The molecular action of insulin is described above, but there is
now a range of modified insulins which vary in their onset and duration of action, and
which can be combined in individualised and flexible treatment regimes. Insulin
molecules tend to assemble themselves into stable hexamers (six insulin molecules
grouped around zinc ions); they are found in this formation in pancreatic islets and
assemble into hexamers in the tissues following injection. The onset of action of injected
insulin then depends upon how quickly individual insulin molecules are released from
the hexamer structure. Insulin analogues are chemically modified to alter the speed at
which insulin dissociates from the hexamers; rapidly acting insulins dissociate the
fastest and long-acting insulins dissociate much more slowly. Pre-mixed insulins are
available, for example combining an intermediate-acting insulin with a short-acting
insulin, which reduce the number of daily injections required.
Short-Acting Insulins
Examples: soluble insulin, insulin aspart, insulin lispro
Short-acting insulins include soluble insulin, which can be used by regular
injections or by infusion. Insulin is absorbed slowly from its injection site due to the
assemblage of insulin hexamers in the tissue. Insulin aspart and insulin lispro are
modified insulins, in which very minor alterations to the structure of the insulin
molecule increase its release from the hexamers without reducing its pharmacological
activity. They therefore act more quickly than soluble insulin but have a shorter duration
of action.
Intermediate Insulins
Examples: isophane insulin
Isophane insulin is produced by treating soluble insulin with zinc and protamine. This
reduces its solubility at physiological pH and slows its release into the bloodstream,
avoiding a peak, and prolongs its action; it can be effective for up to 24 hours. Biphasic
insulin is produced by combining soluble insulin with isophane insulin, giving a
pre-mixed preparation that provides both short- and medium-term blood glucose
control.
Long-Acting Insulins
Examples: insulin glargine, insulin detemir, insulin degludec
Long-acting insulins are insulin analogues with very minor alterations to the amino-
acid structure. These structural changes reduce solubility, slow absorption from the
injection site, and extend their duration of action: this avoids a peak. Insulin glargine
is soluble at pH 4, and when injected into the tissues, which are pH-neutral, it loses
solubility and precipitates out as microcrystals from which it is slowly released into the
bloodstream. It takes over an hour before onset of action, but it is active for 24 hours,
providing the equivalent of naturally secreted basal insulin. Insulin detemir has fatty-
acid residues bound to the peptide structure, which promote hexamer formation and
slow its absorption, and increase its binding to plasma proteins, both of which extend its
duration of action. Insulin degludec also has a fatty-acid residue attached, which
causes it to precipitate out in the tissues after injection and slows its release into the
bloodstream.
Adverse Effects
The most frequent and most dangerous side-effect of insulin treatment, especially
hazardous at night, is hypoglycaemia, which may be caused by insulin overdose,
unexpected exercise, or inadequate food intake. Because the brain is highly dependent
on glucose for fuel, hypoglycaemia causes confusion, impaired cognitive function,
reduced conscious levels, and coma, and is potentially lethal. Falling blood glucose
activates a sympathetic nervous system response and adrenaline release, which usually
acts as an early warning of an impending hypoglycaemic episode, although not all
people experience this and hypoglycaemia can develop without warning. Regular
injections at the same site can cause a local accumulation of subcutaneous fat because of
the anabolic action of insulin. Occasionally, adipose tissue can break down (lipoatrophy)
around a site that is overused for injection, thought to be due to an immunologically
mediated tissue damage. Using a range of injection sites (usually the abdomen and
thigh) and not using the same site repeatedly helps to prevent these permanent tissue
changes. Although initial management of T2D does not include insulin because most
people with this form of diabetes produce some insulin of their own, about 50% of
people eventually require insulin treatment. Insulin in T2D causes weight gain and
increases cardiovascular risk, and its benefits should be weighed against this when
deciding whether to add insulin to a treatment regimen.
Inhaled Insulin
The discomfort and inconvenience of multiple daily injections drove the search for
alternative routes of insulin administration, and most work has been done in inhaled
insulin. The alveoli of the lungs are very thin-walled and richly supplied with blood
vessels and offer a very large surface area for drug absorption. Inhalable insulin is
formulated as a powder to be delivered as an aerosol via an inhaler device. It was first
licensed in the USA in 2006 by Pfizer, but it did not sell well mainly because of its cost.
Although it acts more rapidly than even the fastest-acting injected insulin, which could
in theory increase a patient’s ability to control post-prandial hyperglycaemia, it is
difficult to achieve precise dosing because pulmonary absorption may be unpredictable,
and repeated dosing may be needed to control post-prandial hyperglycaemia. Inhaled
insulin is not currently available on the National Health Service in the UK.
Metformin
Chemically, metformin is a biguanide, derived from a chemical found in French lilac,
extracts of which were used in mediaeval Europe to treat diabetes. A range of biguanides
was developed in the 1920s, but metformin is the only survivor in clinical use, the others
having been discarded because they caused lactic acidosis. Metformin is frequently the
first choice of treatment in newly diagnosed T2D, and globally is the most common oral
antidiabetic drug. It has a range of actions that contribute to its hypoglycaemic action
(Fig. 5.11). It is an insulin sensitiser: that is, it enhances the effects of insulin and so it
is ineffective unless there is some circulating insulin present. It reduces glucose
absorption from the GI tract, inhibits glucose re-absorption in the kidneys, reduces
glucose production in the liver, and increases the uptake of glucose in body tissues,
including skeletal muscle. It enters body cells and accumulates in mitochondria, the
organelles responsible for energy metabolism, where it interferes with energy pathways,
leading to the effects described above. Unlike some other antidiabetic drugs, metformin
does not cause weight gain, so is particularly useful in overweight and obese patients,
and does not cause hypoglycaemia. There is also evidence that metformin reduces total
plasma cholesterol and low-density lipoprotein (LDL) cholesterol, both risk factors for
cardiovascular disease. Such an improvement in blood lipid profiles is likely to be
significantly beneficial in diabetes because of the increased risk of cardiovascular
disease. In many patients, metformin used as monotherapy gives good glycaemic
control, especially in milder disease. It can also be usefully combined with other
antidiabetic agents if required; with time and progressive deterioration of islet function,
second-line agents frequently need to be added to maintain good blood glucose control.
Pharmacokinetics
The half-life of metformin is about 5 hours, and there are modified-release preparations
that extend this up to 8 hours. It is not metabolised in the liver, and is excreted largely
unchanged in the urine, so renal function must be assessed regularly to ensure that the
drug is not accumulating. In severely reduced renal function, it should be avoided
altogether.
Adverse Effects
Metformin reduces the uptake of the waste product lactate from the bloodstream by
liver cells. This can significantly increase blood lactate levels and precipitate lactic
acidosis, most commonly in older adults and in those with poor renal function. Poor
cardiac function also increases risk because it leads to hypoxia, and without adequate
oxygen, cells respire anaerobically, producing excess lactate. Liver disease can also
increase blood lactate and the drug should be avoided in severe liver impairment. Lactic
acidosis is fortunately a rare side-effect, but it can be fatal. Most other side-effects of
metformin are relatively mild and usually improve with time: they include a metallic
taste in the mouth, nausea, and diarrhoea, which may be reduced by taking with food.
Sulphonylureas
Examples: glibenclamide, gliclazide, glipizide
The sulphonylureas are chemically related to the sulphonamides, a group of
antimicrobial agents introduced in 1935 which caused hypoglycaemia as an unexpected
side-effect: this led to the development and introduction of tolbutamide, the first
sulphonylurea, in the 1950s. Tolbutamide remains in current use, although newer
agents like glibenclamide are more potent. Sulphonylureas increase insulin release by
the pancreas by blocking potassium channels in islet β-cells (Fig. 5.7). They are
therefore only effective if there are functioning β-cells in the pancreatic islets, and they
enhance both phases of insulin release: that is, the initial spike of pre-formed insulin
and release of newly synthesised insulin. Although they have traditionally been
considered the first choice when supplementation of metformin treatment is needed and
are generally well tolerated, DPP-4 inhibitors are now considered a superior option
because they have fewer side-effects.
Pharmacokinetics
Sulphonylureas are taken orally and absorbed well, although food in the GI tract slows
absorption. They bind extensively to plasma proteins and compete with other highly
bound drugs (e.g. warfarin, aspirin) for binding sites, and this is an important cause
of sulphonylurea-mediated interactions. Dose adjustment may therefore be necessary
when such drug combinations are used. Different drugs in this class have different half-
lives and duration of action, and increasing either increases the likelihood of
hypoglycaemia. For example, glibenclamide has a long half-life of over 10 hours and
produces an active metabolite, so it is particularly long-acting (sometimes more than 24
hours) and is prone to causing hypoglycaemia, especially at night and in older people.
Glimepiride has a shorter half-life (5–8 hours), but because it too produces an active
metabolite, its duration of action is 12–24 hours. Glipizide has a half-life of only 2–4
hours, produces no active metabolites, and is one of the shorter-acting agents in this
group. Although tolbutamide is the oldest and least potent drug in this group, it has a
short half-life (4 hours), meaning that there is little or no risk of hypoglycaemia. Most
sulphonylureas are excreted by the kidney, so they must be used with caution if there is
any degree of renal compromise.
When sulphonylureas are used with a number of other drugs, including alcohol,
some antifungals, some antibiotics, and non-steroidal anti-inflammatory
drugs, severe hypoglycaemia can occur. The basis of this interaction is probably caused
by inhibition of metabolising enzymes, meaning that metabolism and clearance of the
sulphonylurea is reduced and so its hypoglycaemic effect is enhanced.
Adverse Effects
Most sulphonylureas cross the placenta and appear in breast milk and should not be
used in pregnancy or by breast-feeding mothers. They usually cause weight gain, so they
must be used cautiously in overweight or obese people. Hypoglycaemia is a common
side-effect, especially with longer-acting agents. Long-term use may increase the risk of
cardiovascular disease, particularly concerning in DM. Less severe, although still
troublesome, side-effects include allergic rashes and GI upset.
Meglitinides
Examples: nateglinide, repaglinide
This group of drugs is chemically closely related to the sulphonylureas and work in
the same way, i.e. they block potassium channels in the β-cell membrane and increase
insulin release from the pancreas. They are faster acting and have a shorter duration of
action than the sulphonylureas, with a half-life of less than 2 hours, so they are less
likely to cause hypoglycaemia. Because they are short-acting, they can be used to reduce
post-prandial hyperglycaemia when metformin or the sulphonylureas are not
adequate. Other than hypoglycaemia, their main side-effects include diarrhoea and
abdominal discomfort.
Glitazones
Examples: pioglitazone, rosiglitazone
These are also called thiazolidinediones and are classed as insulin sensitisers because
they increase cells’ ability to respond to insulin. They reduce blood glucose levels by
increasing its uptake into body cells, particularly skeletal muscle and fat. Pioglitazone
is the only member of this group currently used in the UK, other glitazones having been
discontinued due to significant side-effects.
Pharmacokinetics
The onset of action is slow and maximal hypoglycaemic effects may not be seen for up to
8 weeks. Pioglitazone is over 99% bound to plasma proteins and is extensively
metabolised in the liver, producing active metabolites. Its plasma half-life is between 5
and 7 hours, but the active metabolites can significantly extend its duration of action. It
is mainly excreted in the faeces (less than 30% is excreted in the urine). It has a
beneficial effect on blood lipids, increasing levels of ‘good’ high-density lipoprotein
(HDL) cholesterol and reducing levels of ‘bad’ very-low-density lipoprotein. It also
reduces other cardiovascular risk factors and inflammatory markers, which in turn
reduces cardiovascular disease in T2D.
Adverse Effects
The side-effects of pioglitazone can be significant. Common adverse effects include
weight gain, caused partly by fat deposition, but it is also likely that fluid retention,
another of its side-effects, contributes. Fluid retention causes oedema and increases
circulating blood volume. Because of this, pioglitazone can precipitate heart failure,
particularly in older patients and people with pre-existing cardiovascular disease, and
when used in conjunction with insulin. There is also evidence that pioglitazone slightly
increases the risk of bladder cancer, especially in high doses and long-term treatment,
and it should be avoided in patients with risk factors for this tumour.
Pharmacokinetics
The gliptins have a long duration of action because they bind strongly to DPP-4,
inhibiting it over a prolonged period; they are generally given only once daily. For
example, saxagliptin has a plasma half-life of only 2.5 hours, and its active metabolite
has a half-life of about 3 hours, but its hypoglycaemic action lasts for 24 hours.
Side-Effects
The gliptins can cause headaches, nausea, and abdominal pain. There may be an
increased risk of infections, e.g. urinary tract and upper respiratory tract infection, but
the evidence for this is mixed.
Pharmacokinetics
GLP-1 agonists are given by subcutaneous injection, not by mouth, because they are
proteins and are digested in the stomach. The drugs in this group vary significantly in
their duration of action; dulaglutide has a long half-life of approximately 5 days and is
given only once a week, whereas exenatide and liraglutide are given daily.
Adverse Effects
These drugs tend not to cause weight gain because they do not stimulate appetite and
can even help patients lose a little weight. Rarely, they can cause acute pancreatitis, and
patients must be warned to report any relevant signs and symptoms. Commonly, they
cause GI side-effects including abdominal pain, nausea/vomiting, and constipation or
diarrhoea.
Pharmacokinetics
Because they have long half-lives, drugs in this group are given once daily.
Adverse Effects
Glycosuria increases the risk of urinary tract and genital infections, and because the
osmotic pressure of the urine is increased, it draws water with it, causing thirst and
dehydration. There is evidence that some sodium/glucose co-transporter 2 inhibitors
(canafliglozin, dapagliflozin, and empagliflozin) can sometimes cause serious
ketoacidosis, and patients taking these agents should be taught to recognise warning
signs, e.g. deep and rapid respiration, sudden weight loss, or a sweet odour to the
breath. This class of drug has also very recently (2017) been associated with an increased
risk of lower-limb amputation, particularly of the toes. The reason for this is not yet
known, but one study has suggested that they decrease blood volume, thickening the
blood and increasing the risk of blood clots. This would reduce tissue perfusion,
especially in the periphery, already likely compromised in T2D.
Acarbose
Acarbose inhibits the enzyme glucosidase, which in the intestine breaks down larger
carbohydrate molecules into glucose for absorption. This delays (but does not prevent)
carbohydrate digestion following eating, so glucose levels in the intestinal brush border
rise more slowly than normal, glucose absorption into the bloodstream is slowed, and
the rapid post-prandial rise in blood sugar levels is flattened. Hyperglycaemic peaks are
therefore reduced or avoided. It is generally used in conjunction with other antidiabetic
agents because it is less effective at controlling blood glucose than other oral antidiabetic
drugs and is usually insufficient on its own. Very little of an oral dose is absorbed into
the bloodstream; most is excreted in the faeces, and the main side-effects include
diarrhoea, abdominal bloating and discomfort, and flatulence. Its blood glucose-
lowering action is not usually profound enough to significantly increase the risk of
hypoglycaemia, but it does increase the overall risk when used with other hypoglycaemic
agents.
Pharmacokinetics
Oestrogens are well absorbed from a range of sites, extensively metabolised in the
intestinal lining and the liver, and excreted in the urine. Oestradiol metabolism
produces several active metabolites, which extend its duration of action and half-life
to up to 16 hours.
Oestrogen Receptors
ERs are found in the cytoplasm of oestrogen-responsive cells, including breast,
vagina, and uterus, where they mediate the reproductive functions of oestrogens.
However, ERs are found in a range of other tissues, including the bone, skin, heart
and blood vessels, and brain. There are two types of ER: ER⍺ and ERβ. Distribution of
these receptor types differs between tissues, e.g. ER⍺ is the main receptor type in the
breast and is often overexpressed in breast cancer, whereas ERβ is the main type
found in the brain.
Progesterone
Progesterone regulates the female reproductive cycle as described below and is
essential in the maintenance of pregnancy, including breast growth and development
in preparation for lactation. Synthetic progestogens, e.g. norethisterone and
gestodene, are more potent than natural hormones. Some progestogens are
androgenic (i.e. have masculinising action) because they are chemically very similar to
testosterone, bind to ARs, and can cause testosterone-related side-effects like acne
and hirsutism. Newer synthetic progestogens have less androgenic activity because
they do not bind to ARs. There is evidence that synthetic progestogens slightly
increase the risk of breast cancer, especially when combined with an oestrogen. The
reason for this is not clear, because while some studies show that progesterone
stimulates cell division in breast epithelial cells, other studies show inhibition. It is
likely to be due to the interaction of several factors, including oestrogen levels,
duration of exposure, and the woman’s age.
FIG. 5.15 Simplified diagram showing the biosynthesis of the
main sex hormones, including key enzymes. Modified from
Aggarwal NR and Wood MJ (2021) Sex differences in cardiac
diseases, Fig. 2. St. Louis: Elsevier.
Pharmacokinetics
Progesterone is well absorbed from a range of sites, including the GI tract, but when
given orally it distributes rapidly from the bloodstream and is extensively
metabolised, with a plasma half-life of only 5 minutes. Peak plasma levels are reached
in about 2 hours post-dose, but then fall rapidly, which can compromise contraceptive
cover (see below). Progesterone metabolites are mainly excreted by the kidney.
Testosterone
Testosterone is the main androgenic hormone in humans, but it is converted in most
tissues to the more active agent dihydrotestosterone. At puberty in the males, it
promotes the development of secondary sexual characteristics, e.g. the growth of
axillary and pubic hair and deepening of the voice, and maturation of the male sexual
organs. It activates genes that produce muscle proteins and increases the bulk and
strength of skeletal muscle. It increases libido and physical vigour and promotes
sperm production in the testis. At puberty, rising levels of testosterone convert the
cartilage growth plates at the ends of long bones to bone tissue, terminating any
further increase in height. Testosterone and certain synthetic variants, e.g.
nandrolone and trenbolone acetate, are sometimes used to increase muscle mass
and power and athletic performance in professional and amateur sport or for
occupational or cosmetic reasons. These agents are called anabolic steroids and can
cause significant long-term harm, including infertility, acne, masculinisation in
females, and increased risk of prostate cancer.
Pharmacokinetics
Testosterone is well absorbed from a range of sites, and its plasma half-life is very
short (10–20 minutes) because it is rapidly metabolised by the liver. Synthetic
analogues usually have longer half-lives.
Pharmacokinetics
The general pharmacokinetic profiles of the individual hormones are described above.
The natural hormone, progesterone, is not used because it is rapidly and extensively
metabolised by enzymes in the intestinal lining and in the liver, so its plasma levels are
unpredictable and poorly sustained. Synthetic progestogens, first developed in the
1950s, are used instead: examples include norethisterone, desogestrel, and
gestodene. An important drug interaction involves increased hormone metabolism by
several enzyme inducers. Oestrogen and some progestogens are metabolised in the liver
by the cytochrome P450 enzyme family, and drugs that induce cytochrome P450,
including some anticonvulsants (e.g. carbamazepine, phenytoin), some
antimicrobials (e.g. rifampicin, antiretrovirals, griseofulvin), and St. John’s
wort enhance hormone clearance and can reduce contraceptive effectiveness. Women
should be advised to use an additional method of contraception while taking the
interacting drug and for 4 weeks after stopping it.
Adverse Effects
For most women, adverse effects are relatively minor and acceptable when weighed
against the prospect of an unwanted pregnancy: commonly they include nausea, mood
swings, fluid retention, and a tendency to weight gain. Women prone to migraine may
find that this gets worse, and they should be instructed to report any changes in
headache frequency or intensity.
Venous Thromboembolism
Both oestrogen and progesterone increase the risk of venous thromboembolism, e.g.
deep venous thrombosis and pulmonary embolism. Newer (third-generation)
progestogens such as gestodene and desogestrel are more potent and less
androgenic than older progestogens such as norethisterone and levonorgestrel but
have a higher risk of thromboembolism. The risk is highest in the first year of treatment
and in women with pre-existing risk factors, e.g. smoking, overweight, and obesity, but
are not as high as the increased risk associated with pregnancy. Even though
progestogens are pro-thrombotic when used in combination with oestrogen, studies
have shown that the progestogen-only oral contraceptive does not carry an increased
risk of thromboembolism (although there is evidence that there may be a causative
association between injectable progestogens and thrombosis). The reason for this is not
clear.
Myocardial Infarction and Stroke
The increased incidence of venous thromboembolism is probably responsible for the
slightly increased risk of angina, myocardial infarctions, and stroke seen with the COC.
Women with additional cardiovascular risk factors, e.g. increased age, overweight or
obesity, smokers, high blood pressure, diabetes, and a personal or family history are
particularly at risk and should be carefully assessed before a decision to prescribe COC is
made. It may be that a progestogen-only preparation is a better choice in these patients
because it does not increase cardiovascular risk.
Neoplasia
The risks of certain cancers (e.g. breast and cervical) may be slightly increased but the
risks of endometrial and ovarian cancers are slightly reduced in COC users. Changes in
cancer risk are oestrogen-dependent and are not seen with progestogen-only
contraceptives.
Progestogen-Only Preparations
Because oral progestogens are so rapidly metabolised and cleared from the blood, it is
critical that doses are taken strictly at 24-hour intervals; if a tablet is late or missed,
levels become so low that contraceptive cover is compromised. Oral progestogen
contraceptives must be taken daily, with no pill-free break as with the COC. These
agents may be suitable for women for whom oestrogen-containing preparations are not
recommended, e.g. smokers, older women, women with hypertension, or women with
cardiovascular risk factors. They are also useful in short-term cover for times when
oestrogen might be contra-indicated, e.g. in breast-feeding, periods of immobility, and
before and after surgery, when the risk of thrombosis is elevated. Side-effects include
breakthrough bleeding and headaches. Intra-uterine devices impregnated with a
progestogen and long-acting progestogen depot injections are also available. Interaction
with enzyme-inducing drugs, as described above for the COC, can also reduce
contraceptive effectiveness of oral progestogen-only agents and progestogen-only
implants, and a barrier method of contraception should additionally be used.
Medroxyprogesterone acetate, a progestogen used as a depot injection for
contraception, is metabolised by a different enzyme family and so is not subject to this
form of interaction; no additional contraceptive precautions are needed when used with
an enzyme inducer.
Clomifene
Clomifene binds to and blocks oestrogen receptors in the anterior pituitary and
hypothalamus (Fig. 5.18). Unable to detect the presence of oestrogen in the blood, the
hypothalamus and pituitary hugely increase GnRH, FSH, and LH output. This massively
stimulates the ovaries, initiating follicular development, oestrogen secretion, and
ovulation. The likelihood of multiple pregnancies is increased with clomifene treatment.
It is not recommended for more than six cycles because its stimulatory action on the
ovary has been associated with increased risk of ovarian cancer.
FIG. 5.18 The mechanism of action of clomifene.A. The ovary
secretes oestrogen in response to follicle-stimulating hormone (FSH)
and luteinising hormone (LH) from the anterior pituitary. The
hypothalamus and anterior pituitary have oestrogen receptors to
allow them to continually measure blood oestrogen levels and adjust
their own hormone output accordingly. B. In the presence of
clomifene, oestrogen receptors are blocked and the anterior pituitary
and hypothalamus can no longer detect oestrogen in the blood. They
therefore increase gonadotropin-releasing hormone (GnRH), FSH,
and LH output, causing massive ovarian stimulation.
Anti-Androgens
Examples: cyproterone acetate, flutamide, bicalutamide
Anti-androgens, drugs that block testosterone receptors, are used in testosterone-
related disorders. These include prostatic hyperplasia and prostate cancer, early
(precocious) puberty in young boys, and hair loss. They are used in the management of
hypersexuality and compulsive sexual behaviours, which can be highly disruptive to
normal life, cause significant distress and anxiety in the affected male, and contribute to
unacceptable, risky, or deviant behaviours. By directly reducing testosterone action, they
reduce libido and diminish sexual activity. They are also used in the treatment of a range
of cancers in both men and women, although legislation varies in different countries and
much work in this area is in its early stages. Some anti-androgenic agents have shown
efficacy in some triple-negative breast cancers––i.e. breast cancers that do not express
oestrogen, progesterone, or HER2 receptors and for which anti-oestrogen, anti-
progesterone, or anti-HER2 agents are of no use. Triple-negative disease can be difficult
to treat, but a subset of these cancers express androgen receptors, and initial studies
suggest that they may respond to anti-androgens. Likewise, a proportion of ovarian
cancers express ARs, and early clinical trials have confirmed that enzalutamide may
be an effective treatment in some of these patients.
References
1. Deacon C.F, Lebovitz H.E. Comparative review of dipeptidyl peptidase-4
inhibitors and sulphonylureas. Diabetes Obes. Metab. 2015;18(4):333–347.
2. Grisham R, Giri D, Henson M, et al. 38 Phase II study of enzalutamide in
androgen receptor positive (AR+) recurrent ovarian cancer: final results. Int. J.
Gynecol. Cancer. 2019;29:A23.
3. Newson L.R. Best practice for HRT: unpicking the evidence. Br. J. Gen.
Pract. 2016;66(653):597–598.
4. Sola D, Rossi L, Schianca G.P.C, et al. Sulfonylureas and their use in clinical
practice. Arch. Med. Sci. 2015;4:840–848.
Online Resources
Cagnacci, A., Venier, M., 2019. The controversial history of hormone replacement
therapy. Medicina (Kaunas) 55 (9), 602. Available at:
https://doi.org/10.3390/medicina55090602
Collaborative Group on Hormonal Factors in Breast Cancer, . Type and timing of
menopausal hormone therapy and breast cancer risk: individual participant
meta-analysis of the worldwide epidemiological
evidence. Lancet. 2019;394(10204):1159–1168 Available
at:. https://doi.org/10.1016/S0140-6736(19)31709-X.
White, J.R., 2014. A brief history of the development of diabetes medications.
Diabetes Spectr 27 (2), 82–86. Available at:
https://doi.org/10.2337/diaspect.27.2.82
6: Analgesics and Anti-Inflammatory
Drugs
Key Definitions
• Nociceptive pain:
• Hyperalgesia:
• Allodynia:
a pain response to a stimulus that should not normally be painful, e.g. light touch
The axons of the first-order neurones enter the dorsal horn of the spinal cord and
synapse with the second sensory nerve of the pain pathway, called the second-order
neurone. The neurotransmitter here is usually substance P or glutamate. Second-order
neurones cross to the opposite side of the spinal cord and from the dorsal to the ventral
horn and carry pain signals to the thalamus in one of two main pathways (tracts): the
spinothalamic tract or the spinoreticular tract. In the midbrain, neurones from the
spinothalamic tract project to the periaqueductal grey matter (PAG), a region important
in descending inhibition. In the thalamus, the second-order neurones synapse with
third-order neurones, which project to a range of brain centres involved in pain
perception and processing. Some third-order neurones project to the limbic system,
which determines the emotional response to pain. Others project to the somatosensory
cortex, which localises the pain and interprets its intensity and characteristics. Other
centres in the cerebral cortex evaluate the pain in terms of previous experience and
expectations, which in turn help to determine the response to pain.
• Opium:
the juice extracted from the seed head of the opium poppy Papaver somniferum (in
Greek: ‘poppy that brings sleep’)
• Opiates:
• Opioid:
Cave paintings, clay tablets, and other artefacts from a range of ancient civilisations
indicate that humankind has used opium for at least 5000 years, both for recreational
and medicinal purposes. Opium contains several pharmacologically active agents
including morphine, thebaine, and papaverine. Thebaine is too toxic to be given in
its natural form but is used to synthesise certain opioid drugs including oxycodone,
buprenorphine, and naloxone. Opioids are used in modern medicine for a range of
purposes, but most significantly as analgesics: they are potent pain relievers, so effective
in fact that despite their potentially lethal side-effects, they are still the first-line agents
in the management of moderate to severe pain.
The active opiate content of crude opium is highly variable depending on the
conditions under which the poppy was grown, leading to significant variation in the
potency and activity of different opium batches. At the beginning of the 19th century,
opium was widely available and sold over the counter to the general public in a range of
formulations including lozenges, tablets, and tinctures (a drug dissolved in alcohol).
Other than the obvious dangers of such a dangerous preparation being freely available
with no restriction or regulation, the potency of these formulations could not be
controlled or predicted. In 1803, morphine was purified from opium by a German
apothecary called Friedrich Sertürner. This meant that dosages could be accurately
quantified, which in theory should improve safety. Commercial morphine production
began in earnest in the 1820s, and when the hypodermic needle was invented in the
1850s, the popularity of purified morphine skyrocketed. Although it provided previously
unsurpassed benefits for physicians managing their patients’ pain, it also led to
widespread addiction in all strata of society with associated criminality and major family
and societal problems. In response to the growing awareness of such negative opioid-
related repercussions, in 1912, several countries including the US, Russia, and the UK
signed the International Opium Convention. It was the first attempt to control the
distribution, manufacture, and sale of opioids, and in modern times most global
countries have regulatory legislation around their use.
Opioid Receptors
There are three main types of opioid receptors in the CNS and PNS: μ (mu), δ (delta),
and ϰ (kappa), called MOR, DOR, and KOR, respectively. The brain, peripheral nerves,
and some other body tissues produce a family of endogenous opioids, the endorphins
and enkephalins, which act on opioid receptors and provide the body’s internal pain
management system. When the endogenous opioids were discovered in the 1970s, it was
realised that the opium preparations that mankind had been using as analgesics for
thousands of years worked by mimicking the action of the endogenous opioids and
activating internal analgesia mechanisms. In terms of pain relief and most other
clinically observed opioid effects, MOR is the most important opioid receptor type, and
the endogenous opioids and clinically useful opioid analgesics are all agonists here.
However, the KOR and DOR also play a role in analgesia, and the overall effect of an
opioid, including its adverse effects, are produced by the drug interacting with all three
receptor types.
MOR Distribution
MORs are found in high concentrations in pain pathways in the CNS, including in the
dorsal horn of the spinal cord, as well as brain regions involved in emotion, mood,
feeding, hormonal control, respiration, and immune function. They are also found on
peripheral sensory nerves, where they inhibit pain signals. Opioid action here
contributes to their analgesic effect. Ongoing research is directed towards developing
opioids that do not cross the blood–brain barrier and so would be free of central adverse
effects like dependence and respiratory depression, but which could provide effective
analgesia via this peripheral activity. MORs are also present on nerves in the
gastrointestinal system, where they reduce motility and secretion. Additionally, MORs
are found in non-neural tissue, including joint tissues, immune cells, and endocrine
cells.
MOR Signalling
All opioid receptors are linked to inhibitory G-proteins (Gi) and inhibit adenylyl cyclase
(see p. 32 and Fig. 3.7). When an opioid agonist binds to its receptor on a nerve cell
membrane and activates this signalling pathway, potassium channels open, allowing
potassium to enter the cell. This desensitises (hyperpolarises) the nerve and makes it
less likely to fire. In addition, calcium channels are closed, preventing calcium from
entering; calcium entry into nerve cells is needed for activation. By interfering with the
movement of these two key ions across the nerve cell membrane, opioids prevent the
nerve from releasing its transmitter, dampen neuronal transmission, and block synaptic
communication (Fig. 6.3). This is reflected in the key effects of opioids including
blockade of pain signals, sedation, inhibition of smooth muscle contraction, and
respiratory depression.
Actions of Opioids
Opioids produce a range of physiological effects via their action on opioid receptors.
Analgesia
Opioids produce effective analgesia mainly through their action on MORs in both the
CNS and PNS, particularly in nociceptive pain; they are of limited use in neuropathic
pain. They reduce neuronal excitability and transmitter release in pain pathways,
including in the mechanism of descending inhibition (Fig. 6.4). Fig. 6.4 shows the
synapse in the dorsal horn between the first- and second-order pain neurones in more
detail. Incoming pain signals activate this synapse; the excitatory neurotransmitter here
is usually substance P or glutamate, which depolarises the second-order neurone and
transmits the pain signal up the spinal cord towards the brain. The descending nerve
from the PAG synapses here and releases endogenous opioid, which acts on MORs on
the first-order nerve ending. This inhibits release of its excitatory transmitter and blocks
transmission of the pain signal between the first- and second-order neurones. Opioid
drugs mimic the action of the endogenous opioid transmitter by binding directly to the
MORs.
FIG. 6.3 Morphine receptor signalling and inhibition of
neurotransmitter release. Modified from Minneman KP and Wecker
L (2005) Brody’s human pharmacology: molecular to clinical, 4th ed.
St. Louis: Mosby.
FIG. 6.4 Opioid inhibition of pain signal transmission between the
first- and second-order neurone in the spinal cord. MOR, Mu opioid
receptor; PAG, periaqueductal grey matter.
Sedation
Opioids sedate and may induce sleep by inhibiting central pathways responsible for
arousal and wakefulness. This may worsen their respiratory depressant effect but can be
clinically useful because it may relieve distress and anxiety. Alternatively, sedation may
be unwanted: for example, it may limit activities of daily living for people using opioids
to manage chronic pain.
Respiratory Depression
Neurones in the respiratory centre in the brainstem are normally very sensitive to even
slight increases in carbon dioxide levels and respond by increasing respiratory rate and
depth of breathing. Opioids depress this response, allow carbon dioxide levels to rise,
and produce hypoventilation, hypercarbia (high blood carbon dioxide levels), apnoea,
reduced respiratory rate, and potentially respiratory arrest. Opioid-induced respiratory
depression is more marked and therefore most dangerous when brain activity levels are
suppressed, e.g. in sleep, in anaesthesia, or in the presence of other central depressants
like alcohol or benzodiazepines. Habitual opioid use leads to a developing tolerance,
but respiratory arrest is still the most common cause of death in opioid overdose.
Miosis
Opioids act directly on the oculomotor nerve (cranial nerve III), which controls the
muscle of the iris of the eye and therefore pupil diameter. They constrict the circular
muscle in the iris and reduce the size of the pupils (miosis). In overdose, pupil size may
be pinpoint (Fig. 6.5).
Gastrointestinal Effects
The walls of the GI tract contain extensive networks of nerves (the enteric nervous
system) which regulate its function. Enteric nerves release a range of neurotransmitters,
including endogenous opioids, which act on MORs in GI smooth muscle to reduce
motility and secretions and increase sphincter tone. Administered opioids also act on
these MORs and therefore slow gastric emptying, which can significantly delay the
delivery of swallowed medication into the duodenum and delay absorption.
Constipation is a common and very troublesome adverse effect of opioids. However, due
to their ability to put GI smooth muscle into stasis, opioids have been used for
thousands of years to treat diarrhoea. Loperamide is a MOR agonist used by mouth
for this purpose. It is poorly absorbed, so its action is largely restricted to the GI tract,
and it does not cross the blood–brain barrier in high enough concentrations to produce
central effects. Other GI consequences of opioid use include bloating, abdominal
distension and discomfort, and sometimes gastro-oesophageal reflux. Tolerance tends
not to develop to GI side-effects, which persist during treatment, a particular challenge
when opioids are used to manage chronic pain. In addition to non-specific management
(e.g. high-fibre diets, good fluid intake, and laxatives), opioid-induced constipation can
be treated with methylnaltrexone bromide, an opioid receptor antagonist that does
not cross the blood–brain barrier; it therefore blocks peripheral receptors, including
those in the GI tract, without interfering with the central analgesic action.
Cough Suppression
Opioids suppress cough, i.e. they have an antitussive action, but the mechanism by
which they do this is not completely clear. They inhibit central neurones involved in
cough pathways, but there is probably also a peripheral component: for example, they
may decrease the sensitivity of sensory nerves in the airways and reduce their response
to irritants. For example, nebulised diamorphine is used to relieve persistent cough in
bronchial carcinoma. The antitussive action of opioids does not correlate with their
analgesic effect, so that even weak opioid analgesics like codeine effectively suppress
cough.
Immunosuppression
The interaction between opioids and immunity is complex and our understanding is
quite incomplete. There is substantial evidence that opioids, especially in long-term use,
are immunosuppressant, and may increase the risk of infection. Opioids act on MORs
on the cell membrane of a range of important immune cells, including T-cells, B-cells,
and macrophages, and suppress their activity. Immune cells themselves release opioids,
leading to some researchers speculating that activation of the body’s immune system
may also switch on an analgesic effect, which may offer another possible target in
designing new analgesic drugs. Opioids have no anti-inflammatory effect; in fact,
morphine triggers histamine release from mast cells, which can cause itch and
sufficient vasodilation to cause a drop in blood pressure.
Tolerance
Tolerance (and dependence) to opioids develops because continual exposure to the drug
changes the response of the receptors and reduces drug action. This means that the drug
dose must be increased to maintain its effect. Tolerance develops very slowly to GI
effects and to miosis, whereas it develops quickly to opioid-induced respiratory
depression, euphoria, emesis, and analgesia, especially if the drug is used intravenously.
This means that habitual opioid users may require 50 times the standard dose to achieve
a desired euphoric effect without obvious respiratory depression, but still experience
significant constipation and pupillary constriction. Several mechanisms by which
tolerance develops have been identified. One way involves the uncoupling of the
receptor from its signalling pathways, like cutting through the wire of a doorbell; the
drug binds to its receptor but cannot produce an effect inside the cell because the
receptor is no longer connected to its transduction pathways. Other tolerance
mechanisms include increased activation of excitatory nerve pathways to oppose the
inhibitory action of the opioids. There is a degree of cross-tolerance between clinically
used opioids, and switching between different agents when tolerance becomes a
problem with one can help to maintain their usefulness.
Uses of Opioids
Opioids are extensively used as analgesics in both acute and chronic pain, usually for
moderate to severe pain of nociceptive origin. Other uses include:
Morphine
Crude opium usually contains between 9% and 15% morphine, and global supplies of the
drug still rely on extracting it from the poppy seed head because all attempts to
synthesise it have failed.
Pharmacokinetics
Morphine is usually given orally, intravenously, rectally, or intrathecally. Absorption
from intramuscular sites can be slow and unpredictable, and so this is usually not the
route of choice. It has a half-life of 4–6 h and is available in modified release
preparations to extend its duration of action. Morphine is subject to significant first-
pass metabolism, so care is needed in patients with liver impairment in whom the drug
effect can be significantly enhanced. The metabolism of the main opioids, showing
morphine as the main agent, is shown in Fig. 6.7. Opioid metabolism produces several
metabolites, including codeine and active metabolites such as normorphine,
dihydrocodeine, and morphine-6-glucuronide, which contribute significantly to
morphine’s clinical effects.
Diamorphine (Heroin)
This semi-synthetic derivative of morphine was first synthesised in 1874. It is more
potent and more lipid-soluble than morphine and is not licensed for medical use in
many countries because of its significant abuse potential. Reflecting the lack of
legislation at the time around drug regulation and marketing, it was advertised initially
by the drug company Bayer under the trade name Heroin as a treatment for a range of
conditions including respiratory and cardiovascular disorders (Fig. 6.8) and was
available for public purchase. It was also recommended as a safer, non-addictive
alternative to morphine even though it is neither.
Codeine
Codeine is used to relieve mild to moderate pain, as a cough suppressant, and to treat
diarrhoea.
Pharmacokinetics
Codeine is given orally and is well absorbed from the GI tract. It is a pro-drug and relies
upon metabolism by the hepatic CYP2D6 enzyme to convert it to morphine for its
activity. Only around 10% undergoes conversion to morphine, the reason why it is a less
potent analgesic, although it is an effective cough suppressant and causes significant
constipation. It is generally well tolerated, although genetic variations in CYP2D6
activity affects its metabolism to morphine. In rapid and ultra-rapid metabolisers, rapid
conversion of codeine to morphine can worsen opioid adverse effects such as respiratory
depression, whereas in poor metabolisers, the therapeutic effects of the drug can be
delayed and reduced (Fig. 6.9; see also Pharmacogenomics, p. 3).
Fentanyl
Fentanyl is a synthetic opioid structurally similar to pethidine (Fig. 6.6), with potent
MOR agonist activity. It was first produced in 1960 by Janssen and is currently in
widespread use globally to manage severe pain in various clinical settings including
cancer and acute post-surgical pain. It is around 100 times more potent as an analgesic
than morphine. Although it produces the typical range of opioid-induced adverse effects
including respiratory depression and constipation, it is less likely to cause histamine
release, hypotension, and itch than morphine, which has contributed to its rising
popularity. Other drugs in the fentanyl group include sufentanil, alfentanil, and
remifentanil.
FIG. 6.8 Early 1900s Bayer advertisement for heroin. From Koob
GF, Arends MA, and le Moal M (2015) Drugs, addiction, and the
brain, Fig. 5.3. San Diego: Academic Press.
FIG. 6.9 The impact of metabolism on activation of the pro-drugs
codeine and tramadol.Poor metabolisers activate the drugs slowly,
and analgesia may be slow or incomplete. Fast metabolisers activate
the drugs quickly, which can cause toxicity. Modified from Magarbeh
L, Gorbovskaya I, le Foll B, et al. (2021) Reviewing pharmacogenetics
to advance precision medicine for opioids. Biomedicine &
Pharmacotherapy. 142: 112060.
Pharmacokinetics
Fentanyl has a rapid onset of action, acting within 5 minutes, although this depends on
the route of administration – it is faster with intravenous and intranasal administration
but slower with transdermal patches. It has a shorter duration of action (30–40
minutes) than morphine, although this is extended with transdermal or buccal
preparations because the drug is absorbed slowly. Fentanyl is metabolised in the liver by
cytochrome P450 enzymes, and so other agents that inhibit cytochrome P450, including
verapamil, some antifungal agents, and erythromycin, can reduce fentanyl
metabolism and increase plasma levels sufficiently to depress respiration.
Pethidine
Pethidine (also called meperidine) was discovered in 1939 by researchers screening a
range of compounds for antimuscarinic activity. By chance, it was noticed that it also
produced opioid-like activity in the test animals, and it was rapidly developed as an
analgesic. It acts on both MORs and KORs, although its molecular structure is different
from morphine (Fig. 6.6), and as a result, its pharmacological effects are not always the
same as the naturally derived opioids. It is 10 times less potent as an analgesic than
morphine and causes respiratory depression, constipation, and histamine release, but is
much less likely to cause nausea and vomiting and is less sedative. Because it has
antimuscarinic activity, it also causes typical antimuscarinic adverse effects including
dry mouth, blurred vision, and bradycardia.
