c7.pain
C7.Pain and its treatment
Pain is a common presenting symptom in primary care and an important cause of morbidity. Patients with mild to moderate pain self-medicate initially, using familiar analgesics or following pharmacist or lay advice. Those with severe pain normally present to GPs or hospitals. GP referrals to area hospital Pain Clinics are common.
The large psychological and cultural components may cause stoics to ignore a pain until the
condition is difficult to salvage: doctors have dismissed the pain of their own myocardial infarc-
tion as ‘indigestion’! Despite its universality and the existence of effective remedies, journal arti-
cles frequently discuss the poor management of post-operative, chronic and terminal disease
pain.
This chapter discusses the characteristics and pharmacotherapy of various types of pain, to guide best practice. Morphine-like drugs are referred to here as ‘opioids’, although this term strictly describes only synthetic compounds, ‘opiates’ being of natural origin. The term ‘narcotic’ is not synonymous with ‘opioid’: it describes central nervous system depressants that relieve pain, producing narcosis (sedation and unconsciousness) in sufficiently high doses. It is widely used legally for addictive drugs of abuse.
Introduction
Although pain is a universal experience, it is
difficult to define. One possible definition is:
An unpleasant sensory and emotional experi-
ence associated with actual or potential tissue
damage or described in terms of such damage. It
serves biological functions, warning of external
danger, e.g. excessive heat or physical trauma,
and internal pathology, e.g. inflammation or
blockage of a ureter by a kidney stone, enabling
avoidance or treatment. It is inherently self-
limiting when the provoking source is removed
or cured.
This indicates that pain is not simply a phys-
ical sensation. Pain perception also depends on
the patient’s emotional reaction to the stimulus
(see below).
Types of pain
Acute pain usually has a readily definable cause.
Its biological function is protective, acting as a
warning that an external threat is noxious, or
signalling organ malfunction. It has a well-
defined time of onset, often associated with
signs of hyperactivity of the autonomic nervous
system, e.g. tachycardia, hypertension and
pallor, depending on the severity of the symp-
toms and how the patient interprets them. The
best way of managing acute pain is to diagnose
and treat the cause, though this is often clear,
e.g. following any kind of trauma. Temporary
relief with analgesics is valuable while healing and recovery proceed.
Chronic pain is usually considered to be pain
that has lasted for longer than 6 months. It does
not signify a danger that requires immediate
avoidance and a patient may not interpret such
pain as indicating serious disease. Further, adap-
tation by the autonomic nervous system over
time may lead to the absence of objective physical
signs. However, there is often progressive phys-
ical deterioration, with sleep disturbance and
weight loss. In severe cases, patients undergo
serious affective and behavioural changes, e.g.
major depression (see Chapter 6).
Essential components in the treatment of chronic pain are the identification of any organic problem, i.e. accurate diagnosis, and the recogni-
tion and management of significant affective and environmental factors.
Pain threshold and assessment
The patient’s mood, morale and the meaning of
the pain for that patient affect their pain percep-
tion. Thus if a patient has chest pain and a
relative or close friend has recently had an MI,
the patient may interpret his or her pain as a
life-threatening event. This results in the pain
threshold being lowered, i.e. anxiety is algesic,
resulting in less tolerance of the pain or greater
awareness of it. Conversely, if another friend
with a similar pain interprets it as indigestion,
or it is diagnosed as such, this would not be
very stressful, the pain threshold would not be
lowered, and the pain may be tolerated.
However, this may not be a rational response
(see Chapter 3). Although it is possible to
measure an individual’s pain threshold, e.g. by
applying a defined stimulus, usually a constant
heat source at a defined distance and recording
the time from application to withdrawal, this is
purely a research tool, e.g. when assessing the
effectiveness of a new analgesic or for comparing
analgesics. However, this does not help when
assessing a patient’s pain. Further, attention
must always be paid to factors that modulate the
pain threshold (Table 7.1).
Introduction 457
Assessment
Pain is a subjective experience, so only the indi-
vidual affected knows its nature and severity: the
individual patient’s assessment and description
are vital. Useful clues can be gained from a
patient’s response to a particular analgesic, e.g. if
pain is described as ‘severe’ but relief is obtained
from modest doses of paracetamol (aceta-
minophen), it is probable that there is a sig-
nificant emotional component to the pain
perception. Careful, empathetic exploration of
this aspect may be more rewarding than the use
of increasingly potent analgesics. Also, the extent to which a condition interferes with
social and pleasure activities may be a better
guide to severity than absence from work or
school. Other clues are obtained from what a
patient reports and their observable behaviour
(Table 7.2).
The origin of acute pain is usually easy to diag-
nose, unlike that of chronic pain, which is often
of obscure origin, particularly when it is due to
tment
non-malignant disease. A careful assessment
should be carried out before treatment is started
and patients should be re-assessed regularly. The
salient features to be elucidated are given in
Table 7.3.
Figure 7.1 gives some examples of pain assess-
ment tools. The visual analogue scales are rapid
and enable immediate feedback, e.g. advice on
the use of medication or a decision to change
medication. Patients should be given a fresh
tool sheet on each occasion: if they see the
previous sheet it prejudices how they respond.
There are other ways, e.g. a pictorial scale of
faces expressing response to different pain
levels. The Short Form McGill pain question-
naire (see References and further reading, p.
511) can be completed within 5 min and is
well-validated. It is important to document the
results of the assessment, to provide a clear
history of the progression of pain and the progress of analgesia for use by all personnel involved with a patient’s care.
Pathophysiology of pain
An understanding of how the sensation of pain
is generated is essential to an appreciation of
how modification of these pain pathways can
ameliorate the pain.
Gate theory
Various theories have tried to integrate the
anatomical pain pathways and the psychological
and neurological components that contribute to
the perception of pain. The generally accepted
model is the ‘gate control theory’, illustrated
diagrammatically in Figure 7.2. This was first
proposed by Melzack and Wall in 1965, and has
since been modified as knowledge has increased.
The theory proposes that neuronal impulses
generated by noxious stimuli are modified in the
dorsal horn of the spinal cord by a specialized
mechanism (‘gate’), which can tend to either
inhibit or facilitate transmission of the pain
impulse from peripheral organs to the brain.
The gate is not an ‘all-or-none’ mechanism,
and a balance between opposing factors deter-
mines how much of the initial nerve impulse
is transmitted through it.
It has been shown recently that the SNC9A
gene determines the structure of the voltage-
gated sodium channels that are responsible for the transmission of signals along afferent pain fibres. Mutations in the gene may result in an inability to sense pain or excessive pain sensi-
tivity (hyperalgesia). This discovery points the way to a new type of analgesic.
tment
Pain receptors and fibres
Two main groups of skin receptors have been identified:
• High-threshold mechanoreceptors (HTMs),
which detect local deformation, e.g. touch.
• Polymodal nociceptors, which detect a variety
of types of injury (e.g. heat) and noxious
(harmful) stimulation. These do not have a specialized structure and are simply bare nerve endings in the periphery.
Stretch receptors also occur in muscles, the wall
of the gut and the capsules of internal organs.
Three types of nerve fibres are involved in pain
transmission. The A-beta fibres are large, myeli-
nated and fast-conducting (30-100 m/s). They
have a low stimulation threshold and respond to
light touch. The A-delta fibres are small, lightly
myelinated and slower-conducting (5-15 m/s).
They respond to pressure, heat, chemicals and
cooling, and give rise to the sensation of sharp
pain, producing reflex withdrawal and other
prompt action. The C fibres are small and
unmyelinated and therefore slow-conducting
(0.5-2 m/s); they respond to all types of noxious
stimuli and transmit more prolonged, dull pain
signals. The last two of these types of fibres
usually require high-intensity stimuli to trigger a
response.
According to the gate control theory, A-delta
and C fibres transmit pain signals to the dorsal
horns of the spinal cord. Impulses in these fibres
can be modulated by A-beta activity that can
selectively block impulses from being trans-
mitted to the transmission cells in the
substantia gelatinosa of the spinal cord. Such
blockage prevents upward transmission to the
CNS, and no pain sensation is perceived. This
explains why rubbing an injured area, or
applying a ‘counter-irritant’ such as capsaicin,
which stimulates the A-beta fibres, can relieve
the pain caused by an injury to that area, which
stimulates the smaller C-fibres.
The gate control mechanism is believed to
operate continuously, even in absence of an
apparent trigger, because there is a continuous
barrage of impulses from, principally, the C-
fibres, whose receptors are continually active
and react only slowly to stimuli. The effect is to
set a threshold below which there is no effector
response. Action subsequent to stimulation
depends on the numbers of fibres involved, their
firing rate, the proportion of large and small
fibres, and the effect of central control mecha-
nisms. The latter may over-ride the gate control,
as occurs under hypnosis. When the complexi-
ties of this mechanism have been elucidated,
new drugs or techniques of pain control may
emerge.
Pathological pain
It has been suggested that peripheral tissue
damage (e.g. from trauma, surgery or cancer)
causes central sensitization, i.e. neuronal
changes occur that make the spinal neurones
hyper-responsive for a long period to afferent
signals that would not normally trigger the
gating mechanism. One consequence of this
theory is that prophylactic (‘pre-emptive’) local,
regional or opioid analgesia should be given
before surgery or any predictable moderate to
severe pain, to prevent central sensitization
occurring. The concept also accords well with
our experience of treating severe chronic pain:
for effective control it is essential that the pain
should not be allowed to recur (see below).
New classes of analgesics are emerging, e.g.
compounds that block spinal cord receptors for
excitatory amino acids such as N-methyl-D-
aspartate (NMDA). One such drug is dizocilpine,
Pathophysiology of pain 461
which blocks the NMDA ion channel and resem-
bles ketamine, but has a much greater potency
and is more selective for the receptor. Dizocilpine
has been shown to prevent and eliminate
central sensitization in animals. Further,
Swedish research indicates that ketamine, which
was introduced as an IV anaesthetic, is an effec-
tive analgesic at concentrations lower than those
required to produce anaesthesia, or at which
central nervous side-effects (hallucinations and
other transient psychotic effects) are trouble-
some. Ketamine is being used increasingly in
palliative care for difficult-to-control cases and
may give dramatic improvement that is main-
tained for several months. Other general anaes-
thetics are widely used for the relief of obstetric
pain (p. 507).
Neurotransmitters involved in pain
Opioid receptors and endogenous opioids
The important discoveries of stereospecific opioid
receptors (of which several subtypes are known)
and endogenous opioids further increased our
understanding of the biochemical mechanisms
involved in pain transmission and perception.
Several families of endogenous opioids have
been identified including the endorphins,
enkephalins and dynorphins (p. 468). Each
family is derived from a distinct precursor
polypeptide and has a characteristic anatomical
distribution.
Other transmitters and mediators
Physical or chemical insult can stimulate noci-
ceptors. Inflammation, ischaemia or other pain-
inducing stimuli cause the release of noxious
chemicals (e.g. bradykinin, histamine and 5-HT)
in injured tissues. Prostaglandins (PGs),
although not directly producing pain, appear to
sensitize nociceptors to various chemical and
pressure stimuli. This explains why NSAIDs,
which block PG synthesis, are effective analgesics
in some situations.
Substance P (neurokinin-1), a polypeptide
probably released by the small-diameter C-fibres,
is believed to be involved in pain transmission in the dorsal horns of the spinal cord. It is probably
not the actual transmitter, but initiates a series
of events leading to the recruitment of pro-
inflammatory agents. The latter release media-
tors, e.g. PGs, LTs, 5-HT and histamine, which
stimulate the nerve endings and cause sensitiza-
tion. Sensitization involves a lowering of the
trigger threshold, producing hyperalgesia. Exci-
tatory amino acid transmitters, e.g. glutamate
and aspartate, may also be involved.
Pain transmission may be blocked if opioid
receptors have already been occupied by endor-
phins at the spinal level. If successful in passing
through the gating mechanisms, and several are
probably involved in the total pathway, the pain
impulse is transmitted via the reticular activating
system of the pons and midbrain to the thalamus.
From there, they are directed to the appropriate
part of the cerebral cortex where the impulses are
perceived as pain. The limbic system, which is
anatomically close to these areas, is thought to
be responsible for the emotional component of
pain (see Chapter 6). Transmission of the pain
impulse may be modified in the CNS by the
presence of 5-HT and other chemical mediators.
It has been shown recently that pain signalling
is amplified by certain ion channels in neuron
membranes. There are ten isoforms of the
sodium channel protein that share a common
structure but have different amino acid composi-
tions. Gene SCN9A encodes for the Nav 1.7
sodium channel that is expressed preferentially
at high levels in dorsal root ganglion and sympa-
thetic ganglion neurons. It is deployed at the
endings of nociceptive neurons close to the site
of pain initiation and loss of the SNC9A gene
causes inability to experience pain. This opens a
new avenue for analgesia, by blocking either
Principles of analgesic use 463
SNC9A transcription or translation or the Nav
1.7 protein. Preliminary work suggests that this can be done without interfering with
sympathetic nerve signalling.
The exact pathophysiology of pain is
extremely complex and is still not fully under-
stood. Figure 7.3 shows a simplified concept of the pain pathways and ways in which current
treatments are thought to interrupt it.
Principles of analgesic use
The WHO recommendation on how to achieve
effective analgesia is: “Dose patients by the
mouth, by the clock and by the ladder.” These
points, and the general principles that should be
employed when using analgesics, are outlined
below.
Type and characteristics of pain
Because various types of treatment are available
to manage pain (Figure 7.4), it is important
to determine whether the pain is acute or
chronic, to diagnose the cause, and to ascertain
if any psychogenic component is present
before deciding on the appropriate approach to
treatment.
Acute pain generally responds well to anal-
gesics, but chronic pain presents a far more
complex problem. It often requires a multidisci-
plinary approach, employing several different
modes of therapy. While the main emphasis in
this chapter is on analgesics, alternative methods
of pain control will also be discussed briefly, to
place them in context.
Choice of analgesic
The type and severity of the pain will usually
determine the specific drug or regimen to be
used. Initiation of analgesia should be at the step
appropriate to pain severity on the WHO ‘anal-
gesic ladder’ (Table 7.3) and the appropriate dose
and potency of analgesic is found by titrating the
dose upwards, or down, until pain control is
achieved at minimal dosage. Table 7.3 provides
only an outline of the analgesics used, a more
comprehensive listing being given in Table 7.4.
The use of many of these drugs may be a matter
of personal preference or convenience: it is
always preferable to learn to use a few drugs well,
rather than a wide range indifferently. TheUK
Department of Health document “Building a
Safer NHS for Patients ” recommends
using a limited range of opioids.
Individualization of dosage
Dose requirements are affected by many vari-
ables including the severity of the pain, the
patient’s pain threshold, age, weight and the
presence of concurrent disease. Each analgesic
should be given an adequate trial by dose titra-
tion, i.e. modifying the dose until pain control is
adequate or until dose-limiting side-effects
occur, before switching to another drug (see, for
example, Chapter 12 and the BNF, Section
10.1.1).
When using opioids in terminal disease, the
only upper limit is that dose which successfully
relieves the patient’s pain without causing unac-
tment
ceptable side-effects. For example, sedation is
only a problem if it becomes unacceptable to
the patient, but respiratory depression may be
important, especially if respiratory function is
already compromised, or unimportant. A recent
research paper concluded that: “Properly titrated
opioids have no respiratory depressant effect in
adults who are awake”.
The American Pain Society has recently recom-
mended that patients and their families should
be fully involved in pain assessment and
management, including changes in dosage or
routes of administration, and measurement of
outcomes. However, it is common for patients to
tolerate a lower analgesic dose when managed in
a professional environment. This is due partly
to better analgesic management and partly
because, in the home environment, the anxiety
of their families or carers that the patient
should obtain complete relief leads to over-
dosing. Sometimes the patient seeks an exces-
sive dose to spare their families from distress at
seeing them suffer. The anxiety of families and
carers is readily transmitted to patients, either
overtly or inadvertently, and exacerbates the
problem.
Dosage schedule
Traditionally, pain relief has been given on a
‘when required’ basis, so that the patient was
expected to experience a return of pain before
requesting more analgesic. Such an approach
may sometimes be useful in acute pain, to see
whether the symptoms are regressing, but is
inappropriate in treating chronic pain because
pain itself is a potent algesic (pain promoter): if
pain is allowed to recur, a higher dose of anal-
gesic will be required to re-establish pain control.
Thus, regular administration of analgesics is
essential in chronic pain so that the patient’s
memory of the pain is reduced and, hopefully,
gradually obliterated. This controls the psycho-
logical component and associated fear. When
the patient has confidence in prescriber and
treatment, the pain threshold may be raised,
making it possible to reduce the analgesic dose.
The drugs, and their dose level, frequency and
routes of administration, must be reviewed
frequently to ensure not only that analgesia is Routes of administration
adequate but also that doses of concurrent
drugs are appropriate and that side-effects are In selecting the most appropriate route of drug
minimized and managed suitably. administration for a patient, factors such as ‘nil-by-mouth’, gastrointestinal obstruction,
persistent vomiting, limited venous access or
reduced muscle mass have to be taken into
account.
Oral and sublingual
The oral route (per os, PO) is preferred if it is avail-
able although some opioids, e.g. pethidine (meperi-
dine), are poorly absorbed from the gut and
bioavailability can vary widely between patients.
The peak effect usually occurs 30-60 min after
dosing with normal oral formulations. Because
peak activity occurs much later with most
modified-release preparations, it is inappropriate
to initiate pain control with these products.
Ideally, the total daily requirement should be
determined using a standard-release formulation,
i.e. one with rapid release of the drug, preferably a
solution. However, tablets may be used (e.g. with
morphine) if the solution is disliked. When stable
pain control has been achieved, this dose is then
translated to the equivalent dose of a modified-
release preparation to reduce the dose frequency.