Pharmacokinetics
Pethidine is only moderately well absorbed orally and is usually given parenterally, with
a half-life of 2–3 h. It undergoes significant first-pass metabolism in the liver, and care
is needed in patients with impaired liver function, in whom the half-life can be
significantly extended. One of its metabolites, norpethidine, has some analgesic
activity but can cause significant CNS excitation, including hallucinations and seizures,
if the drug accumulates (for instance, in overdose or in renal impairment).
Methadone
Methadone is a synthetic opioid with a long (24 h) half-life because the drug leaves the
bloodstream and accumulates in the tissues where it is slowly released. It binds to MORs
and produces a similar range of pharmacological activity to morphine, including
effective analgesia, euphoria, cough suppression, constipation, and respiratory
depression. Because of its sustained action, it is mainly used in opioid withdrawal
programmes and in maintenance treatment in opioid-dependent people.
Pharmacokinetics
Methadone is well absorbed orally and is also available for injection; its onset of action
is usually between 30 and 60 minutes and its duration of action 4–6 h. It is heavily
plasma protein-bound, which in addition to high levels of tissue binding contributes to
its long half-life. It is metabolised in the liver and eliminated by the kidney.
Tramadol
Tramadol is an interesting opioid analgesic because its action is attributable to at least
two different mechanisms acting synergistically. This synthetic drug is structurally
related to morphine and is a weak agonist at MORs with much lower affinity than
morphine. In addition to its opioid activity, it also acts as a serotonin and noradrenaline
reuptake inhibitor (SNRI). It blocks the reuptake of serotonin and noradrenaline in
serotonergic and noradrenergic nerve pathways, including descending inhibitory
pathways (Fig. 6.2) involved in pain modulation. This increases the level of these
transmitters, and therefore enhances the activity of the inhibitory neurones in blocking
transmission of pain signals from the first-order to the second-order neurones. SNRIs
are used to treat depression and anxiety (p. 58), both associated with chronically painful
conditions, and this action of tramadol may contribute to its analgesic effect in these
disorders. It is useful in a wide range of situations, including neuropathic pain, post-
operative pain, osteoarthritis, and rheumatoid arthritis.
Pharmacokinetics
Because it increases serotonin levels, tramadol may cause serotonin syndrome. It may
also reduce the anti-emetic action of ondansetron, an anti-emetic commonly used to
counteract opioid-induced nausea and vomiting. Ondansetron blocks 5HT3 receptors,
opposing the raised serotonin levels caused by tramadol. There is evidence that higher
doses of tramadol are needed to control pain in the presence of ondansetron, and that
the anti-emetic action of ondansetron is reduced in the presence of tramadol.
Tramadol is a pro-drug and genetic variations in the enzyme that activates it,
CYP2D6, can reduce or increase the rate at which the active agent is released, which in
turn can affect its analgesic action (Fig. 6.9).
New Directions
There would be huge clinical value in new opioid drugs with equivalent analgesic
potency to current agents, but with fewer adverse effects. One promising direction of
research is based on the observation that adjusting the precise molecular shape of the
drug can determine the signalling pathway activated in the cell when it binds to the
opioid receptor. The pharmacological effects of opioids binding to the MOR are due to
activation of signalling pathways within the cell, and recently it has become clear that
different drug effects are caused by different signalling pathways. The analgesic effect of
opioids in pain pathways is due to Gi-protein activation (see p. 32), but respiratory
depression and constipation seem to be the result of the MOR activating a different
pathway in the nerve, called the β-arrestin pathway. Standard MOR agonists, e.g.
morphine, activate both pathways, but it was realised that if an opioid drug could be
designed to activate G-protein signalling but not the β-arrestin signalling pathway, it
could produce an effective analgesic without these additional adverse effects. Research
in this area has produced a group of opioids called biased ligands, because they select
one signalling pathway over another, and which may represent one of the most
significant advances in analgesia development for some time. The first drug in this
group to reach clinical practice, oliceridine (TRV130), was approved by the FDA in
2020, and other agents are currently in development. Oliceridine is a MOR agonist that
preferentially activates Gi-protein signalling over β-arrestin signalling, and so gives
analgesia with fewer adverse effects, particularly respiratory depression. Its mechanism
of action is shown in Fig. 6.11.
Glucocorticoids
The adrenal cortex produces a range of steroid hormones, including mineralocorticoids,
some sex hormones, and glucocorticoids. The main glucocorticoid in humans is
cortisol, which is usually referred to as hydrocortisone when used therapeutically. In
addition, a range of synthetic and semi-synthetic agents is available. Glucocorticoids
have multiple, wide-ranging effects on immunity, inflammation, and energy
metabolism, and so great care is needed when using them.
Table 6.1
FIG. 6.13 Mediators of the inflammatory response and their source
cells.
FIG. 6.14 Typical pattern of cortisol secretion over a 24-h
period. From Hall JE and Hall ME (2021) Guyton and Hall textbook
of medical physiology, 14th ed, Fig. 78.9. Philadelphia: Elsevier.
Glucocorticoids in Therapeutics
Glucocorticoids are used in a wide range of acute and chronic inflammatory and
immune conditions to suppress inflammation and modulate immune responses. They
are essential therapy in adrenal insufficiency or failure or following adrenalectomy and
are used for their immunosuppressant action in malignancies of lymphoid tissue and
leukaemia. Because of their wide-ranging adverse effects, they are used at the lowest
effective concentration and for as short a time as possible. Potency varies significantly
between different preparations, and low to moderate potency agents are used wherever
possible; high-potency glucocorticoids are only used if necessary and with great care.
Box 6.1 lists doses of some commonly used glucocorticoids required to give an anti-
inflammatory effect equal to 5 mg of prednisolone.
Table 6.2
Cushing’s Syndrome
In 1912, Harvey Cushing described the signs and symptoms of excessive glucocorticoid
production by the adrenal gland, and the syndrome produced by prolonged
glucocorticoid excess was named after him. Iatrogenic Cushing’s syndrome, caused by
administered glucocorticoids, is relatively common because of the widespread use of
these agents in medicine, and it consists of a wide constellation of signs and symptoms
listed below and shown in Fig. 6.16.
Glucocorticoid Drugs
Examples: betamethasone, dexamethasone, hydrocortisone, prednisolone,
triamcinolone
The choice of drug is made based on several considerations, including its
mineralocorticoid (sodium- and water-retaining) activity. A glucocorticoid with
significant mineralocorticoid activity is inappropriate in conditions where fluid
retention could be damaging, e.g. in raised intracranial or intraocular pressure, where
increased fluid load would worsen the condition. On the other hand, when adrenal
failure requires replacement therapy, mineralocorticoid activity is useful because it is
part and parcel of the normal physiological function of the missing hormones. Fig. 6.17
illustrates the relative glucocorticoid and mineralocorticoid potencies of a few steroids,
including the mineralocorticoid fludrocortisone. For comparative purposes, cortisol
is assigned the value of 1 for both glucocorticoid and mineralocorticoid activity and the
other agents are evaluated in comparison to this: for example, betamethasone is on
average 35 times as potent a glucocorticoid as cortisol but possesses almost none of its
mineralocorticoid action. Potency, half-lives, and duration of action of some of the main
glucocorticoids are given in Table 6.3.
Table 6.3
Aspirin rapidly became the most popular painkiller worldwide, but its mechanism of
action was not explained until the 1970s when Professor John Vane, working at the
University of London, showed that aspirin inhibits an enzyme called cyclo-oxygenase
(COX). COX produces prostaglandins, a family of inflammatory mediators generated in
large quantities in injured tissues, and which promote all aspects of the inflammatory
response. Inhibiting this enzyme blocked PG production and suppressed inflammation.
Vane was a co-winner of the Nobel Prize in 1982 for this work.
Aspirin’s popularity as an anti-inflammatory analgesic was tempered by the fact that
its adverse effects can be severe and potentially fatal, which drove research to find safer
alternatives including ibuprofen and naproxen. In addition, new uses for this old
drug were being explored: several large studies in the 1990s and 2000s demonstrated
clear benefit of aspirin use in cardiovascular disease and cancers.
Biosynthesis of Prostaglandins
Fig. 6.18 shows the pathway by which PGs are produced. It names two important
enzymes involved: phospholipase A2 and COX.
Phospholipase A2 releases arachidonic acid from the cell membrane into the cell
cytoplasm. Glucocorticoids inhibit this step, one of the many anti-inflammatory
actions of these steroids. Arachidonic acid is the precursor for several inflammatory
mediators, the most important being the PGs and the leukotrienes (see p. 167 and Fig.
8.12). COX converts arachidonic acid into unstable intermediates, PGG2 and PGH2,
which are then converted into the five main PGs – PGD2, PGE2, PGF2⍺, PGI2
(prostacyclin), and thromboxane (TX) A2 – by specific synthases. COX is inhibited by
NSAIDs and also by paracetamol, which is interesting because paracetamol is not
classed as an NSAID due to its lack of anti-inflammatory activity.
Function of Prostaglandins
PGs are important drivers of the inflammatory response and mediate pain and fever, but
in healthy tissues they have a wide range of roles, sometimes called housekeeping
functions, essential to the normal functions of the cell. They include cell growth and
division, thermoregulation, blood clotting and cardiovascular function, appetite and
feeding behaviours, protection of the gastric lining, and smooth muscle contraction, and
are involved in the physiology of pain. Many of the most significant adverse effects of
NSAIDs arise because they block production of housekeeping PGs in healthy tissues, as
well as inflammatory PGs in injured tissues.
Prostaglandins in Fever
The temperature control centre of the brain lies in the hypothalamus, which contains a
network of heat-sensitive neurones that maintain core body temperature at around 37°C
(98.6°F). In response to a range of inflammatory or infective substances circulating in
the bloodstream, the hypothalamus can adjust this internal thermostat to increase body
temperature. Substances that cause fever are called pyrogens. As with other aspects of
the inflammatory response, fever is protective because increased body temperature
usually inhibits microbial growth and enhances the activity of defence cells including
phagocytes. Most pyrogens are derived from microbes and include some bacterial toxins
and lipopolysaccharide (LPS) from bacterial cell walls. In addition, some products of
damaged tissues, and some inflammatory mediators, such as interleukin-6, act as
endogenous pyrogens. Pyrogens increase PGE2 synthesis by COX-2 in the hypothalamus
and this is the key event that sets the hypothalamic thermostat to the higher level and
causes fever. NSAIDs reduce fever because by blocking this pyrogen-induced rise in
PGE2, the hypothalamic thermostat remains at normal core temperature level.
PGE2 and the Stomach Lining
This important housekeeping function is performed by PGE2 secreted in the stomach
mainly but not exclusively by the action of COX-1: COX-2 is also thought to contribute.
PGE2 stimulates gastric mucus production, increases blood flow through the mucosa,
stimulates bicarbonate production, and reduces gastric acid production. All these
measures protect the integrity of the gastric mucosa and enhance its ability to
regenerate damaged cells.
Aspirin
Aspirin is currently used in medical practice mainly as an antithrombotic agent, and its
prior use as an anti-inflammatory has largely fallen out of favour because of adverse
effects. Aspirin is highly selective for platelet COX-1 and accumulates selectively in
platelets. At much lower doses (75 mg/day) than are needed for effective anti-
inflammatory action, it gives prolonged inhibition of platelet activity by irreversibly
blocking platelet COX-1, permanently preventing it from producing the pro-clotting PG
TXA2. This means the effects of aspirin last for the lifespan of the platelet (about 10
days).
Pharmacokinetics
Aspirin (acetylsalicylic acid) is a pro-drug and is rapidly broken down in the plasma and
the liver to release the active agent, salicylic acid. Oral doses are rapidly and completely
absorbed. Because it is an acidic drug, it is not ionised in the stomach and so there is
some gastric absorption; however, most is absorbed in the small intestine. The plasma
half-life of acetylsalicylic acid is less than 20 minutes, and the half-life of salicylic acid is
3–4 h. Salicylic acid is metabolised in the liver, and the main metabolic pathway
involved is saturated at higher doses. This means that the drug can accumulate and the
half-life of the drug increases. This can happen at the upper end of the therapeutic dose
range, and so clearance times in overdose or in liver impairment can be significantly
extended. Metabolites are mainly excreted in the urine, although this is strongly
influenced by urinary pH. Normal urine is acidic, which increases the reabsorption of
salicylic acid from the filtrate back into the bloodstream (see also p. 19). Increasing
urinary pH (i.e. making it more alkaline, for example, by administration of bicarbonate)
increases urinary excretion and is a key element in the treatment of aspirin overdose.
Adverse Effects
Aspirin can cause any or all of the NSAID adverse effects described above. Because of its
antiplatelet action, it can increase bleeding times and the risk of haemorrhage; the risk
of major haemorrhage increases with age. Additionally, it can cause a condition called
Reye’s syndrome in children. This is characterised by encephalopathy and fatty liver
failure and is fatal in a high proportion of cases. It was most commonly seen in children
recovering from a viral illness and who were treated with aspirin, an observation that led
to the recommendation that it should not be used in children under the age of 12.
Implementation of this recommendation led to an immediate and dramatic fall in the
incidence of Reye’s syndrome, which is now rarely seen.
Salicylism
Aspirin toxicity (salicylism) is characterised by headache, nausea, and vomiting. There is
also hyperventilation, because aspirin directly stimulates the respiratory centre; this
increases carbon dioxide loss and causes respiratory alkalosis. Hyperventilation and
vomiting lead to dehydration. Aspirin is neurotoxic and causes a range of neurological
signs and symptoms including tinnitus, nausea, vomiting, seizures, and
cardiorespiratory depression.
Diclofenac
Diclofenac was developed in the mid-1960s as the product of rational drug design, based
on the structures of pre-existing NSAIDs. It reversibly inhibits both COX-1 and COX-2
and is available in oral and parenteral formulations, including rectal preparations and
topical gel. Its plasma half-life is around 2 h, but its duration of action is usually
between 8 and 12 h. It has been associated with higher cardiovascular risks than other
NSAIDs, probably due to a relative selectivity for COX-2 inhibition over COX-1, thus
reducing prostacyclin levels in the cardiovascular system.
Ibuprofen
Ibuprofen, synthesised by Boots in the 1960s, is now globally the most common NSAID.
It is a propionic acid derivative, not a salicylate, and reversibly inhibits both COX-1 and
COX-2. It is well absorbed orally, has a plasma half-life of about 2 h, is over 99% bound
to plasma proteins, and almost completely metabolised in the liver to inactive products.
Naproxen
Naproxen reversibly inhibits both COX-1 and COX-2 and, like ibuprofen, is a propionic
acid derivative. Its plasma half-life is between 12 and 15 h, and so can be given only
twice a day.
Piroxicam
Piroxicam is well absorbed following oral administration, is mainly metabolised in the
liver, and has a long plasma half-life (more than 30 h), which means it needs only once
daily administration. It is associated with more significant GI toxicity and serious skin
reactions than other NSAIDs and is not normally a first-choice NSAID when initiating
anti-inflammatory treatment.
Coxibs
The coxibs are COX-2 selective inhibitors and were developed in the 1990s, when it was
believed that COX-1 and COX-2 had clearly separated roles in prostaglandin production.
COX-1 was clearly associated with housekeeping functions including protection of the
stomach lining. The researchers who initially identified COX-2 could only induce their
experimental tissues to produce it by exposing the tissue to pro-inflammatory stimuli,
and so it was initially thought that COX-2 was only expressed following tissue damage
and that it was exclusively responsible for the production of inflammatory PGs. Several
drugs, including celecoxib, etoricoxib, and rofecoxib, were developed to selectively
inhibit COX-2 and released onto the global market. These agents cause less GI irritation
and bleeding than non-selective NSAIDs and so are better tolerated in longer-term use.
However, they should still be used with care in those with GI disorders because they can
exacerbate inflammatory bowel disease and increase the risk of bleeding from GI ulcers.
They are unlikely to worsen asthma but should be avoided in people with a history of
allergy to NSAIDs. As with the non-selective agents, they increase the risk of
cardiovascular events and impair kidney function, probably because they block COX-2-
mediated prostacyclin production in blood vessels. Roche withdrew rofecoxib in 2004
because clear evidence was emerging linking the drug to increased risk of heart attacks
and strokes. Celecoxib has a plasma half-life of 8–12 h and is given orally. It is well
absorbed and metabolised in the liver. Etoricoxib has a longer half-life of around 20 h
and is given once daily.
Paracetamol
Paracetamol is an effective antipyretic and is analgesic in mild to moderate pain. The
mechanism of action of paracetamol is not fully understood and although it reversibly
inhibits COX, it seems likely that other mechanisms contribute to its pharmacological
activity. It does not block COX in peripheral tissues, so it has no anti-inflammatory
action, does not irritate the stomach, and has no effect on platelets. It inhibits COX-1 in
the CNS, which may explain its antipyretic action. When paracetamol is used in
combination with an NSAID like ibuprofen, the analgesic effect is better than either
drug used alone, suggesting that paracetamol has an additional pain-relieving action
that does not involve COX inhibition. Paracetamol is metabolised in the liver and some
of its metabolites may contribute to its analgesic action by increasing activity in pain
modulation pathways in the brain, and it is possible that further research in this area
will produce new analgesic drugs.
Pharmacokinetics
Paracetamol is well absorbed when given orally or rectally. Its plasma half-life is 2–3 h
and it is metabolised in the liver; some of its metabolites may be responsible for at least
some of the analgesic action of the drug. It is generally well tolerated and is often the
preferred analgesic in children, older people, and individuals unable to tolerate NSAIDs.
Overdose, however, can lead to serious liver injury and fatal liver failure. Paracetamol is
metabolised in the liver to more than one product. Around 10% is metabolised to n-
acetyl-p-benzoquinone imine (NAPQI), which is highly hepatotoxic: it binds irreversibly
to liver proteins, causing cellular necrosis. At therapeutic levels of paracetamol, and with
normal liver function, the liver rapidly and efficiently converts NAPQI to non-toxic
compounds; this reaction requires glutathione. Liver stores of glutathione are limited,
however, and at higher paracetamol doses, the levels of NAPQI exceed available levels of
glutathione, allowing NAPQI levels to rise and cause liver injury (Fig. 6.22). The
standard treatment for paracetamol overdose is n-acetylcysteine (NAC), given orally
or intravenously. NAC increases glutathione levels and increases the liver’s ability to
clear NAPQI. It provides complete protection from liver injury if given within 8 h of
paracetamol ingestion.
Antihistamines
Histamine is an important inflammatory mediator, synthesised from the amino acid
histidine by the enzyme histidine decarboxylase and broken down via two main
metabolic pathways catalysed by diamine oxidase (also called histaminase) and
histamine N-methyl transferase respectively (Fig. 6.23).
Histamine is implicated in a range of inflammatory and allergic disorders including
atopic dermatitis and allergic rhinitis (hay fever).
Histamine
Most body tissues synthesise histamine, which has several actions in regulating
inflammatory and immune functions. It acts via four types of histamine receptor: the H1,
H2, H3, and H4 receptors. The location and distribution of these receptors varies, which
determines the effect of histamine on a specific tissue. The main actions of histamine are
as an inflammatory mediator, a regulator of gastric acid secretion, and a
neurotransmitter in the brain.
FIG. 6.22 Metabolism of paracetamol and the hepatotoxic
metabolite NAPQI. Modified from Ha CE and Bhagavan NV (2023)
Essentials of medical biochemistry, 3rd ed, Fig. 35.9. San Diego:
Academic Press.
Histamine in Allergy
In allergy (immediate or type I hypersensitivity), exposure to a usually harmless antigen
stimulates an excessive and inappropriate immune response. Signs and symptoms
depend on the site of exposure to antigen and on the degree of the immune response.
Local responses, e.g. following an insect bite, include redness, swelling, itch, and pain at
the site. Inhaled allergens, e.g. in hay fever, cause swelling and increased nasal
secretions, sometimes also involving the eyes and ears. This causes runny nose, nasal
congestion, sneezing, and red, itchy eyes characteristic of the disorder. If the bronchial
tree is involved, there is bronchoconstriction and increased secretions, causing wheeze
and cough, as in asthma. Ingested substances in food allergy cause abdominal cramping,
nausea, vomiting, and diarrhoea. In the most severe form, anaphylaxis, widespread
release of histamine gives blood levels high enough to cause systemic and life-
threatening bronchoconstriction and upper airway swelling. In addition, systemic
vasodilation and loss of blood volume because of histamine-induced increased vascular
permeability reduces blood pressure and leads to profound shock, and there is usually a
widespread skin rash, urticaria, or other skin symptoms.
Histamine as a Neurotransmitter
Histamine is a neurotransmitter in the brain, acting on H1, H2, and H3 receptors. It is
released in vomiting pathways linking the inner ear and the brain, triggering nausea and
vomiting induced by motion sickness and vertigo (see Fig. 10.13). It is also released by
a collection of nerves running from the hypothalamus to the cortex and the reticular
activating system, which are involved in regulation of the sleep-wake cycle. These nerves
are active during waking hours and the histamine they release maintains alertness, and
at night their activity stops to allow sleep. Antihistamines that cross the blood–brain
barrier are commonly used as anti-emetics (e.g. cinnarizine and cyclizine) and/or
sedatives (e.g. promethazine).
Antihistamines
Antihistamines antagonise the action of histamine at H1 receptors and are widely used
as anti-inflammatories, especially in allergic inflammation, as anti-emetics, and as
sedatives. Research in the first decades of the twentieth century demonstrated and
explained the role of histamine in allergy and anaphylaxis, and the first antihistamines
were introduced in the 1930s. Most antihistamines are available as oral preparations,
and some are available for topical application to the skin, eye, or ear. They are not useful
in the emergency treatment of anaphylaxis, because histamine is not the only mediator
involved.
First-Generation Antihistamines
Examples: chlorphenamine, cyclizine, cyproheptadine, diphenhydramine, promethazine
Diphenhydramine and chlorphenamine, which are still widely used today,
became available in the 1930s. These and other older drugs are sometimes referred to as
first-generation or sedating antihistamines because they cross the blood–brain barrier
and sedate, impair memory and cognitive function, and can decrease sleep quality.
Promethazine is particularly sedative. Additionally, they are associated with a range
of adverse effects because they block other receptor types including muscarinic
receptors and so can have significant antimuscarinic adverse effects, including urinary
retention, dry mouth, constipation, and tachycardia. First-generation antihistamines
can interfere with cardiac conductivity and cause arrhythmias, including QT
prolongation and torsades de pointes, and can cause hypotension. In general, these
agents are best avoided in older people because newer drugs are considered to be safer
and superior choices. Most have fairly long half-lives: the half-life of chlorphenamine
can vary between 12 and 40 h depending on the individual’s ability to metabolise it.
Cyclizine is used as an anti-emetic.
REFERENCES
1. Bannister K, Dickenson A.H. The plasticity of descending controls in pain:
translational probing. J. Physiol. 2017;595(13):4159–4166.
2. Melzack R, Wall P.D. Pain mechanisms: a new
theory. Science. 1965;150(3699):971–979.
3. Praveen Rao P.N, Knaus E. Evolution of nonsteroidal anti-inflammatory drugs
(NSAIDs): cyclooxygenase (COX) inhibition and beyond. J. Pharm. Pharm.
Sci. 2008;11(2):81s–110s.
4. Steed C. The anatomy and physiology of pain. Surgery. 2016;34(2):55–59.
5. Trang T, Al-Hasani R, Salvemini D, et al. Pain and poppies: the good, the bad and
the ugly of opioid analgesics. J. Neurosci. 2015;35(41):13879–13888.
6. Valentino R.J, Volkow N.D. Untangling the complexity of opioid receptor
function. Neuropsychopharmacology. 2018;43:2514–2520.
Resources
Church M.K, Church D.S. Pharmacology of antihistamines. Indian J.
Dermatol. 2013;58(3):219–224.
Emanuel M.B. Histamine and the antiallergic antihistamines: a history of their
discoveries. Clin. Exp. Allergy. 1999;29(S3):1–11.
Laight D. Accounting for cardiovascular risk when prescribing
NSAIDs. Prescriber. 2018:15–20.
Thangam E.B, Jemima E.A, Singh H, et al. The role of histamine and histamine
receptors in mast-cell mediated allergy and inflammation: the hunt for new
therapeutic targets. Front. Immunol. 2018;9:1873.
7: Drugs and Cardiovascular Function
Introduction
The Myocardium
Contractility of the Myocardium
The Cardiac Conducting System
Autonomic Control of the Cardiovascular System
Sympathetic Activity and Cardiovascular
System Function
Parasympathetic Activity and Cardiovascular
System Function
Drugs and the Heart
Drugs That Affect Cardiac Contractility
Sympathetic Agonists
Sympathetic Antagonists
Calcium-Channel Antagonists
Digoxin
Phosphodiesterase Inhibitors
Cardiac Arrhythmias
The Cardiac Action Potential
The Aetiology of Cardiac Arrhythmias
Anti-Arrhythmic Drugs
Class I Agents
Class II Agents
Class III Agents
Class IV Drugs
Anti-Arrhythmic Drugs Not Covered in the
Vaughan Williams System
Drugs and Blood Vessels
Blood Vessel Anatomy and Blood Vessel Diameter
Factor Xa Inhibitors
Antiplatelet Drugs
Aspirin
Adenosine-Receptor Antagonists
Dipyridamole
Fibrinolytic Drugs
The Fibrinolytic System
Haemostatic Drugs
F OCUS ON
Introduction
Worldwide, cardiovascular disease, including coronary heart disease and stroke, is the
most common non-communicable disease. The morbidity, mortality, and economic cost
imposed on healthcare systems by this disease burden is enormous. In this chapter, the
main groups of drugs used to treat some important cardiovascular disorders are
described and related in the Focus sections to their use in specific conditions. Although
the material here is divided for convenience into drugs acting on the heart and drugs
acting on blood vessels, cardiovascular physiology is an integrated balance of both
cardiac and blood vessel activity, and many drugs discussed below affect the function of
both.
FIG. 7.1 The layers of the heart wall. Modified from Waugh A and
Grant A (2020) Ross & Wilson anatomy and physiology in health
and illness, 14th ed, Fig 5.9A. Oxford: Elsevier.
The Myocardium
The layers of the heart wall are shown in Fig. 7.1. The heart chambers, and the valves
that control flow through the heart and into the aorta and pulmonary arteries, are lined
with a single-cell thick endothelium. This provides a smooth, glossy surface to facilitate
low-friction, non-turbulent blood flow. The outer layer, the pericardium, is a protective
double membrane enclosing the pericardial sac, which contains pericardial fluid to
lubricate the beating of the heart. The middle layer, the myocardium or heart muscle, is
the thickest layer and is composed of branched cardiac myocytes which are structurally
and functionally different from smooth and skeletal muscle cells.
Key Definitions
• Afterload:
the resistance against which the ventricle must push to eject blood into the
circulation
• End-diastolic volume:
where HR is heart rate and SV is stroke volume. Factors, including drugs, that
increase or decrease either Heart rate or SV consequently affect cardiac output, although
when functioning normally, the cardiovascular system (CVS) makes appropriate
adjustments to maintain cardiac output if one parameter changes. For example, in a
warm environment, peripheral blood vessels dilate to increase heat loss through the
skin, which reduces systemic blood pressure (BP). To maintain blood flow to the tissues,
sympathetic stimulation increases heart rate to compensate and maintain cardiac
output. Cardiac output is an important determinant of BP. Falling cardiac output tends
to reduce BP, and rising cardiac output tends to increase it.
Preload is the degree of stretch in the ventricular wall just before it contracts and is
mainly determined by the volume of blood in the ventricle, called the end-diastolic
volume (EDV, Fig. 7.3). The higher the EDV, the more stretched the ventricular wall
and the higher the preload. The EDV in turn is determined by the venous return. The
more blood returning to the ventricle through the venous system, the fuller the ventricle
is and the more work it must do to eject it. In the healthy ventricle, increasing EDV
actually increases the force of contraction because the increased blood volume stretches
the myocardium, and as with other muscle tissue, a degree of stretch increases the force
of muscular contraction. This ensures that the myocardium adjusts its contractility to
match the amount of work it must do to eject the blood from the heart chambers. Drugs
that reduce preload, e.g. β-blockers, reduce cardiac workload but can reduce cardiac
output.
Afterload is the pressure against which the ventricle must push to eject blood into the
arterial system (Fig. 7.3). This increases when arterial BP is high, either because of high
circulating blood volume or because there is increased vasoconstriction in the peripheral
arterial networks. Drugs that reduce afterload, e.g. vasodilators, reduce cardiac
workload and increase cardiac output.
Secondary Pacemakers
The SA node is the primary pacemaker in the heart because it spontaneously discharges
faster than any other area of the conduction system. However, if the SA node fails or if
the conduction pathway carrying its impulses to the AV node are blocked or damaged,
the AV node can take its place, discharging much more slowly (40–60 times per
minute). This gives the heart a backup pacemaker, which preserves life, although an
heart rate so slow greatly limits exercise tolerance. Like the SA node, the AV node is
supplied by the vagus nerve (parasympathetic input), which slows conduction between
atria and ventricles; sympathetic stimulation increases the rate and frequency of AV
conduction. As with the SA node, AV node depolarisation requires increased Ca2+ levels
in its cells. This is achieved by the opening of Ca2+ channels in the nodal cell plasma
membranes, allowing Ca2+ to enter from the extracellular fluid, and so Ca2+-channel
blockers, which prevent Ca2+ entry, effectively delay AV conduction speeds.
Sometimes, an area of tissue somewhere in the heart may develop abnormal
pacemaker activity and, by generating its own action potentials, it can disrupt normal
conduction pathways and interfere with normal cardiac contraction. The abnormal
tissue is called an ectopic focus, and ectopic beats are a common cause of arrhythmia.
Most blood vessels have only sympathetic supply, which usually causes
vasoconstriction (e.g. in the gastrointestinal tract and the skin). In some blood vessel
beds, however, for example the coronary arteries, sympathetic activity causes
vasodilation to supply the myocardium in times of stress.
• Positive/negative inotrope:
• Positive/negative chronotrope:
Sympathetic Agonists
Examples: adrenaline, dobutamine
Sympathomimetics that stimulate cardiac β1-receptors have positive inotropic and
chronotropic effects. β1-receptors in the cardiac myocyte membrane are linked to
biochemical pathways in the cell that generate cAMP, which activates a range of
processes that facilitate contraction, including opening Ca2+ channels in the cell
membrane. The Ca2+ that enters the cell stimulates further Ca2+ release from the
sarcoplasmic reticulum, activating the intracellular contractile mechanism (Fig. 7.6).
Sympathomimetics also cause vasoconstriction, partly by a direct action on ⍺1 receptors
on blood vessels, but also via the vasoconstrictor action of angiotensin II, released by
activation of the RAAS (see Fig. 7.13). This increases BP, which is further enhanced by
RAAS-induced fluid retention and increased circulating blood volume.
Adrenaline acts at both ⍺- and β-receptors and so affects the heart and blood
vessels, and is used in acute cardiac emergencies including cardiac arrest, as well as in
anaphylaxis. Dobutamine is more active on β-receptors than ⍺-receptors, so it has
more effect on the heart than the vasculature and is used to support heart function in a
range of situations including cardiogenic shock.
β2-agonists, used as bronchodilators in asthma and other obstructive airways
disorders, are discussed on p. 164.
Pharmacokinetics
Sympathomimetics should either be avoided completely or used very carefully with
other drugs that increase sympathetic neurotransmitter levels, including amitriptyline
and doxepin, because their common action of increasing sympathetic activity increases
preload and afterload, placing additional strain on the heart. Adrenaline is used by
injection or by infusion. When given intravenously, it is rapidly removed from the
bloodstream and taken up into sympathetic nerves, so it has a rapid onset of action but
its duration of action is short. It is mainly metabolised in the liver and its breakdown
products are excreted in the urine. It is a powerful vasoconstrictor because it also
activates ⍺1 receptors in blood vessels, which helps to slow its absorption and extend its
duration of action when given intramuscularly. Dobutamine has a very short half-life
of around 2 minutes and is given by intravenous infusion.
Adverse Effects
Sympathomimetics increase the risk of cardiac arrhythmias and can cause tachycardia
and hypertension. They increase the workload and the oxygen requirements of the heart
and can precipitate angina in people with pre-existing cardiac ischaemia. Other adverse
effects related to their sympathetic action include palpitations, cold fingers and toes
(Raynaud’s phenomenon), hyperglycaemia, and tremor.
Sympathetic Antagonists
A wide range of drugs is available to block sympathetic effects at both ⍺- and β-
receptors.
Pharmacokinetics
Some β-blockers, including propranolol, pindolol, and labetalol, are more fat-
soluble than water-soluble, and so enter liver cells and are metabolised. Others, like
atenolol and sotalol, are more water-soluble, and so are less well metabolised in the
liver and depend upon renal function to clear them from the bloodstream. The half-lives
of β-blockers is generally short, between 2 and 12 hours, and there are sustained-release
preparations available to extend the duration of action of each dose.
Adverse Effects
Many of the main adverse effects of β-blockers relate to reduced sympathetic nervous
system activity. There may be hypotension and bradycardia. β-blocker-induced
bradycardia can be treated with glucagon, which increases Ca2+ levels in cardiac
myocytes by increasing cAMP levels and has positive inotropic and chronotropic effects.
β-blockers depress myocardial contractility and so should be used very carefully in heart
failure. They interfere with cardiac conduction and can cause arrhythmias and should be
used cautiously if at all in heart block. They can cause cold hands and feet because they
block β2-receptors on blood vessels, which normally mediate vasodilation and maintain
blood flow. This may be less common when cardioselective β1-antagonists are used than
with non-selective agents. They interfere with lipid and glucose metabolism and can
cause weight gain and fatigue. The more lipid-soluble drugs, e.g. propranolol and
metoprolol, cross the blood–brain barrier and cause CNS side-effects, including vivid
dreams, nightmares, and other sleep disturbances. This is less common with the more
water-soluble agents like atenolol.
Non-selective β-blockers
Examples: propranolol, sotalol, pindolol, oxprenolol
Some non-selective β-blockers, including pindolol and oxprenolol, weakly
stimulate β-receptors when they bind; this gives them a degree of sympathomimetic
activity. Although the ability of a β-blocker to mildly stimulate β-receptors may seem
counterproductive, it does mean that the side-effects of these drugs can be attenuated;
bradycardia and cold extremities, for example, can be less marked. Propranolol has an
additional action in the heart: it has an anaesthetic-like action on the sinoatrial node,
which may be important when used to treat cardiac arrhythmias.
Cardioselective β-Blockers
Examples: atenolol, bisoprolol, metoprolol
Cardioselective (β1-selective) agents may cause fewer β2-blockade related side-effects,
including cold extremities. However, they are more likely to prevent the hypoglycaemia-
induced tachycardia that warns many people with diabetes, particularly type 1, of falling
blood glucose levels.
Pharmacokinetics
Amlodipine has a long half-life of 30–50 hours because it is slowly absorbed and
poorly metabolised. Felodipine has a half-life of 11–16 hours because it is heavily
plasma protein-bound. Most drugs in this group, however, have relatively short half-
lives (nifedipine, 2 hours; verapamil, 4 hours), and sustained-release preparations
are available to maintain therapeutic plasma levels. Most are extensively metabolised in
the liver, and enzyme inducers such as rifampicin and St. John’s wort can increase
their metabolism and reduce their effectiveness. On the other hand, enzyme inhibitors
like ketoconazole and grapefruit juice can reduce metabolism of the Ca2+ antagonist
and elevate blood concentrations to potentially toxic levels. The Ca2+-channel blockers
themselves interfere with the metabolism of a large number of other drugs.
Adverse Effects
Most Ca2+-channel blockers, except amlodipine, reduce cardiac contractility and can
precipitate or worsen heart failure. Verapamil is the most likely to do this. There may
be bradycardia and poor exercise tolerance because the heart cannot respond adequately
to the demands of exercise. Relaxation of GI smooth muscle causes gastric acid reflux
and constipation. Vasodilation causes headache, facial flushing, and pooling of blood,
especially in leg veins. This in turn causes oedema of the feet and ankles.
Digoxin
Cardiac glycosides, including digoxin and digitoxin, are a group of plant-derived
organic compounds related to steroids. Digoxin, the main agent in clinical use, was
originally extracted from the purple foxglove (Digitalis purpurea) and is still extracted
from plants of the foxglove family because this is cheaper than manufacturing it. It is
used mainly in heart failure and in some atrial arrhythmias. The doctor credited with
first demonstrating its clinical effectiveness in heart failure was William Withering, who
in 1795 published an account of his use of foxglove extracts in a range of patients,
including those with dropsy (oedema), a common consequence of heart failure.
Digoxin blocks the Na+/K+ pump in the cell membrane. This pump maintains the
ionic gradient across the membrane so that it remains electrically excitable. It
continually pumps potassium ions into the cell in exchange for Na+ ions. By blocking the
Na+/K+ pump, digoxin increases Na+ levels in the cell (Fig. 7.8). This excess Na+ is
pumped out of the cell using a pump which exports Na+ and imports Ca2+. This
therefore increases intracellular Ca2+ levels, which increases myocyte contractility and
the strength of cardiac contraction. Digoxin improves ventricular contractility and
improves heart function in heart failure.
Pharmacokinetics
Digoxin is given orally and is generally well absorbed. Some antibiotics, including
erythromycin and gentamicin, destroy microbes in the GI tract that normally
deactivate oral digoxin and can significantly increase absorption and blood levels.
Digoxin is very water-soluble, and so is poorly metabolised in the liver and has a long
half-life of about 36 hours. Clearance therefore depends on renal elimination, and care
must be taken in people with reduced kidney function. It has a narrow therapeutic
index, and even small increases in plasma levels can precipitate toxicity.
Adverse Effects
Digoxin toxicity is potentially life-threatening and can develop even when plasma levels
are within the therapeutic range. Although digoxin is used to treat certain atrial
arrhythmias, it can cause a range of abnormal rhythms, including ventricular ectopic
beats, because by interfering with the Na+/K+ pump, it increases the excitability of the
myocyte membrane. It may slow down AV nodal conduction enough to cause heart
block, and cause bradycardia because of its inhibition of the SA node. One important
interaction is with diuretics, which may cause excessive K+ excretion and
hypokalaemia and are often co-prescribed with digoxin in heart failure. Hypokalaemia
increases the risk of digoxin-induced arrhythmias, so it is important to ensure K+ levels
remain within the normal range, giving K+ supplements if necessary. Digoxin also has a
range of non-cardiac side-effects, including nausea and vomiting, mainly due to
stimulation of the chemosensitive trigger zone. Other neurological effects include
dizziness, fatigue, and a yellowing of vision, the latter because of inhibition of the
Na+/K+ pump in the light-sensitive cones of the retina.
Phosphodiesterase Inhibitors
Examples: enoximone, milrinone
Phosphodiesterase (PDE) is the enzyme that breaks down cAMP (see Fig. 3.7). When
its action is inhibited, cAMP levels in the cardiac myocyte rise, Ca2+ levels increase, and
contractility improves, without increasing heart rate. PDE inhibitors improve heart
function in heart failure but can trigger potentially life-threatening ventricular
arrhythmias, seen in more than 1 in 10 patients. They are given by infusion. Milrinone
has a plasma half-life of 2 hours and depends on good kidney function for clearance
because it is not metabolised in the liver. Enoximone has a longer half-life of 8 hours
and undergoes hepatic metabolism, so should be used carefully in people with liver
impairment. Both agents cause vasodilation and possibly hypotension.
Drug treatment of heart failure aims to interrupt and delay the progress of the
vicious cycle of deteriorating cardiac function. This includes reducing the workload of
the heart by blocking excessive sympathetic nervous system activity, reducing preload
and afterload, improving coronary artery blood flow to increase oxygen and glucose
supply, and supporting cardiac function with drugs that improve contractility without
increasing oxygen demands. The main drug groups used in the treatment of heart
failure are summarised in Fig. 7.10.
Their pharmacology is described below. For people who cannot tolerate ACEIs,
angiotensin-receptor antagonists (ARAs) can be used; they have a similar range of
effects and their pharmacology is also described below.
Diuretics
Diuretics, e.g. bendrofluazide, furosemide, and spironolactone, are widely
used in heart failure because they promote water and electrolyte loss in the urine,
reducing oedema, pulmonary congestion, and circulating blood volume. By reducing
the volume load of the heart, the efficiency of heart contraction can be improved. Care
must be taken not to over-reduce circulating blood volume, because this reduces
cardiac output, causes hypotension, and may trigger kidney injury if renal blood flow
falls too low. The pharmacology of diuretics is discussed in Chapter 10.
Vasodilators
Vasodilators are useful in heart failure because they:
Cardiac Arrhythmias
A cardiac arrhythmia is a disturbance of rate or rhythm. Some arrhythmias are very
common and do not interfere significantly with the efficiency of the heart’s pumping
action. At the other end of the spectrum, some arrhythmias are immediately life-
terminating. Arrhythmias are described according to their anatomical origin: ventricular
arrhythmias arise from the ventricles, and supraventricular arrhythmias arise either
from the atria or the AV node.
Understanding the physiology of the cardiac action potential explains the mechanism
of action of the main anti-arrhythmic drugs, especially as the classification of anti-
arrhythmic agents relates to their physiological effects on the movement of specific ions
during different phases (Fig. 7.11).
Re-Entrant Rhythms
These are common causes of arrhythmias and arise when an action potential is allowed
to establish a self-perpetuating circuit. In the healthy heart, the impulses travel together
throughout each section of tissue and cannot spread backwards or to adjacent areas
because of the long refractory period following the wavefront. Impulses that do meet
along adjacent pathways extinguish each other (Fig. 7.12A). Re-entrant circuits develop
when two parts of the myocardium are conducting at different speeds, one slower than
the other. As the slower impulse travels down its pathway, it can then excite adjacent
areas of the myocardium that have conducted at normal speed and have repolarised and
become excitable again (Fig. 7.12B). This sends impulses backwards up the normally
conducting tissue, establishing a self-perpetuating re-entrant circuit (Fig. 7.12C). Re-
entrant rhythms are common causes of tachyarrhythmias, i.e. arrhythmias associated
with fast heart rates, including atrial fibrillation and flutter and ventricular tachycardia
following myocardial infarction.
Ectopic Arrhythmias
If an area of heart muscle acquires increased automaticity, it can begin to generate
action potentials independently of normal sinus rhythm, interfering with normal
conduction pathways and co-ordinated myocardial contraction (Fig. 7.12D).
Anti-Arrhythmic Drugs
Because they interfere with electrical activity in the heart, anti-arrhythmic drugs may
themselves generate arrhythmias. By slowing the generation and transmission of
impulses in normal pathways, they may allow an ectopic focus to become dominant, or
may allow a re-entrant circuit to become established (Fig. 7.12). The risks of anti-
arrhythmic drug-induced arrhythmias is increased in people with pre-existing heart
disease, e.g. ischaemic heart disease. In addition, people with increased sympathetic
drive are at higher risk because sympathomimetics are themselves arrhythmogenic; for
example, sympathetic drive to the heart is increased in heart failure in an attempt to
maintain cardiac output. Some anti-arrhythmic drugs, e.g. sotalol, disopyramide,
and digoxin, are particularly associated with the production of arrhythmias.
FIG. 7.12 The genesis of cardiac arrhythmias.A. Normal
conduction pathways. B–C. Re-entrant rhythms. D. Ectopic focus.
Fig. 7.13A shows a typical action potential from a ventricular cell treated with a
class Ic agent, showing the shallower slope of depolarisation caused by the reduced
rate of Na+ entry into the cell. Class Ia and Ic agents are used in both supraventricular
and ventricular arrhythmias, and class Ib drugs are only effective in ventricular
arrhythmias.
Class II
Examples: propranolol, atenolol, metoprolol
Class II agents are the β-blockers, which inhibit the effect of the sympathetic
nervous system on the heart. Adrenaline and other sympathomimetics can cause
arrhythmias by stimulating both SA and AV nodal discharge, by initiating ectopic
beats and by increasing the excitability of the myocardium, and a range of
arrhythmias are at least partly due to excessive sympathetic stimulation, including
atrial fibrillation and post-MI. β-blockers delay AV nodal conduction and control
ventricular tachycardias originating from the atria and suppress discharge from
ectopic foci. Fig. 7.13B shows the effect of β-blockade on an action potential in an AV
nodal cell: the action potential is both delayed and prolonged. Class II agents are used
in both supraventricular and ventricular arrhythmias.
Class III
Examples: amiodarone, sotalol
These agents are K+-channel blockers, and so they inhibit phase 3 of the action
potential and slow repolarisation of the cardiac cells. In so doing, they delay the
restoration of the ion balance across the myocyte membrane and extend its refractory
period. Fig. 7.13C shows the effect of a class III agent on the action potential in a
ventricular myocyte. They are used in both supraventricular and ventricular
arrhythmias.
Class IV
Examples: verapamil, diltiazem
Class IV agents are the Ca2+-channel blockers. They have particular activity on the
Ca2+ channels that open to depolarise cells in the SA and AV nodes, and so reduce the
rate of SA nodal firing and slow impulse transmission through the AV node. For this
reason, they are most useful in treating arrhythmias originating in these parts of the
heart and are not effective in ventricular arrhythmias. Fig. 7.13D shows the effect of
Ca2+-channel blockers on an action potential in an AV nodal cell: depolarisation is
slow because depolarisation in AV and SA nodal cells is due to Ca2+ influx, not Na+
influx, and repolarisation is delayed.
FIG. 7.13 The main effects of each of the Vaughan Williams
classes of anti-arrhythmic drugs on the ventricular action potential
(A and C) and on the action potential in atrioventricular nodal cells
(B and D). Modified from Waller DG and Sampson A (2018)
Medical pharmacology and therapeutics, 5th ed, Fig. 8.3. Oxford:
Elsevier.
Class I Agents
Although all class I agents block Na+ channels and so slow depolarisation in heart cells,
this class is subdivided because some have additional channel-blocking activity, and
some have more extended block than others.
Disopyramide (Ia)
Disopyramide blocks K+ channels (class III activity) as well as Na+ channels, so it not
only delays depolarisation, it slows repolarisation too and extends the action potential
and the refractory period. Along with quinidine, another class Ia drug, it is falling out
of use because of its serious side-effects.
Pharmacokinetics
Disopyramide is given orally and has a half-life of about 6 hours. It is partly metabolised
in the liver, producing a metabolite with marked antimuscarinic activity.
Adverse Effects
Antimuscarinic side-effects (reduced saliva production, bradycardia, constipation,
blurred vision) can be very troublesome. Disopyramide has a significant negative
inotropic action: it can cause or worsen heart failure and produce problematic
hypotension. It is strongly arrhythmogenic.
Lidocaine (Ib)
Lidocaine blocks Na+ channels in a use-dependent manner (see also Local anaesthetics,
p. 69). This means that it is most effective in electrically excitable cells when they are
most active and their Na+ channels are open for most of the time. Lidocaine therefore
blocks rapidly discharging, arrhythmogenic heart cells preferentially to normal tissue.
Pharmacokinetics
Lidocaine is given intravenously because if given orally, it is almost completely cleared
by the liver in first-pass metabolism. It is extensively metabolised in the liver, and some
of its metabolites are active. IV administration allows drug delivery to be rapidly titrated
against its clinical effects. Its plasma half-life is around 2 hours, but this can be longer in
liver or renal failure.
Adverse Effects
Lidocaine depresses nerve activity in the CNS and at high doses may cause drowsiness,
confusion, and convulsions.
Flecainide (Ic)
Flecainide is given orally or intravenously. Although it is used in both ventricular and
supraventricular arrhythmias, it is particularly effective in converting atrial fibrillation
to sinus rhythm.
Pharmacokinetics
Flecainide is slowly but completely absorbed following oral administration. Its plasma
half-life is around 12 hours, and there are extended-release preparations, so its action is
long-lasting.
Adverse Effects
Flecainide is contra-indicated in people who have had a myocardial infarction or have
heart failure, in whom the drug significantly increases the risk of fatal ventricular
arrhythmias. Common side-effects include oedema, dyspnoea, and dizziness.
Class II Agents
The general pharmacology of β-blockers is described on p. 47. A number of β-blockers,
including propranolol and atenolol, are used to treat arrhythmias.
Sotalol
Sotalol is a β-blocker with additional class III activity: that is, it blocks K+ channels
which open to allow the cardiac myocyte to repolarise in phase 3 of the action potential.
It has significant arrhythmogenic action of its own, and so is usually only used in certain
serious arrhythmias, e.g. ventricular tachycardia.
Amiodarone
Amiodarone is widely used in a range of arrhythmias. Although its main action is by
blocking K+ channels and so is categorised as a class III drug, it has activity in all four
classifications. It blocks Na+ channels (class I), has β-blocker activity (class II), and
blocks Ca2+ channels (class IV). It can be given orally or intravenously.
Pharmacodynamics
Amiodarone is highly lipid-soluble and is extensively taken up from the blood and
stored in many body tissues. Its half-life is very long, between 7 and 8 weeks. Because its
half-life is so long, treatment is usually initiated with a large loading dose to bring
plasma levels up quickly. It is metabolised in the liver.
Adverse Effects
The incidence of side-effects with amiodarone is very high: up to 15% of patients report
side-effects in the first year of treatment, rising to 50% with longer-term use. Skin
reactions and nausea are common. Amiodarone contains about 37% iodine by weight,
and so can interfere with thyroid function, causing either hyperthyroid or hypothyroid
states. Assessing thyroid function before beginning treatment is essential. Cardiac side-
effects include arrhythmias and bradycardia. In 10% of patients, corneal deposits of the
drug cause visual symptoms including halos and photophobia. There may be irreversible
pulmonary toxicity, which carries a mortality rate of up to 10%. Neurotoxic side-effects
include peripheral neuropathy and altered taste and smell. Hepatic function should be
monitored because amiodarone can cause liver impairment.
Dronedarone
Like amiodarone, dronedarone has activity in all four Vaughan Williams classes. It
may be preferred to amiodarone because it is better tolerated and patients may be more
likely to comply with treatment.
Pharmacokinetics
Dronedarone was designed to be much less fat-soluble than amiodarone in the
expectation that its distribution throughout body tissues would be less extensive and it
contains no iodine, so it has no thyroid toxicity. It has a much shorter half-life than
amiodarone and is less arrhythmogenic.
Adverse Effects
Dronedarone shares many of amiodarone’s side-effects including pulmonary toxicity
and liver toxicity. It should not be used in heart failure because it increases mortality,
and it can cause heart failure in people with no prior history. GI symptoms, e.g.
vomiting, diarrhoea, nausea, and taste disturbances, are common.
Class IV Drugs
The general pharmacology of Ca2+-channel blockers is described on p. 132.
Adenosine
Adenosine is a small but important molecular mediator widespread in body fluids and
has a range of biological effects. It forms the core of ATP, the cell’s energy storage
molecule. Adenosine is pro-inflammatory and theophylline, an adenosine-receptor
antagonist, is used in asthma (see p. 165). In the heart, adenosine inhibits transmission
in both the SA and AV nodes and is used to terminate supraventricular tachycardia.
Pharmacokinetics
Adenosine has a very short plasma half-life (less than 10 seconds) and its duration of
action is less than a minute, so is given by rapid bolus injection.
Adverse Effects
These are common but short-lived because adenosine is so rapidly cleared from the
bloodstream. There may be bradycardia and AV block. Adenosine is a vasodilator, so
facial flushing and headache may occur. It should not be used in asthma because it can
cause bronchospasm.
Digoxin
The pharmacology of digoxin is described above. It blocks AV nodal conduction and so
is used to reduce transmission of impulses to the ventricles in atrial tachycardias,
including atrial fibrillation and atrial flutter. Because it increases Ca2+ levels in
myocardial cells, it increases excitability and can cause arrhythmias.
Table 7.1
Endothelium-Derived Mediators
Mediator Function Relevant Drugs
Nitric oxide Vasodilator; anticlotting; anti- Nitrate vasodilators increase
inflammatory nitric oxide levels
Prostacyclin Vasodilator and anticlotting Non-steroidal anti-
prostaglandin inflammatory drugs reduce
production (Fig. 6.18)
Endothelin Vasoconstrictor Bosantan blocks endothelin
receptors
Angiotensin II Produced by the enzyme Angiotensin-converting
angiotensin-converting enzyme inhibitors inhibit
enzyme acting on angiotensin production; angiotensin-
I in pulmonary endothelium; receptor antagonists block
vasoconstrictor its action
Cardiac output, the volume of blood ejected from each ventricle per minute, is
discussed above. TPR is determined by the degree of constriction in the arterial bed,
especially the resistance arterioles. The greater the degree of constriction in arterioles,
the higher the resistance to blood flow and the higher the BP. Vasodilation, on the other
hand, opens up the vascular bed and BP falls. Moment-to-moment control of BP is
controlled mainly by the ANS regulating heart rate, cardiac contractility, and peripheral
resistance.
Total blood volume is an additional factor influencing BP. It is usually maintained
within a narrow range, and in health varies little. Excess fluid is excreted, mainly by the
kidney, and if the body water content falls, homeostatic mechanisms conserve fluid, for
example by reducing urine volume, until intake can replace it. This is a slower, more
sustained mechanism in BP regulation because changes in total body water usually take
longer to become significant. The RAAS is fundamental to this process.
Vasodilators
Key Definitions
Vasodilator:
an agent that relaxes smooth muscle in arteries and veins
Venodilator:
Vasodilators relax vascular smooth muscle, increasing blood flow through the vessel.
They are used to reduce BP in hypertension and to reduce afterload in heart failure.
Dilation of systemic vessels improves blood supply to tissues, including the heart in
angina and systemic vessels in peripheral vascular disease and Raynaud’s disease. Most
vasodilators act on both arteries and veins, and so reduce pressure throughout the
vascular system. Exceptions include the nitrate vasodilators, which act preferentially
on veins, and hydralazine, which dilates arteries. Venodilators like nitrates are useful
mainly in heart failure, because by opening venous capacity, more blood remains in the
venous system and less is returned to the heart, reducing preload and relieving cardiac
effort. Arterial dilators are more useful in hypertension, although they reduce BP, which
can trigger reflexive sympathetic tachycardia.
⍺1-Blockers
Examples: prazosin, doxazocin
⍺1-receptor blockade causes vasodilation in arteries and veins, which decreases
afterload, preload, and BP, and these drugs are used in hypertension. They are also used
to treat urinary retention associated with benign prostatic hyperplasia because they
relax the smooth muscle of the bladder neck and improve urine flow. Prazosin has also
been used to improve sleep patterns in post-traumatic stress disorder. Its half-life is
only 3 hours, whereas doxazocin’s half-life is about 12 hours. Hypotension, especially
early in treatment and in older people, is common.
Nitrates
Examples: Glyceryl trinitrate, isosorbide dinitrate
Nitrates have been used as vasodilators since the 1850s to treat hypertension and
angina. Nitroglycerine, the original nitrate, is highly unstable (it is a key component of
explosives), and early workers studying this new class of compound included Alfred
Nobel, who gave his name to the most famous prize in science, and whose first invention
was a detonator to control nitroglycerine explosions. Early work to produce a stable and
convenient form of nitroglycerine included incorporating it into chocolate, which was
nibbled at the onset of anginal pain. Nitrates increase levels of nitric oxide in smooth
muscle in blood vessel walls, producing vasodilation, reducing BP, and increasing blood
flow. Nitric oxide is the main endogenously produced vasodilator, and so has a key role
in the control of systemic vascular resistance. It lowers free Ca2+ levels inside the muscle
cell by increasing Ca2+ uptake into the storage membranes inside the cell and by
blocking Ca2+ uptake by the cell from extracellular fluids. This reduces the amount of
Ca2+ available to activate the actin–myosin contractile proteins and relaxes the muscle.
Nitrate vasodilators cause venous dilation, which reduces venous return and preload,
and arterial dilation, which reduces afterload, relieving the workload of the heart. They
dilate coronary arteries, improving blood and oxygen supply to the heart, and improve
blood flow, particularly to the ischaemic areas of the myocardium.
Glyceryl Trinitrate
GTN is an important drug in the treatment of angina. Given as a sublingual spray or a
tablet tucked under the tongue, it relieves anginal pain within minutes. Tablets lose their
pharmacological activity within 8 weeks of opening their container because GTN is
volatile and evaporates with time, but the spray formulation is stable for up to three
years. It may also be given by infusion or via a skin patch.
Pharmacokinetics
GTN cannot be given orally because it is completely eliminated from the bloodstream by
first-pass metabolism in the liver. Its plasma half-life is between 1 and 4 minutes, and it
is effective over a period of half an hour or so. Tolerance to the vasodilator effect of the
drug develops with extended administration, i.e. with infusion or with patches, and
withdrawal of the drug is needed for it to regain its effectiveness. The cause of the
tolerance is not understood, but it limits sustained use of nitrate vasodilators especially
in chronic angina. The usual management strategy is to build in a nitrate-free period
every 24 hours, usually overnight, to restore the drug’s effectiveness.
Adverse Effects
Because it is such a powerful vasodilator, GTN dilates meningeal blood vessels and
causes headaches, which can be severe. Other side-effects due to excessive vasodilation
include facial flushing and hypotension.
Isosorbide Dinitrate
This nitrate vasodilator can be taken orally, and in addition is available in slow-release
form to prolong therapeutic plasma levels. Tolerance to its vasodilator effects develops
quickly, and it is usually taken in the morning, so that its levels are low overnight and
the tolerance is reversed.
Ca2+-Channel Antagonists
Smooth muscle needs an increase in intracellular Ca2+ levels to activate contraction.
This rise is triggered by Ca2+ influx through Ca2+ channels in the smooth muscle cell
membrane. In the presence of a Ca2+-channel antagonist, e.g. nifedipine or
amlodipine, Ca2+ entry is prevented and vascular smooth muscle is relaxed. This
causes vasodilation and reduces BP. The pharmacology of Ca2+-channel blockers is
described in more detail on p. 132.
K+-Channel Openers
Some vasodilators relax vascular smooth muscle by opening K+ channels in the muscle
cell membrane and allowing extra K+ to enter. This reduces the membrane potential
(makes it more negative or hyperpolarises it, see Fig. 7.11), pulling it further away from
the activation threshold, and the muscle cell relaxes.
Minoxidil
Minoxidil is primarily an arterial dilator. It is well absorbed when given orally and is
metabolised in the liver. Its half-life is about 4 hours, but its vasodilator effect is potent
and long-lasting, which triggers reflex sympathetic compensatory mechanisms. As a
result, there is tachycardia and activation of the RAAS with Na+ and water retention,
which usually require additional treatment with a β-blocker and a diuretic, respectively.
Minoxidil is therefore not a first-line agent and is used to treat severe hypertension that
does not respond satisfactorily to other drugs. One of its metabolites stimulates hair
growth, and hirsutism is an unwanted effect, particularly in women, although the drug is
used topically to treat male-pattern baldness.
FIG. 7.16 The beneficial effects of angiotensin-converting enzyme
inhibitors and angiotensin-receptor antagonists in cardiovascular
disease.HR, Heart rate.
Nicorandil
This vasodilator is given orally to treat angina. It is classed as a K+-channel opener, but
its vasodilator activity is partly due to its action in increasing NO levels in vascular
smooth muscle. It dilates both healthy and diseased coronary arteries, improving blood
flow to the myocardium, and causes systemic vasodilation, reducing both preload and
afterload. It has a short plasma half-life (1 hour), and its main side-effects are similar to
those of the nitrates, including headache, hypotension, lethargy, and fatigue. It may also
cause GI side-effects, including vomiting.
Pharmacokinetics
All ACEIs are given orally. Some, e.g. ramipril and enalapril, are pro-drugs, and rely
upon activation in the liver. Half-lives vary considerably; captopril’s half-life is only 2
hours, but enalapril has a half-life of 35 hours, and its active metabolite enalaprilat is
10 hours. Lisinopril is given as an active drug and needs only once daily
administration because it has a half-life of 12 hours. Caution is needed when co-
administered with a diuretic, especially when initiating ACEI treatment, because this
can result in a sudden and potentially dangerous fall in BP.
Adverse Effects
ACEIs are contra-indicated in pregnancy because they damage the developing fetal
kidney. They can cause a range of side-effects, including allergy, electrolyte imbalances,
and significant liver impairment: liver function should be monitored and the drug
discontinued if there is evidence of liver problems. In up to 35% of patients, a dry,
troublesome cough develops, more commonly in women, which can be bothersome
enough to lead to the patient wishing to discontinue treatment. ACEI-induced cough is
not dose-related and can arise at any time after initiation of treatment. It is attributed to
the accumulation of the irritant inflammatory mediator bradykinin in the airways (Fig.
7.18). In addition to producing angiotensin II, ACE also deactivates bradykinin.
Inhibition of ACE activity allows bradykinin levels to rise, causing cough. This adverse
effect disappears when the drug is withdrawn. Increased levels of bradykinin may also
be the reason why ACEIs can cause urticaria and angioedema (sudden swelling in the
deeper layers of the skin). Although these side-effects are much rarer than cough,
angioedema is sometimes severe enough to cause dangerous tracheal obstruction.
Angiotensin-Receptor Antagonists
Examples: losartan, valsartan
Angiotensin II acts on two different receptor types, AT1 and AT2, and from a clinical
point of view the AT1 receptor is the more important because this is the subtype involved
in the main cardiovascular actions of angiotensin II. Drugs that block the AT1 receptor
therefore have a very similar range of pharmacological activity to the ACEIs, although
they do not cause cough (because ACE is still produced and is still available to break
down bradykinin). Losartan was the first orally active drug in this group, and although
its half-life is short (2 hours), metabolism produces an active metabolite with a half-life
of 10 hours, and losartan can be given once daily. As with ACEIs, ARAs should be used
carefully in people with renovascular disease and not in pregnancy.
Renin Antagonist
The only renin antagonist currently available is aliskiren, licensed for the treatment of
hypertension. It binds to renin and prevents it from converting angiotensin I to
angiotensin II. Angiotensin II levels therefore fall and its effects are significantly
reduced. Aliskiren is given orally, and because of its long half-life (24–40 hours), it is
only given once daily and takes up to two weeks to achieve effective steady state plasma
levels. As with captopril, aliskiren was a product of rational drug design and effectively
reduces BP when used as monotherapy. Used alone, it is generally well tolerated,
although it can cause diarrhoea and hypokalaemia. However, effective control of
hypertension frequently requires concurrent use of two or more drugs, and issues have
been identified when aliskiren is combined with ARAs or ACEIs in patients with
diabetes, cardiovascular disease, or renal disease. In these patients, the rates of serious
side-effects including renal dysfunction and hypotension are high and these drugs
should therefore not be used in combination.
Endothelin-Receptor Antagonists
Endothelin is a powerful vasoconstrictor released by the endothelial lining of blood
vessels and vascular smooth muscle as part of the normal regulation of blood vessel
tone. It acts on two receptor types, ETA and ETB, and it probably causes prolonged, local
vasoconstriction, increases BP, and activates inflammatory mediators mainly through
ETA receptors. It also stimulates proliferation and fibrosis in the blood vessel wall and in
the heart, which may contribute to the remodelling damage seen in the heart and blood
vessels in cardiovascular disease and is likely to be involved in the pathogenesis of
atherosclerosis. Despite endothelin’s well-understood multifunctional role in
cardiovascular pathophysiology, endothelin antagonists have so far only proved useful
in pulmonary arterial hypertension and in treating finger ulcers in systemic sclerosis. In
pulmonary arterial hypertension, endothelin levels are consistently raised, and
endothelin-receptor antagonists improve exercise tolerance, reduce mean pulmonary
artery pressure, and delay the progression of the disease.
Bosentan
Bosentan was the first endothelin antagonist discovered, licensed in 2001, and it blocks
both ETA and ETB receptors equally. It is used to treat pulmonary arterial hypertension,
is given orally, has a half-life of around 5 hours, and is cleared by the liver and the
kidney. It is teratogenic, so is contra-indicated in pregnancy, and can cause liver
impairment, so hepatic monitoring is important and it should be avoided in all but the
mildest degrees of pre-existing liver disease. It commonly causes GI side-effects, and
because it is an enzyme inducer, it can increase the clearance rates of a range of other
drugs including warfarin, opioids, and Ca2+-channel blockers.
F oc us on: Hypertension
Hypertension is a common disorder in which sustained high BP damages the heart
and the arterial system. It is a major risk factor for cardiovascular disease and greatly
increases the risk of stroke, myocardial infarction, heart failure, cardiac arrhythmia,
and kidney disease. It is a major factor in the development of atherosclerosis (see
below) which itself is both a cause and a manifestation of cardiovascular disease. Most
cases are primary (essential) hypertension, sustained high BP with no directly
identifiable cause. Risk factors for essential hypertension include increasing age, male
sex, overweight, obesity, heredity, high salt intake, high alcohol intake, sedentary
lifestyle, and low birthweight. Fewer than 10% are directly related to another
condition; this is secondary hypertension and causes include pregnancy, renal
disease, and a range of endocrine disorders including diabetes.
Regulation of BP in health is a complex process, and a range of factors contribute,
including normal RAAS activity, normal endothelial function, cardiac function,
balanced sympathetic/parasympathetic activity, insulin resistance, and balanced
activity of a large range of hormones, cytokines, and other mediators. There is also
evidence for an inflammatory and immunological component, and it has been shown
that in hypertension, activated immune cells infiltrate the kidney and blood vessels,
both key organs in BP regulation. There is increasing evidence that much of the end-
organ damage seen in hypertension––in the kidneys, the brain, the blood vessels, and
the heart––is due to ongoing inflammation. Research into precisely which immune
cells and which inflammatory mechanisms are involved is ongoing and may lead to
novel treatments for hypertension in the future: this would be particularly useful for
the substantial subset of hypertensive people for whom conventional antihypertensive
treatment does not work.
Atherosclerosis
Atherosclerosis is a chronic inflammatory condition of blood vessels that causes
deposition of semi-liquid fatty material within the blood vessel wall and damages and
roughens the endothelium. The initial changes take the form of a fatty streak on the
endothelium, which enlarges and develops over a period of time to form a mature lesion,
called an atherosclerotic plaque, which progressively obstructs the vessel lumen as it
expands. The fatty streak is initiated by deposition of a form of cholesterol called low-
density lipoprotein (LDL) cholesterol in the arterial wall, which triggers ongoing
inflammation that drives ongoing injury. The main pathological features are shown in
Fig. 7.20A, and Fig. 7.20B is a photomicrograph of a coronary artery showing the
gross appearance of an atherosclerotic plaque. The plaque core is a semi-fluid
accumulation of fatty material, including cholesterol and other lipids, and infiltrated
with inflammatory cells including lymphocytes. Characteristic of these plaques are foam
cells, macrophages that have consumed large quantities of fat, which appear under the
microscope as dense foamy bubbles in the cytoplasm. Smooth muscle cells grow into the
core, and collagen and other fibrous material is deposited in the plaque. As the plaque
grows, the blood vessel lumen becomes progressively narrower and blood flow is
obstructed. The whole plaque is covered with a rough fibrous cap, which lacks the
smooth surface of healthy endothelium and increases the risk of thrombus formation.
The most advanced plaques become calcified and stiff, which makes them more likely to
rupture. If the plaque splits, exposing its fatty contents to the bloodstream, a blood clot
forms over the area of rupture, because the lipid-rich core is highly thrombogenic. This
causes sudden and immediate obstruction of the artery and is the commonest cause of
acute myocardial infarction and sudden coronary death. Fragments of this clot can also
travel in the bloodstream and lodge elsewhere, e.g. in the pulmonary circulation, which
causes a pulmonary embolism.
Vasodilators
The pharmacology of the main vasodilators is discussed above, but their usefulness in
IHD and angina is threefold:
• they dilate systemic arteries, which reduces afterload and cardiac workload.
• they dilate the coronary arteries, increasing myocardial blood flow.
• they are venodilators, which pool the blood in systemic veins and reduce venous
flow to the heart, reducing preload and cardiac workload.
β-Blockers
The pharmacology of β-blockers is described above. In IHD, they slow the heart and
therefore improve coronary artery blood flow by increasing time spent in diastole and
reduce contractility and oxygen consumption. This in turn reduces BP, further relieving
cardiac effort and alleviating the pain of angina. Cardioselective agents include
atenolol, bisoprolol, and metoprolol. Some β-blockers, e.g. pindolol and
carvedilol, also produce a degree of vasodilation, which reduces afterload and reduces
cardiac work. The drugs vasodilate via different mechanisms: pindolol weakly stimulates
β-receptors in blood vessels, and carvedilol blocks vascular ⍺-receptors.
Statins
Examples: simvastatin, atorvastatin, pravastatin
Statins were introduced in the 1980s and inhibit the liver enzyme
hydroxymethylglutaryl co-enzyme A (HMG co-A) reductase. This enzyme catalyses a
crucial step in cholesterol synthesis, so in the presence of a statin, hepatic cholesterol
production is inhibited. This in turn reduces circulating levels of non-HDL (‘bad’)
cholesterol. Statins are used in both primary and secondary prevention of cardiovascular
events like myocardial infarction and stroke: that is, in people who have never had a
cardiovascular event, and in people who have had one or more such event. They protect
against cardiovascular events even in people whose blood lipid profile is normal and are
recommended for use in type 1 diabetes even in the absence of other cardiovascular risk
factors. In addition to their effect on blood lipids, statins have additional biological
activities, including improving the health of vascular endothelium, stabilising
atherosclerotic plaques, and reducing the systemic inflammatory response, all of which
may contribute to cardiovascular protection. Statins are also used to treat primary
hyperlipidaemias and hypercholesterolemia.
Pharmacokinetics
Statins are given orally and most undergo significant first-pass metabolism. Half-lives
vary considerably between these drugs. Simvastatin and pravastatin have short half-
lives of 1–2 hours and are best taken at night because cholesterol synthesis is highest at
night. Atorvastatin has a much longer half-life of more than 30 hours.
Adverse Effects
Statins are generally well tolerated, even at high doses. Muscle pain is commonly
reported, but actual muscle damage caused by statins is rare. This may be due to statin-
induced leakage of Ca2+ from the endoplasmic reticulum within muscle cells, which may
damage the cell. People with pre-existing muscle conditions are more susceptible than
those with no such history, and exercise has been shown to reduce its incidence. Statins
may also cause liver damage, although there is evidence that certain statins may protect
against liver cancer and protect liver function in patients with pre-existing liver disease.
They may trigger diabetes in patients with predisposing factors, who should be
monitored. They are contra-indicated in pregnancy because fetal abnormalities have
been reported.
AntiThrombotic Drugs
Treatment with antithrombotic drugs reduces the risk of clots forming at the plaque site.
Low-dose aspirin inhibits platelet activation via blockade of platelet thromboxane A2
production (p. 152). Clopidogrel (p. 152) reduces clotting risk by irreversibly
preventing platelet aggregation. Anticoagulant therapy may also be used in for example
acutely developing coronary syndromes, in which the risk of thrombosis is significantly
elevated. Antithrombotic drugs are discussed in more detail below.
Haemostasis
Haemostasis, or blood clotting, is the physiological mechanism that converts fluid blood
to a firm, semi-solid thrombus. In health, blood remains fluid unless and until clotting is
needed to stop blood loss, but inappropriate blood clotting is an important
pathophysiological complication in a range of conditions, including myocardial
infarction, some cardiac arrhythmias including atrial fibrillation, atherosclerotic
vascular disease, serious infections and sepsis, pulmonary embolism, and stroke. There
is a range of rare, inherited disorders that increase blood coagulability, but more
common factors that increase the risk of thrombosis include pregnancy, varicose veins,
smoking, surgery, and malignant disease. Antithrombotic drugs are used when the risks
associated with clotting outweigh the risks associated with haemorrhage.
Key Definitions
• Antithrombotic:
a drug that by any mechanism prevents the formation or enlargement of blood clots
• Anticoagulant:
a drug that prevents blood clotting by inhibiting one or more clotting factors
• Antiplatelet:
• Fibrinolytic:
Natural AntiCoagulants
Physiological regulation of the clotting cascade involves a few natural anticoagulants
which control clotting and normally prevent inappropriate clotting, and which also
terminate it once the bleeding episode is resolved. As mentioned above, healthy
endothelium produces anticoagulants, important contributors to the prevention of
inappropriate clotting. These include antithrombin, an inhibitor of both thrombin and
activated FX. The body produces a powerful stimulant of antithrombin activity,
heparin, which is essential to physiological suppression of the clotting process, and
which is in widespread use as an anticoagulant.
AntiCoagulants
Blood flow is slower in veins than arteries, predisposing to thrombosis, and clots that
form here have time to develop and are rich in fibrin. Because most anticoagulants
directly or indirectly reduce thrombin activation and therefore fibrin production, they
prevent thrombus development more effectively in the venous circulation than in the
arterial circulation, where clots contain higher proportions of platelets than fibrin. The
main side-effect of anticoagulant therapy is haemorrhage, and anticoagulants should
therefore be used with great care, if at all, in patients with bleeding disorders and in
those at increased risk of haemorrhage, e.g. peptic ulcer disease or hypertension.
Vitamin K Antagonists
Examples: warfarin sodium, phenindione
Vitamin K is a co-factor in one key step in the synthesis of clotting factors II, VII, IX,
and X. Co-factors are essential participants in a wide range of biochemical reactions,
and the body needs only tiny quantities because they are constantly recycled. The
vitamin K molecule is deactivated each time it is used to make a molecule of clotting
factor and must be converted back to its biologically active form in order to be used
again. Vitamin K antagonists block its reactivation, and so the vitamin remains in its
deactivated form and clotting factor synthesis stops (Fig. 7.22). Their anticoagulant
effect may take several days to develop, because although clotting factor production is
halted, the factors already present in the blood will initially ensure normal clotting.
Warfarin
Warfarin belongs to the coumarin group of anticoagulants. The first coumarin to be
discovered, dicoumarin, was isolated in 1940 from a fungus that infected cattle fodder
and was causing the animals to die from internal bleeding. Warfarin, derived from
coumarin, was originally developed as a rat poison, but found a place in clinical
medicine in the mid-1950s as a therapeutic anticoagulant. Although warfarin is an
effective anticoagulant, its use is in decline because of the development of newer, safer
agents including thrombin inhibitors and Factor X inhibitors.
Pharmacokinetics
Unlike heparin, the only available anticoagulant at the time the coumarins were being
developed, warfarin is given orally. It is well absorbed and very highly bound to plasma
proteins, so it has a long half-life of up to 48 hours. A single dose takes effect in 14–16
hours and lasts for four to five days, and at the end of a course of warfarin it takes two or
three days for clotting times to return to normal, reflecting the time required to produce
adequate levels of new clotting factors. Warfarin crosses the placenta and is both
teratogenic and haemorrhagic in the developing baby, so should be avoided where
possible in pregnancy. Warfarin is associated with a range of drug interactions. Its
antithrombotic effect is additive to that of other anticlotting drugs, e.g. aspirin,
increasing bleeding times, and great care must be taken if these drugs are used together.
Its action is potentiated by a range of drugs, e.g. amiodarone and a range of
antimicrobials, which inhibit the liver enzymes that metabolise warfarin. Its action is
reduced by enzyme inducers such as alcohol and phenytoin, which enhance the liver
enzymes that metabolise warfarin and accelerate its clearance from the bloodstream.
FIG. 7.22 The mechanism of action of the vitamin-K
antagonists. Modified from Padmanabhan S (2014) Handbook of
pharmacogenomics and stratified medicine, Fig. 24.1. San Diego:
Academic Press.
Adverse Effects
The main side-effect is haemorrhage and bleeding times should be routinely monitored.
If bleeding times are excessively long with ongoing bleeding or significant risk of
dangerous haemorrhage, vitamin K (phytomenadione) can be given intravenously
to restore clotting factor production; it takes up to 6 hours for full effect. Alternatively,
blood clotting can be immediately restored to normal by infusion of functional clotting
factors.
Heparin
Examples: dalteparin, enoxaparin (low-molecular-weight (LMW) heparins),
unfractionated heparin
Heparin is the oldest anticoagulant in regular clinical use and is on the World Health
Organisation list of essential medicines. It was discovered in 1918 by a medical student
called L. Emmett Holt, who named it heparin because he was working with liver
extracts, although it is also produced by a range of other tissues including mast cells and
lung and blood vessel endothelium. Chemically, heparin belongs to a family of complex
sugars called glycosaminoglycans (GAGs), which are joined together to form much
larger molecules, the size of which depends on the number of individual GAG molecules
used. Heparin can therefore exist in a range of different molecular weights.
Unfractionated heparin is extracted from a range of animal sources and contains
heparin molecules with a range of molecular weights between 3000 and 30 000 kDa.
LMW heparins are separated from unfractionated heparin and contain molecules with
MW less than 7000 kDa. The molecular weight of the heparin units affects the
pharmacokinetics of the preparation, including absorption.
Heparin exerts its anticoagulant effect by binding to antithrombin. Antithrombin is
one of the key natural brakes on clotting and blocks thrombin and FX activity. Heparin
binds to antithrombin and greatly enhances its activity, further reducing the action of
thrombin, fibrin production, and clot development (Fig. 7.23).
FIG. 7.23 The action of heparin.
Pharmacokinetics
Heparin must be given by injection because heparin molecules are too large and heavily
charged to be absorbed across the wall of the GI tract. It is used intravenously or
subcutaneously, but not intramuscularly because of the risk of haematoma formation.
LMW heparin is usually given subcutaneously and is well absorbed from injection sites.
Its anticoagulant effect is generally predictable when the dose is calculated in relation to
body weight, and routine blood clotting monitoring is usually not required. It does not
bind significantly to plasma proteins and is cleared from the bloodstream by the liver
and the kidneys. Its half-life is longer than unfractionated heparin, and so requires less
frequent administration.
Unfractionated heparin’s absorption is more variable and less predictable than that of
LMW heparin and, depending on dose, its half-life ranges from 30 minutes to 3 hours.
Adverse Effects
Heparin has a narrow therapeutic index. Its main side-effect is haemorrhage, which is
potentially fatal. Bleeding caused by excess unfractionated heparin can be rapidly
reversed by IV administration of protamine sulphate, which binds to heparin in the
bloodstream and deactivates it. Protamine sulphate is much less effective in stopping
haemorrhage induced by LMW heparin, because it binds to it less strongly.
Another potentially lethal side-effect of heparin develops as the result of an
autoimmune reaction against platelets in the presence of heparin. In about 1 in 50
patients, the interaction between heparin and platelets stimulates the production of
antiplatelet antibodies that activate platelets and trigger thrombosis. In addition, the
widespread activation of platelets leads to their clearance from the circulation and
thrombocytopenia. In this situation, heparin must be discontinued and an alternative
anticoagulant, such as danaparoid or a direct thrombin inhibitor, used instead.
Danaparoid
This is a heparin-like agent (a heparinoid) made up of a number of different GAGs
related to heparin. As with heparin, its anticoagulant effect is due to activation of
antithrombin. It has a low MW of 5500 kDa and is completely absorbed from
subcutaneous administration.
Dabigatran
This anticoagulant was licensed in 2008 and was the first new anticoagulant in over 50
years. One important advantage claimed by its manufacturer is that unlike warfarin,
the standard agent in use at the time, monitoring blood levels is unnecessary.
Dabigatran is given orally as an inactive pro-drug, dabigatran etexilate, which is
converted to the active form in the liver. Its onset of action is rapid, and its half-life is
short, about 40 minutes. Life-threatening dabigatran-induced bleeding can be reversed
with idarucizumab, an antibody which binds with high specificity and high affinity to
dabigatran and prevents it from binding to thrombin. Renal function should be assessed
before treatment is begun, because the drug is cleared mainly by the kidney, and
monitoring renal function is particularly important in elderly people, who are most at
risk of life-threatening bleeds with this drug.
Bivalirudin
The use of leeches in ancient medical practice to bleed patients has been revived in
modern medicine in a limited number of clinical situations. Leech saliva contains a
powerful anticoagulant called hirudin, so that when the leech bites its target for a
blood meal, the blood remains fluid to allow the leech to feed. Leeches are sometimes
used in plastic surgery or other situations to prevent clotting and maintain blood flow to
a tissue. Hirudin is a directly acting thrombin inhibitor, and bivalirudin is a stable,
synthetic hirudin analogue with a short half-life of around 25 minutes.
Factor Xa Inhibitors
Examples: apixaban, fondaparinux, rivaroxaban
Activation of FX is the first step in the final common pathway (Fig. 7.21). Inhibiting
activated FX (FXa) therefore blocks clotting initiated by both the extrinsic and intrinsic
pathways. Routine monitoring of bleeding times is not needed, because their clinical
effect correlates very well with dose and so is predictable.
Pharmacokinetics
These agents are given orally and are incompletely absorbed from the GI tract. Their
half-lives are around 12 hours, but the onset of anticoagulant action is quick. They are
only partially metabolised, and much of the administered drug is excreted unchanged in
the faeces and urine.
Adverse Effects
Haemorrhage and anaemia are common but less so than with warfarin. Rivaroxaban
can cause nausea.
AntiPlatelet Drugs
Antiplatelet drugs are most useful in conditions where there is inflammatory endothelial
damage, primarily atherosclerosis, which triggers platelet activation. The endothelium
of arteries is exposed to considerably more shear stress than veins because blood flow
and pressure is much higher in the arterial than the venous system, and so arterial
endothelial damage is more common than venous endothelial damage. Clots that form
in arteries are therefore rich in platelets, and antiplatelet drugs are more effective in
preventing arterial thrombosis than venous thrombosis.
Aspirin
Although aspirin has traditionally been a standard anti-inflammatory drug, its
antiplatelet action has led to its widespread use to reduce thrombotic events in
cardiovascular disease, as well as in primary prevention of thromboembolism in people
without overt disease but with significant risk factors. Aspirin inhibits platelet activation
by irreversibly inhibiting platelet COX, the enzyme that produces prostaglandins.
Platelets synthesise a prostaglandin called thromboxane A2 (TXA2), which activates the
platelet and causes local vasoconstriction by contracting vascular smooth muscle. TXA2
is therefore a pro-clotting substance, and by inhibiting platelet COX and preventing
TXA2 production, aspirin suppresses blood clotting and increases bleeding times (see
also p. 152).
Low-dose aspirin, i.e. aspirin concentrations significantly lower than required for an
anti-inflammatory effect, achieves effective antithrombotic action. The standard
maintenance antiplatelet dose is 75 mg aspirin daily, compared to up to 2.4 g daily as an
analgesic/anti-inflammatory. This is mainly because aspirin irreversibly destroys
platelet COX, permanently abolishing its clotting action, so that restoring full platelet
activity requires the synthesis of brand-new platelets. After a single dose of aspirin,
platelet TXA2 generation recovers slowly, at the rate of about 10% per 24 hours, as new
platelets are released into the bloodstream from the bone marrow to replace the defunct
ones.
Pharmacokinetics
Aspirin (acetylsalicylic acid) has a short plasma half-life of about 20 minutes and is
broken down by plasma esterases to release the active ingredient, salicylic acid, which
diffuses passively into platelets.
Adverse Effects
Because prostaglandins have a wide range of physiological functions in healthy tissues,
COX inhibitors have a range of unpleasant side-effects. The low dose used in
cardiovascular prevention means that it is usually well tolerated, but it may still cause,
for example, GI irritation and should be avoided in people with active peptic ulcer
disease or a history of the same. Haemorrhage and allergy are also common side-effects.
The side-effects of COX inhibitors are discussed in more detail on p. 120.
Adenosine-Receptor Antagonists
Examples: clopidogrel, ticagrelor, prasugrel
ADP is an important pro-clotting factor released by platelets. ADP promotes and
accelerates platelet activation by acting on the P2Y12 receptor found on the platelet
membrane. The more platelets are activated, the more ADP is released to activate more
platelets, a very good example of a positive feedback system. Adenosine-receptor
antagonists bind to the platelet P2Y12 receptors and block them, preventing adenosine
from attaching and activating the platelet (Fig. 7.24). Precursors of the current group of
agents were identified in the 1970s as ADP antagonists only by chance, because
researchers were actually screening for anti-inflammatory drugs, and further work
produced clopidogrel in 1998. Adenosine-receptor antagonists are often used with
aspirin in a range of thromboembolic disorders including ischaemic heart disease and
acute coronary syndromes because the antiplatelet effects of the combination are
additive.
Pharmacokinetics
Clopidogrel and prasugrel are pro-drugs and require activation in the liver. Both
bind irreversibly to the platelet ADP receptors, and so their action is long-lasting even
though clopidogrel’s active metabolite has a half-life of only around half an hour.