The sublingual route is particularly attractive, because it provides a rapid onset of action and avoids the losses from first-pass metabolism, but may be unsuitable for very ill patients who produce little saliva. However, the Expert Working Group of the European Association for Palliative Care advises against the buccal and sublingual routes because there is no evidence for their clinical superiority over other routes
and absorption may be unreliable.
Parenteral
The intramuscular (IM) route is commonly used post-operatively. It has the disadvantages of wide fluctuations in absorption, a 30- to 60-min lag time to peak effect, and a more rapid decline in activity than after oral administration.
In cancer patients, who are often cachectic (i.e.
profoundly ill and malnourished) and have
reduced muscle mass, IM administration can be
painful and normal doses can give abnormally
high peak concentrations. Because many cancer
patients have low platelet counts, IM injection
may cause severe bruising. Further, IM injection
tment
causes some muscle damage and the release of
creatinine kinase, so this route must not be used
within 48 h of a suspected MI (see Chapter 4)
because this enzyme is used as one marker for MI.
The gluteal (buttock) muscles are often used
because of their large mass and it is less likely that
a major blood vessel will be entered accidentally.
IM injections are preferably avoided in children because they may give rise to complications.
A bolus intravenous (IV) dose obviously
provides the most rapid pain relief and avoids
first-pass metabolism. However, rapid action also
means rapid adverse reactions. This is particu-
larly important with the opioids, which cause
respiratory depression. The main determinant of
the time taken to achieve a therapeutic effect
with a centrally acting drug is its lipid solubility,
which determines how quickly the drug leaves
the plasma and is distributed into the CNS.
The IV route does not appear to confer any
advantage for maintenance dosing, and opioid
tolerance may occur more rapidly. However, it
enables rapid titration of the analgesic dose in
patients with acute, severe pain or an acute
exacerbation of chronic pain. The IV infusion
of analgesics after major surgery gives good
post-operative pain control and has been shown
to decrease the recovery period and reduce
post-operative complications.
The oral route is equally effective for treating stable chronic pain, unless the patient is unable to absorb oral medications because of vomiting, dysphagia or bowel disease. In such cases, SC infusion is the preferred parenteral route.
Spinal anaesthesia is used routinely in
obstetrics and increasingly for surgery on
patients who are unsuitable for a general anaes-
thetic, post-surgically and for palliative care in a few terminally ill patients (p. 487).
The continuous SC infusion of opioids is used widely and achieves virtually constant blood
levels (see below).
Rectal
The rectal route (per rectum, PR) is another
alternative to oral administration in patients
who are vomiting or on a ‘nil-by-mouth’
regimen, provided that it is not precluded by
bowel disease. Several analgesics are available as
suppositories (Table 7.4). This route also avoids
loss of available drug by first-pass metabolism.
Rectal administration should be avoided in
patients with a low platelet count due to cancer
chemotherapy or to disease because it may
provoke bleeding which is difficult to control.
Transdermal
This route is used currently only with fentanyl
and buprenorphine. It is useful if the oral route
cannot be used and continuous parenteral infu-
sion is either unavailable or unsuitable. Fentanyl
has a short duration of action if given orally or
by parenteral bolus injection, due to rapid
metabolism and tissue redistribution, whereas
the transdermal patches provide prolonged
dosing.
However, skin reactions may occur with the patches and the BNF advises application to dry, non-irritated, non-irradiated, non-hairy skin of the torso or upper arm, rotation of application sites when a patch is changed and avoidance of the same area for several days.
Guidelines for analgesic use
Accurate diagnosis of the cause of the pain is
very important. However, there may be many
underlying causes, especially in palliative care,
so it may not be possible to identify a specific
cause. The following guidelines are generally
applicable.
• Use the oral route whenever possible.
• Only the patient knows if relief is adequate.
However, treatment goals should be realistic: complete freedom from pain may be difficult to achieve. It may be helpful to set realistic stepwise goals, e.g. freedom from pain, in
collaboration with the patient in the
following ascending order:
- At night.
- At rest.
- On movement.
• If an analgesic fails after a trial at adequate
dose and frequency, move up the ‘analgesic
Principles of analgesic use 467
ladder’ (Table 7.3): a substitute from the same class is unlikely to be more effective. If the patient is not supervised, e.g. in the commu-
nity, it is worth checking that the patient is taking their analgesic(s) and any adjuvant
medicines regularly and at the prescribed dose. Fear of addiction is common.
• If increased analgesia is required, the dose
should be increased but the dosage interval
should remain unchanged. Reducing the
dosage interval below that appropriate for
the drug merely makes life more difficult for
the patient.
• Pain should not be allowed to recur. Drugs
with a short duration of action, e.g. pethidine
(meperidine), are unsuitable for the manage-
ment of sustained severe pain.
• Opioids are not a panacea: due weight must
be given to the use of adjuvants (p. 479), the
management of psychosocial aspects and intercurrent disease, and the control of adverse reactions.
• Tolerance to opioids, and dependence on
them, is not usually a practical problem when
treating severe pain. Many patients receiving
palliative care remain on a uniform dose
throughout much of their illness. Opioid use
should not be dictated by a short or poor
prognosis, but by the needs of the patient.
Physical dependence does not preclude dose
reduction. If the patient’s condition improves
or if other treatments relieve the pain, dose
reduction may be essential to avoid toxicity.
• There is some evidence that a state of hyper-
algesia may occur with high IV or intrathecal
doses of opioids but it is not clear to what
extent this is relevant to clinical practice.
The effect may be due to altered hepatic
metabolism, i.e. a shift from the production
of morphine-6-glucuronide (M6G), a potent
analgesic, to M3G, an opioid antagonist.
• Always keep an open mind and review the
treatment frequently. It may be possible to
reduce the dose following a period of stable, good control. Conversely, a requirement for increasing doses is probably not due to drug tolerance or dependence but may indicate a deterioration in the underlying disease or the onset of another condition.
Analgesic drugs and techniques
Opioid analgesics
Mode of action
Opioid receptor sites
As mentioned earlier, receptor sites exist in the
brain, spinal cord and elsewhere where opioids
such as morphine bind to produce analgesia (and
other pharmacological effects). The body’s
natural ligands for these receptors are the endor-
phin, enkephalin and dynorphin peptides.
Receptor-binding studies have identified at least
three major types of opioid receptors, designated
mu (l; two subtypes, l1, l2), delta (d, two
subtypes) and kappa (j; three subtypes), each
with distinct roles so that the type of pharmaco-
logical effect associated with each receptor is
different. These effects are summarized in Table
7.5. About 65% of the amino acid composition
of the three receptor types in animals are iden-
tical or very similar, as are most of their trans-
membrane regions and intracellular loops, but
most of their extracellular loops differ. A further
sigma (r) receptor has been described, but its
role is uncertain because opioid activity there is
not antagonized by naloxone, a specific opioid
antagonist.
Most of the research in this field has been
carried out in rats and mice, but the evidence
from them is not directly relevant to humans. A
complication is that some of the receptors and
their subtypes have been proposed based on
drug-binding studies, but their pharmacological
properties are ill defined or unknown. Because of
these uncertainties and because further receptors
and subtypes may exist, it is not currently
possible to allocate the actions of drugs with
certainty to specific subtypes in man. However,
this is an active research field and drugs are likely
to emerge that act at specific receptors to achieve analgesia without the unwanted effects.
Morphine and related plant alkaloids have
molecular structures similar to those of the
endogenous peptides, and so activate the same
receptors. Methadone, although chemically unre-
lated, can adopt a similar configuration. Natu-
rally occurring and synthetic opioid drugs are
classified according to the subtypes of receptors
to which they bind, and the type of response
that they thus evoke (Table 7.5). The available
drugs include pure opioid agonists, partial
agonists, agonists-antagonists, and pure antago-
nists. The affinity with which a drug binds to a
receptor is important: analgesics with a high
receptor affinity, e.g. buprenorphine, exert an
analgesic action for much longer than their
plasma half-life would suggest.
Opioid agonists and antagonists
Pure agonists (e.g. morphine) elicit a maximum
response if given in sufficient concentration.
However, a partial agonist (e.g. buprenorphine)
can only produce a partial response irrespective
of the concentration, and there may even be a
decreased response if the optimum concentra-
tion is exceeded. The morphine-like opioids are
thought to exert their agonist effects primarily
at the mu receptor and to a lesser degree at the
kappa receptor (Table 7.5). Partial agonists (Table
7.6) bind with the mu receptor and compete
with the agonists, both naturally occurring and
exogenous. If they are used in combination with a complete agonist, they may act as competitive
antagonists and the level of analgesia may be
reduced, or as partial agonists, so that they
show only limited activity. Mixed agonists-
antagonists, e.g. pentazocine, are antagonists at
the mu receptor but are still effective as anal-
gesics through agonist effects at the kappa
receptor, the agonist effect being either complete
or partial.
At the other end of the spectrum are the pure
antagonists, which are used to reverse respira-
tory depression post-operatively, to treat opioid
poisoning (e.g. naloxone), and to prevent relapse
in detoxified opioid addicts (e.g. naltrexone).
Opioid-sensitive and opioid-insensitive pain
Virtually all acute pain, with the possible excep-
tion of labour pain, falls into the opioid-sensitive
category, the analgesic effect being related both
to the type of agent used (weak or strong opioid)
and the dose. The WHO defines ‘weak opioids’
as those used to control mild to moderate pain
and ‘strong opioids’ are those used for moderate
to severe pain.
Neuropathic pain (e.g. post-herpetic neuralgia
and pain resulting from a stroke), includes de-
afferentation pain, i.e. pain resulting from
damage or interruption of afferent (sensory)
nerve fibres, is partially opioid-sensitive and an
adjuvant, e.g. a tricyclic antidepressant, usually
amitriptyline or nortriptyline, an anticonvulsant,
e.g. gabapentin or pregabalin (see Chapter 6) or
nerve blocks may be required. Opioid-insensitive
pains include most headaches. Pain arising from
muscle spasm is best dealt with using muscle
relaxants, e.g. diazepam and baclofen.
The management of post-herpetic neuralgia is discussed on p. 505.
Other types of pain may also be partially
opioid-sensitive, e.g. that resulting from bone
metastases is best treated with a combination of
NSAIDs and opioids. NSAIDs may be adequate
on their own, e.g. diclofenac in palliative care or
as suppositories post-operatively when patients
are ‘nil-by-mouth’, or possibly in combination
with paracetamol (acetaminophen),. However,
opioids may be needed initially or for continuing
support. Some other types of cancer pain (e.g.
that arising from nerve compression, raised
intracranial pressure and extensive tumour
infiltration of tissues) may require an opioid plus
an adjuvant, e.g. a potent anti-inflammatory
glucocorticoid with minimal mineralocorti-
coid activity (betamethasone or dexamethasone).
Psychogenic pain must always be addressed by
identifying the underlying causes as well as by
drug management, e.g. anxiolytics, antidepres-
sants or other psychotropic agents in addition to
analgesics. Analgesic adjuvants are discussed on
p. 479.
Therapeutic use
Morphine
This has become the standard against which
other opioid analgesics are judged and is gener-
ally the treatment of choice for chronic severe pain in advanced cancer. No other strong opioid, given orally, is consistently superior.
In addition to its central analgesic action,
morphine causes euphoria and a sense of detach-
ment. It also causes drowsiness for up to the first
7 days of treatment. This profile is clinically
useful as it reduces the anxiety and anguish that
are commonly associated with severe pain from
whatever cause, e.g. MI and terminal disease.
However, morphine should not be used primarily
as a sedative. Occasional patients remain drowsy,
in which case a dose reduction and a slower dose
titration can be tried, and coexisting problems
should be sought, e.g. the concurrent use of
other sedatives. Liver impairment has little effect
on the hepatic metabolism of morphine until it is
severe, but renal damage prolongs the duration
of action because 90% is renally excreted, mostly
as glucuronides, and morphine-6-glucuronide
(M6G) has twice the analgesic potency of
morphine.
Morphine aggravates functional gastro-
intestinal pain (e.g. from colonic spasm or constipation), so it is unsuitable in patients with these problems, although the latter can be
managed with stimulant laxatives.
One of the most important advances in the
management of chronic pain has been the intro-
duction of modified-release oral formulations of
morphine, which usually give excellent control
tment
with once- or twice-daily dosing. These contain
morphine bound to an ion exchange resin from
which it is released by inward diffusion of sodium
and potassium ions. Because of their slow onset of
action, the appropriate dose must be established
using quick-release dosage forms, e.g. solutions or
normal tablets, and the first dose of a modified-
release product should be given with the last
dose(s) of quick-release product to cover the
period until the new steady state is achieved.
Adjustment of the doses of both types of product
may be necessary to prevent breakthrough pain
occurring. If breakthrough pain should occur in
the early stages of modified-release medication, it
is controlled with solution or injections (occa-
sionally normal-release tablets) and the dosage
regimen reassessed.
Suppositories are a useful alternative to injec-
tions if vomiting is a problem or if patients are unable to swallow. They are also convenient if a patient’s carer cannot give injections.
The potential hazard of respiratory depression
due to morphine may be increased because it
appears to reduce the sensation of breathless-
ness, so the patient should be in a supervised
environment when large doses are needed.
Papaveretum is a preparation of morphine
hydrochloride (85%), papaverine and codeine,
which is now rarely used in the UK. It no longer
contains noscapine, a centrally acting cough
suppressant, and the reformulated product
contains a lower weight of papaverine but the
same dose of morphine. This has created prob-
lems when dispensing and special care is
required. Papaveretum has also been confused
with papaverine, which is used to treat male
impotence.
Morphine is the antitussive of choice for the
treatment of the distressing cough of terminal
lung cancer, with methadone as an alternative.
Many former traditional morphine mixtures, e.g. the ‘Brompton Cocktail’, opium tincture and camphorated opium tincture (paregoric), are no longer used in the UK because they confer no benefit over morphine alone.
Morphine is sometimes used for topical anal-
gesia when local anaesthetics do not give adequate relief (unlicensed indication).
A guideline to the equi-analgesic doses of opioids is given in Table 7.7.
Other pure agonist drugs
Diamorphine (heroin, 3,6-diacetylmorphine)
Opinions differ over the choice between heroin
and morphine. Because of its abuse potential and
its disputed benefit, diamorphine is not available
(often illegal) in most countries outside the UK.
Diamorphine itself is not an opioid agonist, but
is rapidly metabolized to 6-monoacetylmorphine
(6-MAM) and morphine, which are. Because both
diamorphine and 6-MAM are more lipid-soluble
than morphine they penetrate the blood-brain
barrier more rapidly and so have a faster onset of
action, though the final activity is due to the
morphine produced on hydrolysis. Diamorphine is
generally regarded as more potent than morphine
on injection, both as a euphoriant and analgesic,
and may cause less nausea and hypotension.
However, the oral potencies of the two are
similar because, when given orally, heroin is
completely hydrolysed to morphine before
absorption from the gut.
Diamorphine is unstable in solution, so
morphine is preferred for oral solutions. However,
diamorphine is very soluble in water and so is
preferred in the UK for IM and SC administra-
tion as the injection volume is small. This may be important in very emaciated patients. Solutions are prepared from the sterile powder as required and are not stored.
Pethidine (meperidine)
This moderately potent analgesic is most
commonly used peri-operatively and in obstetric
analgesia. Because it is unpredictably absorbed
and undergoes significant first-pass metabolism,
oral bioavailability is poor and variable, with
some trials showing equivalence with paracetamol
(acetaminophen). However, some patients obtain
satisfactory relief with oral pethidine, and sat-
uration of metabolic pathways may increase
bioavailability with chronic use (but see below).
Given parenterally, it has a swift onset of action as
it is highly lipophilic and crosses the blood-brain
barrier readily. This property makes it useful
for preoperative medication and treating acute
pain. The short duration of action (1-3 h)
normally makes it unsuitable for treating
chronic pain, as approximately 2-hourly dosing
would be required.
Further, pethidine is unsuitable for chronic use
because its toxic metabolite norpethidine accu-
mulates, causing anxiety, agitation, tremors and
seizures. The latter is cleared renally and also
accumulates in renal impairment. Pethidine
probably causes more nausea, vomiting and
hypotension than other opioids, and is said to
have less effect on smooth muscle, so it is some-
times used for renal or biliary colic, though
there is no good evidence to support this: IM
or rectal diclofenac is usually preferred for this
indication.
Although pethidine may produce less euphoria
and sedation than morphine, this is disputed, and
respiratory depression and postural hypotension
are common at effective doses. Most of these
side-effects can be corrected by using it with the
antipsychotic agent haloperidol, which potenti-
ates the analgesia, permitting lower doses to be
used, and is anti-emetic, sedative and possibly
euphoric. However, the toxicity of haloperidol
limits the maximum dose that can be used (see
Chapter 6).
Pethidine interacts with MAOIs to cause either
severe hypertension or hypotension, depending
tment
on the patient and the relative doses of each,
and so should not be used in patients taking
hypotensive drugs or MAOI antidepressants, except under very close supervision.
Methadone
This agent is well absorbed and well tolerated by
mouth and has been used with some success in
pain control, particularly in the USA. However,
its metabolism and excretion are complex, so the
patient’s physical, mental and emotional condi-
tion needs to be monitored closely. Its medicinal
use in the UK is primarily for those intolerant of
morphine and it is used widely to manage opioid
dependence.
Pharmacokinetics. Methadone has a very long
elimination half-life on multiple oral dosing
(about 25 h) and is very highly bound to plasma
and CNS proteins, so that accumulation can
occur with chronic use. This leads to drowsiness
and confusion, which may occasionally be life-
threatening. The half-life is extended con-
siderably by renal or hepatic impairment, so
methadone must be used with special care in
elderly or debilitated patients and in alcohol
abusers. Steady-state blood levels are not
achieved until at least 4-5 days after the initia-
tion of therapy or a change in dose, so loading
doses are sometimes used to achieve rapid
control. Despite the long half-life, the duration
of action is about 6-8 h initially, increasing to
6-12 h with chronic dosing, so several daily
doses must be given initially. Once- or twice-
daily dosing is used when steady-state blood
levels have been achieved, because of the risk of
accumulation and toxicity.