Adenosine-receptor inhibitors are given orally, sometimes with an initial loading dose to
ensure rapid onset of action.
Adverse Effects
Care should be taken when co-prescribing with other drugs that have an antiplatelet
effect: this list includes non-steroidal anti-inflammatory drugs as well as perhaps less
obvious agents such as fluoxetine and sertraline (p. 58). Clopidogrel and
ticagrelor commonly cause GI upset and skin reactions. All agents in this group can
cause haemorrhage.
Dipyridamole
Dipyridamole is used orally, sometimes with aspirin and sometimes as a modified-
release formulation. Its mechanism of action is not entirely clear, and it may inhibit
platelets in several ways: for example, it has been shown to inhibit TXA2 synthesis, and
it enhances the antiplatelet action of prostacyclin.
FIG. 7.24 The antiplatelet action of the adenosine-receptor
antagonists.
Fibrinolytic Drugs
Examples: streptokinase, alteplase, tenecteplase
Clot-dissolving (fibrinolytic) drugs are used to break down a formed clot and restore
blood flow through an occluded vessel. Rapidly restoring blood flow through an artery
following thrombosis, e.g. in a cerebral or a coronary artery, can make the difference
between life and death for a patient, or the difference between significant disability and
complete recovery. Fibrinolytics are used in a range of situations, including deep venous
thrombosis, pulmonary embolism, myocardial infarction, or blocked in-situ cannulas or
stents. The main hazard with using these drugs is haemorrhage. Fibrinolytic drugs are
more effective the younger the clot, because as a clot matures, its fibrin content steadily
increases and it becomes denser, more compact, and less susceptible to drug action.
Treatment outcomes are therefore significantly better when drug infusion is begun as
soon as possible after the onset of symptoms.
Streptokinase
Streptokinase was discovered by chance in 1933 when it was observed that streptococcal
bacteria dissolved blood clotted in test tubes used for an unrelated experiment. The
active enzyme was isolated and named streptokinase as a nod to its microbial origin. It
is now known that streptokinase activates plasminogen, i.e. it acts like naturally
occurring tPA to release clot-dissolving plasmin, but its mechanism of action was not
worked out until the 1940s, when plasmin and plasminogen themselves were isolated.
To clear a blocked coronary artery in myocardial infarction, it is recommended that
treatment starts within 12 hours of the onset of symptoms, but the earlier the better,
because early treatment correlates directly with reduced mortality and improved cardiac
function post-infarction.
FIG. 7.25 The mechanism of action of fibrinolytic drugs.
Pharmacokinetics
Streptokinase is a highly efficient and fast-acting thrombolytic with a half-life of around
25–30 minutes and is given by IV infusion.
Side-Effects
Allergy is common, probably because streptokinase is derived from bacterial sources. In
addition, streptokinase can trigger an immunological response with the generation of
anti-streptokinase antibodies. These antibodies can persist for years and will neutralise
streptokinase if given again. Because of this, streptokinase is only used on one occasion
and if thrombolysis in the same patient is required in the future, a different agent is
chosen. A range of cardiovascular adverse effects, including cardiac arrest, hypotension,
heart failure, and cardiac ischaemia, are all common.
Haemostatic Drugs
Haemostatic drugs promote clotting and stop haemorrhage. Purified clotting factor
preparations are available for the treatment of conditions associated with deficiency; for
example, purified factor XIII is used to treat haemophilia A.
Tranexamic Acid
Tranexamic acid is a synthetic antifibrinolytic used in general surgery and in trauma to
reduce the risk and extent of bleeding and the need for transfusions. It is used to reduce
blood loss in women with very heavy menstrual periods, to stop prolonged nosebleeds,
and to control prolonged bleeding following dental surgery. It works by stopping
plasminogen activation, and so reduces plasmin production and protects the developing
fibrin clot (Fig. 7.25). It is also used to treat hereditary angioneurotic oedema (HAE), a
rare condition caused by a deficiency of a key inhibiting enzyme that restrains the action
of several enzyme cascades, including complement and clotting. Although not licensed
in all countries, there is some evidence that tranexamic acid can reduce the severity of
the attacks of swelling that characterise HAE.
It is given orally or intravenously, and is generally well tolerated, although there is a
risk of seizures with higher doses. It is a small molecule and is excreted unchanged in
the urine, so care must be taken if kidney function is reduced. Its half-life is around 2
hours, but this is extended in renal impairment.
References
1. Awtry E.H, Loscalzo J. Aspirin. Circulation. 2000;101(10):1206–1218.
2. Echt D.S, Ruskin J.N. Use of flecainide for the treatment of atrial fibrillation. Am.
J. Cardiol. 2020;125(7):1123–1133.
3. Lei M, Wu L, Terrar D.A, et al. Systematic review: modernised classification of
cardiac antiarrhythmic drugs. Circulation. 2018;138(17):1879–1896.
4. Marsh N, Marsh A. A short history of nitroglycerine and nitric oxide in
pharmacology and physiology. Clin. Exp. Pharmacol. Physiol. 2000;27(4):313–
319.
5. Norlander A.E, Madhur M.S, Harrison D.C. The immunology of hypertension. J.
Exp. Med. 2018;215(1):21–33.
6. Ruggenenti P, Cravedi P, Remuzzi G. The RAAS in the pathogenesis and
treatment of diabetic nephropathy. Nat. Rev. Nephrol. 2010;6(6):319–330.
7. Sirtori C.R. The pharmacology of statins. Pharmacol. Res. 2014;88:3–11.
Online Resources
Florek J.B, Lucas A, Girzadas D. Amiodarone. In: StatPearls [Internet]. Treasure
Island: StatPearls Publishing; 2023 Available
from:. https://www.ncbi.nlm.nih.gov/books/NBK482154/.
Ornelas A, Zacharias-Millward N, Menter D.G, et al. Beyond COX-1: the effects of
aspirin on platelet biology and potential mechanisms of
chemoprevention. Cancer Metastasis Rev. 2017;36(2):289–303.
8: Drugs and the Respiratory System
CHAPTE R OU TLINE
Introduction
The Respiratory System
The Structure of the Respiratory
Passageways
Drug-Induced Pulmonary Toxicity
Drugs and Pulmonary Defence Mechanisms
Ciliated Epithelium
Mucus
Cough
Obstructive Airways Disease
Asthma
Bronchodilators
Anti-Inflammatory Drugs
Chronic Obstructive Pulmonary Disease
F OCUS ON
Introduction
The respiratory system, conventionally divided into upper and lower parts, brings
atmospheric air into the lungs, from which it extracts oxygen and into which it excretes
waste carbon dioxide. The epithelia lining the respiratory passageways and the alveoli
are constantly exposed to the external environment via this inhaled air, so that
respiratory infections are very common, especially if normal respiratory defence
mechanisms are compromised in some way. Antimicrobial agents are described in
Chapter 11.
Ciliated Epithelium
The trachea and large airways are lined with ciliated columnar epithelium (Fig. 8.5).
Cilia are present on the epithelial lining deep into the respiratory tree, to the level of the
terminal bronchioles. They are thread-like motile structures anchored in the epithelial
cell membrane. Through a co-ordinated beating action, they clear mucus and other
secretions, along with particles trapped on their sticky surface, from the respiratory
passageways and carry them towards the throat for swallowing. This is called the
mucociliary escalator.
Table 8.1
Mucus
Mucus is produced by goblet cells present in the columnar epithelial lining of the upper
airways and forms a thin blanket overlying the cilia. Mucus is a viscous water-based gel
containing mucins, which are complex sugar/protein molecules, and particles present in
inspired air adhere to mucus layer and are trapped. The consistency of mucus is
normally thin enough to allow the ciliary escalator to transport it without difficulty and
clear it efficiently from the respiratory passageways; however, in respiratory infection or
irritation, mucus production increases, and the mucus often becomes thicker and
stickier. This makes the job of the ciliary escalator more difficult and increases the need
to cough to help clearance.
In some situations, mucus viscosity significantly affects clearance. The
parasympathetic nervous system promotes secretion of a fluid mucus that is easily
moved by ciliary action. Antimuscarinic drugs such as hyoscine are used to reduce the
volume of respiratory secretions, e.g. before surgery. However, they thicken the mucus,
which hampers normal clearance and can cause mucus pooling, airway obstruction, and
increased risk of infection. In some respiratory conditions, such as cystic fibrosis,
continual production of thick mucus leads eventually to failure of ciliary action.
Table 8.2
A range of inhalation devices is available, but effective drug delivery with all of
them depends very much on patient technique. If inhalation of drug triggers
coughing, most will be immediately expelled and therefore ineffective. If the patient’s
respiratory effort is poor, e.g. in children or frail elderly people, less drug reaches the
target airways. In addition, drug penetration into the respiratory tree is reduced if the
airways are narrowed by inflammation or bronchoconstriction; this means that
during exacerbations of the disease, inhaled drugs may be considerably less effective
than normal.
Alternative Routes to Inhalation
During exacerbations, drugs can be given orally or intravenously to ensure that high
concentrations reach the lung tissues via the bloodstream. This of course means that
the risk and incidence of side-effects are increased.
Dornase Alfa
Dornase alfa is a synthetic form of human deoxyribonuclease, the enzyme that breaks
down DNA in the body. It is given by nebuliser in cystic fibrosis. The thick mucus
produced in this condition is rich in neutrophils, because of the repeated infections, and
when these cells break down, they release their DNA along with other cellular
constituents. The very long DNA molecules are highly viscous, and dornase alfa snips
them up into much smaller segments, thinning the mucus and facilitating clearance.
Fig, 8.6A shows sputum from a patient with cystic fibrosis stained for DNA. The
elongated DNA molecules are clearly seen. Fig. 8.6B shows the effect of dornase alfa:
the DNA polymers have been effectively degraded, greatly reducing the viscosity of the
sputum.
Mannitol
Inhaled mannitol powder is thought to liquefy mucus by generating an osmotic gradient
between the mucus layer and the epithelial cells lining the airway. Mannitol is not
absorbed and increases the osmolarity of the mucus. This osmotic gradient pulls water
from the airway tissues into the mucus, hydrating it. This is also the basis of its activity
as an osmotic diuretic (p. 174) and in reducing cerebral oedema by pulling water from
brain tissue into the bloodstream. It can cause airway irritation, cough, haemoptysis,
and sore throat.
FIG. 8.5 The epithelial lining of the trachea and large
airways. From Waugh A and Grant A (2018) Ross & Wilson anatomy
and physiology in health and illness, 13th ed, Fig 10.12. Oxford:
Elsevier.
Ivacaftor
Ivacaftor was licenced in 2012 for use in cystic fibrosis (see Focus on: Tailored
Genetic Therapy in Cystic Fibrosis box).
The gene that codes for the CFTR channel is found on chromosome 7, and it is a
large gene, increasing the number of possible mutations that may occur within its
genetic sequence. In fact, there are around 2000 different mutations in this gene,
each of which changes the code in a different way and so produces a wide range of
abnormal amino-acid sequences in the final CFTR protein. Different mutations
interfere with channel production or function at different stages in the process, and
this opens the possibility of designing drugs to specifically target individual channel
abnormalities: a pharmacogenetic approach. The most common mutation in
American and northern European patients is called the F508del mutation and
involves the loss of only one amino acid from the CFTR protein, but even this slight
change means that the protein cannot fold into the correct shape, and the cell destroys
it instead of trafficking it to the cell surface. Nearly 90% of patients with CF have at
least one copy of this common mutation. Other mutations produce channels that
cannot open correctly or do not stay open long enough, or lead to reduced production
of the CFTR protein.
CFTR Modulators
These new agents are low-molecular-weight molecules which target a specific defect
in the CFTR channel and improve its function. Re-establishing correct chloride flow
rehydrates mucus, improves pulmonary clearance, and reduces the likelihood of
obstruction in other systems including the gastrointestinal tract. Identifying the
specific mutation in the patient’s CFTR gene identifies the nature of the defect in the
CFTR protein, and an appropriate CFTR regulator can be prescribed to improve
channel function.
There are three types of CFTR modulators: potentiators, correctors, and amplifiers.
Not all are available in all countries.
• Potentiators, e.g. ivacaftor, increase the length of time that the CFTR channel
remains open, allowing movement of additional chloride.
• Correctors, e.g. lumacaftor and elexacaftor (the latter not currently available
in the UK), allow the CFTR protein to assume the right shape for it to be
trafficked to the cell surface and incorporated into the cell membrane, increasing
the number of channels at the cell surface.
• Amplifiers increase production of CFTR protein and are currently in development.
Understanding the genetic basis of the faulty protein allows tailored therapy to be
offered to patients with certain mutations. For instance, ivacaftor alone is not
effective in patients with the common F508del mutation, because this mutation
results in unstable CFTR protein that is quickly broken down and so little reaches the
cell surface to benefit from the action of ivacaftor. However, using ivacaftor and
lumacaftor together can significantly improve pulmonary function, because
lumacaftor increases the amount of CFTR protein reaching the cell surface, and
ivacaftor keeps the new channels open for longer.
Pharmacokinetics and Adverse Effects
The currently available CFTR modulators are given orally and may be given in
combination: ivacaftor and lumacaftor are used together in the UK and other
combinations are used in other countries. Ivacaftor has a half-life of around 12
hours following oral administration, and its absorption is enhanced if taken with fat-
containing foods. Regular monitoring of liver enzyme levels is recommended because
it can increase liver transaminase levels. It is metabolised by the cytochrome (CYP) 3A
family of liver enzymes, and so interacts with drugs that induce or inhibit these
enzymes. For example, a range of antimicrobials including the azole antifungals
and macrolide antibiotics, which are CYP3A inhibitors, can significantly increase
ivacaftor blood levels.
Cough
Coughing is an essential reflex mechanism for clearing the airways of accumulated
mucus, accidentally inhaled materials, or other obstructions. It is controlled by the
cough centre in the brain stem. Irritation of the respiratory epithelium triggers sensory
impulses that travel to the cough centre via the vagus nerve. The cough centre responds
by activating the respiratory and abdominal muscles to produce the sequence of events
of cough. There is an initial deep inspiration, to bring enough air into the lungs for an
effective cough. The vocal cords in the larynx are then pulled together (closing the
glottis), and the abdominal muscles, diaphragm, and intercostal muscles contract
sharply. This generates a rapid rise in intrathoracic pressure. Then the glottis opens, and
the high intrathoracic pressure generates fast, high-volume airflow out of the airways. A
persistent cough should always be investigated, and the cause identified and treated
where possible.
Antitussives
Antitussives are drugs that suppresses cough and are used to relieve the discomfort and
disturbance caused by persistent dry cough. The cough reflex is suppressed by central
nervous system (CNS) depressants, including alcohol and sedating antihistamines.
This can be clinically helpful, e.g. to treat a dry cough that disturbs sleep, but the
potential for compromising the airway in vulnerable individuals by interfering with this
important protective reflex should always be considered. General anaesthetics
depress cough and can compromise the airway during induction of and recovery from
anaesthesia unless the airway is carefully protected. Antitussives are not used when the
cough is productive, because they predispose to mucus accumulation in the airways and
increase the risk of obstruction and infection.
Opioids
Opioids (Ch. 6) are the most effective antitussive medications currently available and
suppress cough at sub-analgesic doses. They act on μ (mu) receptors in the cough centre
in the brainstem and reduce their sensitivity. Weak opioids such as codeine and
pholcodine have fewer side-effects and are less likely to cause dependence than more
powerful opioids like morphine. However, morphine and diamorphine are used in
palliative care in patients with lung cancer, in whom a persistent, exhausting, and
distressing cough is a frequent symptom. Side-effects include constipation and
respiratory depression. Opioids also thicken sputum and depress ciliary action, and so
care is needed, for example, in chronic obstructive pulmonary disease (COPD).
Asthma
Asthma is characterised by reversible airway obstruction, with periods of good lung
function interspersed with episodes of deteriorating function, airway narrowing, and
airflow obstruction. It is frequently associated with allergy, which stimulates mast cells
and basophils in the airway to release a range of inflammatory mediators and cytokines,
including leukotrienes, platelet-activating factor, and interleukins (ILs). Fig. 8.9 shows
the main inflammatory mediators and cytokines involved, and also shows the targets of
the main drugs used in asthma. The smooth muscle tissue in the bronchial walls is
thickened and hyperreactive and responds inappropriately with exaggerated
bronchoconstriction to a range of triggers. The smaller airways, whose walls contain a
lot of smooth muscle and have no cartilage to keep them open, are disproportionately
affected. Bronchodilators, including β2-agonists, leukotriene-receptor
antagonists, and theophylline help reverse bronchoconstriction and improve
airflow.
Inflammation is a key feature, and the airway wall is swollen and infiltrated with a
range of inflammatory and immune cells including macrophages, mast cells,
eosinophils, and T-lymphocytes. Activated T-lymphocytes release a wide range of pro-
inflammatory cytokines, including Interleukins. The ongoing inflammatory response
damages the epithelium, causing patchy desquamation and exposing the basement
membrane, which contributes to the ongoing airway hypersensitivity. Goblet cells are
hypertrophic, and mucus production increases. Loss of the ciliated epithelium decreases
mucus clearance, so mucus accumulates and plugs airways. With time, the chronic
inflammation leads to structural changes and permanent remodelling of the airway wall.
Glucocorticoids are the mainstay of anti-inflammatory treatment in asthma. Fig.
8.10 shows the main pathological features of an asthmatic airway compared with
normal.
FIG. 8.9 The main inflammatory mediators and cytokines involved
in asthmatic airway inflammation.The sites of action of the main
drugs used to treat asthma are also shown. Modified from Abbas AK,
Lichtman AH, and Pillai S (2021) Cellular and molecular
immunology SAE, 10th ed, Fig. 20.10. New Delhi: Elsevier.
The cause of asthma is not known, although it is likely to be multifactorial. There is
often an inherited component with a strong familial tendency. Allergy is a common and
important component in asthma, and asthma often co-exists with other allergic
conditions such as hay fever and eczema.
Bronchodilators
These drugs reduce airflow obstruction by relaxing bronchial smooth muscle and
opening the airways. They are usually used to relieve symptoms, although sometimes
they are useful prophylactically, for example, before exercise if exercise is a trigger
factor. Because of this, they are sometimes called relievers or rescue agents.
b2-Agonists
Examples: salbutamol, terbutaline, salmeterol
The use of sympathetic drugs to improve symptoms in asthma dates to 3000 BC,
when physicians in ancient China used extracts of the plant Ephedra equisetina to treat
dyspnoea. The active agent, epinephrine (adrenaline), was isolated in 1897, and the
first β-agonist drug, isoprenaline, was synthesised in 1940. It was used in the
treatment of asthma, but it is not selective for β2-receptors and has significant activity
on β1-receptors too, including those in the heart (see Fig. 4.5). This meant that even
when used in the relatively small doses needed by inhalation to improve asthma
symptoms, enough was absorbed into the systemic circulation to cause unwanted
cardiovascular side-effects including hypertension and arrhythmias. The discovery of β1
and β2 subtypes and their distribution in the respiratory and cardiovascular systems led
to the development of drugs relatively selective for β2-receptors, which cause
bronchodilation with minimal activity on the heart. The first β2-selective agonist,
salbutamol, is 29 times more active on the β2-receptor than the β1-receptor and was
introduced in the late 1960s. Since then, a range of similar agents have been produced
including the longer-acting agents formoterol and salmeterol. β2-agonists bind to
and activate sympathetic β2-receptors on smooth muscle cells in the airway wall. They
mimic the action of the sympathetic nervous system, which prepares the body for stress
or activity, and relax bronchial smooth muscle by reducing calcium entry into the
smooth muscle cells. Because calcium is essential for activation of the contractile
proteins in the muscle cell, this leads to bronchodilation, increases air flow, and
maximises oxygen intake. In addition, they improve ciliary function and have an anti-
inflammatory action. β2-agonists and glucocorticoids have a synergistic effect (see later
and Fig. 8.11).
Pharmacokinetics
β2-agonists are given by inhalation for occasional use in well-controlled asthma and may
also be given orally or by intramuscular or intravenous injection if required. Short-
acting agents, e.g. salbutamol and terbutaline, usually take effect within 5 minutes of
inhalation, and their duration of action is usually between 4 and 6 hours. Salmeterol
has a duration of action of around 12 hours, so is useful for managing night-time
symptoms and takes up to half an hour to begin working. Formoterol’s duration of
action can be up to 24 hours, and its onset of action is within 2–3 minutes. Overuse of
these drugs, however, downregulates β2-receptor numbers and reduces their
effectiveness.
Long-acting agents have been associated with increased mortality rates in severe
asthma; the reasons for this are unclear. Current advice is that they should only be used
in conjunction with an anti-inflammatory steroid and should only be added into therapy
if adequate control is not achieved with shorter-acting agents. Patients taking these
agents should be reviewed regularly and the drug withdrawn if no clinical benefit is
observed.
Adverse Effects
Significant systemic side-effects can occur if circulating drug levels are high enough,
including from high doses of inhaled drug. At higher doses, β2-agonists also activate β1-
receptors: i.e. their selectivity at the subtypes of the β-receptor is dose-dependent. This
includes cardiac β1-receptors, leading to tachycardia, palpitations, hypertension, and
arrhythmias. A fine tremor can occur, due to stimulation of β2-receptors on skeletal
muscle. They can cause hypokalaemia by shifting potassium from the blood into cells via
an action on the sodium–potassium pump in the cell membrane. Hypokalaemia in turn
can cause potentially fatal cardiac arrhythmias, and care should be taken in people who
are at risk of hypokalaemia, including concurrent use of theophylline in severe
asthma. Because they reduce blood potassium levels, these drugs are also used to treat
hyperkalaemia. They may cause hyperthyroidism because sympathetic stimulation
increases release of thyroid hormone from the thyroid gland. They increase blood
glucose levels, because sympathetic stimulation increases glycogen breakdown in the
liver, releasing glucose into the bloodstream, which may interfere with blood glucose
control in diabetes.
Theophylline
Theophylline is a methylxanthine, a family of chemicals that includes caffeine; strong
coffee was recommended in 1786 for asthma in a letter written by William Withering (of
digoxin fame, p. 133). As a group, xanthines have bronchodilator, diuretic, and anti-
inflammatory properties, although their exact mechanism of action is unclear. They act
at several targets in human cells and probably produce their pharmacological action via
more than one pathway. One potential mechanism of action is via their blockade of
adenosine receptors on airway smooth muscle cells. Adenosine triggers
bronchoconstriction, so antagonism at their receptor may account at least in part for the
bronchodilator action of the xanthines. In addition, they inhibit one form of the enzyme
phosphodiesterase (PDE), which breaks down cyclic AMP (cAMP) inside cells, including
smooth muscle cells. This increases cAMP levels and relaxes bronchial smooth muscle,
inducing bronchodilation. Other types of PDE inhibitors are used to treat heart disease
(p. 133). They may also have a central effect and stimulate the respiratory centre in the
brainstem. Finally, the anti-inflammatory effect of xanthines may be due to their ability
to increase the activity of anti-inflammatory genes and suppress the activity of
inflammatory cells. Xanthines are usually used to manage acute, severe exacerbations of
asthma.
Pharmacokinetics
Theophylline is given orally. It is well but unpredictably absorbed from the GI tract,
giving fluctuating plasma levels which could increase the risk of side-effects; modified
release preparations are preferred, which are absorbed at a steadier rate and help to
stabilise plasma levels over extended periods. Aminophylline is a preparation of
theophylline and ethylendiamine. Ethylenediamine has no pharmacological activity,
but it makes theophylline much more soluble in water, giving a formulation suitable for
intravenous injection. Once in the body, it releases theophylline, which is responsible for
its pharmacological effects. The plasma half-life of theophylline is 4–8 hours, and it is
heavily plasma protein-bound.
Adverse Effects
Theophylline has a wide range of potentially serious side-effects because it affects such a
wide range of cellular targets important in a range of body tissues. It has a narrow
therapeutic index, and side-effects become increasingly common at plasma levels at the
upper end of the therapeutic range and above. Therapeutic drug monitoring is helpful in
managing dosing. Adverse effects include cardiac arrhythmias, probably due to
adenosine antagonism and seizures due to CNS stimulation, both of which can be fatal.
Gastric acid secretion is increased and so is GI motility, with GI pain, gastro-
oesophageal reflux, diarrhoea, and increased risk of peptic ulcers. Hypokalaemia can
occur, especially when theophylline is used with β2-agonists. Theophylline is
metabolised in the liver, and its effects are decreased in the presence of enzyme
inducers, including smoking, alcohol, phenytoin, and St. John’s wort. Enzyme
inhibitors can reduce theophylline clearance and increase circulating levels: these
include isoniazid, interferons, and carbamazepine.
Antimuscarinic Bronchodilators
Examples: aclidinium, ipratropium, tiotropium
The parasympathetic nervous system causes bronchoconstriction via the action of
acetylcholine on M3 receptors in bronchial smooth muscle (see Fig. 4.8). Drugs that
block M3 receptors therefore cause bronchodilation and are used to relieve
bronchospasm in obstructive airway disease, including asthma and COPD.
Pharmacokinetics
Ipratropium is given by inhalation in maintenance treatment of asthma and in the
management of acute asthma. Its peak action occurs 30–60 minutes after
administration, and it is effective for 3–6 hours. It is also given intranasally to reduce
rhinorrhoea, for example, in hay fever. It is a large, highly charged molecule, so is poorly
absorbed into the bloodstream, and so is generally not associated with systemic side-
effects, although care must be taken not to contaminate the eye directly because of the
risk of glaucoma (see below). Longer-acting agents include aclidinium (duration of
action, 8 hours) and tiotropium (64 hours).
Adverse Effects
Antimuscarinic drugs can cause a range of systemic antimuscarinic side-effects (see p.
51) by blocking muscarinic receptors throughout the body and antagonising the ‘rest and
digest’ functions of the parasympathetic nervous system. Care is needed in people with
glaucoma or at risk of glaucoma because these drugs can increase intraocular pressure
(see Fig. 4.11). Antimuscarinic agents can also block the effects of the parasympathetic
nervous system on the heart and predispose to tachycardia and arrhythmias: the risk is
increased if antimuscarinics are used in conjunction with β-agonists.
Anti-Inflammatory Drugs
Preventing or reversing the inflammatory changes associated with the asthmatic airway
alleviates the signs and symptoms of the disorder and improves airway function. They
should be used on a regular basis even when the asthma is not troublesome to suppress
airway inflammation, and so they are sometimes referred to as preventers.
Glucocorticoids
Examples: budesonide, beclometasone, fluticasone
Glucocorticoids are the mainstay of anti-inflammatory treatment in asthma. They are
superior to other anti-inflammatory medications in the management of recurrent and
chronic disease and prevent or reduce the rate of asthma exacerbations. They block the
activity and proliferation of inflammatory cells in the airway wall and prevent the
production of a wide range of inflammatory mediators and cytokines. Their mechanism
of action is discussed on p. 111. Because they can cause a wide range of potentially severe
side-effects, they should be used at the lowest effective dose and given systemically only
when required to manage severe disease and for as short a period as possible.
Glucocorticoids and β2-agonists demonstrate therapeutic synergism when used
together in asthma. Glucocorticoids increase the number and efficacy of β2-receptors in
the airways, and therefore improve the bronchodilator response to β2-receptor agonists.
β2-agonists in turn have an anti-inflammatory action and enhance the airway response
to glucocorticoids (Fig. 8.11).
Pharmacokinetics
Maintenance glucocorticoid therapy is delivered by inhalation wherever possible to
minimise side-effects. However, because steroid molecules are small and fat-soluble,
they are readily absorbed across the airway wall and can reach physiologically active
levels in the bloodstream if used in high doses to manage severe asthma or used
excessively. For this reason, it is desirable that they are quickly cleared from the
circulation by hepatic metabolism. Bioavailability and half-lives of the commonly used
steroids vary significantly. Fluticasone, budesonide, mometasone, and
ciclesonide are quickly removed from the bloodstream by first-pass metabolism, and
any swallowed drug from an inhaled dose is rapidly cleared. Beclometasone is
metabolised much more slowly and so has a longer plasma half-life. Liver impairment
can therefore increase circulating levels of any inhaled glucocorticoid. Water-soluble
drugs, for example budesonide, dissolve in the watery mucus lining the airways and
are more efficiently cleared by the mucociliary escalator. More lipid-soluble drugs,
including fluticasone and beclomethasone, enter the airway tissues and remain
there for extended periods, extending the duration of their activity.
Adverse Effects
Local side-effects from deposition of inhaled glucocorticoid in the mouth and throat
include hoarseness from the build-up of drug on the vocal cords, and oral thrush
because glucocorticoids suppress local immune defences. It is good practice to rinse the
mouth and/or brush the teeth after inhalation. If the drug reaches physiologically active
levels in the circulation, either because of high inhaled doses or oral/parenteral use in
severe disease, any of the wide range of side-effects may be seen, including growth
suppression in children, osteoporosis, cataract, and adrenal suppression (Cushing’s
syndrome, see Fig. 6.16).
Cromones
Cromones (nedocromil sodium and sodium cromoglicate) are add-on options in
the treatment of asthma and are used as antiallergy medications in some other
situations, e.g. food allergy. They are not helpful in acute asthma, but they may benefit
some patients, particularly children, in maintenance therapy. Their anti-inflammatory
action is less effective than glucocorticoids, and their mechanism of action is not fully
understood. They stabilise mast cells and reduce mast cell release of inflammatory
mediators including histamine, which may be their main mechanism of action in
allergy treatment. They also suppress the activity of a range of inflammatory cells
including neutrophils, eosinophils, and macrophages and reduce the release of a range
of inflammatory mediators and cytokines. Nedocromil sodium is given only by
inhaler, because it is a large, electrically charged molecule not absorbed across the wall
of the GI tract, but sodium cromoglicate can be given orally or by inhaler.
Lipoxygenase Inhibitors
These agents are not available in the UK but are used in other countries including the
US and India. Zileuton was the first agent approved for the treatment of asthma and is
given orally. Its most significant side-effect is elevation of liver enzymes.
Immunosuppressants
The use of monoclonal antibodies against the cells and mediators involved in the
pathophysiology of asthma is the most recent development in asthma treatment. They
are used under specialist care in severe asthma only because suppression of the immune
and inflammatory responses can cause serious side-effects.
Omalizumab
Omalizumab is an antibody to IgE, the immunoglobulin associated with allergy. In
severe allergic asthma, IgE triggers and promotes airway inflammation. By binding to
IgE in the blood and in body tissues, omalizumab prevents it from binding to mast cells
and triggering histamine release and the resultant allergic response. It is therefore used
in severe persistent allergic asthma, as well as certain other allergic conditions. It is
given by subcutaneous injection and the dose is calculated according to the patient’s IgE
levels and bodyweight. It is generally better tolerated than other biologics, although it
can cause a range of side-effects including skin reactions and has been associated with
Churg–Strauss syndrome (see above).
FIG. 8.12 Biosynthesis and activity of the leukotrienes.
Benralizumab
IL-5 is an important inflammatory mediator in asthma. It is released by T-lymphocytes
in the asthmatic airway and binds to its receptors on other inflammatory cells including
eosinophils, which are of particular importance in allergic asthma. IL-5 promotes
eosinophil maturation, activity, and survival, and therefore enhances the allergic
inflammatory reaction. Benralizumab is an antibody to the IL-5 receptor. By blocking
IL-5 receptors on eosinophil cell membranes, benralizumab reduces eosinophil activity
and survival, depletes eosinophil numbers, and suppresses allergic inflammation (Fig.
8.13). Because eosinophils are an important part of the body’s defence against
helminthic infections, it is important to treat any such infection before beginning
benralizumab treatment in asthma.
Bronchodilators
β2-agonists and antimuscarinic agents (see above) may both help relieve
bronchospasm, especially in acute exacerbations, although in general they are less
helpful in COPD than in asthma.
Glucocorticoids
Generally, the inflammatory changes in COPD airways respond poorly to inhaled
steroids, and if these drugs are tried in a patient, the response should be monitored and
the drug withdrawn if they do not help.
Phosphodiesterase Inhibitors
Roflumilast is a PDE inhibitor specific for PDE4, the form of the enzyme found in the
lung. PDE breaks down the important intracellular messenger cAMP. Inhibition of the
enzyme
increases cAMP levels in lung tissues, and this has a range of anti-inflammatory actions,
including preventing neutrophil accumulation of neutrophils and eosinophils in the
airway wall, reducing the inflammatory action of the inflammatory cells present, and
improving the survival of the ciliated epithelium lining the airway. Roflumilast is given
orally and is a pro-drug, converted to its active form by liver enzymes. This means that
any change in liver enzyme activity, either by other drugs that induce or inhibit them or
in liver impairment can affect its action. It should be avoided in moderate to severe liver
impairment and with inducers including rifampicin and a range of anticonvulsants.
References
1. Borghardt J.M, Kloft C, Sharma A. Inhaled therapy in respiratory disease: the
complex interplay of pulmonary kinetic processes. Can. Respir. J. J. Can. Thorac.
Soc. 2018;2018:2732017.
2. Chauhan B.F, Ducharme F.M. Anti-leukotriene agents compared to inhaled
corticosteroids in the management of recurrent and/or chronic asthma in adults
and children. Cochrane Database Syst. Rev. 2012;2014(5):CD002314.
3. Derichs N. Targeting a genetic defect: cystic fibrosis transmembrane conductance
regulator modulators in cystic fibrosis. Eur. Respir. Rev. 2013;22(127):58–65.
4. Hatipoglu M.K, Hickey A.J, Garcia-Contreras L. Pharmacokinetics and
pharmacodynamics of high doses of inhaled dry powder drugs. Int. J.
Pharm. 2018;549(1–2):306–316.
5. Matera M.G, Rinaldi B, Calzetta L, et al. Pharmacokinetics and
pharmacodynamics of inhaled corticosteroids for asthma treatment. Pulm.
Pharmacol. Therapeut. 2019;58:101828.
6. O’Reilly R, Elphick H.E. Development, clinical utility and place of ivacaftor in the
treatment of cystic fibrosis. Drug Des. Dev. Ther. 2013;7:929–937.
Online resources
Camus P. Pneumtox online Available
at:. https://www.pneumotox.com/drug/index/, 2024.
Condren M.E, Bradshaw M.D. Ivacaftor: a novel gene-based therapeutic approach
for cystic fibrosis. J. Pediatr. Pharmacol. Therapeut. 2013;18(1):8–13 Available
at: http://doi.org/10.5863%2F1551-6776-18.1.8.
Scottish Intercollegiate Guidelines Network, . British guideline on the management
of asthma Available at:. https://www.sign.ac.uk/sign-158-british-
guideline-on-the-management-of-asthma, 2024.
9: Renal and Genitourinary Drugs
CHAPTE R OU TLINE
Urine Formation
Diuretics
Osmotic Diuretics
Loop Diuretics
Thiazide Diuretics
Potassium-Sparing Diuretics
Carbonic Anhydrase Inhibitors
Male Sexual Function
Erectile Dysfunction
Phosphodiesterase Inhibitors
Alprostadil
The Nephron
Each nephron is essentially a long, folded tubule, with an expanded end called the
glomerular capsule and the other end opening into a network of drainage tubes called
collecting ducts (CDs), which drain urine towards the renal pelvis. Fig. 9.2 shows the
structure of a typical nephron.
Urine Formation
Key Points
Three processes contribute to the formation of urine (Fig. 9.4).
• Filtration: Water and small molecules are filtered out of the glomerular capillaries
into the glomerular capsule, forming filtrate.
• Reabsorption: As the filtrate passes through the nephron, water, organic
nutrients, electrolytes, and other key substances are reabsorbed from the filtrate
back into the bloodstream.
• Secretion: Unwanted materials, including drugs, hydrogen ions, and other waste
products, are actively pumped out of the bloodstream and into the filtrate for
excretion.
Fig. 9.5 shows the main sites of sodium (Na+), chloride (Cl-), and potassium (K+)
reabsorption in the nephron, and also indicates the region of the nephron where the
main diuretics exert their effect.
Diuretics
Diuretics increase urine production and promote the loss of water and electrolytes,
mainly Na+, from the body. Most work by a direct action on tubular Na+ transport
mechanisms and increase the Na+ content of the filtrate, increasing its osmolarity,
which in turn reduces water reabsorption and increases urine volume. In addition,
diuretics may affect renal handling of other electrolytes because many of the transport
mechanisms in the renal tubule exchange or co-transport more than one ion. Diuretic
therapy can therefore lead to excessive or deficient levels of other electrolytes, most
significantly K+, Cl-, and Ca2+.
FIG. 9.4 Filtration, reabsorption, and secretion in urine
formation.The green arrow represents filtration from the glomerular
capillaries. The turquoise arrows represent reabsorption of
substances from the filtrate into the bloodstream. The purple arrows
represent secretion of unwanted substances from the bloodstream
into the filtrate. Modified from Patton KT, Bell FB, Thompson T,
et al. (2022) Anatomy & physiology, 11th ed, Fig. 42.17. St. Louis:
Elsevier.
Most diuretics must be present in the filtrate in order to work because most target a
Na+-transport mechanism on the epithelium lining the tubule. However, most are
heavily plasma protein-bound, and so are too large to be filtered in the glomerulus and
can only reach the filtrate by active secretion into the tubule by transporters in the PCT
(Fig. 9.6). These transporters secrete a range of substances into the filtrate and have a
finite working capacity. This means that once the diuretic drug plasma concentration is
high enough to saturate these transporters, no additional drug will reach the filtrate, and
increasing the drug dose will have no further therapeutic effect. It also means that if
there is another drug in the bloodstream which is also secreted into the filtrate using the
same transporters, the two drugs will compete at the transporter and reduced levels of
the diuretic may reach the filtrate. This can reduce diuretic action.
Diuretics are useful in cardiovascular disease to reduce circulating blood volume and
blood pressure and are used to reduce fluid load in situations associated with oedema or
fluid retention, like raised intracranial pressure, pulmonary oedema, liver disease, and
nephrotic syndrome. However, in conditions that reduce renal blood flow, e.g. heart
failure, drug delivery to the kidney is reduced and this can reduce drug effectiveness.
Osmotic Diuretics
Example: mannitol
Osmotic diuretics do not directly interfere with renal ion transport mechanisms, but
their presence in body fluids increases the osmotic pressure of that fluid and so
influences water movement between body compartments. The sugar mannitol is the
prototype agent in this group, but other substances with similar actions include urea
and isosorbide. High levels of other solutes in the urine, e.g. glucose, also produce an
osmotic diuresis.
FIG. 9.5 The main regions of the nephron where sodium, chloride,
and potassium are reabsorbed, and the sites of action of the main
groups of diuretic drugs. Modified from Ritter JM, Flower RJ,
Henderson G, et al. (2020) Rang & Dale’s pharmacology, 9th ed, Fig.
30.4. Oxford: Elsevier.
FIG. 9.6 Secretion of diuretics into the filtrate in the proximal
convoluted tubule.Diuretics are heavily plasma protein-bound and so
do not filter out of the blood in the glomerulus. Transporter
mechanisms in the wall of the proximal convoluted tubule actively
extract them from the bloodstream and pump them into the filtrate.
Once in the filtrate, they inhibit their target sodium-transport
mechanism.
FIG. 9.7 The mechanism of action of mannitol: osmotic diuresis.1.
Mannitol in the bloodstream pulls water out of the tissues. 2.
Increased blood volume increases filtration rates in the glomerulus.
3. Mannitol is filtered but not reabsorbed. 4. Mannitol increases the
osmotic pressure of the filtrate and pulls water from the bloodstream
into the nephron.
Adverse Effects
Initially, because mannitol in the blood draws fluid from the tissues into the circulation
and increases circulating blood volume, it can precipitate heart failure. Once diuresis
commences, however, there can be hypovolaemia and hypotension, and pre-existing
heart failure may be exacerbated by depleting circulating blood volume. Cardiovascular
function should therefore be assessed before beginning treatment.
Loop Diuretics
Examples: bumetanide, ethacrynic acid, furosemide, torasemide
Loop diuretics are the most potent diuretics available because they inhibit the
Na+/K+/Cl- co-transport complex in the ascending limb of the loop of the nephron (Fig.
9.8), which normally reabsorbs around 25% of the filtrate’s Na+ content. This complex
pumps Na+, K+, and Cl- from the filtrate into the epithelial cells of the nephron, and the
ions are then pumped out the other side of the cells into the bloodstream, taking water
with them. Loop diuretics bind to the Cl--transporting unit of the complex but are too
large to travel through the transporter, and so they block it. This increases Na+ and Cl-
loss in the urine, which increases its osmolarity and reduces water reabsorption. Loop
diuretics have vasodilator activity, which may be helpful in certain circumstances: for
example, in heart failure because it reduces afterload, and in hypertension because it
reduces blood pressure. In general, however, loop diuretics are not the optimal choice in
hypertension because their duration of action is too short and the diuresis they produce
is excessive. They are usually the diuretic of choice to relieve pulmonary oedema and
fluid retention secondary to heart failure and in renal disease.
FIG. 9.8 The sodium/potassium/chloride co-transporter complex
of the ascending limb of the loop of the nephron. Modified from
Shanbhag TV, Shenoy S, and Nayak V (2021) Pharmacology for
dentistry, 4th ed, Fig. 5.3. New Delhi: Elsevier India.
Pharmacokinetics
Bumetanide and torasemide are better and more reliably absorbed when given
orally than furosemide, and all can be given intravenously if rapid diuresis is needed;
intravenous administration gives peak effect within half an hour. Furosemide’s
plasma half-life is 1.5–2 hours, and with oral administration its effects last around 6
hours; about 50% is metabolised in the kidney, and the remainder is excreted
unchanged. Bumetanide has a plasma half-life of 1 hour and is metabolised by the
liver and kidney. Torasemide has a half-life of 3–4 hours and is mainly metabolised in
the liver. Loop diuretics are tightly bound to plasma albumin and so are not filtered in
the glomerulus, but reach the filtrate by secretion in the PCT; if this is compromised by
poor renal function, diuretic action can be reduced. Several drugs, including non-
steroidal anti-inflammatory drugs and some antimicrobials, inhibit the
transporters that secrete loop diuretics into the filtrate and so impair their actions.
Adverse Effects
As with any diuretic, there may be urinary frequency and urgency, nocturia, and
incontinence. Hypokalaemia is a common occurrence, because blocking Na+
reabsorption in the loop means that the filtrate arriving in the DCT contains high Na+
levels; this stimulates renin release, which in turn stimulates aldosterone secretion from
the adrenal cortex (the RAAS, see Fig. 7.15). This increases activity of the DCT’s Na+/K+
pumps, which reabsorb Na+ in exchange for K+, and K+ is lost in the urine.
Hypokalaemia is potentially dangerous because it can predispose to cardiac
arrhythmias. Diuretic-induced hypokalaemia increases the arrhythmogenic action of
certain anti-arrhythmic drugs, e.g. digoxin, a clinically important interaction. Other
electrolyte imbalances include hyponatraemia and hypomagnesaemia. Ca2+ excretion is
increased, and loop diuretics may be used to bring plasma Ca2+ down in
hypercalcaemia. Uric acid secretion is impaired, and blood uric acid levels rise. The loop
diuretics, especially furosemide, can produce dose-related ototoxicity, giving deafness,
tinnitus, and vertigo, because they affect the delicate hair cells of the inner ear
responsible for hearing and balance; function usually returns to normal when the drug
is withdrawn.
Excessive water and Na+ loss can reduce the circulating blood volume and cause
hypotension. If renal blood flow falls, renal function can be impaired.
Thiazide Diuretics
Examples: chlorothiazide, bendrofluazide, hydrochlorothiazide; chlortalidone,
indapamide, and metolazone have thiazide-like activity but are not structurally related
to thiazides.
The original drug in this group, chlorothiazide, was first introduced to the market
in 1957, and the agent most commonly used today, bendrofluazide, was introduced in
1960. Thiazides are used mainly in hypertension and heart failure. They are heavily
plasma protein-bound, and so are not filtered in the glomerulus and reach the filtrate
via active secretion in the PCT (Fig. 9.6). Thiazides inhibit the Na+ pump in the DCT,
reducing Na+ reabsorption and therefore increasing Na+ and water excretion. This has a
knock-on effect on Ca2+ excretion. Blocking Na+ reabsorption in the tubular epithelial
cell reduces Na+ levels in the cell. This activates a Na+/Ca2+ transporter in the
basolateral membrane which exchanges Na+ for Ca2+. When activated, this transporter
imports Na+ into the cell in exchange for Ca2+, which is transported into the
bloodstream (Fig. 9.9). Thiazide-induced reduction in Na+ reabsorption therefore
reduces urinary Ca2+ and increases circulating blood Ca2+ levels: this is why thiazides
are sometimes used to reduce Ca2+ excretion (and therefore urinary Ca2+ concentration)
in people prone to urinary stones. Elevating blood Ca2+ can also be helpful in people at
risk of osteoporosis. In long-term use, they have a vasodilator effect, which contributes
significantly to their therapeutic use in hypertension.
Pharmacokinetics
Thiazides are less potent than loop diuretics and have a slower onset of action, but their
duration of action is longer. For example, bendrofluazide has a plasma half-life of 3–4
hours but is active over 12–24 hours.
Adverse Effects
As with any diuretic, there may be urinary frequency and urgency, nocturia, and
incontinence. There is a significant risk of hypokalaemia because the filtrate arriving in
the distal section of the DCT and the CDs contains high Na+ levels; this stimulates the
RAAS as described for the loop diuretics above, which increases Na+ reabsorption at the
expense of K+, which is lost in the urine. Electrolyte imbalances including hypokalaemia
and hyponatraemia are more frequent with thiazides than loop diuretics because
thiazides have a considerably longer duration of action. In higher doses, thiazides can
cause erectile dysfunction, which is reversible on stopping the drug; sometimes reducing
the dose can resolve the problem. They compete with uric acid for the transporter which
secretes them into the tubule, so they increase blood levels of uric acid; for this reason,
they should be used with caution in people with, or at risk of, gout.
Thiazides impair glucose tolerance and can cause hyperglycaemia, thought to be
secondary to thiazide-induced hypokalaemia. Low blood K+ levels reduce the ability of
pancreatic β-cells to release insulin in response to falling blood glucose levels. As a
result, blood glucose rises. Thiazides should therefore be used with care in diabetes.
FIG. 9.9 Thiazides increase sodium loss in the urine and calcium
retention.1. Thiazides inhibit the sodium/chloride pump in the distal
tubule, increasing the loss of these ions and water in the urine. 2.
Falling sodium concentrations in the cell activate a sodium-calcium
pump on the basolateral membrane, which imports sodium from the
blood in exchange for calcium. Blood calcium levels therefore
rise. Modified from Hemmings H and Egan T (2013) Pharmacology
and physiology for anesthesia, Fig. 34.8. Philadelphia: Saunders.
Potassium-Sparing Diuretics
Examples: amiloride, eplerenone, spironolactone, triamterene
K+-sparing diuretics act at the distal portion of the DCT and CD, the walls of which
contain pumps and channels that reabsorb Na+ and Cl- in exchange for K+. They are
weak diuretics because most electrolyte reabsorption has already taken place: the DCT
and CD are the sites of any final adjustments to Na+ reabsorption, and relatively small
quantities of Na+ are reabsorbed here. However, they may be usefully combined with a
loop or thiazide diuretic to reduce the risk of hypokalaemia. Although all the K+-sparing
diuretics block this mechanism, preventing Na+ reabsorption and so retaining K+, their
mechanisms of action are slightly different.
Spironolactone and eplerenone are aldosterone antagonists. Aldosterone, the
main mineralocorticoid from the adrenal cortex, binds to DNA in the tubular cell and
increases production of Na+ pumps and channels in the DCT and CD and so enhances
Na+ reabsorption, with water following passively (Fig. 9.10A). Spironolactone prevents
aldosterone from binding to its receptor and so reduces Na+ pump and channel pump
production. With fewer pumps and channels available, less Na+ is reabsorbed,
increasing Na+ and water loss in the urine (Fig. 9.10B). Because Na+ reabsorption
increases K+ loss, the diuresis and increased Na+ loss seen with spironolactone is
accompanied by K+ retention.
Amiloride and triamterene bind directly to Na+ channels in the DCT and CD,
preventing Na+ reabsorption, increasing urinary Na+ and water loss, and retaining K+.
Pharmacokinetics
These drugs are given orally and are generally well absorbed with the exception of
amiloride, whose oral bioavailability is only around 50%, and less if there is food in the
gastrointestinal (GI) tract. Spironolactone is a pro-drug and is metabolised in the wall
of the GI tract and the liver to the active metabolite canrenone (Fig. 9.10B), which
has a longer half-life than spironolactone (16 hours to spironolactone’s 90 minutes).
Both triamterene and amiloride are secreted into the tubule in the PCT. As with
steroid drugs in general, spironolactone and eplerenone can take several days to
achieve full therapeutic effect; triamterene and amiloride, on the other hand, have a
rapid onset of action.
FIG. 9.10 The mechanism of action of spironolactone.A. The
steroid hormone aldosterone enters the distal convoluted
tubule/collecting duct cells and binds to its receptor in the
cytoplasm. The hormone–receptor complex then binds to DNA and
stimulates production of sodium pump protein. This increases
sodium reabsorption and reduces sodium and water loss in the urine.
B. Spironolactone (as its active metabolite canrenone) binds to and
blocks aldosterone receptors. This prevents sodium pump synthesis.
Sodium and water loss in the urine therefore increases but without
additional potassium loss.
Adverse Effects
These drugs can cause hyperkalaemia, and care is needed if there is already a
predisposition to raised K+ levels, for example in renal failure or with co-administration
of β-blockers, angiotensin-converting enzyme inhibitors, or angiotensin-
receptor antagonists, all of which can increase plasma K+. The aldosterone-receptor
antagonists are structurally related to steroids (otherwise they would not be able to bind
to steroid receptors) and can block steroid receptors elsewhere in the body, including
oestrogen, progesterone, and testosterone receptors. Spironolactone is more likely
than eplenerone to cause such side-effects; it may cause gynaecomastia, menstrual
irregularities, and testicular atrophy. Due to its anti-androgen effect, spironolactone
can be used to treat hirsutism in women with polycystic ovary disease and male pattern
baldness.
Erectile Dysfunction
The inability to achieve or maintain an erection sufficient for sexual intercourse (erectile
dysfunction) is common, and prevalence increases with age. There is often a
psychological component, but a range of conditions that may interfere with vascular or
neurological function may be responsible. There is a strong correlation between erectile
dysfunction and cardiovascular disease, including ischaemic heart disease, stroke, and
hypertension. Other recognised risk factors include obesity, diabetes mellitus,
neurological disease such as multiple sclerosis, testosterone deficiency, and benign
prostatic hypertrophy. A wide range of drugs, including recreational substances such as
alcohol and marijuana, can cause erectile dysfunction: the most common include
benzodiazepines, selective serotonin re-uptake inhibitors, thiazide
diuretics, β-blockers, and tricyclic antidepressants.
Phosphodiesterase Inhibitors
Examples: avanafil, sildenafil, tadalafil
The phosphodiesterases (PDEs) are a family of enzymes which break down the
important intracellular signalling molecules cyclic AMP (cAMP) and cyclic guanosine
monophosphate (cGMP) (p.33). Sexual arousal increases the levels of nitric oxide in the
tissues of the penis, which activates guanylyl cyclase and increases cGMP production.
cGMP relaxes the smooth muscle in the erectile tissue of the corpora cavernosa and the
arteries that supply them, increasing blood flow into the penis and facilitating
engorgement and erection. PDE5 breaks cGMP down, reducing cGMP levels and so
reducing engorgement. Sildenafil and the other drugs in this category inhibit PDE5,
increasing cGMP levels, which increases blood flow into the penis and promotes and
prolongs erection (Fig. 9.13). Sildenafil and tadalafil are also used in pulmonary
hypertension: by relaxing pulmonary artery smooth muscle, these drugs reduce
pulmonary artery pressure.
In conjunction with non-pharmacological measures (e.g. reducing alcohol
consumption, stopping smoking, and weight loss if indicated), PDE inhibitors are the
mainstay of erectile dysfunction management.
Pharmacokinetics
Sildenafil is rapidly absorbed from the GI tract and has a duration of action of 3–5
hours, so a single dose must be taken within a relatively short timeframe in advance of
anticipated sexual activity. Tadalafil has a longer duration of action (17–24 hours) and
can be taken as a regular daily dose to allow for spontaneous activity and/or when
sexual activity is regular. PDE5 inhibitors are primarily metabolised by CYP3A4 enzymes
in the liver, which metabolise a range of other drugs. As a result, enzyme inhibitors such
as the azole antifungals (e.g. ketoconazole), Ca2+-channel blockers (e.g.
verapamil), grapefruit juice, and macrolide antibiotics (e.g. erythromycin) are
likely to increase PDE5 inhibitor levels and precipitate toxicity, and enzyme inducers
including some anticonvulsants (e.g. carbamazepine and phenytoin) and St.
John’s wort increase the clearance of PDE5 inhibitors and decrease their effectiveness.
Adverse Effects
Although the PDE5 inhibitors are relatively selective for this form of the enzyme, they
may also inhibit other members of the PDE family in other tissues, giving a range of
side-effects. PDE5 inhibitors relax vascular smooth muscle and can cause systemic
vasodilation, reducing blood pressure and affecting systemic haemodynamics. They
should therefore be used with care, if at all, in individuals with cardiovascular disease or
compromise, including hypotension, heart failure, a history of heart attack or stroke, or
ischaemic heart disease. In addition, they have clinically important interactions with
other hypotensive agents, including nitrates and α-blockers and can cause a
potentially dangerous fall in blood pressure. Rarely, priapism (sustained and painful
erection) occurs. By an action on PDE6 in the rods and cones of the retina, these drugs
can cause vision problems; this is more likely with high-dose sildenafil than with the
other PDE5 inhibitors.
Alprostadil
Natural prostaglandins (PGs) have very short half-lives, usually too brief to be clinically
useful, and so stable synthetic analogues are often more suitable. PGE is an important
vasodilator in normal physiological regulation of cardiovascular function, and the
synthetic PGE1 analogue alprostadil is used to relax penile arterial smooth muscle and
increase penile engorgement in the diagnosis and treatment of erectile dysfunction,
usually as a second-line treatment if a PDE5 inhibitor cannot be used because of side-
effects or a contra-indicating factor. Alprostadil can be applied topically to the penis tip,
given as an intra-urethral pellet, or injected directly into the corpus cavernosum. The
main side-effects include painful erection, and if enough drug escapes into the
circulation, there may be headache and dizziness from systemic vasodilation.
References
1. Koeppen B, Stanton B. Renal physiology. 6th ed. Elsevier: Oxford; 2018.
Online resources
Arumugham V.B, Shahin M.H. Therapeutic uses of diuretic agents. In: StatPearls
[Internet]. Treasure Island: StatPearls Publishing; 2021 Available
from:. https://www.ncbi.nlm.nih.gov/books/NBK557838/.
Ellison D.H. Clinical pharmacology in diuretic use. Clin. J. Am. Soc.
Nephrol. 2019;14(8):1248–1257 Available
at:. https://cjasn.asnjournals.org/content/14/8/1248.
10: Drugs and the Gastrointestinal
Tract
Nabilone
Other Drugs With Anti-Emetic Activity
F OCUS ON
The small intestine is coiled within the abdominal cavity, anchored to the posterior
abdominal wall by folds of the peritoneum, and opens into the large intestine (the colon)
at the ileocaecal valve. The colon arches over the small intestine and opens into the
rectum. The opening from the rectum to the exterior, the anus, is normally under
voluntary control in the adult and ensures faecal continence.
Histamine
Histamine is released by enterochromaffin-like (ECL) cells in the gastric mucosa. It is an
important stimulant of gastric acid secretion and binds to H2 receptors on parietal cells,
which in turn activates the proton pump and increases acid secretion. H2-receptor
blockers, e.g. cimetidine, are widely available to treat conditions associated with
excess acid.
Acetylcholine
ACh released by parasympathetic nerves supplying the stomach acts on M3 receptors on
parietal cells, activating the proton pump and increasing acid secretion. This promotes
digestive function when the body is resting, and parasympathetic activity (‘rest and
digest’) predominates over sympathetic (‘fight or flight’). Antimuscarinic drugs, e.g.
hyoscine and atropine, block M3 receptors and reduce gastric acid secretion as part of
their more widespread inhibitory effects on digestive function.
Gastrin
The hormone gastrin is released into the bloodstream by G cells in stomach glands in
response to stretching of the stomach wall and the presence of protein-rich foods in the
stomach. It does not directly stimulate parietal cells: instead, it binds to receptors on
ECL cells and increases histamine production, which in turn activates the proton pump.
Antacids
Antacids are weak bases, usually prepared as liquid formulations or chewable tablets,
which neutralise gastric acid. They are freely available over the counter and are widely
used on a non-prescription basis to self-manage upper GI tract disorders. The rise in
gastric pH also inhibits pepsin, which works best at pH 2–3.5. Antacids contain calcium,
magnesium, or aluminium salts, which react with and neutralise HCl, increasing gastric
pH. For example, calcium carbonate (CaCO3) reacts with HCl to form H2CO3 and
neutral calcium chloride (CaCl2) according to the following reaction:
H2CO3 quickly breaks down into water (H2O) and carbon dioxide (CO2) gas, which
causes the belching associated with CaCO3 use.
Antacids are best taken with food because gastric emptying is much faster on an
empty stomach, shortening the duration of action of the drug. Sometimes the thickening
agent alginic acid is used with an antacid to increase the viscosity of the gastric fluids
and help retain the drug in the stomach, and/or simeticone, an anti-foaming agent.
Adverse Effects
Taken occasionally and according to manufacturer’s instructions, antacids are generally
well tolerated. There may be significant absorption of calcium, magnesium, or
aluminium which can cause problems particularly in people with impaired renal
function who cannot clear the excess. Magnesium salts can cause diarrhoea and
aluminium salts can constipate, but using a combination can help minimise any
disruption to bowel function. Some antacid preparations contain significant amounts of
sodium and should be avoided in people on salt-restricted diets.
Antacids may increase or decrease absorption of a wide range of co-administered
drugs by several mechanisms (Fig. 10.6). Antacids may chelate other drugs in the GI
tract; that is, the metal ions in the antacid bind to them and form insoluble complexes,
reducing their bioavailability: for example, tetracycline, digoxin, and quinolone
antibiotics are chelated by antacids, significantly reducing absorption. Some drugs,
e.g. itraconazole, are formulated to be released from the drug preparation in the acid
environment of the stomach, and increasing gastric pH can reduce the amount of free
drug reaching the duodenum for absorption, reducing circulating drug levels and drug
efficacy. The coating of enteric-coated formulations is pH sensitive, formulated to
remain intact in the stomach and break down in the higher pH of the duodenum; if
gastric pH rises, this enteric coating may break down in the stomach, exposing acid-
sensitive drugs to gastric fluids and reducing the quantity of active drug reaching the
duodenum for absorption. On the other hand, the absorption of other drugs may be
facilitated by increasing gastric pH. Whenever possible, it is therefore generally advised
to separate administration times of antacids and other drugs.
Proton-Pump Inhibitors
Examples: esomeprazole, lansoprazole, omeprazole
The proton pump is the final common mechanism for all stimulants of gastric acid
production. The first PPI, omeprazole, was introduced in 1989 and is still in
widespread use today. Because these agents reduce gastric acid production irrespective
of the stimulus (histamine, alcohol, smoking, caffeine, etc.) and have proved to be well
tolerated and highly effective, they have become the mainstay of management in acid-
related disorders. Globally they are among the most widely consumed drugs and are
included on the World Health Organisation’s list of essential medicines. They are
usually given orally and are absorbed into gastric parietal cells from the bloodstream.
Here, they bind irreversibly to one of the subunits of the proton pump, permanently
deactivating it (Fig. 10.5). It can take the parietal cell up to 36 hours to synthesise a
new pump to replace it, and the irreversible nature of the binding means that the
biological effect of PPIs far exceeds the plasma half-life, and once daily dosing is
adequate. Both basal secretion and feeding-stimulated secretion are inhibited. The
effectiveness of PPIs improves with repeated administration and is maximal after 5 days
of daily dosing. This is because only around a third of proton pumps are activated when
a meal is eaten. Over the course of the next few days and meals eaten, more and more of
the inactive pumps are recruited in an attempt to maintain acid secretion; after about 5
days, all available pumps are blocked, and acid production is maximally reduced.
FIG. 10.6 The main mechanisms of antacid-mediated drug
interactions.GI, gastrointestinal.
Pharmacokinetics
In general, these drugs have short plasma half-lives; omeprazole’s plasma half-life is
30–60 minutes, esomeprazole’s is between 60 and 90 minutes, and lansoprazole’s
is around 90 minutes. However, because they irreversibly inhibit the proton pump, their
acid-suppressant action is highly effective: a single dose reduces acid production by up
to 90% over a 24-hour period. They are acid-sensitive and oral preparations are enteric-
coated to protect the drug on its passage through the stomach. The increase in gastric
pH reduces absorption of azole antifungals including itraconazole (see also Antacids
above) and some other drugs whose solubility falls at higher pH values, including some
antiviral agents. They are highly plasma protein-bound and all are metabolised to
inactive products in the liver by cytochrome P450 enzymes. This can cause interactions
with other drugs also metabolised by members of this family, including selective
serotonin re-uptake inhibitors, whose plasma levels are increased when taken with
a PPI.
Adverse Effects
PPIs may cause a range of GI symptoms including nausea, vomiting, abdominal pain,
and diarrhoea. There is evidence linking PPI use to increased risk of GI infections, likely
due to improved microbial survival in the less hostile gastric environment. Additionally,
changing gastric pH affects the bowel microbiome, the populations of microbes
inhabiting the tract, which may help pathogenic species establish themselves. The rise in
gastric pH may reduce absorption of calcium and magnesium, and long-term use of PPIs
has been associated with increased risk of osteoporosis, fracture, and magnesium
deficiency. Compared to H2-receptor blockers, PPIs have been shown to slightly increase
the risk of gastric cancer, and the risk increases with dose and duration of treatment.
The reason for this is unclear and it is likely that more than one mechanism is involved.
One possible factor may relate to gastrin levels. When gastric pH rises, which would
normally happen after eating because food and fluids dilute stomach fluids, gastrin
secretion increases in response to aid digestion; gastrin also acts as a growth factor and
may stimulate hyperplasia in stomach tissues. PPI use may also mask the symptoms of a
pre-existing gastric cancer.
H2-Receptor Antagonists
Examples: cimetidine, famotidine, ranitidine
Histamine stimulates gastric acid secretion by stimulating H2 receptors on gastric
parietal cells and activating the proton pump (Fig. 10.5). James Black, a Scottish
pharmacologist, identified the subtype of histamine receptors found in the stomach and
named them H2 receptors because H1 receptors, which mediate the inflammatory and
allergic actions of histamine, had already been named. Standard antihistamines such as
chlorphenamine had been used to treat allergic inflammation since the early years of
the 20th century, but recognising that these drugs did not reduce histamine-induced
gastric acid secretion, Black reasoned that there must be more than one subtype of
histamine receptor and spent several years looking for an agent that would selectively
block histamine’s action on the stomach. The result was cimetidine, which blocked
histamine-induced gastric acid secretion but had no anti-inflammatory action, and
which came on the market in 1976. Black had already had significant research success in
the newly emerging field of receptor subtypes: he had also led the group that in the early
1960s produced the β2-selective antagonist propranolol (p. 131), a highly significant
step in the treatment of cardiovascular disease; he shared the 1988 Nobel Prize in
Physiology and Medicine for his contribution to pharmacological research.
The use of H2-receptor antagonists has steadily fallen since the development of PPIs,
but they are effective suppressants of both basal and stimulated gastric acid secretion:
they reduce 24-hour gastric acid secretion by around 70%. Ranitidine was withdrawn
from global markets in 2019/2020 following concerns that an impurity in its
formulation, called nitrosodimethylamine, a known human carcinogen, could be linked
to increased risk of cancer.
As with PPIs, regular use of histamine-receptor antagonists can mask the symptoms
of stomach cancer.
Pharmacokinetics
H2-receptor antagonists are taken orally and have short plasma half-lives, ranging from
1–3 hours. Famotidine is the most potent member of the group. Cimetidine is a
potent inhibitor of hepatic cytochrome P450 enzymes and can reduce the metabolism of
a range of other medications, including anticoagulants and tricyclic
antidepressants. H2-receptor antagonists are mainly eliminated unchanged by the
kidney and doses may need to be reduced in people with poor renal function.
Adverse Effects
H2-receptor antagonists are usually well tolerated. Common side-effects include GI
upsets such as diarrhoea or constipation and skin reactions, headache, and myalgia.
Rarely, they can cause acute liver injury. Cimetidine has weak anti-androgenic activity
and blocks testosterone receptors; this can cause gynaecomastia and impotence.
Misoprostol
Misoprostol is a stable synthetic analogue of PGE1 with gastroprotective actions and is
used to treat or prevent peptic ulcer disease: it is also given to prevent or minimise
NSAID-induced GI toxicity. Naturally produced gastric PGs are cytoprotective in the
stomach as described above (see also Fig. 10.3) but are too unstable to be useful: oral
misoprostol has a plasma half-life of around 40 minutes and is metabolised in the liver.
It stimulates GI smooth muscle and can cause abdominal cramps, diarrhoea, nausea,
and vomiting. It also stimulates uterine smooth muscle, causing uterine cramping and
potentially uterine rupture. For this reason, it should not be used in peptic ulcer disease
in pregnancy. It has an alternative therapeutic use in termination of pregnancy and
induction of labour.
Bismuth
Bismuth subsalicylate is an antacid with antimicrobial activity. Given orally, it breaks
down to release bismuth, which is toxic to a range of gastrointestinal pathogens
including Escherichia coli and Clostridium difficile but is thought to have minimal
effects on normal gut flora. It is available over the counter to treat the symptoms of
acid reflux and the relief of other gastrointestinal upsets including nausea, flatulence,
and diarrhoea. It is the fourth component of quadruple therapy in peptic ulcer
disease; it blocks the ability of H. pylori to attach to the gastric epithelium and
inhibits some of its key enzymes, including the urease it uses to synthesise its
protective ammonia cloud. At therapeutic doses, less than 1% of bismuth is absorbed,
but systemic side-effects include blackening of the tongue and faeces, encephalopathy
and deteriorating mental status, insomnia, and seizures. Bismuth is excreted very
slowly via the kidneys, so care is needed in renal impairment.
Sucralfate
Sucralfate is a complex of sucrose sulphate and aluminium hydroxide, which in the
acidic environment of the stomach forms a thick gel which coats the ulcer and forms a
physical barrier between it and the gastric juices. Because gel formation depends on
low pH, it is less effective if stomach acid has been neutralised or reduced by other
treatments, so dosing should be appropriately spaced out. In addition, it inhibits
pepsin and increases HCO3- secretion, reducing tissue damage and promoting
healing. Activated sucralfate possesses multiple negatively charged groups, which are
responsible for binding to the protein-rich exudate from the ulcer and essential for its
activity, but which may also bind a range of drugs including digoxin, tetracyclines,
phenytoin, and quinolone antibiotics and reduce their absorption. The most
common side-effect is constipation. Rarely, sucralfate may consolidate into a
hardened mass called a bezoar, which can obstruct the tract.
Opioids
Via their action on μ (mu) opioid receptors (MORs) on enteric nerves, opioids decrease
the motility of GI smooth muscle, delaying gastric emptying, inhibiting forward
peristaltic movement, and halting the propulsion of materials through the tract.
Constipation is a troublesome side-effect when opioid drugs such as morphine and
codeine are used as analgesics. Opioids increase intestinal transit time, allowing more
time for water absorption and reducing faecal volume.
Loperamide
Loperamide is a synthetic opioid widely available as a constituent of over-the-counter
antidiarrhoeal preparations: it has 50 times the antidiarrhoeal effect of codeine and is
more potent than diphenoxylate (see below). It is taken orally and subject to
significant first-pass metabolism, so its bioavailability is very low, usually in the order of
2%. At doses recommended for the treatment of diarrhoea, very little crosses the blood–
brain barrier, so loperamide lacks the central action associated with most opioid drugs.
It has a high affinity for MORs on nerves of the myenteric plexus and is absorbed
directly from the tract into the wall of the intestine: its onset of action is usually within
an hour and it is effective for up to 24 hours. In addition to its action on intestinal
smooth muscle, it increases absorption of water and electrolytes, although it reduces
absorption of nutrients such as glucose. Adverse effects include constipation and
nausea. At high doses, loperamide is sometimes used as a recreational agent to achieve
euphoria and relieve the unpleasant symptoms of opioid withdrawal; supra-therapeutic
doses have been associated with serious cardiac arrhythmias including QT prolongation,
torsades de pointes, and cardiac arrests. Naloxone is an effective reversal agent.
Diphenoxylate/Atropine
Diphenoxylate is an opioid with the expected antimotility action of this group of
drugs, but it crosses the blood-brain barrier in greater quantities than loperamide, so
its potential for abuse is higher. To discourage abuse, it is used with atropine, an
antimuscarinic agent: atropine itself has antimotility action because it blocks
parasympathetic activity in the tract, but it causes a range of other unpleasant
antimuscarinic side-effects, including dry mouth and eyes, nausea, and bloating. Even at
therapeutic doses, the atropine component of the preparation can cause antimuscarinic
side-effects including tachycardia and closed-angle glaucoma.
Racecadotril
This is a pro-drug and is metabolised in the liver to thiorphan. Thiorphan is not a
directly acting MOR agonist like loperamide or diphenoxylate, but it increases the
concentrations of endogenous opioids in the GI tract by blocking the enzyme that breaks
them down. In the tract, endogenous enkephalins, acting on MORs on enteric nerves,
contribute to the regulation of gut function. They are broken down by enkephalinase,
terminating their action. Thiorphan inhibits enkephalinase, prolonging enkephalin
action (Fig. 10.9). Its main therapeutic action in the treatment of diarrhoea seems to be
by reducing excessive intestinal secretory activity rather than reducing gut motility: this
drug does not reduce transit times, and constipation is therefore not one of its side-
effects. Another clinical advantage of not reducing gut motility is that in infection, the
drug does not increase retention time and so does not increase the length of time
infective products are in contact with the GI wall. Thiorphan acts quickly (within half an
hour), and because it does not cross the blood-brain barrier, it does not have central
side-effects. Racecadotril is available in many countries worldwide including India and
several European countries but not currently in the UK.
Smooth-Muscle Relaxants
Examples: alverine citrate, mebeverine, peppermint oil
Drugs that directly relax smooth muscle may be used in GI disorders associated with
spasm.
Alverine Citrate
Serotonergic nerves supplying the GI tract release serotonin (5-HT) which acts on 5-
HT1A receptors on GI smooth muscle to stimulate contraction and cause pain. Alverine
citrate blocks 5-HT1A receptors, and so relieves pain and spasm in GI disorders; it also
effectively relieves the pain of dysmenorrhoea. Side-effects include headache, dizziness,
and nausea.
Mebeverine
Mebeverine is a direct-acting smooth-muscle relaxant used mainly in IBS. It is taken
orally, but little reaches the circulation because it is extensively cleared by first-pass
metabolism in the liver. It is well tolerated with few significant side-effects. Its
mechanism of action is not clear, although it may have antimuscarinic activity. It blocks
sodium channels in the smooth muscle cell membrane and reduces calcium levels within
smooth muscle cells, both of which promote muscle relaxation and relieve intestinal
spasm. Side-effects include skin reactions and facial oedema.
Peppermint Oil
Peppermint oil is the essential oil obtained from the plant Mentha piperita and has been
used for hundreds of years in traditional medicine as an anti-inflammatory,
antispasmodic, and expectorant. It is commonly used to treat the symptoms of IBS. The
major constituent of peppermint oil is menthol, thought to be responsible for its
therapeutic action. Menthol blocks calcium channels in the smooth muscle cells of the
GI tract, reducing calcium influx and inhibiting contraction. In addition, peppermint oil
has antimicrobial, anti-inflammatory, and anaesthetic properties, all of which may
contribute to its therapeutic action. Capsules are enteric-coated to prevent their
dissolution in the stomach, because menthol relaxes the sphincter between the
oesophagus and the stomach and causes acid reflux.
Constipation
Constipation is the passage of stools less frequently than an individual’s normal pattern,
and the faeces is usually dry, hard, and difficult to expel. It is a common problem, and
while it may be a feature of a range of diseases as described above, it is often due to
lifestyle factors including insufficient physical exercise, a low-fibre diet, and inadequate
fluid intake. It may also be a side-effect of a wide range of drugs, including opioids,
antimuscarinic agents, and iron supplements. Because progesterone relaxes
smooth muscle, constipation is also common in pregnancy. Wherever appropriate,
lifestyle advice regarding dietary modifications, drinking more fluid, and taking regular
exercise should be the first approach to managing constipation.
Laxatives
Laxatives promote defaecation, usually either by softening or lubricating the stool or by
stimulating intestinal motility and are used to treat constipation. If laxative use is
required for short-term management, their use should be for as limited a time as
possible. Laxatives may also be used to cleanse the bowel in preparation for endoscopy,
surgery, or radiological procedures. They should not be used in GI obstruction.
Laxatives are classified into four main groups (Fig. 10.10).
Bulk-Forming Laxatives
Examples: bran, ispaghula husk, methylcellulose, sterculia
These are all bulky, indigestible plant extracts which retain fluid and soften and
expand faecal volume. This stretches the intestinal walls, which stimulates reflexive
contraction of its smooth muscle and propels its contents forward. It is important to
ensure an adequate fluid intake with these agents to avoid obstruction. They may take
2–3 days to work, but their action is gentle and they are generally well tolerated,
although they can cause bloating and flatulence.
Softening Laxatives
Examples: arachis oil, docusate sodium, liquid paraffin
These agents coat and soften the faeces, lubricating their passage through the tract.
Arachis oil is given as an enema and liquid paraffin is given orally; both can cause
unpleasant and itchy anal seepage. Accidental inhalation of liquid paraffin can cause
aspiration pneumonia. Docusate sodium can be given orally or rectally, and has an
additional direct stimulant effect on intestinal smooth muscle.
Stimulant Laxatives
Examples: bisacodyl, dantron, glycerol, senna, sodium picosulfate
Stimulant laxatives stimulate enteric nerves, which triggers contraction of intestinal
smooth muscle and stimulates water and electrolyte secretion into the tract, promoting
peristalsis. Because of this, they can cause painful abdominal cramps. Overuse
desensitises the enteric nerves and reduces the efficacy of the drugs, which can lead to
increased laxative consumption. This further desensitises the nerves, establishing a
vicious circle of laxative overuse and deteriorating bowel function, potentially causing
an atonic bowel. In general, stimulant laxatives exert their effects rapidly, usually within
12 hours.
Osmotic Laxatives
Examples: lactulose, macrogol, salt solutions
These agents are concentrated preparations of substances which are not absorbed
from the intestines, creating an osmotic gradient between the contents of the tract and
the walls of the tract. This osmotic gradient draws fluid into the intestinal contents and
traps it there, expanding and softening faeces and stimulating peristalsis. Lactulose is
a semi-synthetic disaccharide which is converted in the colon to organic acids including
lactic acid, acetic acid, and formic acid, as well as methane gas, which can cause
flatulence. Macrogols are large-molecular-weight polymers of ethylene glycol and may
be combined with magnesium or phosphate salts in bowel cleansing preparations or
with a stimulant laxative such as sodium picosulfate. Macrogol 3350 (3350 is the
molecular weight) is taken orally and used both in constipation and to relieve faecal
impaction, and can cause abdominal pain and flatulence.
Opioid-Receptor Antagonists
Examples: methylnaltrexone, naldemedine, naloxegol
Peripherally acting MOR antagonists are used to block MORs on enteric nerves to
prevent opioid-related constipation, generally when standard laxatives are inadequate.
The molecular structure of these drugs is based on the opioid reversal agent
naltrexone, but they are chemically modified to reduce lipid solubility and increase
electrical charge, which prevents them from crossing the blood-brain barrier and
blocking the analgesic and other centrally mediated actions of opioids.
Methylnaltrexone is given by injection, has a half-life of around 10 hours, and is
excreted largely unchanged in the urine and faeces. Naldemedine and naloxegel are
given orally and have half-lives in the order of 11 hours. Their most common side-effects
are GI-related, including abdominal pain, nausea, vomiting, and diarrhoea.
Pro-Kinetic Drugs
Pro-kinetic drugs increase the strength and frequency of GI smooth muscle contraction
and shorten transit times (Fig. 10.11).
Macrolide Antibiotics
Motilin is a hormone secreted by glandular cells mainly in the duodenum. It stimulates
gastric and intestinal motility via action on motilin receptors on GI smooth muscle: it is
most active in the stomach. The macrolide antibiotics, including erythromycin,
azithromycin, and clarithromycin, act as motilin-receptor agonists at doses lower
than required for their antibacterial action, although not all are used for this purpose
and the use may be off-licence: for example, erythromycin is unlicensed for this
indication in the UK but authorised in the US. Erythromycin is the standard agent used
for this purpose but is limited to short spells of treatment (less than 4 weeks), because
with continual exposure, motilin receptors are down-regulated, and the drug becomes
much less effective. In addition, the use of these important antibiotics for non-infective
conditions may contribute to the increasing of multi-drug-resistant microbes. Novel
motilin agonists are in development.
Future Possibilities
Ghrelin is a hormone released by the stomach, which stimulates appetite and GI
secretion and motility. Relamorelin is a ghrelin analogue that increases gastric
emptying and is currently in advanced clinical trials for the treatment of diabetic
gastroparesis.
Antiemetics
Vomiting (emesis) is an important protective reflex to rapidly expel actually or
potentially toxic substances from the stomach. Although the muscular propulsion that
forcibly ejects material comes mainly from the stomach and muscles that compress the
stomach, intestinal smooth muscle can also generate peristaltic contractions to reverse
the normal direction of tract flow and return contents back towards the stomach.
Nausea is the unpleasant sensation that vomiting is imminent, may or may not precede
vomiting, and may occur with autonomic symptoms such as pallor, sweating, and
dizziness.
FIG. 10.12 The mechanism of action of linaclotide.CFTR, Cystic
fibrosis transmembrane conductance regulator; cGMP, cyclic
guanosine monophosphate; GTP, guanosine triphosphate.
Higher Centres
Unpleasant, frightening, or emotional situations may all trigger vomiting because the
cortex, where these higher-order functions are housed, has input to the VC. Sights,
smells, and memories may all induce vomiting. Pain can also be a powerful stimulant of
vomiting. Raised intracranial pressure causes vomiting, although the reason for this is
not clear.
Dopamine Antagonists
Examples: domperidone, metoclopramide, phenothiazines
Dopamine, released in the VC and in the CTZ, is a powerful pro-emetic, and as
described above has direct actions on the GI tract, decreasing motility and increasing
transit time. Dopamine antagonists are therefore effective anti-emetics in a range of
conditions including drug-, pregnancy-, and radiotherapy-induced vomiting and post-
operative nausea and vomiting. However, because dopamine also has important
functions in the nigrostriatal pathway and the regulation of voluntary muscle
movement, dopamine antagonists that cross the blood-brain barrier can produce
extrapyramidal movement disorders similar to those seen in Parkinson’s disease (see
also p. 53), and these drugs should be avoided in people with this disorder. Basal
dopamine release in the tuberoinfundibular pathway suppresses prolactin production,
and dopamine antagonists can therefore increase prolactin levels even in non-lactating
people and cause galactorrhoea and gynaecomastia (Fig. 10.15).
FIG. 10.15 Dopamine antagonists as anti-emetics.DA, Dopamine;
GI, gastrointestinal.
Domperidone
Unlike other D2 antagonists used to treat vomiting, domperidone does not readily cross
the blood-brain barrier and so it is fairly free of central side-effects, including movement
disorders and abnormal lactation, seen with the others in this group. It suppresses
vomiting via its action on D2 receptors in the CTZ; it also blocks D2 receptors in the GI
tract, which increases peristalsis and shortens gastric emptying times. For this reason, it
is also used as a pro-kinetic agent in hypomotility disorders including GORD (see
above). It is given orally and has a plasma half-life of around 7 hours.
Metoclopramide
Metoclopramide is related to the phenothiazines but is not used as an antipsychotic
agent. It does, however, cross the blood-brain barrier, so extrapyramidal movement
disorders and abnormal lactation are seen in its adverse effects profile. Its main action is
via blockade of D2 receptors, both in the CTZ and peripherally in the GI tract, but it also
stimulates 5-HT4 receptors and blocks muscarinic receptors in the intestines, which aids
motility and speeds up transit times. It is given orally or by injection, is well absorbed,
and has a plasma half-life of around 5 hours, which is increased if kidney function is
impaired. Intranasal preparations are available in some countries.
5-HT3 Antagonists
Examples: granisetron, ondansetron, palonosetron
5-HT3 receptors are found in the GI tract and in the CTZ. 5-HT3 antagonists are used
in post-operative nausea and vomiting, hyperemesis gravidarum, and drug-induced
vomiting including with chemotherapy. The first 5-HT3-receptor antagonist developed
as an anti-emetic, ondansetron, came on the market in the mid-1980s. It is given
orally, rectally, or by intravenous or intramuscular injection and has a plasma half-life of
3 hours. Palonosetron has a longer half-life of up to 40 hours. Unlike antihistamines
and phenothiazines, these drugs are non-sedating and are generally well tolerated;
common side-effects include headache and constipation, the latter due to blockade of 5-
HT3 receptors in the GI tract (Fig. 10.14). Rarely, they may cause arrhythmias
including QT interval prolongation.
Neurokinin-Receptor Antagonists
Examples: aprepitant, fosaprepitant, netupitant (used in combination with
palonosetron)
These drugs are anti-emetic because they block binding of substance P on NK1
receptors in the GI tract (Fig. 10.14), VC, and CTZ, inhibiting input into and activation
of the VC. They are used mainly to manage chemotherapy-induced vomiting and in
some countries are also licensed for post-operative nausea and vomiting.
Fosaprepitant is given intravenously and is converted to aprepitant in the liver;
aprepitant has a half-life of 9–13 hours.
In addition to their anti-emetic action in blocking NK1 receptors, neurokinin-receptor
antagonists enhance the anti-emetic effect of 5-HT3 antagonists. Netupitant has a long
half-life (around 90 hours) and is used in combination with the 5-HT3 antagonist
palonosetron to treat chemotherapy-induced nausea and vomiting (CINV),
particularly with regimens that include cisplatin, which is highly emetogenic. The
acute phase of CINV in the hours immediately following administration of the cytotoxic
drug is thought to be mainly due to 5-HT release in the GI tract, so both drugs
contribute therapeutically at this time. The delayed phase of CINV, which manifests 25–
120 hours after cytotoxic treatment, is thought to be mainly due to substance P acting on
NK1 receptors, and netupitant blocks this.
Adverse effects of these drugs include appetite suppression, constipation, and
abdominal discomfort.
Antihistamines
Examples: cinnarizine, cyclizine, doxylamine, promethazine
The most familiar use of antihistamines is in allergy (p. 122). The older
antihistamines, the so-called first-generation agents, have significant antimuscarinic
properties and cross the blood-brain barrier. They block H1 and muscarinic receptors in
the vestibular nuclei and muscarinic receptors in the VC itself. They relieve vomiting
from most causes, although they are particularly helpful in preventing and treating
nausea and vomiting associated with motion sickness and middle ear disorders such as
Ménière’s disease. The newer second- and third-generation antihistamines do not cross
the blood-brain barrier and do not prevent or relieve motion sickness. When treating
motion sickness, antihistamines are more effective as preventive treatment, taken in
advance, than stopping established nausea and vomiting. Cyclizine is also used in
palliative care, especially to counteract opioid-induced vomiting. It has a half-life of 20
hours.
Antihistamine anti-emetics block muscarinic receptors throughout the body and
cause a range of antimuscarinic side-effects, including dry mouth, tachycardia, closed-
angle glaucoma, and blurred vision. Sedation is a significant side-effect because
histamine is also an important transmitter in central pathways that maintain daytime
alertness (see also p. 124). Advice should always be given regarding likely drowsiness,
e.g. avoiding driving and cycling; alcohol should be avoided because it enhances
sedation.