Although the degree of sedation and respira-
tory depression reflect the absolute amount of methadone in the body, the total body load does not influence the magnitude of analgesic response, possibly owing to different affinities for the different receptor subtypes.
Use in opioid dependence. Methadone is
widely used in drug abuse clinics as a replacement
for heroin, to help recreational drug users main-
tain a stable lifestyle. When a drug user has
learned alternative strategies to heroin use for
coping with stressful situations, the methadone
may be withdrawn gradually. However,
methadone may itself lead to a morphine-like
dependence and the potential for abuse is similar
to that for morphine. Although withdrawal symp-
toms are less intense than those with morphine,
and it may relieve the physical withdrawal
symptoms without giving ‘highs’ for the same
length of time, the onset of withdrawal symp-
toms is slower (24-48 h) and they are more
prolonged. Opioid misusers are usually given a
very gradually reducing methadone dosage
regimen, the level of which is carefully titrated
to the patient’s needs.
However, buprenorphine is now more widely
used to manage opioid detoxification (with-
drawal). Further, the value of methadone in
treating opioid withdrawal has been challenged:
it is not a cure and is often regarded more as a
method of social control, to minimize the crim-
inal activities often associated with drug misuse,
rather than as therapy.
In these days of opioid-addicted mothers, neonatal opioid dependence should not be
overlooked.
Hydromorphone
This potent analgesic, about 7.5 times as potent as morphine, is used in the UK when patients are intolerant of other opioids. It has a rapid onset of action but a relatively short half-life. Oral
dosing is required 4-hourly, or twice daily with the modified-release preparation.
It is more widely used in the USA and Europe,
probably because diamorphine is not available
there. Like the latter it is very soluble in water
and injection volumes are small, so it may be
useful in syringe drivers (see below). However,
the injectable preparation is not licensed in the
UK.
Dipipanone
This agent is usually given orally only. Although it has been given by SC or IM injection in the past, it should not be given by the IV route
because this may produce a dramatic fall in
blood pressure.
The only available tablet in the UK is formu-
lated with cyclizine (an antihistamine) as an anti-emetic. This makes the product unsuitable for chronic use, e.g. in palliative care.
Opioid analgesics 473
Oxycodone
This is effective orally and is used widely in
North America and elsewhere as a modified-
release oral product and, combined with aspirin
or paracetamol (acetaminophen), for moderately
severe pain. In the UK it is available as normal-
release capsules, modified-release tablets and as
an injection for slow IV injection or SC use. It is
sometimes used in syringe drivers for patients
intolerant of other opioids, but the low concen-
tration of the existing formulation limits the
dose volume that can be given by this route.
Fentanyl and its congeners
These agents are used primarily for intra-
operative analgesia. Opioids are widely used in
low doses to supplement general anaesthesia
with nitrous oxide-oxygen and a neuromuscular
blocking agent. The muscle relaxants, e.g.
suxamethonium, pancuronium and atracurium
(there are many others), relax the diaphragm
and abdominal muscles and may permit light
anaesthesia to be used. In addition, they relax
the vocal cords and so facilitate the passage of an
endotracheal tube to assist in passing anaesthetic
gases or oxygen. Patients who have received a
muscle relaxant must always have assisted or
controlled respiration when fentanyl is used.
Alfentanil, fentanyl and remifentanil have a rapid onset of action (1-2 min) and are used to reduce the induction dose of an anaesthetic,
especially in poor-risk patients.
Remifentanil can be used intra-operatively as an
IV infusion in adults and young children. Because
it is rapidly metabolized by blood enzymes it has
a very short duration of action and does not accu-
mulate, so post-operative respiratory depression
is unlikely. Because of its very short duration of
action, additional analgesia is usually needed
post-operatively.
Alfentanil and fentanyl may also be used intra-
operatively as an IV infusion or as IV bolus
injections. These may cause severe respiratory
depression and cardiovascular side-effects,
especially with fentanyl. Because respiratory
depression may occur for the first time post-
operatively, patients who have received either
drug need to be observed carefully for some
hours after recovery. Sufentanil (not licensed in
the UK) is used similarly in the USA.
The side-effect of respiratory depression with alfentanil and fentanyl is used to advantage in intensive care patients on assisted respiration to manage respiration without interference from their endogenous respiratory drive. The opioid effect is reversed with naloxone when respiratory depression is no longer required.
Fentanyl is used widely as transdermal patches
(Chapter 13), each of which lasts for 72 h, for the
control of stable chronic pain when the oral
route is unavailable or unsuitable. Because it
takes about 12 h for the patch to produce
adequate analgesia, it should be applied at an
appropriate time, e.g. simultaneously with the
last modified-release morphine dose when trans-
ferring from morphine. The equivalence between
the fentanyl patches and morphine is that fentanyl
25 lg/h for 72 h is equivalent to morphine
hydrochloride 90 mg daily.
Unfortunately, the patches are often used
poorly in the community. Because of the lag time
in reaching an effective concentration after a
change in dose there may be too rapid titration;
titration by increments that are too large; or inap-
propriate use in unstable pain, all of which may
cause toxicity.
There is a buccal formulation, supplied with a
special applicator, for breakthrough pain, but
many patients find it difficult to use. An
intranasal formulation is under trial for break-
through pain when using patches, as a more
convenient alternative to the buccal and IV
formulations.
Fentanyl is used by SC infusion in palliative care, mainly for patients with renal failure because its metabolites are inactive.
Opioid rotation
Although oral morphine is the potent analgesic of
choice, there is a minority of patients in whom
their pain is inadequately controlled despite
large doses. It is unclear why this situation occurs.
Postulated mechanisms are the complex metabo-
lism and pharmacokinetics of morphine, with
active metabolites that may accumulate, and
down-regulation of receptors or other receptor
change.
These patients may benefit from a change
in the route or method of administration, e.g.
SC injection, patient-controlled analgesia (p.
tment
489) or epidural anaesthesia (p. 508). Nerve
blocks (Figure 7.3 and p. 487) may also be
appropriate.
However, a satisfactory result may be achieved
by a change of opioid, i.e. opioid rotation (also
known as opioid switching). It is preferable to
use a pure agonist, e.g. hydromorphone, methadone
or oxycodone. Transdermal fentanyl or buprenor-
phine is unlikely to be useful in this context
because of the pharmacokinetics of the patches.
An initial dose reduction is often advocated, and
is safe practice, with access to medication for
breakthrough pain, taken when necessary. The
dose can then be titrated according to patient
response.
The reasons for benefit from rotation are not clear, but include differing metabolism, different receptor subtype responses and variable sensitivity to side-effects.
Opioid rotation is not a universal panacea, and
frequent changes are undesirable. It is important
to evaluate possible reasons for loss of pain
control, e.g. new symptoms or intercurrent
disease, and to consider alternative approaches
to pain control.
Codeine and its congeners
Codeine phosphate itself is a relatively weak anal-
gesic, but is converted to morphine and norcodeine in the liver. Although 10% of Caucasians cannot carry out this change the relevance of this to
clinical practice is not known.
It is used for mild to moderate pain. It has a
‘ceiling’ effect, i.e. if the normal maximal dose of
60 mg fails to control the pain, further dose
increases do not produce more analgesia. The
side-effects of drowsiness, nausea and constipa-
tion often become intolerable at the ceiling
dose. It is appropriate to co-prescribe a laxative
(e.g. lactulose) in patients taking regular doses.
It is widely used as a cough suppressant and as
a component of compound mild analgesics, e.g.
with aspirin or paracetamol (acetaminophen).
There is little evidence for the efficacy of these
compound products, because the codeine doses
are usually sub-therapeutic, i.e. 30 mg, and
there is a considerable increase in side-effects.
However, they are firmly entrenched in clinical
practice.
Codeine formulations are best avoided in all
children under 12 and should not be used in
infants: the common belief that codeine is an
effective sedative and hypnotic has resulted in
fatalities.
It has the potential to cause dependence and pharmacists need to be vigilant when clients are using regular supplies of OTC products containing it.
Dihydrocodeine tartrate is possibly a more potent analgesic than codeine and is used for moderate to severe pain. However, both drugs are on Step 2 of the WHO ladder. Like codeine it is used in compound analgesic products and has similar disadvantages, including dependence. It is sometimes prescribed for dental pain.
Partial agonist and agonist-antagonist drugs
These were developed in an attempt to over-
come opioid dependence problems. Although
not totally devoid of abuse potential, they have
less than that of morphine and other similar
agonists.
Nalorphine was the prototype of this group but is no longer used owing to an unacceptably high incidence of psychotomimetic side-effects. Subsequently, two types of agonist-antagonist drugs have been developed that are classified
according to their activity relative to morphine or nalorphine (Table 7.6).
Agents of the nalorphine type characteristically act only as competitive antagonists at the mu-
receptor but have varying affinities and intrinsic activities at all receptor types. The mixed
agonist-antagonists of the morphine type have a high affinity for mu-receptors, but a low intrinsic activity there.
Morphine-like opioids
Buprenorphine is a partial agonist that has a 6- to
8-h duration of action and is effective in the
relief of moderate to severe pain unresponsive to
non-opioid analgesics. Its potency is similar to
that of pethidine. It is available in a sublingual
formulation (but see above) and is sometimes
used for premedication and peri-operative anal-
gesia. For acute pain, the onset of analgesia of
the sublingual tablets is about 30 min. However,
the use of buprenorphine in chronic severe pain is
Opioid analgesics 475
problematic. It has a ‘ceiling effect’, like pethidine
(meperidine) and codeine, and a low therapeutic
index, like pentazocine, so increasing the daily
dose above about 3 mg is unlikely to be benefi-
cial. It is used to manage opioid withdrawal (see
‘methadone’ above). The sublingual tablets are
popular among drug abusers and some health
authorities have introduced a voluntary ban on
prescribing these.
The transdermal patches have a lower inci-
dence of side-effects than the sublingual formu-
lation and are suitable for controlling moderate
to severe chronic pain in palliative care and
other patients. It takes about 24 h to reach a
steady-state plasma level with the patches that
are replaced after 72 h. Patches for replacement
after 7 days are also available, the steady-state
plasma level being achieved during the first
application. However, the dose should be deter-
mined with the 72-h patches before switching
to the 7-day formulation. Because buprenorphine
is a partial agonist-antagonist, breakthrough
pain can only be managed appropriately using
the buprenorphine sublingual tablets: the use of
other opioids gives unpredictable effects. The
analgesic response should not be assessed before
24 h, but any dose adjustments should be made when the patches are changed. Due to its long terminal half-life of about 30 h, patients who
suffer side-effects necessitating removal of a patch need to be monitored for a further
24-30 h. Unlike the pure opioid agonists, the
effects, and side-effects, of buprenorphine are only partially reversed by naloxone.
Side-effects. Because of its high receptor
affinity, large doses of other opioids may be
required to displace buprenorphine from the
receptor. This may lead, in those patients who do
not obtain adequate pain relief with buprenor-
phine, to a confused situation of inadequate anal-
gesia, despite a large opioid dose, but enhanced
toxicity. If given to a patient receiving other
opioids chronically, buprenorphine may precipi-
tate pain and withdrawal symptoms. Thus it
should be used alone. In common with most other
opioids, buprenorphine causes dose-related respira-
tory depression. Because of its high receptor
affinity its effect is not readily reversed by
naloxone, making it more hazardous in overdose.
Buprenorphine is highly emetogenic in some patients but appears to be less likely than other opioids to cause constipation.
Meptazinol has about one-tenth of the anal-
gesic potency of morphine. It is unusual in that it
is thought to have two central mechanisms of
action: a partial agonist-antagonist effect at
opioid receptors, plus effects on central cholin-
ergic receptors. It has a variable onset of action
(0.25-3 h orally, 0.5 h rectally), and its duration
of action is also variable (2-7 h). Thus dosing
is required every 3-6 h, depending on patient
response. It undergoes extensive first-pass metab-
olism, so blood levels after oral dosing are low
and this route is better suited to the short-term
relief of moderate pain, e.g. peri-operatively. For
moderate to severe pain, meptazinol is best
given by IM or slow IV injection. It is likely to
cause nausea and vomiting.
Meptazinol is claimed to cause less respiratory
depression than other opioids, possibly because
of its cholinergic effects or its preferential
action at the mu-receptor, and may be a useful
analgesic to consider in patients with compro-
mised respiratory function, but there is a diver-
gence of opinion on this point. Some clinicians
advocate the cautious use of morphine, despite
its respiratory depressant effects, in patients
with compromised lung function, believing
that pain itself acts as a respiratory stimulant
and reduces the risk of administering a known
respiratory depressant. Further, should respira-
tory depression occur following morphine
administration, this can readily be reversed
by administering naloxone, whereas complete
reversal of the effects of a mixed agonist-
antagonist such as meptazinol cannot be
achieved readily with naloxone and additional
measures are needed, e.g. assisted respiration
with oxygen (see Chapter 5) and possibly a
respiratory stimulant.
The potential for abuse is probably less than that of morphine because its euphoric effects disappear with increasing dose.
Nalorphine-like opioids
Pentazocine is a moderately potent analgesic
that is used infrequently nowadays in the UK
as it shares the hallucinogenic potential of
nalorphine and causes a high incidence of
tment
confusion and hallucinations. Like buprenor-
phine, pentazocine may precipitate a withdrawal
reaction in patients who are opioid-dependent.
It is unsuitable for pain associated with MI
because, unlike morphine, it can increase the
cardiac workload.
The oral efficacy of pentazocine is poor (slightly
less potent than codeine) and, due to its low ther-
apeutic index, doses cannot be increased greatly
to treat severe pain without also markedly
increasing the incidence and severity of side-
effects. However, it is more potent when admin-
istered by any parenteral route than both codeine
and dihydrocodeine.
Tramadol
This has both opioid and non-opioid modes of
action, enhancing adrenergic and serotonergic
actions. It is a metabolite of the antidepressant
trazodone, so it is not surprising that it inhibits
noradrenaline (norepinephrine) re-uptake and
the stimulation of serotonin release at synapses.
These secondary effects are responsible for the
psychiatric side-effects seen in some patients,
but they are also believed to facilitate the
pathways that inhibit pain perception.
Tramadol is a weaker analgesic than most other
opioids and appears to cause less respiratory
depression. Because it causes less constipation it
is often used when toileting difficulties create
post-operative problems. There may also be a
reduced potential for dependence, but tramadol
should not be used if there is a history of drug
dependence or convulsions. It has been used for
obstetric and peri-operative pain and in MI, but
is unsuitable for intra-operative analgesia during
light anaesthesia.
Side-effects. Apart from those referred to
above, it may also cause occasional hyperten-
sion, anaphylaxis, hallucinations and confu-
sion, particularly in elderly patients. Because it
is not classed as a controlled drug in the UK,
it is probably used more frequently than is
justified.
Summary of opioid side-effects
Actions of morphine other than those described
above are usually considered to be side-effects Nausea and vomiting
Morphine and its derivatives stimulate the CTZ
(Chapter 3) and may cause nausea and vomiting,
although this tends to be transient, wearing off a
few days after initiating therapy or an increased
dose. These effects may be avoided by prophy-
lactic co-administration of an anti-emetic (see
Chapter 3) over this period. Anti-emetics are also
appropriate in patients who are already vomiting
owing to drug use or who have a history of
vomiting with opioids, and may be given
initially either rectally or parenterally to bring
existing vomiting under control.
Anti-emetics are not indicated in patients who
are not currently nauseated and so should not be
used routinely: good practice is not to prescribe
a drug unless there is a positive indication.
However, it is appropriate to prescribe a small
quantity of an anti-emetic for use ‘as required’,
in anticipation of possible need. Because dipi-
panone is only available combined with an
anti-emetic (cyclizine), it is not recommended
for use in palliative care.
Because the incidence of nausea and vomiting is higher in ambulatory patients, it is thought
that a vestibular component is also involved. It is often helpful for the patient to lie quietly if
this problem occurs.
Constipation
Opioids cause an increase in gastrointestinal
sphincter tone and a decrease in propulsive peri-
stalsis. This causes delayed gastric emptying and,
almost inevitably, constipation. The regular co-
administration of a stimulant laxative plus a
stool softener is nearly always used in anticipa-
tion of the problem. Dantron, as co-danthrusate
(dantron plus docusate) or co-danthramer (dantron
plus poloxamer ‘188’), is probably the most effec-
tive agent. Dantron is specifically licensed in the
UK only for the treatment of constipation in
terminal care, but it has also been used for
patients with cardiac failure or MI, to avoid
cardiac stress due to bowel strain. The licence is
restricted because studies in rats have indicated a
potential carcinogenic risk. Products containing
dantron may colour the urine red, and patients
should be warned of this apparently alarming
effect. Dantron may also cause a rash in the
buttock area in incontinent patients, so it should
Opioid analgesics 477
be reserved for use when other laxatives are
ineffective.
The equivalent product in the USA is a combi-
nation of casanthranol (a natural anthracene)
and docusate (dioctyl sodium sulphosuccinate).
Other smooth muscle effects. Morphine also
increases the tone in the sphincter of Oddi,
which leads to increased pressure in the biliary
system and occasional biliary colic. Other
actions on smooth muscle include increased
urethral tone, causing difficulties in micturition,
and very rarely bronchoconstriction after large
doses.
Respiratory depression
Morphine and other opioids can significantly
depress respiration, and this is usually the cause
of death from overdose. Respiratory rate, tidal
volume and response to hypercapnia or hypox-
aemia are all reduced. However, like many of its
other side-effects, respiratory depression is not
usually a limiting factor in patients who are
experiencing severe pain, because pain is a
potent arousal mechanism. Nevertheless, opioids
must be used with great care in patients with
advanced respiratory disease or otherwise
depressed respiratory function. Opioid doses
should be reduced if other procedures, e.g. nerve
block (Figure 7.2 and p. 487) or radiotherapy,
reduce pain successfully.
Effects on the eye
Stimulation of the oculomotor nerve causes
constriction of the pupil, which is often a diag-
nostic aid in cases of morphine overdose and
addiction. Thus, opioid analgesics are generally
avoided in patients with head injury, because
the opioid-induced pupillary changes, nausea
and general CNS clouding may mask the signs
induced by trauma and confuse the neurological
examination.
Cardiovascular effects
The usual doses of opioid analgesics generally do
not have major cardiovascular effects in patients
with normal cardiac function. However, mor-
phine may cause venous pooling and postural
hypotension, through its venodilator action. The
consequent reduction in cardiac preload (see
Chapter 4) is an additional benefit to analgesia,
and its euphoric action may help immediately following MI, to relieve severe anxiety.
Hypersensitivity
Occasionally, allergic-type reactions occur with
opioid agents, and both local reactions at the site
of injection and systemic allergic symptoms have
been reported. If a patient is hypersensitive to
morphine, both codeine and diamorphine (heroin)
are also contra-indicated as they are structurally
similar. However, methadone and pethidine
(meperidine) are suitable alternatives, being
chemically unrelated.
Dependence
Opioid analgesics can produce both physical and
psychological dependence, although the latter
appears to be a rare event when opioid analgesics
are used to relieve pain. If a patient asks for
increased dosages of analgesics because their
pain has worsened or not been controlled, this
should not be perceived automatically as drug-
seeking behaviour or evidence of dependence.
Physical dependence does occur, and can be
managed by reducing the dose of opioid slowly
when it is no longer needed for pain, rather than
stopping abruptly, which causes unnecessary
withdrawal symptoms.
Naloxone. Adequate doses of this antagonist
will reverse completely all the actions of pure
opioid agonists, so it is a complete antidote for
both the actions and side-effects of morphine-like
drugs. However, care should be taken when
using naloxone to reverse opioid-induced respira-
tory depression, as patients who have been using
opioid agents chronically are extremely sensitive
to antagonists and too high a dose of naloxone
can precipitate a withdrawal reaction and recur-
rence of severe pain. Further, it will not fully
antagonize the action of partial agonist and
agonist-antagonist drugs (see above).
Because the duration of action of naloxone is
shorter than that of morphine and other opioids,
repeated injections or IV infusion may be
required. The naloxone dose varies widely, being
dictated by the patient’s condition and response.
Oxygen and the respiratory stimulant doxapram
(hospital use) may be needed to spare the naloxone
dose and so maintain adequate analgesia.
tment
Less potent analgesics
These are mainly used for the treatment of acute or chronic pain resulting from trauma, surgery and chronic systemic diseases such as arthritis. They include low-potency, centrally-acting morphine-like compounds (‘weak opioids’), e.g. codeine and dihydrocodeine, and drugs that act on peripheral pain pathways, e.g. aspirin, salicylates and NSAIDs (see Chapter 12). Paracetamol (aceta-
minophen) also has central effects, but at
different receptors from the opioids, and its toxicity is discussed in Chapter 3.
Weak opioids
These drugs have a ceiling to their analgesic
effect, usually because of dose-limiting adverse
reactions, and therefore have a limited efficacy
relative to the strong opioids. Therefore
combinations of these drugs with paracetamol
(acetaminophen), provided that it is not
contra-indicated (i.e. liver function is not compro-
mised), or aspirin (if tolerated), may be expected
to have an additive, possibly synergistic, anal-
gesic effect. However, the BNF states that
“Compound analgesics are commonly
used, but the advantages have not been substan-
tiated”. Despite this, they are prescribed very
widely. Their continuing popularity, with both
patients and prescribers, may represent the
triumph of experience over theoretical good
practice.
Codeine is chemically related to morphine and is
metabolized to morphine in the liver, so it shares
its pharmacological actions. It is thought to have
less abuse potential but is too constipating for
long-term use.
Dihydrocodeine is mainly used for moderate
pain. It has a flat dose-response curve, so there is
no advantage in increasing the dose above that
normally recommended: if analgesia with dihy-
drocodeine is inadequate, a change to a strong
opioid is indicated.
Dextropropoxyphene (propoxyphene) resembles
methadone structurally, and is less potent than
codeine. There has been considerable contro-
versy over its widespread use owing to serious problems if taken in overdose. In the UK, dextro-
propoxyphene has been used principally in combi-
nation with paracetamol (acetaminophen) as
co-proxamol, which is unfortunately commonly
used as a agent for suicide. It has been suggested
that as little as 15-20 tablets of this combination
can prove fatal, especially if alcohol is impli-
cated. The main cause of death in overdose with
dextropropoxyphene alone is respiratory depres-
sion, but in overdose with co-proxamol this is
compounded with the hepatotoxicity of parac-
etamol (acetaminophen; see Chapter 3). Acute
over-dosage with co-proxamol requires prompt
administration of naloxone, to antagonize the
dextropropoxyphene, resuscitation treatment and
management of paracetamol overdose. If naloxone
is not used, patients may die of cardiovascular
collapse before reaching hospital.
Because of these serious toxic effects in over-
dose, and the fact that it is one of the most
common suicide agents in the UK, co-proxamol is
regarded as unsuitable for prescibing in the NHS
and is being withdrawn from use in the UK.
Like other opioid analgesics, dextropropoxyphene can lead to dependence, the likelihood being
about the same as with codeine.
Nefopam is structurally unrelated to the
opioids and is sometimes useful when the pain
has not responded to other analgesics. Its main
advantage is that it does not cause respiratory
depression, but its sympathomimetic and
antimuscarinic side-effects, notably restlessness,
dry mouth, urinary retention and, less often,
blurred vision, tachycardia, insomnia, headache
and confusion, may be troublesome. Thus,
nefopam must be used with caution in the elderly
and if there is evidence of renal or hepatic
impairment, because it is extensively metabo-
lized in the liver and largely excreted in the
urine. Because of its adverse cardiovascular and
CNS effects, nefopam is contra-indicated in MI
and if the patient is liable to convulsions.
Non-steroidal anti-inflammatory drugs
NSAIDs appear to act peripherally at the pain
receptor level, and so do not produce the phys-
ical dependence often associated with opioid
analgesia. They are particularly useful in treating
Analgesic adjuvants 479
patients with chronic disease accompanied by both pain and inflammation, e.g. RA (see Chapter 12) and for the short-term treatment of mild to moderate acute pain, including musculoskeletal injuries and bone pain. Particular indications include the relief of pain accompanying dysmen-
orrhoea, and that associated with neoplastic bone metastases. In the latter case, combinations of an opioid with an NSAID are likely to be considerably more effective than an opioid alone.
The topical and other uses of NSAIDs are discussed in Chapter 12.
Analgesic adjuvants
These include three types of agent:
• Co-analgesics (secondary analgesics), for
example an anticonvulsant, e.g. clonazepam, gabapentin or pregabalin, or a low-dose tricyclic antidepressant, e.g. amitriptyline.
• Other psychotropic agents, e.g. normal dose
antidepressants, to treat the depression that
frequently accompanies moderate to severe chronic pain.
• Corticosteroids, e.g. dexamethasone, to reduce
oedema around a tumour and so prevent
pressure on adjacent nerves or other tissues.
Other drugs that are used to prevent or treat
the adverse effects of the primary analgesic, e.g.
antiemetics or laxatives, are not discussed here.
Analgesic adjuvants tend to be used primarily
in treating the chronic pain of neoplastic disease
(Table 7.8). Their inclusion in a drug regimen
may enhance pain relief, or it may be possible to
reduce the dose of opioid, and consequently its
side-effects. Other categories of drugs are also
used, e.g. antispasmodics, neuroleptics and anxi-
olytics. The mechanisms by which these agents
exert their effects are not clearly established, but
are unrelated to the opioid receptor system.
First-generation antihistamines, e.g. ali-
memazine, chlorphenamine and promethazine are
used primarily for their sedative properties and
may also help to relieve nausea and skin irrita-
tion. However, many of these are markedly
hypnotic, notably alimemazine and promethazine,
and the latter has a long duration of action
(about 12 h). Great care is needed when using
the sedative antihistamines with other
psychotropic drugs, especially opioids, because
profound sedation may result. They also have
marked antimuscarinic activity and may cause
urinary retention, glaucoma and pyloroduo-
denal obstruction. Patients vary considerably in
their response to antihistamines and children
and the elderly are very susceptible to their
side-effects.
Stabbing or shooting pains (neuralgia) caused
by nerve inflammation or damage appear to
respond particularly well to anticonvulsant
drugs, which are thought to act by suppressing
abnormal spontaneous activity in traumatized
nerve fibres. Carbamazepine is generally the most
successful agent, although side-effects can be
troublesome, especially in the elderly. If carba-
mazepine is ineffective, it is worth trying phenytoin
or another anticonvulsant before abandoning
this line of treatment. Therapy should be initiated
gradually, increasing the dose carefully until relief
is obtained or unacceptable side-effects are
encountered (see Chapter 6).
Tolerance to the side-effects of all adjuvants is improved by starting with low doses and titrating the dose slowly, especially in frail or elderly patients.
The best results with analgesic adjuvants are
often obtained if they are introduced early on in
the disease process, before demyelination (loss
of the myelin sheath of nerves) has occurred.
Demyelination may result from infiltration or
sustained pressure by a tumour, producing nerve
block, slowing of nerve conduction or nerve irri-
tability and inflammation. It also makes nerves
more liable to viral infection. The end result of
these processes may be paraesthesias, partial
paralysis, painful spasm, etc.
Damage to central or peripheral nerves, either
as the prime cause of neuropathic pain (see
above) or due to the secondary effects from
a tumour is recognized by abnormal sensitivity
in an area of autonomic, sensory or motor
dysfunction.
Psychotropic drugs
Drugs from disparate therapeutic groups are
used, and their modes of action in the treat-
tment
ment of pain is controversial. It is postulated
that they block the re-uptake of certain neuro-
transmitters in the pain pathway, e.g. 5-HT and
noradrenaline (norepinephrine), thus inter-
fering with pain impulse transmission or its
modulation. However, their pro-analgesic effect
does not simply result from their psychotropic
actions because they are effective at much
lower doses, and act more rapidly, than in the
treatment of depression (see Chapter 6). If
depression needs to be treated, full antidepres-
sant doses should be used. They may raise a
pain threshold that had been lowered by the
understandable anxiety and depression associ-
ated with chronic pain. Some studies have
suggested that tricyclic antidepressants with an
intact tertiary amine group (e.g. amitriptyline
and imipramine) are the most effective. Anti-
depressants are thought to be most effective
against the ‘burning’, deafferentation pain
associated with sensory nerve damage, which is unresponsive to opioids.
The benefits of an antidepressant may be
further increased by combination with a small
dose of a neuroleptic drug, such as a buty-
rophenone (e.g. haloperidol) or a phenothiazine
(most commonly perphenazine). However, these
are rarely used in palliative care and such combi-
nations should only be used by prescribers
experienced in their use, because cardiac
arrhythmias, postural hypotension, excessive
sedation, dyskinesias (e.g. TD, p. 420), dystonias
(abnormalities of muscle tone), myelosuppres-
sion and enhanced antimuscarinic side-effects
may be a problem. Therefore neuroleptics are
best avoided.
Anxiolytics. Diazepam, is especially useful because it has muscle relaxant properties in addition to its anxiolytic effect, and so is used frequently. If anxiety is not a problem and diazepam is too sedating, then baclofen is a suitable alternative for muscle spasm.
Cholecystokinin (CCK), the hormone
involved in food digestion that is released by
enteroendocrine cells in the gut (see Chapter 3),
is also produced in the brain where it acts as a
neurotransmitter involved in feelings of anxiety.
It has recently been shown that CCK antago-
nists, e.g. devazepide and proglumide, reduce the
sensation of anticipated pain. Although this points to a novel mode of analgesia, no product of this type has been marketed.
Glucocorticosteroids
These have a wide application in advanced
neoplastic disease because they reduce the
inflammatory swelling around tumours, and
hence the pressure on nerves and in bones, thus
alleviating the pain. These actions are additional
to any growth-suppressant action on tumour
cells, but the effect may be only temporary.
Glucocorticoids also suppress the release of
mediators such as histamine and kinins and are
euphoric, thus raising the pain threshold. It is
usual to start with a high dose to bring the symp-
toms under control and then reduce it as quickly
as possible to the lowest effective dose, or to zero
if the corticosteroid is ineffective (see also
Chapters 5, 12 and 13). Slow dose reduction is
not necessary if treatment has been less than
3 weeks.
Dexamethasone, and to a lesser extent
betamethasone, lowers intracranial pressure in
cerebral oedema and is especially useful for
relieving the headache and associated symptoms.
Low-dose corticosteroids (e.g. 2 mg dexametha-
sone) also improve mood and, possibly, appetite,
but co-analgesia requires higher doses - about ten
times that dose.
Anabolic steroids have been advocated to
improve food utilization and increase muscle
bulk and strength in patients with oesophageal,
gastric and small bowel tumours who have prob-
lems with swallowing and nutrient absorption.
An increase in muscle bulk would also reduce the
discomfort of repeated injections. However,
there is no good evidence for clinical benefit.
Feeding by a nasogastric tube or percutaenous
endoscopic gastrostomy (PEG), with a cannula
giving direct access to the stomach to bypass
swallowing problems, is preferred.
Analgesic adjuvants 481
Cannabis and cannabinoids
The use of cannabis as monotherapy or as an
adjunct to other analgesics is hotly debated
because of its current rescheduling in the UK as
a Class C Controlled Drug, to which special
licensing conditions apply. Marijuana is the
most widely used, or abused, psychoactive
substance. It is regarded by most governments
as having no therapeutic uses, although there
are numerous anecdotal reports of its benefit,
e.g. in relieving spasticity in multiple sclerosis.
The UK Medicines and Healthcare Products
Regulatory Agency (MHRA) has recently stated
that it has “not objected to importing Sativex
oromucosal spray for use in multiple sclerosis
patients”. Clearly, further research is needed,
but this seems to imply that a UK licence is
likely.
Only one cannabinoid, nabilone, is currently used in the UK, for the management of nausea and vomiting caused by cytotoxic chemotherapy uncontrolled by other drugs, and this has
numerous side-effects.
The use of cannabis, especially at a young age, is a definite risk factor for the development of schizophrenia (see Chapter 6).
A major barrier to progress is that cannabis
smoke contains more than 60 cannabinoids
and no standardized product exists. One
component, D-9-tetrahydrocannabinol (D-9-
THC), seems largely to reproduce effects similar
to smoking cannabis. However, it would be
rash to assume that other cannabis compo-
nents are not adjuncts or that they do not
have potential value as co-analgesics. The
precise effects of D-9-THC vary with the imme-
diate environment, dose and route, and the
psychological attributes of the user, similarly to
alcohol.
Cannabinoid receptors are widely distributed in the brain. Why this is so, and the functions of endogenous ligands, are unknown but are of
considerable interest.
sensation of anticipated pain. Although this points to a novel mode of analgesia, no product of this type has been marketed.
Glucocorticosteroids
These have a wide application in advanced
neoplastic disease because they reduce the
inflammatory swelling around tumours, and
hence the pressure on nerves and in bones, thus
alleviating the pain. These actions are additional
to any growth-suppressant action on tumour
cells, but the effect may be only temporary.
Glucocorticoids also suppress the release of
mediators such as histamine and kinins and are
euphoric, thus raising the pain threshold. It is
usual to start with a high dose to bring the symp-
toms under control and then reduce it as quickly
as possible to the lowest effective dose, or to zero
if the corticosteroid is ineffective (see also
Chapters 5, 12 and 13). Slow dose reduction is
not necessary if treatment has been less than
3 weeks.
Dexamethasone, and to a lesser extent
betamethasone, lowers intracranial pressure in
cerebral oedema and is especially useful for
relieving the headache and associated symptoms.
Low-dose corticosteroids (e.g. 2 mg dexametha-
sone) also improve mood and, possibly, appetite,
but co-analgesia requires higher doses - about ten
times that dose.
Anabolic steroids have been advocated to
improve food utilization and increase muscle
bulk and strength in patients with oesophageal,
gastric and small bowel tumours who have prob-
lems with swallowing and nutrient absorption.
An increase in muscle bulk would also reduce the
discomfort of repeated injections. However,
there is no good evidence for clinical benefit.
Feeding by a nasogastric tube or percutaenous
endoscopic gastrostomy (PEG), with a cannula
giving direct access to the stomach to bypass
swallowing problems, is preferred.
Analgesic adjuvants 481
Cannabis and cannabinoids
The use of cannabis as monotherapy or as an
adjunct to other analgesics is hotly debated
because of its current rescheduling in the UK as
a Class C Controlled Drug, to which special
licensing conditions apply. Marijuana is the
most widely used, or abused, psychoactive
substance. It is regarded by most governments
as having no therapeutic uses, although there
are numerous anecdotal reports of its benefit,
e.g. in relieving spasticity in multiple sclerosis.
The UK Medicines and Healthcare Products
Regulatory Agency (MHRA) has recently stated
that it has “not objected to importing Sativex
oromucosal spray for use in multiple sclerosis
patients”. Clearly, further research is needed,
but this seems to imply that a UK licence is
likely.
Only one cannabinoid, nabilone, is currently used in the UK, for the management of nausea and vomiting caused by cytotoxic chemotherapy uncontrolled by other drugs, and this has
numerous side-effects.
The use of cannabis, especially at a young age, is a definite risk factor for the development of schizophrenia (see Chapter 6).