Antimuscarinic Agents
Examples: hyoscine hydrobromide
Drugs with central antimuscarinic activity are anti-emetic because ACh stimulates
vomiting via an action on muscarinic receptors in the VC and the vestibular nuclei. As
discussed above, phenothiazines and first-generation antihistamines owe at least some
of their anti-emetic activity to their ability to block muscarinic receptors in vomiting
pathways. Hyoscine (scopolamine) and the closely related drug atropine are found in
a range of poisonous plants including deadly nightshade and henbane. Hyoscine was
first isolated in 1880; it is highly lipid-soluble, crosses the blood-brain barrier, and is a
potent CNS depressant. Two formulations are currently in use, the main difference
between them being their lipid solubility and therefore their ability to cross the BBB (see
also hyoscine butylbromide above). Hyoscine hydrobromide is used as an anti-
emetic because it crosses the blood-brain barrier and blocks muscarinic receptors in
central vomiting pathways. Other central antimuscarinic side-effects include sedation,
which may be beneficial in certain circumstances: for example, in the treatment of
motion sickness in children, in pre-medication, and in reducing respiratory secretions in
palliative care. Peripheral antimuscarinic side-effects include dry mouth, urinary
retention, and blurred vision. Its plasma half-life is around an hour, although this is
extended if the drug is given subcutaneously or by transdermal patch.
Nabilone
Nabilone is a synthetic cannabinoid closely related to tetrahydrocannabinol, the
main psychoactive substance in cannabis. Both the peripheral and central nervous
systems possess cannabinoid receptors and several endogenous substances, collectively
called the endocannabinoids, have been identified. This is a relatively new area of
study, but the known functions of endocannabinoids relate to anti-inflammatory activity
and modulation of pain. It is not certain why nabilone has anti-emetic activity, but it
may inhibit 5-HT release in the VC. It is well absorbed orally and has a plasma half-life
of around 2 hours. It is used to relieve CINV when standard anti-emetics have not been
effective. Adverse effects include sedation, confusion, sleep disruption, psychotic
symptoms, and GI upsets.
References
1. Abrahami D, McDonald E.G, Schnitzer M.E, et al. Proton pump inhibitors and
risk of gastric cancer: population-based cohort study. Gut. 2022;71(1):16–24.
2. Camilleri M, Atieh J. New developments in prokinetic therapy for gastric motility
disorders. Front. Pharmacol. 2021;12:711500 art. no.
3. Denholm D, Gallagher G. Physiology and pharmacology of nausea and
vomiting. Anaesth. Intensive Care Med. 2021;22(10):663–666.
4. Ogawa R, Echizen H. Clinically significant drug interactions with
antacids. Drugs. 2011;71(14):1839–1864.
5. Strand D.S, Kim D, Peura D. 25 years of proton pump inhibitors: a
comprehensive review. Gut. Liver. 2017;11(1):27–37.
11: Antimicrobial Drugs
CHAPTE R OU TLINE
Antiviral Drugs
Inhibitors of Viral Nucleic Acid Synthesis
Inhibitors of Viral Uncoating and Release
Inhibitors of Viral Protein Synthesis
Antifungal Drugs
Antifungals That Interfere With Cell Wall Integrity
Echinocandins
Antifungals That Interfere With Cell Membrane
Integrity
Nystatin
Amphotericin
Terbinafine
Azoles
Antifungals That Interfere With Cell Division
Griseofulvin
Flucytosine
Antiparasitical Drugs
Protozoal Infections
Antimalarial Drugs
Helminthic Infestation
Antihelminthic Drugs
Arthropod (Insect) Infestations
Insecticides
F OCUS ON
Key Definitions
• Microbe/micro-organism:
These terms are used interchangeably to mean an organism so small that it can only
be seen with a microscope.
• Pathogen:
• Antimicrobial:
Microbial Adaptability
Microbes are the most varied and adaptable forms of life on Earth. Underlying their
evolutionary success throughout their billions of years of existence is their seemingly
inexhaustible ability to constantly adapt to changing environments. They survive and
flourish in the face of a wide range of intensely hostile conditions: in ice, around
volcanic craters, deep in the ocean, and in deserts. Some bacteria are even resistant to
radiation and survive for years in the inhospitable environment of space. Despite
reproducing asexually, i.e. each new daughter cell in theory should be an exact copy of
the parent cell, microbes constantly mutate and evolve, sometimes through random
genetic mutations, sometimes through errors in copying genetic material during cell
division, and sometimes in response to changing environmental conditions. The ability
of microbes to produce and survive quite significant degrees of genetic mutation means
that often not all microbes in a colony of apparently identical microbes are genetically
identical. This gives the colony, as a whole, a survival advantage. If a mutation in a
microbe’s genetic material improves its ability to survive and reproduce in its current
environment or in changing environmental conditions, it is more likely than its less
well-equipped neighbours to divide and proliferate, passing its beneficial mutation to its
daughter cells. In this way, if a colony is exposed to a toxin or other stressor, susceptible
microbes will perish, but better adapted ones survive to generate a new colony capable
of thriving in their new environment (Fig. 11.3). This adaptability is the basis for the
development of antimicrobial drug resistance.
FIG. 11.2 Electron micrograph of tobacco mosaic virus. From
Williams RC and Fisher HW (1974) An electron micrographic atlas of
viruses. Charles C. Thomas, Springfield, Il, with permission.
Antimicrobial Stewardship
From the first half of the 20th century, clinical medicine has had access to an expanding
range of effective antimicrobial drugs, leading to a steady increase in global use. The
bulk of antimicrobial use, however, is not in human medicine, but in plant agriculture,
animal husbandry, food production, veterinary medicine, and a range of industrial
applications, and significant quantities of these drugs are washed into the natural
environment, including soil and water. This has led to widespread microbial exposure to
antimicrobial drugs, contributing to the consequent widespread emergence of multiple
resistant strains. This phenomenon was recognised relatively quickly in the
antimicrobial era, with warnings in the literature as early as the 1950s that antibiotic
efficacy was at serious risk. However, optimism that the continual production of new
drugs would offset the issue, as well as resistance from agriculture and industry to any
regulation of antibiotic use, has meant that the worldwide approach required to tackle
the issue has been slow and piecemeal.
FIG. 11.3 Genetic mutations can give microbes a survival
advantage in changing environmental conditions.1. A microbe is
exposed to an environmental stressor, e.g. a natural toxin or an
antimicrobial drug. 2. One daughter cell develops or acquires a
mutated gene that protects against the stressor. 3. Daughter cells
produced from this line inherit the protective gene and are more
likely to survive and proliferate, producing a colony with a selective
survival advantage. 4. Daughter cells lacking the mutated protective
gene are much less likely to survive.
Antibacterial Drugs
There are more antibacterial drugs in clinical use than any other category of
antimicrobial agents, because bacterial cells are sufficiently different in structure and
function to human cells to offer a range of potential drug targets. The term ‘antibiotic’ is
used throughout this section to mean any antibacterial drug, natural or synthetic, used
to treat bacterial infections.
Although the definitions above are straightforward, the action of individual drugs can
be either bactericidal or bacteriostatic depending on drug dose, bacterial numbers, and
how favourable the growth medium is; most antibiotics can be either, depending on the
conditions. Clearing an infection is the result of a combined action of effective
antibacterial treatment and the efforts of the host’s own immune system; therefore,
treating infections in immunocompromised people is often harder than in individuals
with functional immunity. For this reason, bactericidal antibiotics may be preferred if
the patient has weakened immunity.
Key Definitions
Bacterial Metabolism
Like human cells, bacteria contain thousands of different enzymes to catalyse the wide
range of biochemical reactions required for life. Although microbial and human
biochemistry share similar pathways, it is possible to find metabolic processes present in
bacterial cells but not in human cells, including their handling of folic acid.
Trimethoprim and the sulphonamides are examples of antibiotics that interfere
with folic acid metabolism.
AntiBacterial Resistance
If a population of bacteria is exposed to an antibiotic, all susceptible cells will die, but
resistant cells will proliferate, so that eventually all bacteria in the population will be
resistant to that drug. Broad-spectrum antibiotic use induces the development of
resistance more effectively than narrow-spectrum antibiotics because it affects a wider
range of bacteria. Some bacteria develop resistance more readily than others: for
example, Staphylococcus aureus, a common bacterium often found living harmlessly on
skin and mucous membranes, but which causes a range of skin, wound, and other tissue
infections, rapidly develops resistance to nearly all known families of antibiotics.
Methicillin-resistant S. aureus (MRSA) infections are a major global problem,
particularly in healthcare settings and in vulnerable people, e.g. the very old and the
very young. Bacteria can develop or acquire protective genes against a drug, i.e. develop
resistance to that drug, in a range of ways. Once the bacterium possesses one or more
genes that protect it against a drug, it copies them, and in the ways described below, the
gene(s) can be spread through a rapidly dividing bacterial population in a matter of
hours (Fig. 11.6).
FIG. 11.7 Bacterial conjugation.A. Bacteria can pass genes that give
antibiotic resistance to each other, spreading resistance in a
population. B. False-coloured micrograph of bacteria, showing pilus
bridges between the large bacterium (bottom left) and its neighbour,
and the bacterium (middle right) and three of its
neighbours. Modified from (A) VanMeter KC, Hubert RJ, and
VanMeter WG (2010) Microbiology for the healthcare professional,
Fig. 25.2. St. Louis: Mosby and (B) Fitzgerald-Hayes M and
Reichsman F (2010) DNA and biotechnology, 3rd ed, Fig. 4.6. San
Diego: Academic Press.
Spontaneous Mutation
As mentioned before, bacteria are genetically unstable: that is, unlike human cells,
which have rapid and robust mechanisms for eliminating errors from their genetic
material, they tolerate significant mutations in their DNA. The mutations are random,
but because bacteria replicate so fast, they occur very frequently. Some will be beneficial
and will appear in all cells descending from the original (Fig. 11.3).
The Gene May Code for an Enzyme That Breaks Down the Drug
For example, when exposed to penicillin, bacteria with the gene that produces β-
lactamase survive because β-lactamase destroys the penicillin molecule.
The Gene May Change the Cell-Surface Receptor That Allows the Drug to
Bind to the Bacterium
For example, when exposed to penicillin, bacteria that do not make penicillin-binding
receptors survive, because penicillin cannot latch onto the bacterial cell surface and
penetrate it.
The Gene May Increase the Bacterium’s Ability to Excrete the Drug
For example, bacteria that produce pumps to excrete a drug survive, because they can
keep the drug levels in their cytoplasm too low to cause damage. Pseudomonas
aeruginosa, a microbe responsible for a wide range of clinically important infections
and which readily evolves into multi-drug resistance strains, quickly generates efflux
pumps to clear antibiotics from their internal environment.
The Gene May Change the Permeability of the Bacterial Cell Wall, Blocking
Drug Entry
For example, structural changes to pores in the bacterial cell membrane can allow the
bacterium to exclude particular drugs: this is a common mechanism and accounts for
resistance to a wide range of bacteria.
Combination Therapy
Using more than one antibiotic simultaneously (combination therapy) can be
advantageous in two main ways (Fig. 11.8).
Prevention of the Emergence of Drug-Resistant Bacteria
Combination therapy is used in hard-to-treat, serious, and chronic infections such as
TB, or to eradicate gastric Helicobacter pylori infection, which is strongly associated
with peptic ulcer disease (p. 188). It is also used in immunocompromised or vulnerable
individuals, whose own defence mechanisms are unable to contribute significantly to
clearing the infection. The principle is that by using more than one drug, a larger
proportion of bacteria are cleared as fast as possible at the beginning of treatment,
increasing the chances that the host immune system can eradicate the remainder (Fig.
11.8A). There are however risks to this approach, including the increased incidence of
side-effects from multiple drugs, and the likelihood that if treatment is not successful
and the infection is not cleared, surviving organisms will be multi-drug resistant.
Antibiotic Synergism
In some situations, antibiotics work synergistically, and their combined antibacterial
effect is greater than the sum of their individual action. Fig. 11.8B shows a hypothetical
example, where antibiotics A and B individually have a 35% kill rate for a particular
infection, but because of synergism, when combined, their total kill rate is 80%. For
example, trimethoprim and the sulphonamides work synergistically because
although they both interfere with bacterial folate use, they inhibit different steps in the
biochemical pathway. Ampicillin and gentamicin work synergistically when treating
enterococcal infections, e.g. infective endocarditis, because ampicillin increases bacterial
uptake of gentamicin (although this synergy does not work for all aminoglycosides, such
as amikacin).
FIG. 11.8 The value of combination therapy in treatment of
complex or chronic infections.
Superinfection
There are about the same number of bacteria living in or on the healthy individual as
there are cells in the body: a huge number, running into the tens of trillions. The
umbrella term given to these organisms is the human microbiome, and the importance
to human health of this symbiotic relationship is becoming increasingly clear. One
benefit of the healthy microbiome is that by colonising both internal and external body
surfaces, they prevent other, potentially pathogenic organisms from establishing
themselves. Introducing an antibiotic into the body, especially if broad-spectrum, kills
not only the target pathogen, but also normal body flora, and can allow pathogens to
colonise and proliferate. This is called super-infection. A common example is
candidiasis (thrush), caused by the yeast Candida albicans.
Allergy
Allergies, often manifested as skin reactions, are common, especially with certain groups
of drugs including the penicillins and cephalosporins. Many antibiotic-triggered
allergies are due to a type I hypersensitivity reaction, with histamine release and rapid
onset of symptoms, including itchy, raised blisters (urticaria). Other antibiotic-related
allergies are due to a type IV (delayed type) hypersensitivity reaction and may not
appear until up to 2 weeks after the antibiotic has been started, often after the course is
finished. This is sometimes called post-antibiotic rash and is more common in children.
The immunology of drug-related allergy is described in more detail on p. 37.
Initiation
In initiation (Fig. 11.9A), an mRNA molecule, containing the code for the new protein,
binds to the 30S subunit so that its code can be read and a new protein molecule
assembled. The first amino acid to be added is usually an N-formylmethionine residue
and is called the initiating amino acid. This amino acid is not found in human proteins.
It binds to what is called the P site. The next amino acid slots into an adjacent binding
site, called the A site, and the two amino acids are joined together with a peptide bond
(Fig. 11.9B and C).
Elongation
As soon as the two amino acids in the A and P sites are linked with a peptide bond, the
ribosome slides along the mRNA molecule to the next coding section. The A site is now
free for the next amino acid; a third amino acid is therefore added according to the
mRNA code, joined to the second with a peptide bond, and the ribosome shifts along
again. This is called elongation (Fig. 11.9D). The new protein emerges from the
ribosome through an exit tunnel, pushed out as new amino acids are added via the A
site. This proceeds until the ribosome has read the entire code along the mRNA
molecule and the new protein is complete.
Termination
Once the ribosome has finished reading the mRNA code, the new protein is released.
The mRNA itself, its job done, is degraded by enzymes in the cytoplasm.
Aminoglycosides
Examples: streptomycin (the first), gentamicin, tobramycin, neomycin, amikacin
Streptomycin was isolated in 1944 from the soil bacterium Actinomyces griseus. Its
use now is almost exclusively in TB treatment, and gentamicin is the most frequently
used aminoglycoside. Aminoglycosides bind irreversibly to the 30S ribosomal subunit,
preventing new amino acids from accessing the A binding site (Fig. 11.9B). The new
protein is forced to terminate while only partially complete and is therefore non-
functional. Anaerobic organisms are not susceptible to aminoglycoside action because
the uptake mechanism needed to absorb the drug into the microbe is oxygen-dependent
and not present in anaerobes. They are broad-spectrum agents, with higher activity
against Gram-positive than Gram-negative organisms, and are usually reserved for
serious infections such as sepsis because they have highly unpleasant side-effects.
Pharmacokinetics
Aminoglycosides are large, highly charged molecules and so are not absorbed across the
wall of the GI tract; they are usually given by intravenous or intramuscular injection.
Tobramycin is also available for nebulisation in respiratory infections. Neomycin is
too toxic for systemic use, but it is used for topical infections; because it is not absorbed,
it is given orally to sterilise the GI tract before surgery. Aminoglycosides have relatively
short half-lives of 2–3 hours and cross the placenta but not the blood–brain barrier.
They are not metabolised, mainly because they are too large and electrically charged to
enter liver cells, so their clearance depends largely upon renal excretion. Active drug can
accumulate very quickly in the renal tubules if kidney function is impaired, and
renotoxicity is one of these drugs’ most dangerous side-effects.
Adverse Effects
Aminoglycosides have significant side-effects, which are generally dose-related, and
therapeutic drug monitoring is recommended to guide dosing. Renal toxicity is
described above, and kidney function must be monitored and the drug stopped at the
first sign of impairment. Ototoxicity, leading to permanent hearing loss, occurs in up to
one-third of patients. The drugs accumulate in the fluids in the cochlea of the inner ear,
where they may persist for extended periods of time and permanently damage the
sensitive hair cells responsible for hearing. High-pitched sounds are lost first, and
progressive hearing impairment can occur over several months following drug
administration because of the slow clearance from inner ear fluids. Neomycin is
particularly toxic to the hearing apparatus of the inner ear and so is not used
systemically. There may also be toxicity to the vestibular system, causing dizziness and
problems with balance. Gentamicin and streptomycin are more vestibulotoxic than
cochleotoxic. Aminoglycosides should not be used in myasthenia gravis, and care should
be taken with neuromuscular blockade in surgery, because they block acetylcholine
release at the neuromuscular junction and can cause paralysis, including of the
respiratory muscles.
Chloramphenicol
Chloramphenicol was isolated in 1947 from the bacterium Streptomyces venezuelae. It
binds to the 50S subunit of the ribosome and prevents peptide bond formation between
amino acids in the growing protein (Fig. 11.9C). Microbial resistance is usually due to
the acquisition of a gene that codes for an enzyme that breaks down the drug.
Pharmacokinetics
The plasma half-life of chloramphenicol is 2–3 hours. It is broad-spectrum and active
against a wide range of Gram-positive and Gram-negative bacteria. It is well absorbed
orally, but because of its significant toxicity, systemic use is limited to life-threatening
infections that do not respond to other, safer agents. Topically, it is widely and safely
used to treat bacterial conjunctivitis. Chloramphenicol is an enzyme inhibitor and
reduces metabolism of a range of other drugs, including phenytoin (p. 73) and the
coumarin anticoagulants (p. 150).
Adverse Effects
The most dangerous side-effect of chloramphenicol is irreversible bone-marrow
suppression, which occurs in 1 in 10,000 people and is always fatal. In newborn babies,
it can cause ‘grey baby syndrome’, which carries a 40% fatality rate. The characteristic
ashen colour of affected babies is due to progressive circulatory collapse caused by
chloramphenicol interfering with the ability of myocardial cells to use oxygen to produce
energy. The younger the baby, the greater the risk, probably due to liver immaturity and
a reduced capacity to metabolise the drug.
Fusidic Acid
Fusidic acid was first isolated from the fungus Fusidium coccineum and prevents the
new protein from sliding along the ribosome to free up the A site for an incoming amino
acid (Fig. 11.9D). It is bacteriostatic and narrow-spectrum, effective only in
staphylococcal infections. Because resistance develops rapidly, it is used in combination
with other agents when treating systemic infections. It is chemically related to steroids,
so is very fat-soluble and distributes very well through all body tissues (including bone
and joints), crosses the placenta, and appears in breast milk.
Pharmacokinetics
It is narrow-spectrum, and its plasma half-life is 10–14 hours. It is well but
unpredictably absorbed from the GI tract and is better absorbed from tablet
formulations than from suspensions. It is used topically for a range of staphylococcal
skin and eye infections and can be given intravenously if required.
Adverse Effects
Topical preparations can cause local irritation, including skin rashes and eye dryness
and discomfort. Side-effects from oral administration include GI upset, dizziness and
drowsiness, and impaired liver function.
Macrolides
Examples: erythromycin, clarithromycin, azithromycin
Erythromycin was first isolated from the soil bacterium Streptomyces erythraeus in
1950. It is a first-generation macrolide, and a range of second-generation agents
including clarithromycin and azithromycin were developed from erythromycin to
produce drugs more stable in stomach acid. They have broad-spectrum activity and are
effective in a wide range of important infections, but widespread use led to widespread
bacterial resistance, and in recent decades research effort has focussed on development
of novel macrolides. Third-generation agents entered the market in the 1990s but have
largely been withdrawn because of serious side-effects; fourth-generation agents are
currently in clinical trials. Macrolides block the exit tunnel in the ribosome through
which the new protein emerges (Fig. 11.9D). As a group, they are more active against
Gram-positive bacteria than Gram-negative because they are bulky molecules which do
not easily penetrate the complex outer cell membrane of Gram-negative bacteria (Fig.
11.5). Different members of the group have different activity profiles; for instance,
azithromycin is more active than erythromycin against Haemophilus influenzae.
Pharmacokinetics
Erythromycin is degraded by stomach acid, poorly absorbed across the gut wall
especially in the presence of food, and normally almost completely metabolised in the
liver. Azithromycin and clarithromycin are acid-stable and better absorbed.
Erythromycin and clarithromycin are both enzyme inhibitors, and so decrease the
activity of metabolising enzymes in the liver; this interaction can increase the plasma
concentrations of a wide range of other drugs.
Adverse Effects
Erythromycin can interfere with liver function, giving abnormal liver enzyme levels
and sometimes cholestatic jaundice. Both erythromycin and clarithromycin block
potassium channels in cardiac muscle, and high doses can cause arrhythmias, including
QT prolongation.
Oxazolidinones
The two antibiotics in this group, linezolid and tedizolid, released in 2000 and 2014,
respectively, are synthetic drugs most active against Gram-positive bacteria, including
MRSA. Tedizolid is more potent with a wider range of activity than linezolid and has
a longer half-life, meaning that it needs to be given only once daily. These drugs bind to
the P binding site on the ribosome (Fig. 11.9A) and prevent initiation of protein
synthesis. Currently, microbial resistance is not a major problem but is increasing,
mainly due to bacterial alterations in ribosome structure, preventing the drugs from
binding. They are well absorbed orally and can be given intravenously. Both agents can
cause GI upsets, headache, and rash. Linezolid can cause irreversible optic neuritis and
potentially dangerous blood disorders, including low blood platelet levels; patients
should be advised to report any visual symptoms, and full blood counts performed
regularly.
Tetracyclines
Examples: tetracycline, doxycycline, minocycline, oxytetracycline
Tetracyclines are produced by soil-dwelling bacteria of the Actinomycetes species.
They were first discovered in 1948 and widely used in the years following their
introduction because they are broad-spectrum agents effective in a very wide range of
infections. However, although development of second- and third-generation
tetracyclines produced newer and more effective drugs, widespread resistance has
limited their usefulness. Resistance is spread mainly by plasmids (Fig. 11.7), which
often carry genes for resistance to other antibiotics as well as tetracyclines, increasing
multi-drug resistance. These drugs work by blocking access of new amino acids to the A
site on the ribosome, preventing elongation of the new protein (Fig. 11.9A).
Pharmacokinetics
Following oral administration, many tetracyclines are irregularly and unpredictably
absorbed, although doxycycline and minocycline are slowly but completely
absorbed. All tetracyclines form insoluble complexes with iron, calcium, magnesium,
and aluminium, significantly inhibiting absorption. Dairy products, iron supplements,
and antacids, which frequently contain aluminium, calcium, or magnesium salts,
should therefore be avoided.
Adverse Effects
Tetracyclines can cause photosensitivity, super-infections, and GI disturbances. They
bind to calcium, are taken up into bones and teeth, and can permanently stain growing
teeth. The presence of the drug in developing bone and tooth tissue can also interfere
with its normal structure, leading to weakening or deformity. Tetracyclines are therefore
contra-indicated in pregnancy or breast-feeding women and not given to children. In
common with several other drugs, tetracyclines can cause drug-induced autoimmunity.
The drug binds to specific proteins, including collagen, in any of a range of tissues,
including skin and liver, and triggers an immune response against that tissue.
Minocycline is the most strongly implicated, with clear causative links to systemic
lupus erythematosus-like syndromes and autoimmune hepatitis.
Cephalosporins
Examples: cephalexin, cefuroxime, cefotaxime
The first cephalosporin, cephalosporin C, was identified in 1953 from the fungus
Cephalosporium, grown from sewage. It contained a β-lactam ring, but unlike the
penicillins, which by this time were in widespread use and had caused widespread
resistance, it was less susceptible to β-lactamase. Thousands of semi-synthetic
derivatives have been produced, leading to the introduction of several generations of
cephalosporin antibiotics, each giving improved activity against Gram-negative
infections and increased stability to β-lactamase. The newest agents have good activity
against both Gram-negative and Gram-positive organisms and do not lose activity when
exposed to bacterial β-lactamase.
Pharmacokinetics
Absorption in this group is variable: some can only be given intramuscularly or
intravenously because they are not absorbed across the wall of the GI tract (e.g.
ceftriaxone), whereas some, e.g. cephalexin, can be given orally. They distribute
well, including into the cerebrospinal fluid, especially the newer agents. As with the
penicillins, they are not significantly metabolised in the liver and rely on renal excretion
for clearance from the body.
Adverse Effects
Like their close cousins the penicillins, the cephalosporins are generally well tolerated.
There is penicillin cross-allergy, so a penicillin-allergic individual has an increased risk
of cephalosporin allergy too. Antibiotic-associated colitis (see above) is more common
with the cephalosporins than most other antibiotics.
Carbapenems
Examples: imipenem, meropenem
These β-lactam antibiotics were developed in the 1980s from thienamycin,
produced by a bacterium of the Streptomyces family.
Pharmacokinetics
Carbapenems are not absorbed across the wall of the GI tract and so must be given
intravenously. Imipenem, the original, is an effective broad-spectrum drug resistant to
β-lactamase but is rapidly deactivated by enzymes in the kidney. It is therefore given
with an inhibitor of these enzymes, cilastatin, to preserve it from renal metabolism and
prolong its activity. Carbapenem half-lives range from 1–5 hours. They are not
metabolised significantly in the liver and are therefore cleared from the body mainly by
the kidneys.
Adverse Effects
GI side-effects are very common. These drugs can also cause neurotoxicity (especially
imipenem) and renal toxicity.
Bacitracin
Bacitracin is produced by the bacterium Bacillus subtilis. It interferes with cell wall
synthesis by blocking the biochemical pathway that transports peptidoglycan molecules
into the growing cell wall structure. It is toxic in systemic use, including nephrotoxicity
and allergy, and is not licensed for use in all countries. Topical preparations are
available in some countries to treat eye and skin infections.
Pharmacokinetics
Colistimethate is not absorbed across the wall of the GI tract because it is highly charged
and is given intravenously for systemic infections. It is also used by nebuliser for
respiratory tract infection, usually in cystic fibrosis in children, and can be given orally
to clear the bowel of Gram-negative organisms prior to surgery. It is not significantly
metabolised and is excreted unchanged by the kidney. Care is therefore needed in
reduced renal function, which may significantly extend the normal half-life of 2–3
hours.
Adverse Effects
Polymyxins can cause a range of unpleasant side-effects, including neurological and
renal toxicity.
Metronidazole
Metronidazole is a pro-drug. It diffuses passively into cells, including host cells and
microbes. Some protozoa and some anaerobic bacteria, e.g. C. difficile and H. pylori,
possess an enzyme that converts metronidazole to a very reactive and toxic metabolite.
This metabolite binds to DNA and breaks it up (Fig. 11.14). This is a bactericidal action,
because the damaged DNA can neither be used to make new proteins nor can it be
copied to allow the microbe to divide. Aerobic cells, including human cells, lack this
enzyme and so do not produce the toxic metabolite. It also means, of course, that
aerobic microbes are resistant to metronidazole. Metronidazole is broad-spectrum and
used to treat a range of anaerobic infections, including vaginal, pelvic, and intestinal
infections, but formerly susceptible organisms are steadily developing resistance, mainly
due to reducing their uptake of the drug. Tinidazole is in the same class and has a
similar spectrum of action to metronidazole.
FIG. 11.14 Activation of metronidazole in anaerobic organisms.
Pharmacokinetics
Metronidazole is available in a range of formulations and is usually given orally,
intravenously, rectally, or vaginally. It is well absorbed when given orally, with a
bioavailability of over 90%. It distributes into the CSF, crosses the placenta, and appears
in breast milk. It is not highly plasma protein-bound and has a half-life of 6–9 hours.
Most is metabolised in the liver and the metabolites excreted in the urine, so doses may
need to be reduced in reduced liver function.
Adverse Effects
Metronidazole can give an unpleasant metallic taste in the mouth, muscle pain, and GI
upsets. Current advice is to avoid alcohol while taking this drug because a disulfiram-
like reaction has been reported, although the validity of this is increasingly being
questioned because the evidence is weak.
Nitrofurantoin
Nitrofurantoin is a synthetic agent and a pro-drug. Susceptible bacteria possess enzymes
that convert it to toxic metabolites that interfere with certain key biochemical processes,
including synthesis of DNA, RNA, and protein. Cells lacking these enzymes, including
animal cells, do not produce the toxic metabolites. The drug can cause serious side-
effects and so is not used as a first-line agent. Nitrofurantoin is concentrated in the urine
in its active form because it is actively secreted into renal tubules, and so is used in
urinary tract infection. It is highly effective against Escherichia coli, which is responsible
for 80% of uncomplicated urinary tract infections, and resistance rarely develops. It
must, however, be used with caution or avoided altogether, in renal impairment,
because the drug can quickly accumulate and cause toxicity, including peripheral
neuropathy.
Pharmacokinetics
The half-life is short, less than 1 hour, and nitrofurantoin is very heavily plasma protein-
bound (up to 90%). Up to half of a dose is excreted unchanged in the urine. It is well
absorbed orally, especially if taken with food.
Adverse Effects
Mild GI side-effects are common, but nitrofurantoin can cause a range of serious
adverse effects, including acute pulmonary disease and bone marrow failure.
Quinolones (Fluoroquinolones)
Examples: ciprofloxacin, levofloxacin, ofloxacin
These broad-spectrum synthetic antibiotics are used in a wide range of infections,
including serious respiratory and enteric infections. They inhibit DNA gyrase and
topoisomerase (Fig. 11.13) and so prevent correct coiling and uncoiling of bacterial
DNA, in turn preventing DNA replication and bacterial cell division. Resistance is
usually due to the bacteria altering their binding site for the drug, or due to production
of efflux pumps which pump the drug out of the bacterial cell. The original drugs in this
group, including nalidixic acid, are narrow-spectrum, but incorporation of a fluoride
atom into the quinolone molecule produced fluoroquinolones, which are more potent
and broad-spectrum.
Pharmacokinetics
The quinolones are usually given orally or intravenously and distribute well throughout
body tissues. Levofloxacin is available for nebulisation, e.g. to treat chronic
respiratory infection in cystic fibrosis. Foodstuffs, iron preparations, and antacids in
the GI tract reduce absorption. Some of the drugs in this class, e.g. ciprofloxacin, are
enzyme inhibitors, decreasing the metabolising activity of cytochrome P450 in the liver
and inhibiting the metabolism of some other drugs, including warfarin.
Adverse Effects
Fluoroquinolones are generally well tolerated, but their more severe side-effects include
tendinitis, with possible rupture, especially in older people and most often affecting the
Achilles tendon. Central nervous system effects include headache, dizziness, and, rarely,
convulsions. They can also cause GI upsets, skin rashes, and cardiac arrhythmias.
Pharmacokinetics
The most common sulphonamide, sulphamethoxazole, has a plasma half-life of 10
hours and is mainly excreted in its unchanged form by the kidney. Care is therefore
needed in reduced renal function. It is well absorbed after oral administration and
distributes well, crossing the blood–brain barrier and the placenta.
Adverse Effects
Sulphonamides commonly cause allergic reactions, including potentially fatal
anaphylactic events. Serious hypersensitivity reactions include Stevens–Johnson
syndrome (see Fig. 3.13) and other serious skin reactions and can trigger or worsen
systemic lupus erythematosus. They can cause folic acid deficiency and so should be
used with caution in people predisposed to this, e.g. in alcohol dependency, pregnancy,
or malabsorption syndromes. Use is contra-indicated in the first trimester of pregnancy
because its antifolate action can cause developmental abnormalities in the fetus. Other
significant side-effects include bone marrow suppression, blood abnormalities, and
renal toxicity.
Trimethoprim
The clinical pharmacology of trimethoprim is very similar to that of the sulphonamides,
unsurprisingly so because of their closely related activities. Trimethoprim is also broad-
spectrum and is more frequently used as monotherapy than the sulphonamides. Rarely,
it can precipitate serious blood disorders, and patients on long-term therapy should be
monitored. Its half-life is between 8 and 10 hours, which can be prolonged in renal
impairment because a significant proportion of the drug is excreted in its unchanged
form.
Adverse Effects
Isoniazid is generally well tolerated. Liver impairment, renal impairment, and
ototoxicity are among the most serious side-effects. It can cause peripheral
neuropathy because it inhibits cellular use of vitamin B6 (pyridoxine), essential for a
wide range of important metabolic pathways including myelin synthesis. Co-
administration of vitamin B6 supplements prevents this and is important in people at
particular risk of nutritional deficiency, including in those who are alcohol-
dependent, in malnutrition, and in older people. It reduces the efficacy of L-DOPA in
the treatment of Parkinson’s disease because it inhibits the enzyme dopa
decarboxylase, which converts L-DOPA to dopamine (p. 54).
Rifamycins
This group of drugs includes rifampicin and rifabutin; rifampicin (rifampin) is the
main one used in TB. It is broad-spectrum, bactericidal, and particularly useful in
mycobacterial infections, including leprosy. It inactivates the bacterial enzyme RNA
polymerase so that the bacteria are unable to make RNA, including mRNA. Because
mRNA is the molecule used to transfer the genetic code from DNA to the ribosome for
protein synthesis, this prevents the microbe from producing essential proteins, and it
dies. It is used in short courses for a range of infections, including meningococcal
meningitis, but in TB it is given for up to 6 months.
Pharmacokinetics
Rifampicin is well absorbed when given orally and is taken up and concentrated
within macrophages and other immune cells, explaining its ability to target the
intracellular pathogen M. tuberculosis. It is a strong inducer of a range of liver
enzymes, and so it increases the rate at which a range of other drugs are cleared from
the plasma. Drugs affected include oestrogen (so rifampicin can cause contraceptive
failure), warfarin, phenytoin, and sulphonylureas. It is metabolised in the liver
and has a plasma half-life of around 3.5 hours. Care is needed in patients with
reduced liver function, in whom the drug can accumulate.
Adverse Effects
Rifampicin commonly causes nausea and vomiting. It distributes well, including into
the CSF, and can cause an orange discolouration of body fluids, including urine and
tears, which can stain soft contact lenses. It can cause blood disorders, and blood
counts should be monitored regularly in long-term therapy.
Ethambutol
Although the mechanism of action of this bacteriostatic agent is not known, it is
believed to block production of a substance called arabinogalactan, which
mycobacteria use in their cell wall. Because other types of microbes do not use these
pathways, the drug is only effective in mycobacteria. In the presence of ethambutol,
dividing bacteria cannot produce an intact cell wall and do not survive. It is given
orally and work is currently under way to develop a dry powder formulation for
inhalation. This would reduce the drug dose required and therefore limit systemic
toxicity. Some of the drug is metabolised in the liver, but about 50% is excreted
unchanged by the kidney. In people with normal renal function, its half-life is 3–4
hours, but because of the large contribution made by the kidney to clearing
unchanged drug from the plasma, renal impairment can significantly extend this. It
enters the CSF, so is effective in tuberculous meningitis. Its most significant side-
effect is a dose-dependent neuropathy of the optic nerve, leading to reduced visual
acuity and red–green colour blindness. Although some restoration of vision can occur
if the drug is stopped as soon as vision problems are detected, there is often residual
and permanent vision loss.
Pyrazinamide
Pyrazinamide is bacteriostatic and a pro-drug. It is taken up into infected
macrophages and enters the cell’s lysosomes, where the macrophage has enclosed the
mycobacteria in an effort to destroy them. The environment in these lysosomes is
acidic, which activates the drug. Once activated, the drug inhibits microbial synthesis
of important fatty acids, disrupting membranes within the cell. Energy production
takes place on internal membranes, so interfering with membrane structure interferes
with energy production and depletes the mycobacterium’s energy stores, without
which it cannot survive. Pyrazinamide is given orally and has a long half-life of 10
hours. It distributes well, including across the blood–brain barrier, so is effective in
treating tuberculous meningitis. About 70% of a dose of the drug is excreted
unchanged in the urine, so care must be taken in reduced renal function. It can cause
liver impairment, and liver function should be checked before commencing
treatment. Other side-effects include reduced appetite, skin reactions, and
development or worsening of gout.
AntiViral Drugs
Compared to antibiotics, there are relatively few antiviral drugs because viruses are
obligate intracellular parasites: that is, they enter host cells and insert viral DNA into the
host cell DNA. Directed by viral genes, host cell organelles and biochemical pathways
produce new viral proteins and nucleic acids, which the host cell then assembles into
new viral particles. Because each step in viral replication is performed by host cell
machinery, finding a selective drug target and avoiding poisoning the host cell is a
challenge. Many antiviral agents are significantly toxic and cause serious adverse effects,
especially in systemic use.
Replication of Retroviruses
Retroviruses are a type of RNA virus and contain RNA, which they convert to DNA using
an enzyme called reverse transcriptase which they carry with them in their capsid. This
DNA can then be inserted into the host cell DNA. Because reverse transcriptase is a
uniquely viral enzyme, it is an ideal selective target for drug action. Clinically important
retroviruses include HIV, and their biology is discussed in the section on HIV treatment.
AntiViral Resistance
As with antibacterial agents, viral resistance to antiviral drugs is spreading,
compromising the treatment of a range of infections. Viruses mutate readily and, unlike
animal cells, can remain viable even following significant alterations in their genetic
material. This further complicates efforts to find effective antiviral drugs, as well as
effective vaccines: if the virus constantly mutates, it presents the equivalent of a moving
target. Some clinically important viruses, such as rhinoviruses, responsible for the
common cold, exist as many serotypes, i.e. variants of the virus, all possessing slightly
different surface proteins. This means that even if a drug (or a vaccine) is developed
against one serotype, it is not necessarily effective against others. It also explains why it
is possible to catch the common cold on multiple occasions: antibodies produced against
the serotype causing this week’s cold will not protect against a different serotype
prevalent in 2 weeks’ time.
FIG. 11.17 The replication cycle of a DNA virus. Modified from Ryu
W-S (2017) Molecular virology of human pathogenic viruses, Fig.
3.1. Boston: Academy Press.
Latency
Latency is associated mainly with viruses of the herpes family, which have evolved this
strategy to resist elimination from their host organism and presents a significant
challenge to effective antiviral therapy. Herpes viruses are DNA viruses that include
herpes simplex (cold sores, genital sores, eye and central nervous system infections),
varicella zoster (chickenpox and shingles), Epstein–Barr virus (infectious
mononucleosis and a range of malignancies including Hodgkin’s disease), and
cytomegalovirus (a range of infections including hepatitis, and stillbirth and congenital
abnormalities). In latency, the virus scales all metabolic activity down to a very low level
but retains all its genetic material and is capable of rapid re-activation. Concealed within
its host cell, the virus effectively evades immune mechanisms, and because it is
metabolically inactive, is unaffected by antiviral drugs.
Antiviral Drugs
Most antiviral drugs are only effective when the virus is replicating because they inhibit
the synthesis of viral nucleic acids. Other mechanisms of action include prevention of
the uncoating step and inhibition of viral protein synthesis. Because of the pared-down,
relatively simple nature of viral biology, there is a limited range of viral-specific enzymes
available as drug targets.
Nucleoside Analogues
Examples: acyclovir, famciclovir, ganciclovir, ribavirin
These are sometimes called false nucleosides and are structurally very similar to the
nucleoside units assembled into new viral DNA. Acyclovir, famciclovir, ganciclovir,
and ribavirin all contain the normal base guanine (and so are sometimes referred to as
guanosine nucleosides), but instead of a normal sugar, they are attached to a different
chemical group which does not bind to incoming nucleotides. DNA polymerase mistakes
the drug molecule for the true nucleoside and inserts it into the growing nucleotide
chain. However, once the drug molecule is incorporated into a new nucleotide chain, no
further nucleotides can be added, and synthesis is terminated (Fig. 11.18C). Viral DNA
synthesis cannot be completed and so the production of new viral particles stops. These
drugs are most useful against herpesviruses, including cytomegalovirus. Ribavirin is
also used in hepatitis C and a range of respiratory infections.
FIG. 11.18 The mechanism of action of nucleoside inhibitors.A.
Nucleoside and nucleotide structure. B. Nucleotides linked in a
section of DNA. C. Nucleoside analogues, e.g. acyclovir, ribavirin,
and ganciclovir, are so similar in structure to the true analogue that
the polymerase enzyme builds new nucleic acid using them instead
of the true nucleosides. They cannot attach to incoming nucleosides,
and so the chain synthesis is terminated.
Acyclovir (Aciclovir)
This was the first drug in this class to be developed and entered the market in the mid-
1970s. When used topically, it is remarkably free of side-effects even though it blocks
DNA synthesis. Its selective toxicity is based on the fact that it is thirty times more active
against viral DNA polymerase than human DNA polymerase. In addition, it is a pro-
drug: it is only activated in host cells that have been infected with a virus. The result is
that it has very little effect on healthy host cell DNA production but is highly effective
against viral DNA synthesis in infected cells. However, viral resistance is an increasing
problem, usually because the virus produces slight variants of its DNA polymerase not
affected by the drug. Acyclovir is used topically, e.g. to treat cold sores, but needs to be
applied early to achieve effect. It is also given intravenously and orally to treat systemic
infections, and distributes well throughout body fluids, including crossing the blood–
brain barrier. Its very high selectivity for viral DNA polymerase means that even with
systemic use, it is usually well tolerated and does not cause significant adverse effects,
although encephalopathy has been reported. In systemic use, a good fluid intake is
important, because the drug can crystallise out in renal tubules, leading to inflammation
and renal impairment.
Foscarnet
Foscarnet binds to the active site on viral DNA polymerase and blocks its ability to add
nucleotides to a growing nucleotide chain. It is 100 times more selective for the viral
enzyme than the human enzyme, and so effectively blocks viral DNA production at
concentrations much lower than are needed to affect the host cell. It is most effective in
herpesvirus infections and is usually used in cytomegalovirus infections that have not
responded to a nucleoside analogue such as ganciclovir. It is poorly absorbed from the
GI tract and is given by slow intravenous injection. It causes a range of side-effects,
including serum electrolyte changes, which in turn can cause neurological abnormalities
including paraesthesia and seizures.