A major barrier to progress is that cannabis
smoke contains more than 60 cannabinoids
and no standardized product exists. One
component, D-9-tetrahydrocannabinol (D-9-
THC), seems largely to reproduce effects similar
to smoking cannabis. However, it would be
rash to assume that other cannabis compo-
nents are not adjuncts or that they do not
have potential value as co-analgesics. The
precise effects of D-9-THC vary with the imme-
diate environment, dose and route, and the
psychological attributes of the user, similarly to
alcohol.
Cannabinoid receptors are widely distributed in the brain. Why this is so, and the functions of endogenous ligands, are unknown but are of
considerable interest.
Mode of action
These agents depend on the ability of the
lipophilic aromatic moiety of their molecules to
dissolve in and attach to Na÷ channels and pene-
trate the lipoid nerve membrane. They prevent
the large transient Na÷ flux across excitable
nerve membranes by interacting directly with
the intracellular components of voltage-gated
sodium channels. The result is that the
excitability threshold rises and nerve conduction
slows and eventually fails. Although the drugs
also bind to potassium channels and inhibit
them at higher concentrations, this is not
thought to contribute to their action.
The smaller the nerve fibre, the more sensi-
tive it is to the action of these agents, so there is some selectivity at the concentrations used: they block transmission by small pain fibres but leave inhibitory (large) pain fibres, touch and
movement relatively unimpaired.
Local anaesthetics also have important effects
on calcium flux across cell membranes, so lido-
caine (lignocaine) is the drug of choice for treating
the ventricular arrhythmias that may accompany
MI, heart surgery and digitalis intoxication. This
effect also contributes to their systemic toxicity.
Use
Except for procaine, the penetration of local
anaesthetic agents through ophthalmic and
mucous membranes is greater than through the
skin, permitting effective local anaesthesia in the
eye, nostrils, throat, urethra and rectum. This
property is used to facilitate the passage of endo-
scopes, catheters, etc. into body cavities and
enables even relatively major operations (e.g. for
cataract) to be carried out with minimal trauma
as day-care procedures.
Local anaesthetics are not normally injected
intravenously, except for the use of IV lidocaine
(lignocaine) for treating ventricular tachycardias.
However, IV regional anaesthesia (Bier’s block) is
used to anaesthetize a limb, intravascular spread
being prevented by using a tourniquet. The use
of local anaesthetics by IV infusion to produce
general anaesthesia is hazardous and requires
Local anaesthetics 483
expert advice, so this route is rarely used. Regional anaesthesia can also be achieved by spinal use (see below).
Skin anaesthesia
This is difficult to achieve through intact
skin, owing to poor penetration, though a
lidocaine-prilocaine cream may be used under an
occlusive dressing (to increase penetration) for
1 h before painful procedures. This is a eutectic mixture that has a low melting point and is an oil that is able to penetrate intact skin at normal temperatures. It has been used before injecting very nervous children.
A tetracaine gel can also be used similarly
before venepuncture or venous cannulation.
These must not be used on wounds, abrasions or
inflamed skin because absorption may be rapid
and extensive and lead to systemic side-effects.
Ophthalmic procedures
Oxybuprocaine and tetracaine are used extensively.
Because tetracaine produces deeper anaesthesia, it
can be used for minor surgery. Proxymetacaine
and lidocaine-fluorescein eye drops are used for
conventional tonometry (measuring the intraoc-
ular pressure by pressing a membrane against the
cornea) as one of the procedures for diagnosing
glaucoma. However, non-contact methods, e.g.
firing a jet of compressed air at the cornea, are
used increasingly. Oxybuprocaine has a similar
activity to tetracaine, but is less irritant.
Otolaryngology
Lidocaine (lignocaine) is the preferred agent.
Nose, throat, eye and ear procedures are the
only ones in which cocaine is still used. It is
preferably avoided because of its powerful CNS
stimulant action and potential for abuse.
Because of its sympathomimetic effects, due
to blocking of noradrenaline (norepinephrine)
at synapses, cocaine should not normally be
used with adrenaline (epinephrine) and similar
drugs, but some surgeons believe that combi-
nation with adrenaline reduces cocaine absorp-
tion and is operatively beneficial. Cocaine
rapidly produces effective surface analgesia that persists for at least 30 min, depending on the
concentration used.
Spinal anaesthesia
Bupivacaine, levobupivacaine and ropivacaine are
used to block nerve transmission at any point up
to spinal cord level (see Figure 7.3), providing
temporary pain relief. This topic is dealt with on
p. 487. It is likely that levobupivacaine will replace bupivacaine because the L-isomer is the biologi-
cally active form and can be given at higher
doses with fewer side-effects.
Prolongation and enhancement of effect
Prolongation and enhancement of local anaes-
thesia is usually done by combination with
vasoconstrictors. Most local anaesthetics cause
vasodilatation and so are often formulated
with adrenaline (epinephrine), which produces
local vasoconstriction and so prevents rapid
distribution into the surrounding tissues and
the circulation.
This effect prolongs the duration of action and
potentiates the anaesthetic effect, owing to the
increased local concentration of agent. Also, the
reduced rate of systemic absorption enables drug
metabolism to keep pace with administration,
thus reducing the peak plasma level and so
systemic toxicity. Peak plasma concentrations
occur after about 10-25 min, so patients should
be monitored carefully for 30 min after injection
to ensure that no serious side-effects arise.
However, adrenaline (epinephrine) is contra-
indicated if the anaesthetic is to be used in or near the fingers, toes or other appendages, because the intense vasoconstriction of their small blood vessels may result in ischaemic necrosis.
Felypressin, a synthetic derivative of the ADH
vasopressin, may be a suitable alternative to
adrenaline (epinephrine) and is used for dental
procedures if the latter is contra-indicated in a
particular patient. However, vasoconstrictors are
themselves toxic and the same considerations
apply as with adrenaline. Further, they must not
tment
normally be used with cocaine, which has intrinsic vasoconstrictor and mydriatic proper-
ties. An alternative is to use a viscous vehicle,
usually containing dextran 110, and this approach has been used to prolong the local
action of lidocaine (lignocaine) for up to 10 h, though the effect is very variable.
Bupivacaine has a long duration of effect (up to
10 h), and may be useful if adrenaline or fely-
pressin are contra-indicated. However, it has a
slow onset of action (30 min), so lidocaine is
usually used initially, followed by bupivacaine or
levobupivacaine. The time of onset of action of
bupivacaine depends on the concentration, dose
and route used. It is used for peripheral nerve
blocks and infiltration nerve blockade, and is the
principal drug used for spinal anaesthesia in the
UK (Figure 7.3). Bupivacaine is contra-indicated
for use in IV regional anaesthesia, because of
the risks of its long action, cardiovascular and
central nervous toxicity, and because prolonged
restriction of the blood supply would be
required. Although bupivacaine is used widely for
spinal obstetric anaesthesia, the concentration
and circumstances of use in this setting require
expert advice.
Side-effects of local anaesthetics
As usual, inflammation and trauma increase
penetration markedly, so significant systemic
absorption may then occur. On occasion, this
may lead to toxicity, especially with tetracaine
(amethocaine). This drug must never be used on
mucous membranes, e.g. before bronchoscopy
and cystoscopy: lidocaine is safer. The lidocaine-
prilocaine cream used on the skin is irritant to the
eyes and is ototoxic, and so should not be used
on or near these organs.
Local anaesthetics are generally very safe, and
adverse events are rare when they are used
properly. However, apart from the use of the
lidocaine-prilocaine cream and tetracaine gel
mentioned above, they should generally be
avoided for topical application to the skin
because sensitization may occur. Systemic
complications may also occur, usually when
large doses are used to produce spinal nerve
blocks. This is usually from accidental intravas- Physical methods
cular injection, which is particularly hazardous if
injections contain adrenaline (epinephrine).
Traction, to relieve pressure on spinal nerve roots
Hypersensitivity occurs occasionally: com-
pounds of the same chemical type tend to give cross-reactions, but these do not occur between the ester and amide groups (Table 7.8).
Topical agents
Liniments and rubs have a long medical history
and were once the only available treatments for
localized joint and soft tissue damage. The
active ingredients are mostly essential oil prod-
ucts, e.g. camphor, menthol, methyl salicylate,
turpentine, sometimes with capsicum oleoresin
or nicotinates.
More recently, several NSAIDs have been
presented as gels or creams. These have the
advantage that if only one or two joints are
affected they can be applied to those localized
areas. This avoids the need to take larger
amounts of drug orally and some of the associ-
ated side-effects. Because of this, and possibly
because patients are involved in the treatment,
topical NSAIDs are popular. Although penetra-
tion of the drug into joints and soft tissues has
been demonstrated, there is little localized effect
and the therapeutic benefit is debatable.
The modes of action of most topical agents, apart from any specific NSAID activity, are alleged to be:
• Increased blood flow to the area, induced by
the massage and vasodilatation.
• Counter-irritation, i.e. stimulation of C
neurons inhibits local release of substance P
and so attenuates the transmission of pain
impulses. This mode of action has been demonstrated for capsaicin cream, which is used for the alleviation of osteoarthritis and pos-herpetic neuralgia (PHN; p. 505).
A 5% lidocaine (lignocaine) patch is licensed in the USA for the treatment of post-herpetic neuralgia and is occasionally imported into the UK on a named-patient basis.
(see Chapter 12), and manipulation and mobi-
lization of muscles and joints are the traditional
approaches of physiotherapy (PT), osteopathy
(OT) and chiropractic, and are of undoubted
value in some patients. PT also uses the applica-
tion of heat (including short-wave diathermy),
cold, ultrasound, electrical muscle stimulation
and laser therapy. Ultrasound has been advo-
cated for soft tissue injuries, PHN, facial neuritis
and phantom limb pain, i.e. that apparently felt
in an amputated limb.
Ice packs are widely used in the early treatment
of soft tissue injuries, especially sport and similar
traumas. The mnemonic ‘RICE’, i.e. Rest, Ice,
Compression, Elevation, is used by first-aiders.
Care is needed not to cause ‘ice burns’ by using
a cloth between the skin and the cold pack.
Techniques recruiting endogenous inhibitory mechanisms
In addition to increasing the understanding of
how conventional analgesics act, the gate theory
has stimulated interest in alternative methods of
pain relief. The emphasis has shifted from nerve
destruction or blocking of pain conduction to
the recruitment of the body’s own inhibitory
systems. This is the principle underlying the use
of vibration, percussion, massage, and counter-
irritation with rubefacients. The cooling
produced by pain-relieving sprays may also stim-
ulate pain trigger points (see below) or, if the
cooling is sufficiently intense and prolonged, a
degree of local anaesthesia may be produced.
However, skin ‘burns’, or even frost-bite, must be
avoided.
These techniques are not always successful,
but they may provide acceptable pain relief to
some patients with otherwise intractable condi-
tions. Some authorities insist that these simple
methods should be used first, and persevered
with for several weeks, before either a maximum
effect is achieved or the approach is abandoned.
Table 7.9 lists the principal alternative methods of pain control.
Neuromodulation by electrical stimulation
The most commonly used method in this cate-
gory is transcutaneous electrical nerve stimula-
tion (TENS). This is normally used for the relief of
chronic peripheral pain, though it has also been
used to relieve acute pain, e.g. of operative inci-
sions. Electrodes are placed over the painful area,
or at the periphery of a very sensitive area, though
the optimum placing needs to be found by careful
trial by the patients themselves. Acupuncture
sites are occasionally used (see below).
Patients wear a battery-driven pulse generator and vary the frequency, pulse width and power, normally to produce a pleasant non-painful tingling sensation. Patients usually know whether the method is going to be successful
within 5-15 min. The pulse characteristics can be adjusted and the frequency of use may vary from three 1-h sessions daily upwards.
The conditions most likely to respond are post-
herpetic neuralgia, low back pain, phantom limb
pain and post-operative scar pain. The mode of
action is believed to be stimulation of the large,
inhibitory A-beta nerve fibres, closing the pain
gate in the dorsal horn of the spinal cord and
reducing or abolishing upward transmission of
the stimulus.
Invasive techniques have also been used
occasionally. Electrodes connected to a minia-
ture radio receiver have been implanted
around peripheral nerves, near the posterior
columns of the spinal cord, or even in the
brain, and stimulated by a patient-controlled
radio transmitter.
Although complications are rare, electrical neuromodulation is contra-indicated for psycho-
neurotic or emotionally unstable patients, and for opioid addicts. The most common problem is contact dermatitis from the electrodes.
Acupuncture
Therapeutic acupuncture, i.e. for the relief of
chronic pain rather than as an operative anaes-
thetic, is used both by practitioners who follow
the traditional Chinese system, with its over 300
sites for insertion of the needles, and some
Western doctors, who may follow an empirical
system. Needles are inserted to a precise depth
and may be left in place for up to 30 min, or
rotated, moved up and down, or electrically
stimulated. Western practitioners sometimes
insert the needles into sensitive trigger points,
i.e. sensitive sites in muscles associated with
fibromyalgia or myofascial pain that cause acute
pain or muscular spasm when touched.
The mode of action is hotly disputed, but may
involve interference with nerve depolarization,
stimulation of inhibitory nerve fibres, release of endorphins, enkephalins or 5-HT, or hypnotic suggestion. The success of the treatment is certainly influenced profoundly by cultural and psychological factors. One estimate is that about 10% of patients are responders, a further 10% are non-responders, and the remainder experience varying degrees of benefit.
Chemical nerve blocks
Regional nerve blocks
These may be:
• Field blocks, i.e. SC injections at various sites
around sensory nerves in the area of the
procedure.
• Peripheral blocks involve a similar procedure,
but the local anaesthetic is often injected
around a nerve plexus, e.g. the brachial
plexus, an intricate network of nerves
emerging from the spinal cord at the base of
the neck (between the lower cervical and
uppermost thoracic vertebrae) which supply
the whole arm, or the lumbosacral plexus,
which supplies the lower back and limbs.
• Central blocks can be:
- Epidural (extradural, peridural), outside
the spinal cord between the dura mater
and the inner wall of the vertebral canal.
- Caudal, in the lumbar or sacral regions of
the spinal cord, where it divides to form
the cauda equina.
- Intrathecal (subarachnoid), into the CSF
between the pia mater and the arachnoid.
With intrathecal anaesthesia the level of the
nerve block, and so the area and organs affected,
depends on the position of the patient and the
specific gravity (SG) of the anaesthetic solution.
Isobaric solutions have the same SG as the CSF
and exert their effect at the level of injection.
Hypobaric solutions are lighter than CSF and
act higher than the injection site, depending on
the positioning of the patient. Hyperbaric solu-
tions are heavier than CSF and flow towards the
bottom of the spinal cord. They are used
Chemical ner ve blocks 487
primarily for operations in the genital area and on the legs.
Central and other anaesthetic blocks
Local anaesthetics
These agents (see above) may be used to produce
a reversible block of afferent nerves at any
point up to spinal cord level (see Figure 7.3),
including the local segment of the spinal cord.
This provides temporary pain relief or anaes-
thesia for operations on patients in whom
general anaesthesia is unsuitable, e.g. due to
cardiac and respiratory problems or when central
nervous sedation is undesirable. However, local
anaesthetics also block other types of sensation
and motor impulses, though usually to a lesser
extent. The degree and extent of the anaes-
thetized area depends on the concentration and
volume of solution injected.
Epidural (extradural) and intrathecal opioids
are sometimes used for the relief of post-operative
and chronic pain. Injections are usually given
via a catheter located at the correct segment of
the spinal cord. The effect is on the sensory,
dorsal horn nerves. The release of substance P
and other neurotransmitters from primary
afferent nerves is inhibited by pre-synaptic
opioid receptors, and effects on post-synaptic
receptors reduce activity in ascending spinal
tracts. However, conduction in motor and auto-
nomic nerves is unaffected by opioids, so motor
functions and blood pressure are generally not
affected by the spinal use of opioids. The anal-
gesia produced is normally insufficient for intra-
operative pain so spinal opioids are used as
adjuncts to general anaesthetics. Spinal morphine
(often diamorphine in the UK) may provide
8-16 h of analgesia and can give months of low-
dose analgesia in cancer patients without the
side-effects of oral or other parenteral use.
However, continuous infusions of morphine are
safer and technically simpler and are widely
used. Alfentanil infusions have also been used.
Such reversible nerve blocks are also used in
diagnosis and to determine which nerves are
involved in a particular pain process and the
probable result of a permanent nerve block because the production of the latter by nerve
ablation with neurolytic agents can often be
inaccurate and may not achieve the desired result. Levobupivacaine is used intrathecally for this purpose and is the agent of choice in the UK. The use of morphine with a low concentration of bupivacaine is synergistic.
Permanent nerve blocks
Once the correct site has been identified, an
irreversible block produced by the injection of
such agents as alcohol, phenol, chlorocresol
or urethane may provide much longer-lasting
relief. Unfortunately, this procedure may be too
lengthy for patients who have severe intractable
pain.
Alcohol produces total neurolysis, but the
extent of blockage produced may be varied
by using an appropriate concentration of a
phenolic agent. These injections may be
subarachnoid, extradural, subdural, autonomic
or peripheral depending on the site of ablation
that provides satisfactory relief. Surgery or heat
(radiofrequency ablation) may also be used.
These are most useful in treating patients
with well-defined localized pain. As expertise
and knowledge increases, a wider range of
destructive methods, peripheral, central or
autonomic, is becoming available. Because the
peripheral nerves are easily accessible, they are
often chosen as sites for destruction, although
nerve regeneration can occur with consequent
return of the pain. Alternatively, the sensory
root can be destroyed. This should theoretically
result in permanent pain relief, as axonal
regeneration should not occur if the nerve
fibres are interrupted proximal to the sensory
ganglion. Unfortunately, this is not always
successful.
The procedures are potentially hazardous, and
partial loss of sensation and function, throm-
bosis, spinal cord infarction, and even death
have occurred: a skilled and experienced anaes-
thetist or neurosurgeon is required to conduct
such procedures successfully. Because of these
hazards, permanent nerve blocks are procedures
of last resort.
Injection of alcohol has sometimes been used
to destroy the pituitary gland, and so prevent
tment
hormone release, in patients who have wide-
spread bilateral cancer pain, especially if the
tumour is hormone-dependent. Because the
alcohol spreads around the floor of the third
ventricle and into its cavity, it is possible that part
of the success of this technique is from direct
hypothalamic injury. About 70% of patients
benefit, more than half obtaining complete relief.
However, nerve blocks do not usually give
complete pain relief, so adjuvant oral or
parenteral opioids are usually still required,
although it may be possible to use lower doses.
Neurosurgical approaches
The most common surgical procedures for pain
relief are cordotomy (see below) and insertion
of epidural, intrathecal and intraventricular
catheters for opioid or local anaesthetic delivery
directly to specific areas of the CNS. Subarach-
noid catheter injection can sometimes be
successful in cases where conventional opioid
administration has failed to reach central opioid
receptors.
Whereas drug therapy may not completely
remove pain, an effective surgical nerve block
can do so. The main problem is that it may not
be possible to block the appropriate pathway
without impairing other nerve pathways. It is
usual to identify the nerve to be treated using
low-power radiofrequency stimulation and to
follow this with high-power pulses to ablate the
nerve.
Cordotomy involves the interruption of the
anterolateral quadrant of the spinal cord in
the cervical or thoracic region. This may be
done percutaneously using a diathermy probe,
or sometimes by open surgery, and is most
useful in treating unilateral pain below the
shoulders. The method is used primarily in
patients who have a limited life expectancy
(about 2 years), because the development of
alternative nerve pathways often allows pain
to return after some time. Thus, cordotomy is
used primarily in patients with advanced, irre-
versible disease and severe intractable pain, of
whom over 80% obtain complete relief with
such treatment.
Psychotherapy and hypnosis intravenously, though the epidural route may
also be used. When the patient experiences
unacceptable pain, they press a button on the
The role of psychotherapeutic drugs has already been mentioned (see above). These behavioural approaches may be effective if there is no organic basis for the pain (i.e. it is psychogenic), if there is a minor cause but the pain is grossly aggravated by psychological factors and if anxiety and depression exacerbate pain significantly.
Meditation and relaxation training help
patients divert their attention away from pain
and facilitate their tolerance of it. Hypnosis
sufficient to induce a light trance is a recognized
form of treatment and some patients are able to
self-hypnotize. It may relieve pain completely in
the (highly suggestible) 20% of responders, and
be a useful adjunct to other forms of treatment
in many more. Hypnosis is unsuitable for
patients with psychiatric illnesses who may be
impossible to hypnotize or who behave in a
bizarre fashion under hypnosis.
Biofeedback involves linking a patient to
equipment that monitors and displays parame-
ters such as heart rate or blood pressure (which
are increased by pain) or muscle tension (which
may cause pain). The patient is taught relaxation
techniques and sees how this modifies their
physiological responses and reduces their pain
level; thus they learn to control their symptoms
appropriately.
Syringe drivers and patient-controlled analgesia
Definition
This is a technique for continuous, regular or
intermittent dosing with opioids, and occasion-
ally with epidural local anaesthetics, according to the patient’s perception of their own needs
and pain severity. The method is used for controlling acute pain, e.g. post-operatively, following trauma and in burns patients.
The equipment usually comprises a micro-
processor-driven syringe containing the drug
solution, which is administered parenterally via
a catheter. This may be done subcutaneously or
control unit and a predetermined dose of drug is administered.
Advantages
The advantages of PCA are:
• Patients are in control of their pain, not vice
versa, and feel more secure. This alone
improves the quality of analgesia obtained.
• The blood level is maintained more closely
within the therapeutic range than can be
achieved with injections given on demand.
This should improve analgesia and minimize
side-effects, but PCA has to be stopped some-
times due to severe nausea and other side-
effects. Unacceptable side-effects may be due
to overdosing by the patient (see below).
• Doses can readily be titrated to cope with
wide variations in pain severity and patient
needs.
• There is minimal delay between the percep-
tion of intolerable pain and obtaining a dose
of analgesic. There is also less demand on
nursing time and pharmacy provision.
• The system is very versatile and can some-
times be used in a domiciliary setting.
However, some patients tend to overdose and
a syringe driver delivering a SC opioid at a
constant rate, which is not patient-controlled,
may occasionally be used in palliative care.
Disadvantages and limitations
The following points are additional to those
usually associated with opioid or local anaes-
thetic use:
• Highly trained staff are required to set up the
equipment and supervise the patient.
• The method is only suitable for maintenance
analgesia, not for dose titration. The patient’s needs should be stable and established clearly, or readily estimated, and a suitable loading dose given before starting PCA.
• Some patients reject the technique.
• A patient’s understanding of how to use the
equipment may be limited by:
- Extreme age.
- Language or comprehension problems.
- Sedation or confusion.
• Poorly controlled blood pressure may
compromise perfusion of a SC injection site
and the IV route has to be used.
• Changes in renal or hepatic function may
cause significant variation in the plasma
concentrations of drug, as for all routes of
administration.
Modes of use
Two PCA arrangements are possible:
• Patient-controlled analgesia only.
• Continuous background infusion plus a
patient-controlled bolus analgesic.
PCA only
This technique is best suited to acute analgesia. If excessive demand is made and the patient
becomes sedated, this is a self-regulating situa-
tion, because the patient then makes less demand for analgesia until the sedation wears off.
Continuous background infusion plus PCA
This is particularly appropriate for chronic pain relief, e.g. severely burned patients, who often need opioids for long periods. It can be given as 30-50% of their predetermined hourly dose as a background infusion, the PCA function being used for breakthrough pain.
Continuous background infusion only
Strictly, this is not PCA, because there is no
control by the patient. It is more useful in
managing chronic pain, especially in patients
who have stable analgesic needs. Further, they
may be unable to manage the equipment
correctly because they are confused or have little
understanding of the equipment and the princi-
ples of use. It is less suitable for acute pain
because of the inability to manage break-
through pain, the risk of excessive sedation and
other adverse reactions.
tment
Technical and clinical aspects
Technical and clinical points include the following:
• The bolus dose delivered on demand in
PCA must be adequate, but must not cause
unacceptable side-effects.
• The time over which the dose is injected
following patient triggering is adjusted
according to the route used. Thus, for IM or
IV injections the time may need to be length-
ened to 5 min to avoid stinging or inadequate
clearance from the site. Also the ‘lock-out
interval’, i.e. the time after the end of a
dose during which another bolus cannot be
obtained, may need to be lengthened. Alter-
natively, the solution concentration may be
increased so that a smaller volume is injected.
• The lock-out interval should be long enough
to avoid adverse drug reactions. It is usually
5-8 min.
• The maximum dose allowed in a given
period should be controlled, e.g. 30 mg
morphine in 4 h.
• Inadequate analgesia. If usage is:
- Two or less doses/hour, counsel the patient
about fears of opioid use, etc. and advise more frequent use.
- More than three bolus doses/hour, increase
bolus concentration.
- Review other possibilities, e.g. pain is not
opioid-responsive or may be a new com-
plication of the disease and/or surgery.
• Nausea and vomiting:
- Use anti-emetics (see Chapter 3, p. 107),
e.g. rectal prochlorperazine or a 5-HT3 antag-
onist. Transdermal hyoscine (scopolamine)
may be used occasionally in patients under
60 years if injections are undesirable. This is not suitable in the elderly because it may cause urinary retention and confusion.
- Decrease bolus volume or increase the
duration of delivery of each bolus, to give lower peak plasma concentrations.
- Change the route, for example to IM, or
possibly the opioid.
• Urinary retention: relieve with indwelling or
intermittent catheterization.
• Pruritus: this is not usually a problem with IV PCA, but may be with spinal or epidural dosing.
- Change the route or opioid.
- Use a non-sedating antihistamine.
ches, migraine and facial pain 491
- Use low-dose naloxone if the pruritis is
intractable, but care is required because the
analgesia may be reversed if the naloxone dose is too large.
Some common pain situations
The application of the principles discussed above is illustrated by discussing the management of some common pain situations.
Headache, migraine and facial pain
Epidemiology and aetiology
Headache is probably the most common of all
the pain syndromes, and about 80-90% of the
population have at least one headache each year.
Of these, about 15% experience recurrent
episodes of all kinds that interfere with their
normal daily activities, and it is estimated that headaches cause an annual loss of about 70 million working days in the UK.
Peak GP consulting rates occur in the 10- to 40-year age group, the female : male ratio being about 2.5 : 1 overall. The principal causes of
chronic daily headache are given in Table 7.10. There are estimated to be over 5 million
migraine sufferers in the UK, though at least half have mild to moderate pain and do not consult their doctors. Pure tension-type headache is an unusual cause for a visit to a GP.
Analgesic abuse is an important cause of headache, recognition of which requires a high index of suspicion.
The main concern of pharmacists when a
client requests advice is to distinguish between
benign headaches and those requiring medical
intervention (Table 7.11). Headache accompa-
nied by the following features requires referral to
the patient’s GP.
• New pain of sudden onset, especially in those
aged 60 or over.
• A marked change in the character or timing of
recurrent headaches.
• Fever
• Neck stiffness.
• Impairment of cognition, motor function or
vision lasting for more than 24 hours, e.g.
- Understanding or reasoning.
- Abnormality of gait, posture or writing.
- Limb weakness or clumsiness.
tment
- Eye problems, e.g. flashing lights, jagged
lines, blurred vision, loss of part of the
visual field (scotoma).
- Disturbance lasting 24 h is defined as a
transient ischaemic attack (TIA; see
Chapter 4) and still requires referral, espe-
cially in those already taking low-dose
aspirin.
- Sensitivity to touch in the area of the pain
(may be temporal arteritis; see Chapter 12).
- A family history of migraine.
Classification
The International Headache Society has issued
modified guidelines for classifying chronic daily
headache (Table 7.11). Despite this formidable
list, most headaches are benign. Only the four tion (provided that this has been confirmed
most common types are discussed below, the
pain patterns associated with these and other
types being illustrated in Figure 7.5.
Tension-type headache
Clinical features
This common form of headache may arise from
sustained muscle contraction in the cervical
(neck) region or scalp, or be psychogenic in
origin (e.g. caused by stress or depression).
Headaches may be episodic, i.e. occurring on less
than 15 days/month and having no persistent
symptoms. However, they are often chronic, e.g.
on more than 15 days/month for more than
6 months, present at similar times each day,
every morning or evening, or on the same days each week (Table 7.12). The pain is:
• Mild to moderate.
• Typically has a bilateral ‘hatband’ or more
generalized distribution (Figure 7.5(a)).
• Non-throbbing, no burning or pressure
sensations.
• Not aggravated by head movement.
There are few abnormal signs, but the scalp and neck muscles may be tender. Bilateral pain, absence of vomiting and tendency to be chronic distinguish it from migraine (see Table 7.12), which is episodic by definition.
Management and pharmacotherapy
Treatment of this condition is often unsatisfac-
tory because no specific pathology can be iden-
tified and it may be difficult or impossible to
modify employment, social and personality trigger factors. Management and symptomatic treatment involve:
• Possible extensive investigations to eliminate
serious pathology (e.g. cerebral abscess or
cancer, stroke), to reassure the patient.
• Avoidance of any identifiable causes, if
possible.
• Physical and psychological treatments may
help, e.g. relaxation therapy, psychotherapy,
hypnotherapy, ice packs or cervical manipula-
as a safe procedure in that patient).
• Analgesics appropriate to severity, e.g. para-
cetamol (acetaminophen), aspirin (in the over-
16s, if tolerated) or combinations of these
with codeine or dihydrocodeine, but these
may be abused. Combinations with dextro-
propoxyphene are no longer used in the UK.
• NSAIDs. Diclofenac, ibuprofen and naproxen,
sometimes flurbiprofen or tolfenamic acid (unlicensed indication).
• Antidepressants (see Chapter 6) if indicated.
Low-dose amitriptyline, 10-25 mg nightly,
increasing up to 75 mg daily if required is
widely used (unlicensed indication). Anxi-
olytics are undesirable because they may
lead to habituation. Treatment is more effec-
tive if patients present soon after the onset of
symptoms. Antidepressants are withdrawn
gradually after about 6 months’ sustained
improvement.
• It has recently been shown that performing a
brain scan in people who suffer chronic daily
headache relieved anxiety in the short term
and reduced the use of medical resources
significantly, with an associated reduction in
costs.
Analgesic abuse headache
This may accompany any other form of
headache. Because it occurs daily it is frequently
associated with tension headaches. The normal
pattern is that a patient complaining of headache
will manage well initially by self-medicating with
simple analgesics as the need arises. There may
then be a progression to regular daily simple
analgesic use, seeking advice for ‘something
stronger’, consulting their doctor and regular use
of a compound analgesic (e.g. co-codamol or co-
dydramol in the UK), even a potent opioid,
without obtaining satisfactory relief. This adds
the side-effects of the analgesic, i.e. any of the
side-effects of opioids (see above), to the effects
of the headache. Misuse of antimigraine drugs
can cause similar problems. Opioids should not
be required in tension-type headache.
The correct course of action is to stop the daily
analgesic medication. This may cause rebound
exacerbation of headache for 1-2 weeks and • Migraine without aura (common migraine).
withdrawal symptoms may need to be treated with anti-emetics and sedatives. Cognitive therapy (see Chapter 6) usually helps, but failure to improve subsequently should prompt a neurological or psychiatric investigation.
Migraine
Definitions
The International Headache Society has devel-
oped the following criteria (ICHD-II; 2004) with
the intention of simplifying the diagnosis of
migraine and defining those patients who are
likely to respond to different treatments. The
character of the headache should not be explic-
able by possible CNS damage. Two main types of
migraine are defined:
Repeated headache attacks, lasting for 4-72 h, with the following features: recurrent
moderate to severe, throbbing headache, usually unilateral but sometimes bilateral, accompanied by intolerance of light or noise, nausea, and sometimes vomiting. The pain should comply with at least two of:
(a) Normal physical examination.
(b) No other reasonable cause for the
headache.
(c) At least two of:
- Unilateral pain.
- Throbbing pain.
- Aggravation of pain with head
movement.
- Moderate to severe intensity of pain.
(d) At least one of:
- Nausea or vomiting.
- Photophobia and sonophobia.
• Migraine with aura (classical migraine). Two
or more headache attacks that comply with
three of the following characteristics:
(e) One or more fully reversible aura
symptoms indicating cerebral cortical or brainstem dysfunction.
(f) At least one aura symptom developing
over more than 4 min, or two or more
symptoms occurring in succession.
(g) No aura symptom should last for more
than 1 h.
(h) Headache follows aura with a pain-free
interval of less than 1 h.
‘Borderline migraine’ is often diagnosed when one of the criteria is not met.
The pain scale for migraine (migraine index)
is a product of duration and intensity, where
intensity is graded from 0 (none) to 3 (severe).
The aura consists of warning of an impending
attack, usually with visual symptoms, but audi-
tory, smell and motor limb disturbances may
also occur, lasting 4-60 min (see below). The
headache usually follows within 60 min, or may
accompany these symptoms.
Epidemiology
For epidemiological purposes, migraine patients (migraineurs) are defined as having had at least five attacks without aura, or two with aura.
Migraine is common worldwide, reported
variously as affecting 5-25% of women and
2-20% of men. This spread is due to different
definitions and trial methods. The highest inci-
dence of migraine without aura is at 10-11
years of age in males and 14-17 years in
females. That of migraine with aura is at about
5 years in males and about 12-13 years in
females. There is then a slow increase in preva-
lence in women up to age 40 years, but the
prevalence of all forms declines after the age of
45-50. Onset after age 60 of an exceptionally
severe headache unlike any in a patient’s
previous experience is very unusual: this should
raise the possibility of significant pathology, e.g.
subarachnoid haemorrhage or temporal arteritis
(see Chapter 12). There is a genetic predisposi-
tion in some patients, but no simple Mendelian
inheritance.
tment
Some 10% of the population are ‘active
migraineurs’, 5% have 18 or more migraine days annually; 1% have one each day or week. The average duration of an attack is about 24 h, but may be 2-3 days in 20% of patients.
Aetiology and pathology
The aetiology has yet to be elucidated in humans.
The traditional view that symptoms are simply
the result of alterations in cerebral blood flow is
probably incorrect. The haemodynamic changes
observed during all phases of the attack cannot
alone account for the symptoms. Rather, the
vascular changes reflect cranial disturbance, e.g.
vasodilatation of cranial or meningeal blood
vessels, or oedema.
The anatomy of the cerebral circulation is
complex with a high degree of redundancy, probably because:
• There is an absolute requirement for a contin-
uous supply of glucose and oxygen.
• The brain is highly active metabolically,
consuming about 20% of total blood oxygen at
rest.
• Even a brief interruption of the blood supply
may cause unconsciousness; 1-2 min depriva-
tion impairs brain cell activity and 4-5 min causes permanent damage.
There is a complete circle of interconnecting
arteries at the base of the brain (the circle of
Willis), derived from the four main ascending
arteries, two vertebral, two internal carotid. This
minimizes the risk of ischaemia, e.g. due to a
small clot, because the circle can be supplied by
any of its ascending arteries. The cerebral veins
have no valves, have very thin walls, and no
muscle layer. Thus they cannot be causal in
migraine but can account for the pounding
nature of the headache, reflecting systolic heart-
beats. There are also large venous sinuses that
drain pooled venous blood from the brain
and skull. Numerous anastomoses connect the
arterial circle and this venous system.