Pharmacokinetics
Nucleoside HIV reverse-transcriptase inhibitors (NRTIs) are given orally. Zidovudine
is a pro-drug and is activated in the infected cell. It is well absorbed and distributes
widely, including across the placenta and into the CSF.
Adverse Effects
NRTIs as a group are very toxic and cause significant side-effects that can involve any
body organ, probably by interfering with host cell energy pathways in the
mitochondria. Adverse effects include reduced white blood cell counts, bone marrow
disorders, and liver impairment. Regular full blood counts are needed, and care is
needed in reduced renal function, because the drug may accumulate if renal excretion
is impaired. The side-effects can be severe enough to require withdrawal of treatment.
Non-Nucleoside HIV Reverse-Transcriptase Inhibitors
Examples: efavirenz, etravirine, Rilpivirine
Non-NRTIs (NNRTIs), like NRTIs, inhibit HIV reverse transcriptase and prevent
the conversion of viral RNA into DNA. Their mechanism of action is, however,
different. They do not resemble normal nucleotides and are not assembled into new
DNA strands; instead, they bind to the enzyme, changing the shape of its active site
and deactivating it. The final result is the same: production of new DNA from viral
RNA is halted, and without it the virus cannot replicate. NNRTIs are less toxic and
more potent than NRTIs.
Pharmacokinetics
As a group, the NNRTIs have long half-lives of 2 days or more. Efavirenz is given
orally despite unpredictable GI absorption, and distributes widely, including crossing
the blood–brain barrier and causing central side-effects such as insomnia.
Etravirene is well absorbed when given orally, although absorption is more effective
following a meal, and is useful in treating HIV infections resistant to other NNRTIs.
Adverse Effects
NNRTIs can cause a range of adverse effects including hypersensitivity reactions and
GI upsets. Etravirine increases the risk of myocardial infarction, likely because it
interferes with glucose metabolism, can induce diabetes, and increases blood lipids.
HIV Protease Inhibitors
Examples: atazanavir, ritonavir, darunavir, saquinavir
The mechanism of action of protease inhibitors is described above (Fig. 11.19).
Saquinavir, approved in 1995, was the first protease inhibitor licensed to treat HIV
infection. It is given with ritonavir because ritonavir inhibits the liver enzyme
CYP450, which breaks saquinavir down. In this way, ritonavir increases the half-life
and plasma levels of saquinavir, and for the same reason is often used in low
concentrations with other protease inhibitors.
Pharmacokinetics
Inhibiting metabolism of these agents with ritonavir extends their half-lives
sufficiently that most require only once-daily dosing.
Adverse Effects
The protease inhibitors cause a range of adverse effects, including peripheral
neuropathy, seizures, and hypersensitivity reactions including Stevens–Johnson
syndrome (Fig. 3.13).
Antifungal Drugs
Key Definitions
• Mycosis:
an infection caused by a fungus
• Yeast:
• Mould:
a fungus that grows as multicellular colonies in the form of long, thread-like hyphae
There may be as many as five million species of fungus on Earth, of which only a few
hundred cause human disease. Most fungal infections are superficial, involving the skin,
nails, or mucous membranes. Very few fungi are pathogenic enough to cause systemic
infection in otherwise healthy people, and in general, those with functionally effective
immune systems rarely develop systemic fungal infection, except in certain
circumstances, for example following broad-spectrum antibiotic treatment. In the
vulnerable, fungal infections can be life-threatening: for example, Cryptococcus
neoformans, a very common yeast, causes no problems in healthy individuals but
produces cryptococcal meningitis in immunocompromised people. In recent decades,
the rate of invasive fungal infections is rising, because the numbers of
immunocompromised people are rising: an ageing population, whose immunity is
naturally declining, along with rising rates of certain chronic disorders, e.g. cancers and
HIV infection, are significant contributors to this. Globally, the burden of fungal disease
is disproportionately borne by resource-limited populations, with poor living conditions
and limited diagnostic and treatment facilities.
A much wider range of fungi can cause systemic infections, which may establish in a
range of body organs. Frequently affected is the respiratory system, because inhalation
of fungal spores is a common route of infection. Fungi can also access the internal
environment via wounds, or the GI or genitourinary tracts, and cause sepsis,
endocarditis, meningitis, arthritis, osteomyelitis, peritonitis, and pyelonephritis. Many
plant fungi produce toxins, which may be stable to food processing and storage
processes especially if not carried out to high-enough standards, and then consumed in
contaminated foods. For example, aflatoxins produced by Aspergillus species
contaminating cereals can cause hepatitis and liver cancer.
Echinocandins
Examples: caspofungin, micafungin, anidulafungin
These are semi-synthetic agents, based on antimicrobial substances produced
naturally by a range of fungi. The first of this family of drugs, caspofungin, was made
available in 2001. Their target is an enzyme that produces glucans, essential
components of fungal cell walls. Because glucans are not used in mammalian biology,
this pathway provides a selective drug target. Echinocandins are only active against
Candida and Aspergillus species.
Pharmacokinetics
As a group, the echinocandins have long half-lives: caspofungin’s half-life is up to 50
hours. They are heavily plasma protein-bound and although they distribute well
throughout tissues, very little enters the CSF. They are not absorbed across the wall of
the GI tract and must be given intravenously.
Adverse Effects
These drugs cause a range of side-effects but are generally well tolerated, better so than
other classes of systemic antifungals. They can trigger histamine release from mast
cells, which can cause bronchospasm, bradycardia, hypotension, itch, and angioedema.
Antifungals that Interfere with Cell Membrane Integrity
The cell membrane controls the entry and exit of substances in and out of the cell, and
damage leads to microbial cell death.
Nystatin
Nystatin, one of the WHO’s essential medicines, was isolated in 1950 from a soil-based
bacterium. Although it is broad-spectrum, it is highly toxic in systemic use so is mainly
used topically to clear Candida infections. It can be given orally to clear infections of the
GI tract, including oral thrush, because its molecules are large, bulky, and highly ionised
and so are not absorbed, avoiding systemic effects. Nystatin binds directly to ergosterol
and generates damaging oxidising free radicals, which punch holes in the membrane.
This immediately allows rapid and uncontrolled movement of fluid and other
substances in and out of the fungal cell, killing it. Resistance to nystatin is very rare,
because ergosterol is so fundamentally important to fungal cell membrane biology that
it is preserved throughout generations. However, reports of resistant isolates are
increasing.
Amphotericin
Amphotericin works like nystatin. It is however used systemically for a range of serious
fungal infections, including invasive candidiasis, aspergillosis, and cryptococcus
infections. It must be given intravenously and is very toxic because although its favoured
target is fungal ergosterol, it also binds to cholesterol in host cell membranes, giving
widespread adverse effects. Amphotericin is both nephrotoxic and hepatotoxic, and
kidney and liver function should be monitored during treatment. The drug should be
immediately stopped if liver function tests begin to show liver impairment.
Terbinafine
Terbinafine prevents ergosterol synthesis by inhibiting the enzyme that produces it.
Without an adequate supply of ergosterol, the fungal cell cannot grow and divide
because it cannot make a functionally intact cell membrane. Terbinafine is strongly
keratinophilic and accumulates in skin and nails, which are rich in this protein, and is
very useful in skin and nail infections. It is broad-spectrum and can be given orally or
topically. It is highly lipophilic, so distributes very well throughout body tissues, and is
generally well tolerated even in extended oral therapy for stubborn nail infections,
which can be up to 6 months’ duration. It can however cause severe hypersensitivity
reactions including Stevens–Johnson syndrome (see Fig. 3.13) and hepatotoxicity,
which requires discontinuation of the drug.
Azoles
Triazoles: fluconazole, itraconazole, voriconazole
Imidazoles: ketoconazole, clotrimazole, econazole
The triazoles and imidazoles are closely related synthetic antifungals used in a wide
range of infections. Clinically, the triazoles are all used systemically whereas the
imidazoles are generally used topically. Although ketoconazole was the first antifungal
azole licensed for oral use, it was withdrawn from systemic therapy in the UK in 2013
because of hepatotoxicity and is rarely used systemically elsewhere because there are
safer drugs available. As a group, the azoles act by blocking ergosterol synthesis. This
prevents the fungal cell from producing new cell membrane, so that it cannot grow or
divide.
Pharmacokinetics
Orally administered agents have long half-lives; for example, fluconazole has a half-
life of 25 hours and itraconazole’s half-life is 21 hours. Fluconazole is well absorbed
with a bioavailability of over 90%, distributes into the CSF, and is one of the least toxic
azoles. Itraconazole is erratically absorbed from the GI tract. Absorption of the liquid
formulation is better than the capsules and is even better when gastric pH is low. It is
therefore best taken on an empty stomach because food and drink in the stomach dilute
the contents and increase pH. The azoles are metabolised in the liver and itraconazole
is the only agent to have an active metabolite.
Adverse Effects
Systemic use can cause a range of potentially significant side-effects, including GI
disturbances, blood disorders, and skin reactions. Azole antifungals are also associated
with multiple drug interactions via a range of mechanisms. One important cause of
interactions is azole-mediated inhibition of the important metabolising liver enzyme
CYP3A4, which metabolises a wide range of drugs, including anticonvulsants, non-
steroidal anti-inflammatory drugs, immunosuppressants, benzodiazepines,
antidepressants, antivirals, anticoagulants, and many others. When co-
administered with an azole antifungal, increased plasma levels of these drugs should be
anticipated and managed appropriately. Even when applied topically, absorption
through the skin can lead to plasma levels high enough to cause systemic side-effects,
including these drug interactions. Because azoles are better absorbed when gastric pH is
low, co-administration with drugs that reduce stomach pH, e.g. proton-pump
inhibitors and antacids, reduces their absorption. Itraconazole should be used with
caution in patients with heart disease because it has a direct, negative inotropic effect on
the myocardium: that is, it reduces cardiac contractility, although the mechanism by
which it does this is uncertain. Posaconazole can cause drowsiness and impair motor
skills such as driving.
Griseofulvin
Griseofulvin is produced naturally by Penicillium species and is thought to interfere with
the function of microtubules in the cell. Microtubules are hollow threads made of
protein, used to give internal structure, to aid in motility in motile cells, and to guide
and transport materials, granules, and organelles around within the cell. They form the
mitotic spindle when a cell is dividing, which acts as tramlines to direct chromosome
movement to each end of the cell. Disruption of these guiding pathways prevents mitosis
and blocks fungal cell division. Griseofulvin is given orally, although its absorption is
poor; fatty foods in the GI tract improve this. It has a high affinity for keratin and
accumulates in hair, nails, and skin, and is used in tinea infections. Its half-life can be up
to 21 hours and it should not be used in serious liver disease. It is teratogenic in animal
studies, and effective contraception should be practiced if sexually active people are
being treated with the drug. It is a potent inducer of liver cytochrome P450 enzymes and
may therefore reduce the effect of a wide range of other drugs, including oral
contraceptives and warfarin. It commonly causes GI side-effects and may cause
serious skin reactions and peripheral neuropathy.
Flucytosine
Flucytosine is a synthetic agent mainly active against Candida and Cryptococcus
species. Within the fungal cell, it is converted to 5-fluorouracil (5-FU, used in the
treatment of some cancers, see also p. 236). 5-FU binds to and inhibits the enzyme
thymidylate synthase, which builds RNA and DNA. Therefore, in the presence of the
drug, the fungal cell can neither produce new proteins nor copy its DNA for cell division
and does not survive. Resistance develops rapidly if it is used alone, and it is usually
paired with amphotericin to prevent this. It is usually given intravenously and
distributes well throughout body tissues, including into the CSF. Little is metabolised in
the liver, so most is excreted unchanged by the kidney and the dose must therefore be
reduced in patients with poor renal function. Flucytosine can inhibit blood cell
production, probably because it exerts its antiproliferative action in the rapidly dividing
tissues of the bone marrow, and blood counts should be made weekly.
Antiparasitical Drugs
All living creatures are susceptible to colonisation by parasites, organisms that establish
themselves within the body of the host and survive and proliferate at the host’s expense
without providing any benefit in the relationship. Viruses by definition are therefore
parasitic and are considered above. Parasites may enter the body via insect bites,
contamination of wounds, via the GI tract in contaminated food or water, or from
mother to child across the placenta. The main groups of organisms that parasitise
humans and cause disease include protozoa, helminths (worms), and arthropods
(insects).
Protozoal Infections
Protozoa, a diverse group of single-celled organisms that vary hugely in size, preferred
habitats, feeding habits, and methods of locomotion, include Plasmodium, Leishmania,
and Trichomonas species.
Antimalarial Drugs
Malaria is caused by five different species of Plasmodium, which spend part of their
lifecycle in the Anopheles mosquito and part in their human host. The WHO estimated
the total number of cases worldwide in 2019 was 219 million, 94% of which occurred in
Africa.
Chloroquine
Chloroquine has been used to treat malaria since the 1940s and is still used today,
although resistance is a growing problem. Plasmodium spends part of its life cycle in the
infected person’s erythrocytes, where they ingest haemoglobin, the haem portion of
which is toxic to the parasite. Normally, the parasite converts haem to non-toxic
metabolites, but chloroquine and the related agents, quinine and mefloquine inhibit
this. Accumulation of toxic haem poisons and kills the parasite. Mefloquine is given
orally, and quinine and chloroquine are given orally or intravenously. These drugs
cause GI upsets, and a range of more significant problems: for example, at higher doses,
chloroquine binds to melanin in the retina and damages the photoreceptors, leading
to permanent loss of vision. They interfere with cardiac conduction and should be used
with care in patients with known conduction disorders.
Primaquine
Although structurally related to chloroquine, primaquine interrupts the Plasmodium
life cycle at a different point. Prior to entering host erythrocytes, the parasite spends
time in the host liver, and while it is at this stage, primaquine poisons it by inhibiting its
mitochondrial ATP production. It is given by mouth, is well absorbed, and is usually well
tolerated although it can cause GI upsets. Care should be taken in patients who are
deficient in glucose-6-phosphate dehydrogenase, and all patients should be tested prior
to beginning treatment. This enzyme is important for protecting the plasma membrane
of red blood cells, and a degree of deficiency is relatively common in the general
population. Primaquine worsens the effects of this deficiency and can cause serious
haemolysis.
Helminthic Infestation
Helminths (worms) may be transmitted orally, from contaminated food, water, or soil,
or by invasion through the skin. Some helminths, e.g. tapeworms (Fig. 11.23) and
roundworms, live in the host’s GI tract, and others, e.g. Schistosoma species (flukes),
invade other body tissues, e.g. the eye or the liver. Helminthic infections are of global
importance: WHO 2020 data estimates that 24% of the world’s population are infested
with soil-transmitted helminths, most of which occur in tropical and sub-tropical areas.
Antihelminthic Drugs
Ivermectin
Ivermectin is given orally to treat a range of helminthic infestations and is also effective
against the scabies mite Sarcoptes scabei. It interferes with calcium entry into the
parasite’s nervous and muscle tissue, paralysing it and causing death. It does not enter
the CSF in humans, thought to be the main reason why it causes few side-effects.
Benzimidazoles
Examples: mebendazole, albendazole
These drugs inhibit motility in helminths by blocking their ability to take up glucose.
Starved of their main source of energy, the helminth is paralysed and dies.
Mebendazole is given orally, and little is absorbed across the wall of the GI tract. Most
of what is absorbed is destroyed by first-pass metabolism in the liver, so this drug has
little systemic toxicity in humans and is mainly used to treat infestations of the GI tract.
Insecticides
The two main agents used to treat these infestations are permethrin and malathion,
applied topically, usually as creams and lotions.
Permethrin
Permethrin is a neurotoxin, blocking nerve conduction in the insect’s nervous system. It
is poorly absorbed through the skin, so has no measurable effect on the host’s
neurological function, although it can cause local irritation at the site of application.
Resistant variants in insect populations have been reported.
Malathion
Malathion is an organophosphate insecticide, a neurotoxin that blocks the enzyme
acetylcholinesterase. This enzyme clears the neurotransmitter acetylcholine from nerve
endings, including at the neuromuscular junction (the synapse of a motor nerve at
skeletal muscle tissue). Acetylcholine therefore accumulates, and its action is
significantly prolonged causing lethal interference with normal neurological function.
When applied topically, some drug is absorbed across the skin, but it is rapidly
metabolised, and does not cause measurable change in host nervous system function.
Resistance has been reported, but less frequently than permethrin.
References
1. Bhat Z.S, Rather M.A, Maqbol M, et al. Drug targets exploited in Mycobacterium
tuberculosis: pitfalls and promises on the horizon. Biomed.
Pharmacother. 2018;103:1733–1747.
2. Dinos G.P. The macrolide antibiotic renaissance. Br. J.
Pharmacol. 2017;174(18):2967–2983.
3. Gest H. The discovery of microorganisms by Robert Hooke and Antoni van
Leeuwenhoek, Fellows of the Royal Society. Notes Rec. R. Soc. 2004;58(2):187–
201.
4. Lobanovska M, Pilla G. Penicillin’s discovery and antibiotic resistance: lessons
for the future. Yale J. Biol. Med. 2017;90(1):135–145.
5. Rehman K, Kamran S.H, Akash M.S.H. Toxicity of
antibiotics. In: Hashmi M.Z, ed. Antibiotics and Antimicrobial Resistance Genes
in the Environment. Oxford: Elsevier; 2020.
6. Rubinstein E, Lagace-
Wiens P. Quinolones. In: Cohen J, Powderly W.G, Opal S.M, eds. Infectious
Diseases. fourth ed. Oxford: Elsevier; 2017.
7. Wanger A, Chavez V, Huang R.S.P, et al. Antibiotics, antimicrobial resistance,
antibiotic susceptibility testing and therapeutic drug monitoring for selected
drugs. In: Wanger A, Chavez V, Huang R.S.P, et al., eds. Microbiology and
Molecular Diagnosis in Pathology: A Comprehensive Review for Board
Preparation, Certification and Clinical Practice. Oxford: Elsevier; 2017.
Online resources
National Institute for Health and Care Excellence, . Antimicrobial stewardship:
systems and processes for effective antimicrobial medicine use: NICE guideline
[NG15] Available at:. https://www.nice.org.uk/guidance/ng15, 2015.
Preston S.L, Drusano G.L. Penicillins. Antimicrobe. 2017 Available
at:. http://www.antimicrobe.org/d24.asp.
Very good review of the pharmacology of the penicillins.
Reygart W.C. An overview of the antimicrobial resistance mechanisms of
bacteria. AIMS Microbiol. 2018;4(3):482–501 Available
at:. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6604941/.
Uthayakumar A. Cutaneous adverse reactions to
antibiotics. DermNet. 2018 Available
at:. https://dermnetnz.org/topics/cutaneous-adverse-reactions-to-
antibiotics/.
Clear and well-illustrated guide to skin reactions caused by antibiotics.
World Health Organisation. Global Tuberculosis Report: Executive
Summary. 2020 Available at:. https://www.who.int/docs/default-
source/documents/tuberculosis/execsumm-11nov2020.pdf?
sfvrsn=e1d925f_4.
12: Cytotoxic Drugs
Introduction
Introduction
The literal meaning of ‘cytotoxic’ refers to an agent which damages or kills a living cell,
and in its widest sense includes adverse effects of drugs in physiological systems and
antimicrobial agents used to kill or halt division of pathogenic micro-organisms. Its
common use, however, has come to mean a drug used to treat cancer, and because
cancer is such a common disease, the study and development of cytotoxic drugs is one of
the most rapidly advancing areas in pharmaceutical research. Cancer is a disease that
predates the history of our species. The oldest known case of cancer is an osteosarcoma
in a toe bone of an earlier relative of Homo sapiens and is around 1.7 million years old
(Fig. 12.1). Ancient Egyptian mummified remains show lesions indicative of bone
cancer, and cases of what is clearly breast cancer are described in ancient Egyptian
writings. The word ‘cancer’ is believed to have been first coined by Hippocrates (460–
370 BC), the Greek physician sometimes also referred to as the father of medicine. It
derives from the Greek word for ‘crab’, and likely originates because of the finger-like
projections of abnormal tissue extending from a superficial breast tumour and which
distort the surface of the breast. Chemotherapy, the treatment of cancer with cytotoxic
agents, is the most recent of the three main therapeutic approaches, the others being
radiotherapy and surgery.
The prospect of using drugs to treat cancer first emerged in the 1940s. Following the
observation that the highly toxic chemical warfare agent sulphur mustard (mustard gas)
caused a drastic fall in white blood cell counts, it was trialled in the treatment of
lymphoma. In the 1950s, antifolates including methotrexate were used to treat
leukaemia and other cancers. Since those early days, as understanding of the genetic,
immunological, hormonal, and molecular basis of malignancy rapidly advanced, it has
become clear that cancer is a collection of at least 200 different types. Based on this, a
raft of new therapies has been developed, and the concept of targeted treatments has
become a standard approach in many cancers. Cytotoxic drugs are amongst the most
noxious therapeutic agents in clinical pharmacology because of their harmful actions on
healthy tissues. However, our expanding understanding of the biochemistry, genetics,
and molecular biology of cancer cells is driving the development of novel drugs which
selectively target some aspect of malignant cell biology, reducing toxicity and improving
the tolerability and efficacy of treatment regimens. Reflecting the ingenuity of the
researchers engaged in this work, a bewildering and expanding array of drugs with a
wide range of highly specific mechanisms of action is now available, many of which are
only helpful in a small number of cancers because of their targeted action. This chapter
does not aim to provide a comprehensive list of chemotherapeutic agents: indeed, with
the speed with which the field is advancing, it is likely that any such list would be
incomplete at publication anyway. However, it explains the scientific principles
underpinning the pharmacology of the main groups of anticancer drugs and discusses
important examples in more detail.
With improved diagnostics and treatment options, survival times and cure rates for
many cancers have improved dramatically, but globally cancer remains the second
leading cause of death.
Key Points
All tumours originate from a body cell whose DNA has suffered one or usually more
mutations, the consequence of which is to release the cell from normal growth
controls and allow it to replicate in a disorderly and unregulated fashion. The cell
population produced from this malignant transformation becomes progressively more
genetically unstable as the tumour expands, accumulating more and more mutations
and becoming more and more abnormal. Cancers can be classified according to their
tissue of origin:
• Carcinomas originate in epithelial tissues. Epithelial tissues cover and line body
organs and surfaces, e.g. the skin and the lining of the gastrointestinal tract, and
cell division rates are high. Multiple cell divisions increase the likelihood of
copying errors occurring in the cell’s DNA, leading to mutations appearing in the
cell line. Eighty to ninety percent of cancers are carcinomas.
• Sarcomas originate in connective tissues like bone, cartilage, fat, and muscle.
• Blastomas originate in precursor cells (blasts). These cancers are most common in
children.
Ninety-eight percent of the DNA in a healthy human cell does not code for a protein
and is referred to as ‘non-coding’. Some of this DNA regulates gene expression, but most
of it has no known function. The sections of DNA that do carry the code for a protein are
called genes and are found at specific locations on a particular chromosome: for
example, the gene that codes for insulin is found on chromosome 11. The human
genome, the complete set of genes within the cell, contains between 20,000 and 25,000
genes.
DNA Structure
The DNA molecule is formed of two chains twisted around each other into a double helix
like a twisted ladder (Fig. 12.2). The ‘uprights’ of the ladder are formed of alternating
phosphate and sugar (deoxyribose) groups. Each sugar group has a base attached to it,
and the bases on one chain are bound to the bases on the other chain with hydrogen
bonds, stabilising the molecule and forming the ‘rungs’ of the ladder. Each sugar/base
group is collectively called a nucleoside (see Fig. 11.18), and the sugar/base/phosphate
unit is called a nucleotide (Fig. 12.2).
There are only four bases in DNA, thymine, adenine, guanine, and cytosine, and they
display specific pairings. Thymine always binds to adenine and guanine to cytosine.
Therefore, knowing the base sequence on one DNA strand accurately identifies the base
sequence on the second strand. This is called complementary base pairing. The genetic
code is written in the base sequences. Mutations in the base sequence of genes generate
errors in the code and lead to the production of faulty, often non-functional protein.
Chemically speaking, adenine and guanine are purines, and thymine and cytosine are
pyrimidines. Pyrimidine analogues, e.g. fluorouracil (FU), and purine analogues, e.g.
mercaptopurine, are long-standing members of the chemotherapeutic drug armoury,
and act by substituting for the true base when a cell is producing new nucleic acids.
Nucleic acid containing the fake bases is non-functional.
FIG. 12.2 The structure of DNA.DNA is formed from two chains of
alternating sugar and phosphate groups, linked in a ladder-like
structure by bases attached to the sugar units. The molecule is then
twisted into a double helix. From Thibodeau G and Patton K (2019)
Anatomy and physiology, 10th ed. St. Louis: Mosby.
DNA Replication
In body cells preparing for mitosis, the cell’s DNA is duplicated, the fundamental event
in the S phase of the cell cycle (Figs 12.3 and 12.5). DNA replication is a complex event
involving several important enzymes, some of which are important targets for cytotoxic
drugs. The DNA molecule is first uncoiled by DNA helicase, and the hydrogen bonds
between the bases of the two strands are broken. This gives two complementary single
strands of DNA, each of which is used to make a new double-stranded molecule: two
identical daughter molecules are therefore produced from the original parent molecule.
The enzyme which assembles the second strand on each original strand is called DNA
polymerase. DNA topoisomerase stabilises and repairs DNA during DNA replication,
and DNA ligase stitches newly synthesised segments of DNA together.
Because malignant cells usually divide more quickly than normal cells, DNA
replication is an established chemotherapeutic target. Anticancer drugs can interfere
with DNA synthesis in a number of ways. For example, cytotoxic anthracyclines such as
doxorubicin and idarubicin insert themselves between the base ‘rungs’ of DNA
(intercalation), damaging the molecule and preventing DNA replication.
Cell cycle-checkpoint defects are characteristic of cancer cells, leading to repeated and
uncontrolled cell division. Each time a damaged cell is allowed to proceed round the cell
cycle and divide, the number of mutated cells increases, and because one mutation
predisposes the cell to developing others (genetic instability), it leads to an expanding
population of cells accumulating multiple mutations and increasing risk of malignant
transformation. Some of the most common genetic mutations leading to checkpoint
failure are discussed below; they are an important target in anticancer drug research.
Key enzymes called cyclin-dependent kinases (CDKs) drive the cell cycle; CDKs need
to bind to a regulatory protein called cyclin for activation. Both are potential targets in
cancer drug development.
Apoptosis
Apoptosis is programmed cell death, sometimes referred to as cell suicide. It is an
essential part of embryonic and fetal development, during which many more cells are
produced than are actually needed, and the extra cells are eliminated by apoptosis to
bring cell numbers down to within the desired range. Old, damaged, or mutated cells are
also disposed of this way, as are cells that have arrested during mitosis, including as a
result of chemotherapy. Apoptosis is therefore essential for growth and repair and in
maintaining healthy function of cell populations in adult life. The key enzymes that drive
apoptosis are called caspases. During apoptosis, the cell undergoes controlled but rapid
disintegration: typically, half an hour is all that is needed for a cell to destroy itself. As
the cell breaks up, macrophages rapidly scavenge the fragments to prevent the cellular
contents from stimulating an inflammatory response. Identifying drugs that selectively
stimulate apoptosis in cancer cells is an active area in anticancer drug development.
Fig. 12.6 shows advancing stages of apoptosis in a malignant pig kidney cell exposed to
the chemotherapeutic drug etoposide.
FIG. 12.4 Transcription and translation: the role of DNA, RNA,
and ribosomes in protein synthesis. From Waugh A and Grant A
(2020) Ross & Wilson anatomy and physiology in health and illness,
14th ed, Fig. 17.5. Oxford: Elsevier.
Telomerase
At each end of a chromosome is a section of DNA which is there to protect the
chromosome end and prevent it from damage, a bit like the short plastic sleeves
protecting the ends of shoelaces. These protective caps are called telomeres. With each
cell division, the telomere gets shorter. When it becomes too short to protect the
chromosome end, cell division is arrested, and the cell may even have its apoptosis
genes activated. This limits the number of divisions that a cell can undergo and ensures
that older cells are shunted out of the cell cycle. Telomerase is the enzyme that builds
telomere DNA onto the ends of chromosomes. Healthy body cells do not produce this
enzyme, but cancer cells frequently do. This allows them to constantly repair the
telomere caps on their chromosomes, theoretically allowing them to continue dividing
indefinitely, producing expanding populations of malignant cells. Telomerase offers a
selective target for cancer drugs; however, although telomerase inhibition strips the
cancer cells of their immortality, it does not disrupt their ability to divide or exert direct
cytotoxic effects. Although research in this area is ongoing, it has so far produced limited
success and only one telomerase inhibitor, imetelstat, has so far reached the market.
Angiogenesis
When a tumour is very small, less than 2 mm in diameter, nutrients and oxygen can
reach its centre by simple diffusion. As the tumour grows, diffusion is inadequate to
supply its metabolic requirements, and it needs its own blood supply. Cancer cells
produce growth factors like vascular endothelial growth factor (VEGF) that stimulate
blood vessel growth (angiogenesis), developing a vascular network within the growing
tumour and supporting its expansion. Several anticancer drugs, including aflibercept
and sorafenib, inhibit angiogenesis.
Motility
During periods of normal tissue growth, e.g. embryogenesis and wound healing, newly
divided cells may be motile, allowing them to migrate along pre-determined pathways to
their destination. This is a tightly regulated process, and once at its destination, the cell’s
motility is inhibited, and the cell takes up its position in the developing tissue. Cancer
cells display increased and unhindered motility which allows them to invade locally and
to cross basement membranes and other physiological barriers, permitting distant
spread. Inhibition of cancer cell motility is a potential target in cancer therapeutics and
several agents currently under investigation have shown promise in animal models.
FIG. 12.6 The progression of apoptosis in a malignant pig kidney
cell exposed to etoposide. Courtesy L.M. Martins and K. Samejima,
Wellcome Trust Institute for Cell Biology, University of Edinburgh,
UK.
Genetic Instability
Genetic instability is the increased likelihood of further mutations when a mutated cell
divides: that is, once one mutation is present, that cell will accumulate more and more
mutations as it continues to divide. This is an important feature in cancer cells because
cancer requires more than one, often multiple, mutations to be present. This can be
exploited in chemotherapy because the more mutations accumulate, the likelier it is that
the cell will be tripped into apoptosis and destroy itself. Poly-ADP ribose polymerase
(PARP) inhibitors such as niraparib prevent a tumour cell from repairing DNA breaks
and so push it towards cell suicide.
Proto-Oncogenes
Proto-oncogenes are healthy genes important in embryonic development, in growth, in
healing and repair, and in replacement of cells that have reached the end of their useful
life. They code for proteins which stimulate cell division and differentiation, and which
inhibit apoptosis. Mutations in a healthy proto-oncogene can produce an abnormal
oncogene (Fig. 12.8), which produces excessive quantities of a growth-stimulating
protein, driving the cell through the cell cycle and allowing it to escape normal growth
checks and halt signals. This is a common feature in cancer, and oncogenes are an
important molecular target for the design of new cytotoxic drugs.
One example illustrating how understanding the molecular genetics of an oncogene
has translated into clinical therapeutics is the story of trastuzumab (Herceptin), which
came on the market in 1998. In the mid-1980s, researchers discovered an oncogene,
called HER2, which produces a protein called human epidermal growth factor receptor
(HER) 2. HER2 belongs to a family of cell-surface receptors called epidermal growth
factor receptors (EGFRs). Epidermal growth factor (EGF) is a common growth factor
which stimulates cell division in a range of cell types by binding to EGFRs. It was then
shown that the HER2 gene is overactive in a subset of women with breast cancer,
increasing HER2 protein expression on the cell surface and increasing the cells’
responsiveness to growth stimuli and promoting tumour growth. This type of breast
cancer was designated HER2-positive. Trastuzumab is a monoclonal antibody which
binds to and blocks the HER2 protein at the cell surface, blocking the incoming growth
stimuli and preventing the cell from dividing. Introduction of trastuzumab into the
treatment regimes for HER2-positive primary and metastatic breast cancers
significantly improves survival and reduces relapse rates: it has been a game-changing
medical advance for this cohort of patients. It has also since been shown that the HER2
oncogene is involved in other cancers including some stomach and oesophageal
malignancies, and trastuzumab is now also used in these cases (Fig. 12.9).
Tumour-Suppressor Genes
Genes that produce proteins with anti-proliferative properties are called tumour-
suppressor genes. These proteins act as brakes on the cell cycle, suppress cell division,
and induce apoptosis in old and damaged cells. Mutations in one or more tumour-
suppressor genes are commonly found in cancer cells. Important tumour-suppressor
genes include TP53, which is mutated in over half of human cancers. The protein it
codes for, p53, is essential for normal function of the G1/S restriction checkpoint in the
cell cycle. Mutation of the TP53 gene and loss of p53 allows damaged and abnormal cells
to progress through this checkpoint, proceed through the cell cycle, and divide. Other
tumour-suppressor genes directly linked to cancer include BRCA1 and BRCA2.
Mutations in the BRCA (Breast Cancer) genes increase the risk of a range of
malignancies, including breast, prostate, pancreatic, and ovarian cancers. The proteins
coded for by these genes are enzymes that ensure that damage to the cell’s DNA is
accurately repaired and so maintain DNA integrity. Loss of these enzymes leads to DNA
instability, which predisposes the cell to malignant transformation. An understanding of
the basic science underlying BRCA function led to the development of the PARP
inhibitors, like niraparib (see below).
Teratogenicity
Women of childbearing age are always counselled to avoid pregnancy during
chemotherapy because chemotherapeutic agents are almost always teratogenic in
animal studies. The anti-proliferative and growth-suppressive activity of most
chemotherapeutic agents confer significant risk of birth defects, especially in the first
trimester.
Future Malignancies
Many chemotherapy agents kill or eliminate cancer cells by damaging DNA so are
themselves carcinogenic. Their action also extends to healthy tissues, and even if the
cancer is cured, there is the likelihood that mutations in normal cells can persist and
themselves cause secondary cancers, usually haematological, sometimes years down the
line.
Reproductive Consequences
Cytotoxic drugs may eliminate or critically damage stem cells in the testes and
permanently damage the oocytes of the ovary, leading to reduced or lost fertility.
Freezing of sperm or ova in advance of chemotherapy may be an option in patients who
may want to have a family in the future.
Taxanes
The first taxane, paclitaxel, was derived from fungi living in the bark of the Pacific yew
tree (Taxus brevifolia) and was first tested in cancer in the early 1970s. The newer
taxanes are semi-synthetic taxane derivatives. These drugs prevent tubulin proteins
from dissociating from each other (depolymerisation), which prevents the normal
dynamic shortening and lengthening of spindle fibres essential for pulling chromosomes
around within the cell (Fig. 12.11C). In addition to inhibiting mitosis, taxanes activate
caspases, the enzymes that drive apoptosis, stimulating programmed cell death. The
taxanes are metabolised in the liver. On infusion, they commonly cause mild to severe
hypersensitivity reactions including urticaria, angioedema, and bronchoconstriction,
and an antihistamine or corticosteroid is usually given in advance to reduce the risk or
severity of this.
Fluorouracil
Fluorouracil is a pyrimidine analogue which is taken up more extensively in malignant
cells than in healthy cells and binds to and blocks thymidylate synthetase (Fig. 12.12).
Thymidylate synthetase converts 2-deoxyuridylate to 2-deoxythymidylate. This is a key
step in the synthesis of DNA, and FU therefore blocks the cancer cell’s ability to replicate
its DNA and prevents it from dividing. It was first produced in 1957 and is still widely
used today in a range of solid tumours, including some GI and breast cancers, and in
pre-malignant and malignant skin conditions such as actinic keratosis. It has a short
plasma half-life of 10–20 minutes and is metabolised mainly in the liver. Among others,
it causes the standard range of cytotoxic adverse effects (see above) and may cause
cardiotoxicity. Capecitabine and tegafur are pro-drugs which are converted in the
tumour to FU.
Mercaptopurine
Mercaptopurine is used to treat leukaemia and is used for its immunosuppressant action
in some autoimmune disorders including inflammatory bowel disease and severe
psoriasis. It is an adenine analogue, is mistaken for a true adenine nucleoside, and is
incorporated into DNA and RNA in cancer cells. It can cause the standard range of
cytotoxic adverse effects and may cause significant hepatotoxicity: liver function should
be monitored while the drug is being used.
Topoisomerase Inhibitors
Examples of topoisomerase I inhibitors: irinotecan, topotecan
Examples of topoisomerase II inhibitors: etoposide
Topoisomerase I and II are essential enzymes in DNA synthesis and repair. They
cause the general cytotoxic adverse effects described above. Topoisomerase II inhibitors
increase the risk of developing acute myeloid leukaemia later in life.
Topoisomerase I Inhibitors
Topoisomerase I stabilises and repairs DNA during transcription and replication, when
the DNA strands are separated and vulnerable to damage. Irinotecan and topotecan
are derived from camptothecin, which was first isolated from the bark of the Chinese
tree Camptotheca acuminata and has been used in traditional Chinese medicine for
thousands of years. They bind to and block topoisomerase I, causing double-strand
breakages in the DNA molecule which are not repaired and lead to cell death. They are
not absorbed when given orally and so are administered intravenously. Care should be
taken in patients with hepatic or renal impairment. Irinotecan is used in advanced
colorectal cancer and topotecan is used in ovarian, cervical, and small-cell lung cancer.
Topoisomerase II Inhibitors
Topoisomerase II stitches together (ligates) newly synthesised fragments of DNA in
DNA synthesis. Topoisomerase II inhibition causes double-strand breaks in DNA, which
are not repaired and cause cell death. Etoposide is derived from the mandrake root
Podophyllum paltatum. It binds to and inhibits topoisomerase II, preventing DNA
ligation and leaving the cell with non-survivable DNA damage. It is given orally, is partly
metabolised in the liver and partly excreted unchanged, and has a plasma half-life of 4–
11 hours. It is used in testicular cancer, in lymphoma, and in small-cell lung cancer.
Folate Antagonists
Examples: methotrexate, pemetrexed, raltitrexed, fluorouracil
Vitamins are co-factors, essential ‘helper’ substances without which an enzyme cannot
function. Folic acid (vitamin B9) is essential for several key enzymatic steps in the
biochemical pathways that synthesise purine and pyrimidine nucleosides and some key
amino acids, including methionine, serine, glycine, and histidine. Folic acid deficiency
therefore inhibits DNA, RNA, and protein synthesis and so is particularly important in
actively dividing cells. This makes folate metabolism a potential target in cancer therapy
because cancer cells usually divide at a faster rate than normal cells.
Dietary folic acid is converted to dihydrofolate and then to tetrahydrofolate by the
action of the enzyme dihydrofolate reductase (Fig. 12.12; see also Fig. 11.15).
Dihydrofolate reductase is inhibited by methotrexate. Fig. 12.12 shows that
tetrahydrofolate is a co-factor for the enzyme thymidylate synthetase, a key enzyme in
the production of thymine nucleosides for DNA synthesis. This reaction converts
tetrahydrofolate back to dihydrofolate, but dihydrofolate is then recycled back to
tetrahydrofolate by dihydrofolate reductase, keeping tetrahydrofolate availability high.
FU (see above), raltitrexed, and pemetrexed bind to and block thymidylate synthase,
thus blocking the recycling of tetrahydrofolate and blocking DNA, RNA, and protein
production.
Methotrexate
Methotrexate has been used as an anticancer drug since the 1950s and is still widely
used in a range of malignant diseases including some leukaemias, non-Hodgkin’s
lymphoma, and some breast, lung, and ovarian cancers. It is also used as an anti-
inflammatory and immunosuppressant agent in rheumatoid arthritis, inflammatory
bowel disease, and psoriasis. Methotrexate inhibits several enzymes involved in folic
acid metabolism, including dihydrofolate reductase and thymidylate synthase,
preventing the tumour cell from manufacturing DNA, RNA, and key proteins and
leading eventually to apoptosis of the cancer cell (Fig. 12.12). It is given orally or by
injection and causes the standard range of cytotoxic side-effects (see above) and others,
including neurotoxicity, confusion, respiratory problems, and hepatotoxicity. It is poorly
metabolised and is mainly excreted unchanged in the urine, so great care is needed in
people with impaired renal function. It is given once weekly and the half-life is dose-
dependent: it is shorter (3–10 hours) in low-dose treatment, which extends to up to 15
hours in higher-dose treatment and even longer in renal impairment. Clearance varies
significantly between patients.
FIG. 12.12 The cytotoxic action of fluorouracil and methotrexate.
Folinic acid (also called leucovorin) is used as a rescue medication and is given 24
hours after high-dose methotrexate to help reverse methotrexate-induced
myelosuppression.
Cyclophosphamide
Cyclophosphamide is used to treat a wide range of cancers in addition to severe
rheumatoid arthritis with systemic complications. Its plasma half-life is between 3 and
12 hours. It is given orally and is a pro-drug activated in the liver and body tissues to the
cytotoxic substances acrolein and phosphoramide. Phosphoramide is thought to be
mainly responsible for alkylation and cross-linking, and acrolein is responsible for most
of cyclophosphamide’s toxicity; it produces large quantities of reactive oxygen species,
which injure cell organelles including the cell membrane, damage DNA, and impair
enzyme function. Acrolein is particularly toxic to the bladder, causing haemorrhagic
cystitis especially at higher doses of cyclophosphamide.
Nitrosoureas
Examples: carmustine, lomustine, streptozocin
Unlike most other alkylating agents, carmustine and lomustine cross the blood–brain
barrier and are used to treat central nervous system tumours. In addition, they are used
in Hodgkin’s lymphoma, and lomustine is used in melanoma and some lung cancers.
They have short plasma half-lives of usually less than 30 minutes, but both are
metabolised in the liver to active metabolites which extend their biological activity.
Intravenous carmustine can cause delayed pulmonary toxicity, which is dose-related
and may be fatal. Streptozocin is not absorbed orally and is given by infusion. It does
not cross the blood–brain barrier; its plasma half-life is usually less than 15 minutes and
it is metabolised mainly in the liver. It has an affinity for pancreatic islet cells and so is
used to treat pancreatic tumours and is particularly renotoxic.