There are several pathophysiological theories
of migraine symptomatology, reflecting current
uncertainties. Migraineurs probably have a
genetically determined, or congenital, reduced
CNS excitation threshold, i.e. they fall into an
intermediate group between epilepsy and familial hemiplegic migraine), and paralysis of
normality, but a genetic origin for this has not
been demonstrated. However, mutations have
been identified in genes for the voltage-gated
calcium and sodium channels and for the alpha2
subunit of the Na÷/K÷ pump in some types of
familial migraine.
Thus the concept that vascular changes may
account for the prodrome (see below), and that
the subsequent headache is caused by vasodi-
latation is no longer tenable. However, opening
of arteriovenous anastomoses may be expected
to expose the cerebral veins to arterial pressures
for which they are not designed, and this could
explain the pounding nature of the headache,
because the thin-walled veins would be exposed
to the systolic pressure with every heartbeat.
They would then expand, putting pressure on
the brain, and relax during diastole. Further, it is
known that stimulation of cranial nerves, espe-
cially the trigeminal, causes neurogenic plasma
extravasation in the dura mater and the release
of pro-inflammatory mediators.
Dilatation of the carotid arterial circulation
causes stretching of arterial walls and so
thickening of the meninges, producing pain.
Serotonin (5-HT), released by vascular nerves or platelets, is clearly implicated in the patho-
genesis of migraine. The recent advances in treatment have been derived from the observa-
tion that injection of 5-HT can abort migraine attacks, but causes substantial side-effects. Addi-
tionally, the introduction of an NSAID that is a potent inhibitor of PG and LT B4 synthesis has also focused attention on the pathogenetic
role of these eicosanoids. Nitric oxide is also
implicated in CNS vasodilatation.
Clinical features
Different patterns occur in individual sufferers,
ranging from occasional headaches that are
almost indistinguishable from tension headache
to frequent disabling episodes (Figure 7.4(d-j)).
Symptoms may mimic those of TIAs (which
resemble a stroke, but the patient recovers within
24 h) and are clearly associated with cerebral ischaemia. There may be dizziness or partial or complete blindness (basilar migraine), partial or
complete hemiparesis (one-sided paralysis,
the eye muscles (ophthalmoplegic migraine) or
facial muscles. It is important to distinguish these
symptoms, which arise gradually, from those
of thromboembolic TIAs, which are usually of
sudden onset, because both treatment and prog-
nosis are very different for the two conditions.
These atypical migraines are uncommon but are
important because they are associated with
ischaemia; the 5-HT1 agonists, because they are
potent vasoconstrictors, are contra-indicated for
their treatment. These forms of migraine are now
believed to be due to genetically determined
abnormalities of the cerebral calcium channels
and will not be discussed further here. However,
the possibility is raised that this is the inherited
basis of some forms of migraine.
Migraine is known to be associated with an increased risk of ischaemic stroke, and a large European study has found that this risk is increased threefold in young women with migraine. Up to 40% of strokes in this study
developed from a migraine attack, and factors such as oral contraceptive use, hypertension and smoking further increased the risk.
Common migraine
This is the usual form of migraine, affecting
about 75% of sufferers. The symptoms resemble
those of classical migraine, but tend to comprise
only headache, malaise and nausea. Occasion-
ally, aura occurs without other symptoms. There
may be considerable difficulty in distinguish-
ing between common migraine and tension
headache. Both syndromes are very common
and so may be concurrent, or tension headache
may exacerbate common migraine at some stage.
Migraine with aura
Triggers for attacks are listed in Table 7.12. There may be three distinct phases to an attack:
• Feelings of well-being, yawning, food rejec-
tions or cravings, etc., which may last for up
to 24 h (the prodrome).
• These are followed by an aura; and
• Finally the headache and associated features
occur (Table 7.13).
The aura develops over 5-20 min and lasts for
4-60 min. The aura is usually visual, including
spots (scotomas), blurred vision, flashing lights and jagged lines. ‘Pins and needles’, facial tingling, speech difficulties and unusual smells may also occur, and there may be nausea.
This is followed by a severe throbbing headache
that may start unilaterally and be localized, but
may later become more general. Nausea increases
and vomiting usually follows. Sufferers are irri-
table, photophobic and sonophobic and usually
retreat to a dark, quiet bedroom. The attack lasts
from hours to days and often terminates with
sleep, often followed by a longish, variable pain-
free period. Because attacks are associated with
smooth muscle inhibition there is often urinary
retention followed by a delayed diuresis towards
the end of an attack.
Investigation
A typical history and a normal neurological examination are usually conclusive. The pres-
ence of atypical features should prompt further examination to exclude meningitis, subarach-
noid haemorrhage, TIAs, partial epilepsy, stroke, a brain tumour or other serious pathology. Patients are normal between attacks.
tment
The possible involvement of all of the factors listed in Table 7.12 should be assessed, possibly with a patient diary.
Management
Apart from the mildest form of common
migraine, this requires pharmacotherapy, but
some general measures are important, as follows:
• Effective identification and treatment of any
associated diseases.
• Counselling:
- For identification and avoidance of
possible trigger factors (Table 7.12), espe-
cially changing or stopping a combined
oral contraceptive in young women.
- Strong reassurance that the condition is
benign, because many patients think that
they have a brain tumour or are going
‘mad’.
- The importance of prompt treatment at
the first sign of an impending attack.
- Rest, usually in a quiet, darkened room.
The principles of treatment of are outlined in
Table 7.13.
Acute attacks icantly, not least because patients may regard it
as treatment failure. Recurrence occurs in about
General considerations
35% of responders, for unknown reasons.
Gastric stasis and generally poor gut peristalsis
develops during an attack, so drug absorption
from the gut may be compromized with all drugs
in migraine, especially with conventional tablets
and capsules. Absorption is improved by using
effervescent oral formulations and by using an
anti-emetic and prokinetic drug, e.g. metoclo-
pramide or domperidone, 30 min before an anal-
gesic if possible, or simultaneously. Routes of
administration that avoid first-pass metabolism,
e.g. buccal dosage forms (sublingual tablets and
oral aerosols), nasal sprays and injections, are
pharmacokinetically preferable, because many
of the drugs used undergo extensive first-pass
metabolism. Suppositories are useful if the
patient is nauseated and vomiting. Patient
dislike of suppositories and repeated injections
may be outweighed by the rapidity and extent of
benefit derived from a product.
It may take experimentation over a few attacks
to establish the optimum drug, dosage and route
of administration. Patients do not always require
the same treatment: mild attacks may respond to
simple analgesics, while severe ones in the same
patient may require specific treatment. The rela-
tive responses to different drugs may be different
in different attacks, but the response to specific
treatment seems to be more consistent than that
to simple analgesics.
Opioid analgesics should be avoided, because they are ineffective, exacerbate nausea and vomiting and, if taken regularly for attacks, may cause analgesic overuse headache.
Drugs should not be used during the aura,
which is unresponsive to treatment and may not
always be followed by headache. However, if it is
known that headache always supervenes, simple
analgesics should be taken before the headache
is established.
Specific anti-migraine drugs, the 5-HT1 agonists (triptans), should be reserved for established headache, on grounds of toxicity, in patients who find simple analgesics and anti-emetics ineffec-
tive. Although all triptans are effective, the response varies between patients, as do the incidence and severity of side-effects.
Headache recurrence following response to
treatment limits the benefits of treatment signif-
The ‘therapeutic gain’ (TG) is the response rate to treatment minus the response to placebo or comparator, and is used below. However, trial results are difficult to compare due to different end-points, time of measurement of the response, use of ‘escape medication’ and comparison in only one attack or over several attacks.
There are two approaches to management: the
traditional stepped-care model (ladder), i.e.
simple analgesic to compound analgesic to a
triptan (see below) or ergotamine, or to decide on
the appropriate treatment for a particular patient
depending on their symptoms, the stratified-
care model. The latter is gaining ground with
the introduction of the triptans, which are
effective and less toxic than ergotamine.
Simple and compound analgesics
These have been discussed above and in Chapter
12. Adequate doses should be used, i.e. aspirin
600-900 mg every 4-6 h, provided that it is
tolerated and not contra-indicated, or paracet-
amol (acetaminophen) 1000 mg every 4-6 h (to a
maximum of 4 g daily). Effervescent aspirin is pre-
ferred and is sometimes as effective as sumatriptan.
Non-steroidal anti-inflammatory drugs
Tolfenamic acid is an NSAID that is specifically
licensed for the treatment of acute migraine
attacks. It has the general properties and side-
effects of NSAIDs and may also cause dysuria
(mostly in men), tremor, euphoria and fatigue,
but it seems to be well tolerated. Tolfenamic acid
is reported to be as effective as the 5-HT1 agonists
and is formulated as a tablet that disintegrates
rapidly and has good bioavailability. Other
NSAIDs that are licensed for migraine treatment
are ibuprofen (1.2-1.8 g daily in divided doses),
diclofenac, flurbiprofen and naproxen, at equiva-
lent dosage. Flurbiprofen and diclofenac are also
available as suppositories, which are useful if the
patient is vomiting.
Serotonin agonists (5-HT1 agonists, ‘triptans’)
All of these are effective in relieving migraine
headaches, but they are not suitable for prophy-
laxis. Unlike simple analgesics and NSAIDs, there
is evidence that they are most effective when
used at the first signs of the headache and not during the aura.
The 5-HT1B/1D-receptor agonists licensed in the
UK are sumatriptan and the newer almotriptan,
eletriptan, frovatriptan, naratriptan, rizatriptan and
zolmitriptan. These drugs act highly selectively
on specific subsets of 5-HT1 receptors, 5-HT1D
and, to a lesser extent, 5-HT1B. They have a much
lower affinity for other 5-HT1 receptors and are
inactive at 5-HT2, 5-HT3, adrenergic, dopamin-
ergic, muscarinic and benzodiazepine receptors.
The role of 5-HT1D and 5-HT1B receptors is uncertain. They are believed to be pre-synaptic ‘autoreceptors’ that control the release of
neurotransmitters at the nerve terminal.
Because of their vasoconstrictor action, 5-HT1
agonists are likely to aggravate any condition
caused by arterial blockage, spasm or inflamma-
tion and are contra-indicated in such situations,
e.g. IHD, previous MI, atypical angina, uncon-
trolled or severe hypertension and intermittent
claudication (see Chapter 4), temporal arteritis
and Raynaud’s disease (see Chapter 12).
Non-responders. Zolmitriptan is the only
antimigraine drug that can be repeated for the
same attack by those who fail to respond to a first
dose; this should be not less than 2 h after the
first dose. For all other triptans non-responders
should not repeat the dose. This also minimizes
the risk of analgesic-induced headache. Non-
responders will not gain any benefit from a
second dose of the same or any other triptan,
because this will increase or prolong vasocon-
striction and the risk of ischaemia and throm-
boembolic stroke, without relieving the migraine.
Further, the maximum dose of all triptans that
may be used within 24 h is limited.
However, the same drug, given by a different
route or in a higher dose, may be successful in
another attack. Non-responders are more likely
to benefit from a change to a different drug class.
If a small dose has proved to be inadequate, a
larger dose may help in a subsequent attack.
Partial responders. If an attack responds to a
first dose only one repeat dose of a triptan should
be used for the same attack if there is inadequate
relief or in those in whom the headache remits
tment
and recurs. The precise details vary with the drug and dosage form: the BNF and patient information leaflets should be consulted.
Although more expensive than previous treat-
ments, the cost-benefit ratio for triptans is prob-
ably no worse than for older drugs. There is less time off work and a better quality of life.
Sumatriptan is poorly absorbed from the tablets
(14% bioavailability), taking 2 h to reach a peak
plasma concentration which is 75% of that
obtained by SC injection. The latter is the
preferred mode of use, which avoids first-pass
metabolism. An autoinjector device has been
produced (peak plasma concentration at 12 min,
97% bioavailability; TG in 51% of patients
initially, 71% at 1 h).
The oral route is not suitable if there is nausea
and vomiting, and it clearly gives slower relief
than the injection. However, some patients who
do not wish to inject find it adequately effective
(TG 33%, 58% of patients, 100-mg dose). Alter-
natively, a nasal spray is available for those not
wishing to inject or who are vomiting and gives
rapid absorption, but the peak plasma concen-
tration (at 1-1.5 h) is only about 20% of that
from the injection, partly due to pre-systemic
metabolism. Some patients find the nasal spray
inconvenient and irritant: it is also expensive.
The terminal elimination half-life is about 2 h.
Sumatriptan may also help in the management of cluster headache (see below).
Side-effects include drowsiness, a transient
increase or reduction in blood pressure, brady-
cardia or tachycardia and, occasionally, fits.
However, these side-effects are usually mild and
fairly brief. Caution is required in renal impair-
ment and, following reports of chest pain and
coronary vasoconstriction, they are contra-
indicated in patients with ischaemic syndromes
(see above).
Interactions. Because of the similar modes of
action, sumatriptan must not be used with ergot-
amine: nor within 24 h of stopping ergotamine,
which must not be used within 6 h of taking
sumatriptan. Use with MAOIs, SSRIs and lithium
increases the risk of CNS toxicity, and use of
sumatriptan with these must be avoided.
Second-generation triptans have better bio-
availability, greater potency at active receptor sites and longer half-lives than sumatriptan. of ergotamine, despite its complex actions and
Because they are more lipophilic, CNS penetra-
tion is also improved and they reduce neuroex-
citability, especially in the trigeminal ganglion.
Naratriptan is similarly effective to sumatriptan, and is available as tablets. Used at the lower dose (2.5 mg) it may have fewer side-effects than sumatriptan and has similar interactions, but the TG is also lower. At the higher dose (5 mg) it resembles sumatriptan. Because it has a longer duration of action it may be useful in patients who regularly suffer from relapses.
Zolmitriptan is another recent introduction in
tablet form, designed to be a potent 5-HT1 partial
agonist and more lipophilic than sumatriptan,
giving better CNS penetration. It has a slightly
higher TG than sumatriptan but a higher inci-
dence of adverse reactions. It is also available as
orodispersible tablets and as a nasal spray. It is not
clear whether the orodispersible tablets are supe-
rior to normal ones and are clearly unsuitable if
they cause a dry mouth.
Rizatriptan is available as tablets and ‘melt’
wafers that are dissolved on the tongue and swal-
lowed. It is consistently effective and well toler-
ated, and is cost-effective. Rizatriptan metabolism
is reduced by propranolol. Patients taking propra-
nolol must not take rizatriptan within 2 h of a dose
of propranolol and should take only the minimum
dose of rizatriptan.
Frovatriptan has the longest half-life of the 5-
HT1 agonists and may be particularly useful in menstrual migraine. It is somewhat less effective, and cheaper, than other triptans.
Possible side-effects with all these agents include drowsiness and transient hypertension; dry mouth, unpleasant sensations in any part of the body, and muscle pain and weakness may also occur. Absolute contra-indications for this entire group are given above.
Arrhythmias due to accessory cardiac
conduction pathways are an additional contra-
indication for zolmitriptan. It is also contra-
indicated in those who have had a TIA or a stroke.
There is inadequate information on their use in the elderly.
Second-line drugs
These include ergotamine and isometheptene
mucate. Low cost has ensured the continued use
long list of side-effects. The BNF states that both of these are “less suitable for prescribing”, and their use is declining.
Ergotamine tartrate is the oldest antimigraine
drug, with ergot preparations having been in
use for at least 2000 years. The isolation and
pharmacological characterization of the ergot
alkaloids in the mid-20th century (pure ergota-
mine was isolated by Stoll in 1920) was a major
event in the development of modern pharma-
cology. It is an amino acid alkaloid derived from
the lysergic acid nucleus. Preparations contain
about 40% of the relatively inactive ergotami-
nine, due to spontaneous epimerization. Because
of its long history, well-conducted controlled
trials have not been carried out, so there is no
objective evidence for its level of benefit. Actions
of ergotamine include:
• At 5-HT1 and 5-HT2 receptors:
- Partial agonist in many blood vessels (it is
a powerful vasoconstrictor).
- Mixed agonist/antagonist actions in the
CNS.
- A non-selective 5-HT antagonist in many
smooth muscles.
• At alpha-adrenergic receptors:
- Partial agonist/antagonist in blood vessels
and smooth muscles, promoting uterine
contraction and vasoconstriction.
- Antagonist in the central and peripheral
nervous systems.
• At dopaminergic D2 receptors:
- Powerful stimulation of the CTZ, espe-
cially with injections, causes nausea and
vomiting.
While the vasoconstrictive effects of ergotamine
contribute to its benefit in migraine, these non-
selective, wide-ranging actions mean that it also
has a formidable array of side-effects. These, and
the risk of habituation, which limits its use to not
more than twice per month, have contributed to
its increasingly rare use. It is less effective than
sumatriptan and probably also less so than
naproxen.
Ergotamine is available as compound tablets
with cyclizine, as an anti-emetic, and with
caffeine, allegedly to promote absorption. The
benefit from caffeine is debatable and it may even cause analgesic headache (see above). Oral
bioavailability is poor, about 1-3%, with exten-
sive first-pass metabolism and large inter-
individual variations in absorption. The use of a
buccal aerosol or sublingual tablets may provide
improved absorption, but the aerosol has a very
nauseating taste. Suppositories give about a
20-fold increase in peak plasma concentration,
and this is probably the preferred route. Phar-
macokinetic data are rather imprecise due to
assay difficulties (peak plasma concentrations
are only about 10-500 pg/mL).
Because ergotamine binds irreversibly to recep-
tors it has a longer duration of action than the
triptans. This may be beneficial in patients with
lengthy attacks or those in whom headache
recurs frequently after triptans. However, there are
restrictions on dose frequency because of its strong
peripheral vasoconstricting action, i.e. not more
than four tablets in 24 h, not to be repeated within
4 days, and not more than eight tablets/week.
Patients have lost fingers because of failure by GPs and pharmacists to adhere to these dosage restrictions, or to recognize inappropriate
prescribed dosage.