Miscellaneous Agents
Examples: busulfan, mitomycin C
Busulfan is rapidly metabolised in the liver and has a plasma half-life of less than 3
hours. It is given orally, is well absorbed, and crosses the blood–brain barrier. In 6–8%
of patients, chronic inflammatory and fibrotic pulmonary changes can occur, often after
the drug has been withdrawn; the risk increases with dose. It is sometimes used in
leukaemia, but its main use is in conditioning treatment before stem cell or bone
marrow transplantations.
Mitomycin C is an old cytotoxic antibiotic derived from Streptomyces caespitosus.
It is mainly metabolised in the liver and can cause chronic lung inflammation and
pulmonary fibrosis.
Anthracyclines
Examples: daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone
The anthracyclines are intercalating agents which have a range of cytotoxic actions.
They bind (intercalate) between base pairs, distorting the normal shape of the DNA
molecule, preventing DNA replication and the action of RNA transcriptase, the enzyme
which reads the DNA code and produces the mRNA needed for protein synthesis.
Additionally, they produce toxic free radicals when metabolised, which breaks DNA and
damages the cell membrane and cellular proteins. They are used in a range of
haematological malignancies and solid tumours and are given intravenously because
their oral absorption is poor.
Doxorubicin
Doxorubicin is produced by Streptomyces peucetius. Its mechanisms of action include
those described above. Additionally, by inserting itself between the DNA strands, it
prevents topoisomerase II (see above) from stitching together newly synthesised strands
of DNA, and so halts cell division. Its half-life is 20–40 hours and it does not cross the
blood–brain barrier. It should be avoided if possible in those with a current or past
history of heart disease because it is cardiotoxic.
Bleomycin
Bleomycin is a mix of antibiotics from Streptomyces verticillus. It binds metal ions into
a complex on the DNA strand, which generates reactive oxygen free radicals and
produces both single- and double-strand breaks. The damage bleomycin inflicts on DNA
is visible under the light microscope as abnormal and fragmented chromosomes (Fig.
12.14). Bleomycin is not given orally because it is not absorbed and is given by injection
or infusion. Its plasma half-life is around 2 hours, and it is metabolised mainly in the
liver. It should be avoided in people with pre-existing lung disease because it is highly
toxic to the lungs, causing potentially fatal pulmonary fibrosis (Fig. 8.2). Incidence of
bleomycin-induced lung toxicity may be as high as 1 in 10 patients. Other adverse effects
include hypersensitivity-type reactions, fever, and skin toxicity including
hyperpigmentation.
PARP Inhibitors
Examples: niraparib, olaparib, rucaparib
The enzyme poly-ADP ribose polymerase (PARP) binds to and repairs single-strand
breaks in DNA. Inhibition of this enzyme means that strand breaks are not re-joined;
this destabilises the DNA, makes it more susceptible to damage by DNA mutagenic
agents such as radiation, and increases the risk of further damage including double-
strand breaks. This normally triggers apoptosis and cell death. The first PARP inhibitor
to reach the market, olaparib, was approved in 2015, and these drugs are mainly used
in breast and ovarian cancer.
PARP inhibitors have been found to be up to 1000 times more effective in their
anticancer activity in cancers associated with BRCA mutations than those with normal
BRCA function. The BRCA genes are tumour-suppressor genes (see above) that produce
proteins important for DNA repair. When these genes are mutated and non-functional,
the cell is more reliant than normal on DNA repair carried out by PARP. Therefore,
inhibiting PARP in cancer cells without functional BRCA genes means that DNA break
repair either cannot proceed at all or, if a repair is made, it is prone to error. Either way,
the cell experiences genetic instability, predisposing it to further DNA damage and
eventual cell death (Fig. 12.15). PARP inhibitors are therefore used to treat BRCA-
associated tumours not only of the breast and ovary but also, for example, of the
prostate, uterine tube, and peritoneum.
PARP inhibitors are well absorbed following oral administration and mainly
metabolised in the liver. Their half-lives are variable (olaparib, 6 hours; rucaparib,
26 hours) and they cause general cytotoxic side-effects. They increase the risk of
developing secondary myelodysplastic syndrome and acute myeloid leukaemia, usually
within 8 months to 2.5 years.
Hormone-Responsive Cancers
Cancers developing in sex-hormone-responsive tissues such as breast and prostate may
be hormone-dependent, and so drugs that block the hormone have anti-tumour activity.
Breast cancers that express oestrogen receptors are designated oestrogen (ER)-positive
and those that express progesterone receptors are designated PR-positive. Tumours may
have both (ER/PR-positive), either, or neither (ER/PR-negative). Most breast cancers
are hormone-sensitive, so determining the receptor status of a tumour is an early
standard part of breast cancer management because it dictates treatment decisions.
Antioestrogens
Oestrogen has a range of proliferative actions on breast tissue, and lifetime exposure to
oestrogen is a recognised risk factor for breast cancer. About 75% of breast cancers are
ER-positive, and drugs that block oestrogen receptors are therefore an important
treatment option.
Tamoxifen
Tamoxifen was first developed in the search for oral contraceptives, and although it has
no contraceptive activity in humans, it was trialled in the 1970s as chemotherapeutic
agent in breast cancer. It competes with oestrogen for the ER and blocks its proliferative
effect. It is taken orally, and its plasma half-life is between 5 and 7 days, but it produces
active metabolites with much longer half-lives. It increases the coagulability of the
blood, predisposing to clots, and should not be used in women with a history of
thromboembolism. It can cause endometrial changes including malignant change.
Patients should be advised to report vaginal bleeding or discharge, menstrual
irregularities, or pelvic pain or pressure for prompt investigation. In pre-menopausal
women, it causes menopausal side-effects such as hot flushes.
Tamoxifen may also be used prophylactically in women who have a strong family
history of breast cancer.
Aromatase Inhibitors
Oestrogens are synthesised from testosterone by the action of the enzyme aromatase
(see Fig. 5.14). Aromatase inhibitors therefore block the conversion of testosterone to
oestrogen and reduce oestrogen levels, which inhibits growth of an oestrogen-dependent
tumour. In addition, some breast cancers express elevated aromatase levels and so are
particularly sensitive to the effect of aromatase inhibition.
Anastrozole
Anastrozole is an aromatase inhibitor, and it may be preferred to tamoxifen in ER-
positive breast cancer because it has fewer side-effects. Common side-effects include
osteoporosis, hair loss, vaginal dryness, and hot flushes. Letrozole is a similar agent;
neither should be used in pre-menopausal women.
AntiAndrogens
Examples: abiraterone acetate, darolutamide, flutamide
Eighty to ninety percent of prostate cancers express androgen receptors (ARs) and are
testosterone-dependent in the earlier stages of their development. Androgen-
deprivation treatment using antiandrogens is therefore an important treatment; these
drugs bind to the AR and block testosterone binding, depriving the tumour cell of this
important growth signal. They are given orally.
Glucocorticoids
Glucocorticoids have profound anti-inflammatory and immunosuppressant effects (see
also Ch. 6) and are sometimes used as adjuvant treatment in cancer. Agents such as
prednisolone and dexamethasone are used for their immunosuppressant effect in
lymphomas, myelomas, and some leukaemias: they suppress the proliferation and
activity of the malignant white blood cells and enhance cell death. In many cancers,
however, glucocorticoids are not used as cytotoxic agents, but as supportive drugs to
take the edge off the side-effects of chemotherapy and make the treatments more
tolerable. For example, they may be given to reduce the severity of hypersensitivity
reactions common with agents such as pemetrexed and cisplatin. Their anti-
inflammatory action can help reduce the swelling and inflammation around a tumour,
especially important when an expanding mass is compressing adjacent normal tissues,
i.e. in the confined space of the skull. In this situation, dexamethasone is preferred
because it has no fluid-retaining activity. Glucocorticoids are anti-emetic and stimulate
appetite, so are useful in countering chemotherapy-induced nausea and vomiting and
encouraging food intake. They can have a positive effect on mood and induce feelings of
well-being and contentment, always a beneficial contribution in what is likely to be a
difficult set of life circumstances for anyone dealing with cancer.
Adverse Effects
In general, mAbs are less toxic than older chemotherapeutic agents, but they can induce
severe allergic-type reactions, including skin eruptions, hypotension, fever, chills, and
GI upsets. Some can cause target-specific adverse side-effects; for example, drugs which
interfere with VEGF, an agent important in blood-vessel growth and development, can
cause thromboembolism, bleeding, poor wound healing, and hypertension (see also
bevacizumab below).
Rituximab
Rituximab binds to a protein called CD20 which is found on healthy B-cell membranes,
but which is expressed in higher-than-normal quantities by B-cells in chronic
lymphocytic leukaemia and some forms of non-Hodgkin’s lymphoma, making it a
relatively selective target. Binding of the antibody to the B-cell activates complement
and triggers cell lysis, causing a rapid and sustained reduction in B-cell counts. It is also
sometimes used in severe rheumatoid arthritis and other autoimmune disorders in
which B-cells are thought to contribute to the disease pathology. It has a long half-life,
which extends with repeated dosing and can exceed 30 days. Side-effects are numerous
and can be severe. Severe infusion reactions, which can be fatal, have been reported:
widespread cytokine release causes hypotension, cardiac arrhythmias, pulmonary
infiltration, and acute respiratory distress syndrome and shock. Another serious and
potentially fatal reaction is acute renal failure due to the sudden release of B-cell
breakdown products into the bloodstream. Other agents with similar mechanisms of
action to rituximab include ofatumumab and alemtuzumab.
Trastuzumab
Trastuzumab blocks EGFRs on some types of cancer cells and so silences this growth-
stimulating signal. Its mechanism of action is described in more detail above. It is used
mainly in HER2-positive breast cancer but also in other cancers that over-express the
HER2 receptor, including some gastric cancers. Only about 15%–25% of breast cancers
are HER2-positive and therefore respond to trastuzumab, but these tumours tend to be
faster growing and more aggressive than HER2-negative disease, and this drug has
significantly improved prognosis and survival in HER2-positive cases. Its most
significant side-effect is cardiotoxicity, and it can cause arrhythmias, heart failure, and
coronary artery disease, which are usually reversible when treatment stops. The reason
behind this is not completely clear, but it is known that healthy cardiac myocytes
express HER2, and that blocking this receptor may inhibit the myocardial cells’ capacity
to detoxify harmful products of cellular metabolism, including reactive oxygen species,
which then accumulate and damage the cell. Cardiac function should be monitored
during and after treatment. Other HER2 receptor inhibitors include pertuzumab.
Because trastuzumab and pertuzumab block different sites on the HER2 receptor, they
have a synergistic action and can usefully be combined.
Binding to Chemical Factors Essential for Cancer Cell Growth and Survival
Monoclonal antibodies may be targeted against growth factors or other cytokines on
which the cancer cell is dependent. When the mAb binds to the growth factor, it can no
longer bind to its receptor.
Bevacizumab
As a solid tumour expands, it needs its own network of blood vessels to ensure an
adequate supply of oxygen and nutrients, and most tumours produce increased levels of
vascular endothelial growth factor (VEGF) to promote blood vessel growth and survival.
Bevacizumab is a mAb directed against VEGF; it binds to and neutralises the VEGF
molecule, depriving the tumour of this key mediator and inhibiting tumour
angiogenesis. It is used in several cancers including breast, colon, and reproductive tract
tumours and is currently also being trialled in the treatment of severe post–COVID-19
respiratory complications including acute lung injury, in which VEGF seems to
contribute to the disease process. It has an average half-life of 20 days, although this can
be more than doubled in some patients. It is associated with a wide range of side-effects,
including the consequences of blocking VEGF in normal tissues (see above).
Immunotherapy
The most familiar role of the immune system is in the prevention and control of
infection, but part of its role is the constant surveillance of tissues to identify and
destroy mutated or abnormal cells. Cancer cells develop ways to conceal and protect
themselves from the unwanted attentions of T-cells and other body defences, including
producing tumour-derived immunosuppressant factors which dampen down normal
immune activity. Immunotherapy is an umbrella term meaning any treatment which in
some way enhances the immune system’s capacity to destroy tumour cells, and these are
among the newest available cancer therapies. The mAbs which coat cancer cells and
stimulate complement (e.g. rituximab, see above) are one example of immunotherapy.
Other examples of immunotherapy include:
Treatment vaccines
Although research in this area is still at a very early stage, it offers the exciting prospect
of personally tailored cancer treatment. Unlike vaccination to prevent infection, cancer
vaccines are not intended as prophylactic therapies. The principle here is that
identification of tumour-specific proteins expressed on the surface of an individual’s
cancer cells can be used to make a vaccine specific to those cancer cells. Administration
of the vaccine to that individual then stimulates an immune response specific to the
cancer cells with resultant destruction of the tumour.
Virus Vaccines
Viruses are intracellular parasites. They enter a cell and hijack the cell’s organelles to
produce new viral proteins and nucleic acids, which are then assembled into new viral
particles (see Fig. 11.17). Depending on the virus, these new viral particles may
accumulate in the host cell until the host cell literally bursts (lysis), releasing the viral
particles into the local vicinity to infect neighbouring cells. One novel approach to
cancer therapy is to use viruses to infect and destroy cancer cells.
Talimogene Laherparepvec
This therapy is a genetically modified preparation of herpes simplex virus type 1. It is
used in malignant melanoma and injected directly into the tumour. The virus is
genetically engineered to selectively enter and replicate within tumour cells, causing the
cell to lyse and release large quantities of melanoma-tumour antigens into the local
environment. This activates defence and immune cells, which multiply and produce a
local and systemic immune response directed against the tumour. The released viral
particles infect neighbouring cancer cells, and the process is repeated (Fig. 12.19). Care
is needed in immunosuppressed patients, for example those on steroid treatment,
because although the pathogenicity of the genetically engineered virus has been
substantially reduced, it may still cause systemic herpes infections in people whose
immune function is impaired.
Proteasome Inhibitors
Examples: bortezomib, carfilzomib, ixazomib
The proteasome is a complex of proteolytic enzymes responsible for destroying
unwanted, abnormal, or misfolded proteins inside the cell. Proteasomes are found in
cells throughout the body, ensure that potentially harmful proteins do not accumulate,
and allow the cell to maintain levels of key regulatory proteins within normal ranges.
Proteasome inhibitors bind to one or more of the enzymatic sites within the proteasome
and block them. This leads to the accumulation of a range of unwanted and abnormal
proteins in the cell, interfering with the cell’s ability to regulate its internal pathways
and triggering apoptosis. These drugs are used in multiple myeloma and can cause a
wide range of side-effects, including cardiovascular events and reactivation of latent
viral infections including herpes zoster and hepatitis B.
Asparaginase
The enzyme asparaginase breaks down the amino acid asparagine. Therapeutically, it is
available in different formulations derived from different sources: for example,
crisantaspase is the asparaginase produced by Erwinia chrysanthemi and there are
formulations prepared from Escherichia coli asparaginase. Pegaspargase is a
PEGylated formulation. Asparagine is needed for DNA and protein synthesis, and
asparagine depletion inhibits the proliferation of tumour cells. Asparaginase is used in
acute lymphoblastic leukaemia.
References
1. Drew Y. The development of PARP inhibitors in ovarian cancer: from bench to
bedside. Br. J. Cancer. 2015;113(Suppl. 1):S3–S9.
2. Pufall M.A. Glucocorticoids and cancer. Adv. Exp. Med. Biol. 2015;872:315–333.
3. Tadesse S, Caldon E.C, Tilley W, et al. Cyclin-dependent kinase 2 inhibitors in
cancer therapy: an update. J. Med. Chem. 2019;62(9):4233–4251.
4. Wang, J., Fang, Y., Fan, R.A., et al., 2021. Proteasome inhibitors and their
pharmacokinetics, pharmacodynamics and metabolism. Int. J. Mol. Sci. 22 (21),
11595
Online resources
Chial H. Proto-oncogenes to oncogenes to cancer. Nat.
Educ. 2008;1(1):33 Available
at:. https://www.nature.com/scitable/topicpage/proto-oncogenes-to-
oncogenes-to-cancer-
883/#:∼:text=Often%2C%20proto%2Doncogenes%20encode%20proteins,maintena
Emory Winship Cancer Institute, . Cancer biology Available
at:. https://www.cancerquest.org/cancer-biology, 2024.
Sun Y, Liu Y, Ma X, et al. The influence of cell cycle regulation on
chemotherapy. Int. J. Mol. Sci. 2021;22(13):6923 Available
at:. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8267727/.
Zahavi D, Weiner L. Monoclonal antibodies in cancer
therapy. Antibodies. 2020;9(3):34 Available
at:. https://www.mdpi.com/2073-4468/9/3/34/htm.
Index
Page numbers followed by “f” indicate figures, “b” indicate boxes, and “t” indicate tables.
abacavir, 4, 219b–220b
abemaciclib, 239
abiraterone acetate, 241
absence seizures, 72
cutaneous, 38f, 39
delayed, 40
end-of-dose, 40
hepatic, 38
immunologically mediated, 37–38, 37f–38f
mechanisms of, 37–38
nervous system, 39
pharmacologically mediated, 38
red bone marrow toxicity, 39
renal, 38–39
reproductive function, 39
affinity, 35
afterload, 128, 128f
agomelatine, 59
agonist, 28b
albendazole, 224
alcohol, 90
for asthma, 165–166
causing erectile dysfunction, 179
for cough, 162
insomnia and, 60
origins of, 1–2
respiratory depression and, 57–58
warfarin and, 150–151
withdrawal, 41
aldesleukin, 243–244
aldosterone, 173
amiodarone, 136
causing delayed reactions, 38
class III agents, 139
renal excretion of, 18–19
warfarin and, 150–151
amitriptyline, 2–3, 56–57, 103, 131
amlodipine, 132, 148
pharmacokinetics of, 69
propofol, 69
thiopental, 69
local, 69b–71b
mechanism of action of, 66–67
stages of, 66–67, 67t
analgesics, 101–125 , 101b
anthracyclines, 238
antiandrogens, 100, 241
anti-arrhythmic drugs, 4, 136–139
class I agents, 138
class II agents, 138
class III agents, 138–139
class IV agents, 139
antibiotics
acting on bacterial cell membrane, 210
acting on bacterial DNA, 210–213, 210b
metronidazole, 211–212
nitrofurantoin, 212
polymyxins, 210
quinolones, 212–213
cutaneous adverse drug reactions to, 39
excretion of, 17
gastrointestinal tract and, 181
general adverse effects of, 204
inhibit protein synthesis, 204–207
aminoglycosides, 206
chloramphenicol, 206
fusidic acid, 206
macrolides, 206–207
oxazolidinones, 207
tetracyclines, 207
interfere with cell wall integrity, 207–210, 207b
β-lactam antibiotics, 208–210
bacitracin, 210
cell wall structure, 207–208
teicoplanin, 210
vancomycin, 210
interfering with folic acid metabolism, 213–216, 213b
sulphonamides, 213–214
trimethoprim, 214–216
intrathecal administration of, 24
synergism, 203–204
anticancer drugs, 21
See also cytotoxic drugs
anticoagulants, 5, 150–152
foscarnet, 218
nucleoside analogues, 217–218
protein synthesis, inhibitors of, 218–220, 218f
renal excretion of, 18–19
resistance to, 216–217
uncoating and release, inhibitors of, 218
anxiety disorders, 59–60
apixaban, 150
apomorphine, 54
apoptosis, 229–230, 232f
aprepitant, 183, 195
arachis oil, 191
aripiprazole, 66
aromatase inhibitors, 241
arthropod (insect) infestations, 224
asparaginase, 244
aspirin, 90, 116–117, 119, 121, 152
distribution of, 24
immediate hypersensitivity reactions to, 37
irreversible binding of, 31
metabolism of, 15–16
plasma protein binding and, 14, 14f
transfer across cell membranes, 8
unwanted/dangerous interactions of, 24–25
aspirin-induced bronchoconstriction, 38
asthma, treatment of, 163–168, 164f, 169f, 169b
atazanavir, 219b–220b
atropine, 185
augmented reactions, of drugs, 39
autonomic nerve supply, 130f
avanafil, for erectile dysfunction, 179
avibactam, 208
axon, 42, 43f
azacitidine, 236
azithromycin, 192, 206–207
azoles, 222–223
adverse effects of, 222–223
erectile dysfunction and, 179–180
pharmacokinetics of, 222–223
benzimidazoles, 224
benzocaine, 69b–71b
benzodiazepines, 46, 74
biologics, 3–4
bipolar disorder, 62–63
bisacodyl, 191
bismuth, for peptic ulcer, 188b–189b
bisoprolol, 148
bivalirudin, 152
cabazitaxel, 235
caffeine
for asthma, 165
insomnia and, 60
calcitonin, 83
calcium (Ca2+), 44
calcium-channel blockers, 128–129, 132–133, 142, 145, 145b–146b
cefuroxime, 210
celecoxib, 122
cell cycle, 228–229, 231f
stages of, 235–240
cell division, 227–230
cell membrane, 6–8, 7f
clomipramine, 57
clonazepam, 61–62, 61b–62b
clonidine, 47–48
clopidogrel, 148–149, 152–153
clotting, 149–150, 149f
clozapine, 65–66
cocaine, 69b–71b
codeine, 106, 107f, 108, 109f, 189
beneficial interactions of, 24
for cough, 162–163
pharmacokinetics of, 108
colistimethate sodium, 210
collecting duct, reabsorption in, 173
combined oral contraceptive (COC), 96–98
adverse effects of, 97–98, 97f
pharmacokinetics of, 97
competitive antagonism, 29t, 31, 31f
constipation, 191
contact inhibition, loss of, 231
contraceptives
formulations of, 21
topical administration of, 22
contrast media, intrathecal administration of, 24
corticosteroids, adverse reactions of, 39–40
corticotropin-releasing hormone (CRH), 79
cortisol
physiological effects of, 112, 114t
coxibs, 122
CRH, See corticotropin-releasing hormone
cromones, for asthma, 166–167
curcumin, 230
Cushing’s syndrome, 115, 116f
cutaneous adverse drug reactions, 38f, 39
cyclic adenosine monophosphate pathway, G proteins and, 33, 34f
cyclin-dependent kinases (CDKs), 229, 239
cyclizine, 52, 105, 124, 196
cyclophosphamide
for cancer, 238
metabolism of, 15, 15b
danaparoid, 151–152
dantron, 191
dapagliflozin, 92
darolutamide, 241
darunavir, 218, 219b–220b
daunorubicin, 238
decitabine, 236
deflazacort, 117t
degradative enzymes, 44
delayed reactions, of drugs, 40
dendrites, 43f
deoxyribonucleic acid (DNA), 227–230, 227b
agents that directly damage, 238–239
alkylating agents, 237–238, 238f
drugs that act on, 236–239
mRNA and protein synthesis, 228, 230f
replication of, 228, 229f
structure of, 227–228, 228f
dependence, opioids and, 106
depression
biology of, 56–57, 57t
signs and symptoms of, 56
treatment of, 56, 57f
descending inhibition, 102–103, 103f
desflurane, pharmacological actions of, 67t
desmopressin, 82
desogestrel, 97
type 2, 88
diamorphine (heroin), 106–108, 108f
for cough, 162–163
diazepam, 61b–62b
excitatory and inhibitory receptor response of, 29
for status epilepticus, 72
diclofenac, 121–122
in red bone marrow toxicity, 39
dicoumarin, 150
digitalis, 2
Digitalis purpurea (foxglove), 2, 2f
digitoxin, 133, 133f
digoxin, 133, 134b–135b, 136
excretion of, 17–19
formulations of, 21
in membrane transport, 35
metabolism of, 14
monitoring of, 21
for peptic ulcer, 188b–189b
reabsorption of, 18
diphenoxylate/atropine, 189
dipyridamole, 153
direct thrombin inhibitors, 152
disopyramide, 138
distal convoluted tubule (DCT), reabsorption and secretion in, 172–173, 177f
distribution, of drugs, 11–14
in body fat partitioning, 12–13
plasma protein binding, 13–14, 13f–14f
sanctuary compartments of, 12
in tissues and fluids, 11–12, 11f
volume of, 11–12, 11b, 12f
diuretics, 133, 134b–135b, 144, 173–178, 175f
loop, 175–176, 176f
osmotic, 174–175, 175f
pharmacologically mediated adverse drug reactions to, 38
potassium-sparing, 177–178
proximal convoluted tubule and, 172
thiazide, 176–177, 177f
transfer across cell membranes, 8
DM, See diabetes mellitus
formulations of, 21
half-life of, 19–21, 19f
heart and, 130–135, 130b
affect cardiac contractility, 130–135
function, 126–154
interactions of, 24–25
ionisation of, 8–10
female, 95–99
male, 100
respiratory system and, 155–170 , 157f, 157t
steady state of, 20–21, 20f
therapeutic dose range of, 20–21
thyroid function and, 82–85
hyperthyroidism, 84–85
hypothyroidism, 83–84
drug action
assessment of, 35–37
affinity, efficacy, and potency, 35
dose-response relationships, 36–37, 36f
therapeutic index, 36–37, 36f–37f
levels of, 27–28, 28f, 29t
principal targets of, 28–35, 29f
cell-surface receptors in, 32–33
competitive antagonism in, 29t, 31, 31f
drug receptors in, 28–33, 28f, 28b
enzymes, 35
excitatory and inhibitory receptor responses in, 29–31, 30b
echinocandins, 222
ectopic arrhythmias, 136
efavirenz, 219b–220b
efferent arteriole, 172
efficacy, 35
empagliflozin, 92
enalapril, 15b, 144
endocannabinoids, 196
endocrine control, 77, 77b
endocrine function, drugs and, 77–100 , 77b
hypothalamic and pituitary hormones in, 80–82
hypothalamic-pituitary axis, 78
endocrine system, 77, 78f
end-of-dose reactions, of drugs, 40
eplerenone, 177–178
erectile dysfunction, 179–180
eribulin, 235
erythromycin, 192, 206–207
of digitoxin, 133
erectile dysfunction and, 179–180
esketamine, for depression, 59
esomeprazole, 187
ethacrynic acid, 175
ethambutol, 214b–215b
ethanol
dependence to, 40
metabolism of, 15–16
reproductive function and, 39
ether, 66
ethinyloestradiol, 96–97
ethosuximide, 75
for absence seizures, 72
etomidate, 67t, 69
etoposide, 229–230
etoricoxib, 122
etravirine, 219b–220b
excitatory receptor responses, 29–31, 30b
excretion, of drugs
faecal excretion, 17
renal, 17–19, 18f
exenatide, 91
expectorants, 157–162
ezetimibe, 148
famciclovir, 217
famotidine, 124, 187
receptor selectivity in, 30
fat solubility, transfer across cell membranes, 7–8
felodipine, 132
felypressin, 82
female reproductive cycle, 95–96, 96f
fenofibrate, 148
fentanyl, 107f, 108–109
pharmacokinetics of, 109
topical administration of, 22
fever, prostaglandins in, 119
fexofenadine, 123–124
fibrinolytic drugs, 153–154, 153f
fibrinolytic system, 153–154
filtration, 172b
first-generation antihistamines, 124, 181
first-order (linear) kinetics, 19–20
flecainide, 138
Fleming, Alexander, 3, 3f
fluconazole, 222
flucytosine, 223
flumazenil, 62
fluorouracil
for cancer, 236, 237f
transfer across cell membranes, 8
fluoxetine, 58, 153
flupentixol, 65
flutamide, 241
fluticasone, for asthma, 166
focal seizures, 72
folate antagonists, 237
folic acid pathway, 213, 213f
folinic acid, 237
follicle-stimulating hormone, 82, 99
formoterol, for asthma, 164
fosaprepitant, 195
fungi, 220
biology of, 220–222, 220f
infections, pathology of, 220–222
furosemide, 134b–135b, 172, 175
renal adverse drug reactions to, 38–39
renal excretion of, 18–19
unwanted/dangerous interactions of, 24–25
fusidic acid, 206
glibenclamide, 86, 90
glimepiride, 90
glipizide, 90
Gliptins, 86–87, 91
glitazones, 91
glomerular filtration, 171, 173f
glomerular filtration rate, 171, 174f
glucagon, 85
glucocorticoids, 111–116
administration, routes of, 114–115
adverse effects of, 115–116
for asthma, 163–164, 165f, 166
for cancer, 241
for chronic obstructive pulmonary disease, 168
drugs, 116, 116f, 117t
mechanism of action of, 112–114, 115f
stress response and, 111–114
in therapeutics, 114–115, 115b
glutamate, 46, 56
glycerol, 191
glyceryl trinitrate (GTN), 142
metabolism of, 17
pharmacokinetics of, 142
GnRH, See gonadotropin-releasing hormone
gonadotropin-releasing hormone (GnRH), 79, 82, 99
GORD, See gastro-oesophageal reflux disease
goserelin, 82
gonadorelin, 82
guaifenesin, 157–159
guanylate cyclase-C receptor agonists, 192–193, 193f
H
heroin, 107–108
hirudin, 152
histamine, 46, 52–56, 52t, 77, 113t, 122–124, 123f, 185
in allergy, 124
asthma and, 166–167
gastric acid secretion and, 124
inflammation and, 123–124
motion sickness and, 56
as neurotransmitter, 124
sleep-wake cycle and, 52
synthesis and breakdown, 52b
histamine receptor family, 30
histamine receptors, 52–56, 52t
hormonal contraception, 96–98, 97f
hormone levels
control of, 79–80, 80f
negative feedback and, 78
hormone replacement therapy (HRT), 99b
hormone-responsive cancers, 240–241
5-HT3 antagonists, 195
5-HT3 receptors, 46
hydralazine
immune-complex mediated reactions to, 38
metabolism of, 16
hydrocortisone, 111, 116, 117t
nuclear receptors in, 33
5-hydroxytryptamine, synthesis and breakdown, 46
ibuprofen, 122
as enzymes, 35
idarubicin, 238
idarucizumab, 152
imatinib, 240
imipenem, 210
immediate hypersensitivity, 37
immune checkpoint inhibitors, 243–244
immune system modulators, 243–244
immunotherapy, 243–244
imunosuppressants
for asthma, 167–168
opioids and, 106
incretin (glucagon-like peptide 1) agonists, 91–92, 91f–92f, 91b
incretins, role of, 87f
indirect negative feedback control, 79–80, 80f
infertility, female, hormonal treatment of, 99
inflammation
histamine and, 123–124
prostaglandins in, 118–119
inflammatory mediators, 111, 113f
inflammatory response, pathophysiology of, 111
inhalation, of drugs, 23
inhaler devices, for respiratory disorders, 158t, 158b–159b, 159f
inhibitory G proteins, 33
inhibitory neurotransmission, 43
inhibitory receptor responses, 29–31, 30b
insecticides, 224
insomnia, 60
insulin, 85, 88–89
action of, 86–88
adverse effects of, 89
adverse reactions of, 39
ipilimumab, 243
ipratropium, 51b
for asthma, 166
iproniazid, 58
irinotecan, 236
iron salts, transfer across cell membranes, 8
irreversible drug-receptor binding, 31
ischaemic heart disease, 147–148
drugs used to treat, 148
myocardial infarction and, 147–148
isoflurane, pharmacological actions of, 67t
isoniazid, 214b–215b
for asthma, 165–166
metabolism of, 16
isophane insulin, 88
isoprenaline, for asthma, 164
labetalol, 131–132
lactulose, 191–192
lamivudine, 219b–220b
lamotrigine, 62, 75–76
for focal seizures, 72
lanolin, formulations of, 21
lanreotide, 81–82
lansoprazole, 187
lapatinib, 240
latency, 217
laxatives
bulk-forming, 191
for constipation, 191–192, 191f
osmotic, 191–192
softening, 191
stimulant, 191
levothyroxine, 83–84
adverse effects of, 84
pharmacokinetics of, 84
lidocaine, 69b–71b, 136, 138
ligand, 28b
lignocaine, as ion channel blocker, 34
linaclotide, 192–193, 193f
linagliptin, 86–87, 91
linear kinetics, 19–20
liothyronine, 84
lipid-modifying drugs, additional, 148
liposomes, 21, 21f
lipoxygenase inhibitors, for asthma, 167
liquid paraffin, 191
liraglutide, 86–87, 91
Lister, Joseph, 198–199
lithium, 62–63
local anaesthetics
absorption of, 24
efficacy, factors affecting, 70–71
side-effects of, 71
lomustine, 238
loop diuretics, 175–176, 176f
adverse reactions of, 39
in membrane transport, 35
loop of nephron, reabsorption in, 172
loperamide, 106, 189
loratadine, 124
lorazepam, 61b–62b, 196
losartan, 140, 144
low-dose aspirin, 145b–146b
lung tissue, histology of, 157f
luteinising hormone, 82
macrogols, 191–192
macrolides, 192, 206–207, 214b–215b
adverse effects of, 207
erectile dysfunction and, 179–180
pharmacokinetics of, 207
malaria, 223
malathion, 49–50, 224
male sexual function, 178–180, 178f
mannitol, 159–161
in non-mammalian cell targets, 35
in osmotic diuretics, 174, 175f
MAO inhibitors, 46
binding to channel, 34
excitatory and inhibitory receptor response of, 29
minocycline, 207
minoxidil, 142–143
miosis, opioids and, 105, 105f
mirtazapine, 59
misoprostol, 120, 184–185, 187–189
mitomycin C, 238
mitoxantrone, 238
moclobemide, 58
modified ethers, 68–69
molecular weight, transfer across cell membranes, 7
mometasone, for asthma, 166
monoamine
adverse effects of, 58–59
breakdown, drugs that block, 58–59, 58f
pharmacokinetics of, 58–59
receptor blockers, 59
monoamine oxidase, 55, 55f
monoclonal antibodies
for cancer, 241–243, 241f
immediate hypersensitivity reactions to, 37
monotherapy, 73
mood-stabilising drugs, 62–63
MOR distribution, 104
MOR signalling, 104, 104f
morphine, 2, 103, 189
metabolism of, 17
opioid receptors for, 56
pharmacokinetics of, 107
in receptor desensitisation, 35
targets of, 28–29
motility, cancer and, 231–232
mould, 220b
mucolytics, 157–162
mucus, drugs and, 157–162
muscarinic receptor, subtypes of, 50t
Mycobacteria, 214f, 214b–215b
mycosis, 220b
myelin sheath, 43f
myocardial infarction
combined oral contraceptive and, 98
ischaemic heart disease and, 147–148
myocardium, 127–129
afterload, 128, 128f
blood supply to, 127
cardiac conducting system, 128–129
nabilone, 196
N-acetylcysteine (NAC), 122
N-acetyl-p-benzoquinone imine (NAPQI), 38
naldemedine, 192
nalidixic acid, 212
naloxegol, 192
naloxone, 24, 103, 110, 110f, 189
naltrexone, 31, 110
naproxen, 122
narrow-spectrum antibiotics, 201b
nateglinide, 90
natural anticoagulants, 150
natural prostaglandins (PGs), 180
nausea
cytotoxic agents and, 234
opioids and, 105
nedocromil sodium, for asthma, 166–167
neomycin, 206
neoplasia, combined oral contraceptive and, 98
neostigmine, 44
nephron, 171
loop of, reabsorption in, 172
regions of, 171
reabsorption and secretion in the different, 172–173
structure of, 173f
nerve action potential, 44, 44f
ombitasvir, 218
omeprazole, 184, 186–187
adverse drug reactions to, 37
formulations of, 21
irreversible inhibition of, 31
in membrane transport, 35
oseltamivir, 218
osmotic diuretics, 174–175, 175f
osmotic laxatives, 191–192
oxaliplatin, 238
oxazolidinones, 207
oxprenolol, 132
oxybutynin, 51b
oxytetracycline, 207
oxytocin, 82
palbociclib, 239
palonosetron, 195
pancreatic function, drugs and, 85–93
diabetes mellitus, 85–93
glucagon in, 85
insulin in, 85
pancuronium, 66
papaverine, 103
paracetamol, 5, 122, 123f
beneficial interactions of, 24
biochemically mediated adverse drug reactions to, 38
drug action, 27
hepatic adverse drug reactions to, 38
metabolism of, 15
paracrine control, 77
paracrine substances, 77
parasympathetic activity, 130
parathyroid hormone, reabsorption and secretion of, 172–173
Parkinson’s disease
dopamine and, 53f, 53b–55b
drug treatment in, 53–55
signs and symptoms of, 53–55
Pasteur, Louis, 198–199
pegaspargase, 244
pembrolizumab, 243
pemetrexed, 237, 241
half-life of, 19
immediate hypersensitivity reactions to, 37
immunologically mediated adverse drug reactions to, 37
isolation of, 3, 3f
metabolism of, 14
modifications and altered activity of, 209b
pharmacokinetics of, 209b
renal excretion of, 18–19
transfer across cell membranes, 8
Penicillium notatum, 209b
penis, 178–179
peppermint oil, 190
peptic ulcer, treatment of, 188b–189b
permethrin, 224
pertuzumab, 242
pethidine, 109
PGE2, 119
PGI2, 119, 119f
pH, 8–10, 8f–9f
partitioning, 9–10, 10f
absorption in, 24
of calcium channel antagonists, 132–133
of digitoxin, 133
distribution in, 24
drug administration in, 21–24
drug disposition in, 10–19, 10f
absorption, 10–11
distribution, 11–14
excretion, 17–19
metabolism, 14–17
drug movement, 6–10
excretion in, 24
interactions of, 24
of loop diuretics, 176
of phosphodiesterase inhibitors, 179–180
of potassium-sparing diuretics, 177–178
special situations of, 25
of sympathetic agonists, 131
of sympathetic antagonists, 132
of thiazide diuretics, 176–177
pharmacology, 1–5
from bench to bedside, 4–5
clinical trials, 5
drug development process, 4–5, 5f
drug names, 5
origins of, 1–4, 1f
biologics, 3–4
pharmacogenomics, 3–4, 4f
pre-clinical development of, 4
phenelzine, 44, 58
metabolism of, 15–16
phenindione, 150
phenothiazine antipsychotics, 2–3, 195
phenoxymethylpenicillin, 209b
quinine, 198
in red bone marrow toxicity, 39
quinolones, 212–213
adverse effects of, 212–213
for peptic ulcer, 188b–189b
drug action, 27
ranitidine, 187
rapid-eye movement sleep, 60
rational drug design, 4
reabsorption, 172, 172b
receptor desensitisation, 35
receptor ‘families’, 29t, 30–31, 30f
repaglinide, 90
reproductive function
of drugs, 39
female, 95–99
hormonal contraception, 96–98
hormonal control of, 95–96
male, 100
rilpivirine, 219b–220b
risperidone, 62, 66
ritonavir, 218, 219b–220b
rituximab, 242
rofecoxib, 122
roflumilast, for chronic obstructive pulmonary disease, 168–170
ropinirole, 54
salbutamol
for asthma, 164
drug action, 27
G proteins and, 33
regulation of, 31
targets of, 28–29
salicin, 116–117
salicylates, zero-order kinetics of, 20
salicylism, 121
salmeterol, for asthma, 164
saquinavir, 219b–220b
saturation kinetics, 19–20, 20f
saxagliptin, 91
schizophrenia, 63–64
Sertürner, Friedrich, 3f
sex hormone, 94f, 94b–95b
sildenafil
for erectile dysfunction, 179–180
G proteins and, 33
mechanism of action of, 179f
simvastatin, 148
sinus rhythm, 135
sleep, physiology of, 60
smooth muscle, of respiratory passageways, 155–156
smooth-muscle relaxants, in gastrointestinal motility, 190
sodium (Na+), 44
sodium cromoglicate, for asthma, 166–167
sodium metabisulphite, formulations of, 21
sodium picosulfate, 191
sodium valproate, 62, 75
for absence seizures, 72
hepatic adverse drug reactions to, 38
zero-order kinetics of, 20
sodium/glucose co-transporter inhibitors, 92, 93f
sodium-potassium pump, 44
softening laxatives, 191
soluble insulin, 88
somatostatin, 81–82
analogues, 81–82
somatropin, 81
sorafenib, 240
as enzymes, 35
streptomycin, 206
renal adverse drug reactions to, 38–39
streptozocin, 238
stroke, combined oral contraceptive and, 98
subcutaneous/intradermal administration, of drugs, 22f, 23
substance P, 101–102, 113t
intestinal contraction and, 183
sucralfate, for peptic ulcer, 188b–189b
sulfasalazine, 213
sulphamethoxazole, 2–3, 213
sulphonamides, 90, 213–214
adverse effects of, 214
causing delayed reactions, 38
cutaneous adverse drug reactions to, 39
immune-complex mediated reactions to, 38
mechanism of action of, 213f
metabolism of, 16
pharmacokinetics of, 213
in red bone marrow toxicity, 39
teicoplanin, 210
telomerase, 231
temazepam, 61–62
tenecteplase, 154
teratogenicity, cytotoxic agents and, 234
terbinafine, 222
terbutaline, for asthma, 164
terfenadine, 124
terlipressin, 82
testosterone, 94b–95b
tetracaine, 69b–71b
tetracyclines, 207
absorption of, 24
adverse effects of, 207
gastric emptying and, 11
for peptic ulcer, 188b–189b
pharmacokinetics of, 207
thalidomide, 243–244
thebaine, 103
theophylline, 163, 165–166
adverse effects of, 165–166
G proteins and, 33
pharmacokinetics of, 165
therapeutic index, 36–37, 36f–37f
thiamazole, 84
thiazide diuretics, 145b–146b, 176–177, 177f
causing erectile dysfunction, 179
cytotoxic reactions to, 37–38
thiazides, 177, 177f
in membrane transport, 35
thiopental, 69
pharmacological actions of, 67t
for status epilepticus, 72
third-generation antihistamines, 124
thromboembolism, hormone replacement therapy and, 99b
vomiting, 193
chemosensitive trigger zone and, 193–194
cytotoxic agents and, 234
gastrointestinal tract during, 193, 194f
higher centres and, 193
opioids and, 105
physiology of, 193–196, 194f
vestibular nuclei during, 194
voriconazole, 222
xenon, 66
yeast, 220b
yohimbine, 47–48
zaleplon, 62
zanamivir, 218
Z-drugs, 62
zero-order (saturation) kinetics, 19–20
zidovudine, 219b–220b
metabolism of, 15b
zileuton, for asthma, 167
zolpidem, 62
zopiclone, 62