Although injections of ergotamine have been
used, they usually contain a mixture of ergot alka-
loids. They therefore have numerous side-effects
and, in addition, are highly emetogenic (20% of
tment
patients) and are often poorly tolerated. The side-
effects, contra-indications, etc. of ergotamine are given in Table 7.14.
Dihydroergotamine preparations have been
withdrawn in theUKbut the nasal spray is
widely used inNorth Americaand continental
Europe.
Isometheptene, an indirect sympathomimetic
agent, is marketed in theUKcombined with
paracetamol (acetaminophen). It may cause circu-
latory disturbances, dizziness, rashes and, rarely,
blood dyscrasias. Its numerous side-effects,
cautions, contra-indications and interactions are
considered to outweigh its clinical usefulness.
Prophylactic pharmacotherapy
This may not be necessary if attacks are infre-
quent or of mild to moderate severity and if this
situation is well tolerated by the patient and is
well controlled by acute drug usage. However,
opinion differs as to when prophylaxis should be
considered. In theUK, it is often recommended if
there is more than two moderate to severe attacks
a month, if they suffer increasing headache
frequency or significant disability despite suitable
treatment and if they cannot take appropriate
migraine treatment. In theUSAthe borderline is
drawn at three or more attacks per month. Much
depends on what level of pain and disruption to marked drowsiness initially, so the minimum
their lives, balanced against side-effects, patients are able to tolerate.
The drugs used comprise:
• Beta-blockers: propranolol, metoprolol (possibly
atenolol, nadolol, timolol).
• 5-HT antagonists: pizotifen (pizotyline),
methysergide.
• Tricyclic antidepressants: e.g. amitriptyline
(whether the patient shows depressive
symptoms or not; unlicensed indication).
• Sodium valproate and CCBs (unlicensed
indication).
The modes of action of these drugs in migraine are unclear, especially because the effects of other beta-blockers and anticonvulsants are similar to those of placebo.
The use of methysergide for resistant cases is
restricted to hospital consultants in the UK,
because of its toxicity, especially retroperitoneal
fibrosis.
All of these drugs are, at most, 50% effective in
only about half of the patients, but one can
usually be found useful by trial and error.
Further, it is difficult to be sure of the true
benefit because there is a large placebo effect, up
to about 40%. Each of the drugs needs to be tried
for at least 2-3 months before it is discarded as
ineffective. If the patient responds, treatment
should continue, with 6-monthly medication
reviews: complete remission is common.
It has also been suggested that trials of high-
dose riboflavin or NSAIDs might be worthwhile.
If NSAIDs are used (this is an unlicensed indica-
tion), the drug selected would have to be rela-
tively free of side-effects (e.g. ibuprofen) because
long-term use is involved. As usual, there needs
to be a balance between benefit and harms.
Beta-blockers, usually propranolol, are usually regarded as the drugs of choice for migraine
prophylaxis. Their utility is limited by their side-
effects (see Chapter 4) and, in the past, by their interaction with ergotamine, because both drugs cause peripheral vasoconstriction. This interac-
tion is less of a problem if one of the newer 5-HT1 agonists is being used to treat attacks.
Pizotifen (pizotyline) is probably the second
choice, but good evidence of benefit is lacking.
This also has antihistaminic properties and causes
dose should be taken at night and increased grad-
ually, as tolerance is achieved, to the minimum dose that gives satisfactory control. Increased appetite and weight gain often make it unaccept-
able, especially to women. Its weak antimus-
carinic properties may cause urinary retention and closed angle glaucoma, so it may not be suit-
able in middle-aged to elderly patients. Another 5-HT1 agonist/antihistamine, cyproheptadine, is sometimes used in refractory cases.
Clonidine has been used but is regarded as
unsuitable in theUKbecause it may cause or
aggravate severe depression, with an increased risk of suicide. Migraine sufferers may often be depressed due to the condition
The highly desirable introduction of more
effective prophylactic drugs presumably awaits a better understanding of migraine
pathophysiology.
Cluster headache (migrainous neuralgia)
This name derives from the fact that episodes
tend to occur in clusters of attacks, lasting
several weeks, interspersed by remissions of
months to years. Despite its synonym, it is unre-
lated to migraine, though many of the same
drugs are used.
Clinical features
There are abrupt episodes of excruciating, unilat-
eral pain (‘suicide headache’) which affect the
eye, temple or forehead and increase over about
30 min and may last for several hours. The
affected eye waters copiously and there is often
flushing of the same side of the face (Figure
7.5(k)), though this may vary. Attack frequency is between eight per day and one on alternate
days, usually once or twice a day for a few weeks or months, often at night and at predictable
times. The prodromal signs of classical migraine and the aura do not occur, and the attacks do not usually cause vomiting.
Sufferers are mostly men aged 30-50 (male :
female ratio, 10 : 1), but remission tends to occur
by the age of 60. There is no persistent major
deficit. The cause is unknown, but alcohol may provoke attacks, especially during a cluster. The fact that high-dose oxygen may abort an attack points to the possibility of oxygen starvation in part of the CNS.
Pharmacotherapy
Sumatriptan, by SC injection, is the only drug
licensed for cluster headache. It may be taken in
anticipation of an imminent attack (unlicensed
indication), because the timing is usually consis-
tent once a cluster has started. High-flow (100%)
oxygen often provides relief within minutes.
Pizotifen and verapamil (also methysergide,
hospital-only; see above) are used for prophy-
laxis throughout a cluster. Regular oral lithium
(see Chapter 6) may help chronic sufferers.
Trigeminal neuralgia
Definition
This is a neuropathy of the fifth cranial (trigem-
inal) nerve that causes episodes of agonising,
lancinating (stabbing or ‘electric shock’) pain, usually on one side of the face. Each episode lasts for a few seconds.
Clinical features
The trigeminal (Vth cranial) nerve is mostly
sensory and has three branches:
• Ophthalmic; carrying sensory fibres from the
anterior half of the scalp, forehead, the eye
and surrounding structures, the nasal cavity and side of the nose.
• Maxillary; contains fibres serving the lower
eyelid, nose, palate, upper teeth and lip, and
parts of the pharynx.
• Mandibular; serving the anterior tongue (not
taste), lower teeth and jaw, cheek and side of
the head in front of the ear.
Trigeminal neuralgia is usually of unknown
cause, but it can also occur in multiple sclerosis
and due to a fifth nerve tumour. It may also be
a form of post-herpetic neuralgia, when it
usually affects the ophthalmic branch. Other
tment
neuropathies can affect the trigeminal nerve, but these are usually chronic and distinct from trigeminal neuralgia.
The severe spasms of pain usually affect the
mandibular division and may spread upwards to
involve the other branches. The characteristics
and localized distribution of the pain are
diagnostic: no neurological abnormality can be
detected.
Spasms may occur several times a day, usually
in response to trivial triggers, e.g. cold wind,
touching, shaving or washing the face, chewing
or tooth brushing. Episodes remit spontaneously
for anything from months to years, but always
recur. Middle-aged and elderly patients are
mostly affected.
Management
Most patients respond to pharmacotherapy with carbamazepine, taken at the commencement of an attack. This usually reduces the severity, duration and frequency of attacks and is not
beneficial in other forms of headache.
Carbamazepine is a rather toxic antiepileptic
drug with a long list of side-effects and interac-
tions, being a liver enzyme inducer (see Chapter
3). Consequently, it is usual to start with a low
dose in a first attack and build up slowly (fort-
nightly) until symptoms are controlled. This
is especially necessary if dizziness occurs. Like
phenytoin and some other anticonvulsants, carba-
mazepine has a narrow therapeutic window, so
plasma-level monitoring should be instituted if
high doses are used.
Phenytoin may be effective in those not responding to carbamazepine. If the side-effects of high-dose carbamazepine are not tolerated, a combination with phenytoin is sometimes used, the doses of each being reduced appropriately. However, these interact (both are liver enzyme inducers) and such combinations are rarely justified (see Chapter 6).
The dose and build-up in subsequent attacks depend on the patient’s reaction and tolerance to the drugs.
Tricyclic antidepressants are more useful in
post-herpetic facial neuralgia and in non-specific
facial and jaw pain associated with depression
(see below).
Surgery or nerve ablation with alcohol injec-
tions may be required in those not responding to pharmacotherapy.
Post-herpetic neuralgia
Definition
This is a chronic pain syndrome in the
dermatome (the skin area served by a single
sensory nerve) affected by herpes zoster, an acute
skin infection (shingles) due to reactivation of
varicella-zoster provirus (VZV, see Chapter 8).
Clinical features
VZV causes chickenpox, a common acute skin
infection. Most patients (90%) are children under
the age of 10. Infections in older people are gener-
ally severe and occasionally fatal, and are usually
associated with immunosuppression, e.g. drug
treatment (transplant, autoimmune and cancer
patients), radiotherapy, some neoplastic diseases
(especially lymphomas), and AIDS, or waning
immunity in old age.
During recovery from chickenpox the virus
tracks up sensory nerve axons to the local dorsal
root ganglia and becomes incorporated in the
nuclear DNA there as a provirus. This location is
protected from immunological defence mecha-
nisms, so the provirus persists until it is reacti-
vated by a reduction in host immunity. The virus
then tracks back down the nerve axon to cause a
skin infection in the dermatome innervated by
that sensory nerve. This pattern may be
repeated. There are usually three distinct phases:
• Prodromal, with malaise, unilateral nerve
pain or paraesthesia lasting 3-5 days (range
1-14 days), the skin being very sensitive to touch, and sometimes mild fever.
• Active, vesicles appear over 3-5 days and crust
over during several days to 3 weeks, accompa-
nied by nerve and skin pain which seems excessive relative to the skin involvement.
• Chronic post-herpetic neuralgia (PHN)
lasting months to years.
Post-herpetic neuralgia 505
Occasionally, the nerve may be affected without any skin eruption. The affected area is unilateral, sharply demarcated at the mid-line front and back, and may involve a few adjacent dermatomes. The principal sites are the thorax (50% of cases), head and neck (20%) and lumbosacral area (15%). Involvement of the eye or ear requires specialist advice.
Management and pharmacotherapy
Shingles
Mild cases
Only simple or compound analgesics and soothing and drying lotions (see Chapter 13) are required.
Moderate to severe cases
These require prompt treatment, especially if the patient is immunocompromised:
• Early antiviral treatment, to minimize the risk
of PHN and complications, e.g. eye or ear
involvement. However, diagnosis may be difficult because the prodromal symptoms may mimic migraine, heart disease or acute abdominal problems. The antivirals (see also Chapter 8) used include:
- Aciclovir orally, or by IV infusion in the
immunocompromised, plus topical appli-
cation to the eye or ear if these are affected.
Alternatives are famciclovir and valaciclovir,
oral prodrugs of penciclovir and aciclovir
respectively, with superior bioavailability.
- Foscarnet, occasionally amantadine, for resis-
tant strains of VZV (unlicensed indications in theUK).
• Antibacterials for bacterial opportunistic
infections of the rash (see Chapter 8).
• Pain control with:
- Analgesics, including opioids (see below) if
needed, though most have not been
assessed. Side-effects may become unac-
ceptable before effective analgesia is achieved.
- Local anaesthetics (p. 482), especially
topical lidocaine, or nerve block (p. 487) if pain is severe.
- A sedative tricyclic antidepressant, e.g.
amitriptyline or trimipramine, at night, as
an analgesic adjunct, hypnotic and anti-
depressant.
- Anticonvulsants, i.e. gabapentin or prega-
balin, reduce the pain.
- There is conflicting evidence for the
benefit of capsaicin, and this may cause
skin reactions (see below).
- TENS (see p. 486).
• Soothing soaks to the affected skin.
Chronic post-herpetic neuralgia
Severe pain in the prodromal or early active
phases indicates the likelihood of severe PHN,
so early aggressive antiviral treatment is indi-
cated. Prolonged PHN requires any of the anal-
gesic treatments listed above (depression is a
feature of moderate to severe PHN). There is no
evidence for most opioid analgesics in PHN,
but tramadol and oral oxycodone are effective
and the risk of dependence must be weighed
against the possible benefit for this condition,
which is not life-threatening although very
debilitating.
Capsaicin cream, a counter-irritant, is also used
after the rash has healed. This is very irritant
and must not be applied until the lesions have
healed completely. It should be applied not less
than three, nor more than four times daily. If
applied less frequently the transient burning
sensation may be more severe and prolonged;
the skin needs to become habituated. More
frequent application also causes skin irritation.
Application sites must not be occluded and the
hands must be washed thoroughly immediately
after application. The area around the eyes must
be avoided.
Corticosteroids are contra-indicated in the acute phase because they may promote wide-
spread viral dissemination across the skin. They may help when the rash has healed and a short trial course of prednisolone is worthwhile. Intrathecal methylprednisolone is also helpful, but its safety has not been established.
Non-drug measures (e.g. TENS, p. 486) may help to minimize the dose of analgesic and the risk of opioid dependence.
tment
Prophylaxis
A varicella vaccine is available. Use of the vaccine in non-immune individuals, especially those at risk of varicella infection, may prevent an attack of shingles in later life.
Shingles is not contagious, because the
syndrome develops only after a previous chick-
enpox infection and reactivation of dormant
provirus. However, chickenpox may occur in a
non-immune patient due to contact with a case
of shingles.
Some special pain situations
Certain categories of patient present particular problems in pain control. This can be from the type of pain itself or may relate to the constraints that their disease state may impose on any choice of therapy.
Opioids and other sedatives in surgery
Premedication
The objectives of pre-anaesthetic medication are
to:
• Relieve anxiety without excessive drowsi-
ness, so that the patient remains cooperative
and the various preparative procedures can proceed smoothly.
• Give an amnesic effect and so avoid
unpleasant memories.
• Relieve any preoperative pain. • Minimize:
- the dose of the general anaesthetic, usually
inhalational.
- the undesirable effects of anaesthesia,
e.g. vomiting, headache, coughing and
excessive secretions.
- post-operative stress.
Several agents may be required to achieve these ends.
Surgery is often preceded by organic or
traumatic pain, and itself obviously causes
moderate to severe pain. Even mild preopera-
tive pain interferes with the smooth induction of general anaesthesia and increases the amount of anaesthetic required.
Oral benzodiazepines are widely used. They are
sedative, anxiolytic and amnesic, but have no
analgesic effect. Short-acting ones are preferred,
e.g. lorazepam, midazolam or temazepam, but
diazepam, which is long-acting, is also used in
adults.
Diazepam is used to provide mild sedation, but
it is unsuitable for children because its effects in
them are unreliable and it may cause paradoxical
excitation. Alimemazine, a sedative antihista-
mine, may be used in children, but this may cause
post-operative restlessness. It is unsuitable for use
in elderly patients because of its antimuscarinic
effects, e.g. urinary retention and blurred vision.
Opioids are no longer widely used for
premedication. The choice of opioid is deter-
mined by their duration of action and the inci-
dence of side-effects. Morphine, and sometimes
pethidine in obstetrics, are used occasionally, but
their side-effects are particularly undesirable in
surgical patients, e.g. prolonged recovery time,
nausea and vomiting, constipation and urinary
retention, bradycardia causing hypotension,
respiratory depression (especially in those
patients with an asthmatic tendency or frank
asthma) and spasm of the bile duct and ureters.
However, premedication with an opioid may
minimize the occurrence of agitation during
recovery. They are more usually used at induc-
tion and to reduce the dose of a general anaes-
thetic required by about 15%. They also enhance
analgesia during general anaesthesia. Drugs with
a rapid onset and short duration of action are
now preferred because any side-effects resolve
rapidly. Alfentanil and fentanyl can be adminis-
tered by IV injection or infusion. Remifentanil is
given by IV infusion only. They are also used as
analgesics to prevent the pain due to procedures,
e.g. dressing changes. Their side-effect of respira-
tory depression is beneficial in ventilated
patients, to prevent spontaneous respiration
interfering with the ventilation provided by the
equipment.
Patients with severe adrenal suppression due
to corticosteroid use may have a dramatic fall in
blood pressure due to operative stress, so a high-
dose glucocorticoid with mineralocorticoid
properties, e.g. hydrocortisone and especially
Some special pain situations 507
fludrocortisone, may be used in anticipation of intra-operative shock.
Antimuscarinic drugs, e.g. hyoscine (scopo-
lamine) hydrobromide, are now rarely used for
premedication, whether alone or in combination
with an opioid. They reduce the excessive
bronchial and salivary secretions that occur with
some inhaled anaesthetics, due to airway intuba-
tion. Hyoscine produces some sedation and
amnesia and reduces vomiting, but may cause
undesirable bradycardia and CNS side-effects,
e.g. excitement, hallucinations and drowsiness,
especially in the elderly.
However, modern anaesthetic techniques and
the increasing use of day-case surgery largely
avoid the need for traditional premedication
routines.
Induction agents
Propofol provides rapid recovery without hang-
over and causes little undesirable muscle activity.
It is an oil at room temperature: 1% injections
are given intravenously, but 2% injections are
emulsions and are given by IV infusion. Propofol
is also used as a sedative for short surgical and
diagnostic procedures, but there is a significant
incidence of bradycardia, allergic reactions and
convulsions.
Obstetric pain
The use of analgesics to control the pain of
labour presents several problems and poses risks to both mother and fetus. The ideal agent should meet the following criteria:
• Provide adequate pain relief.
• Interfere minimally with the course and
duration of labour.
• Have little effect on fetal vital signs during
labour and after birth.
Pethidine (meperidine) is the most common
obstetric analgesic as it is short-acting and
generally meets these criteria. However, prob-
lems still occur, notably respiratory depression
in the neonate, the incidence of which can be
reduced by giving the drug early in the course
of labour and using the IM route, the IV route being associated with more neonatal respiratory depression.
Common alternative forms of analgesia
include the epidural administration of local
anaesthetics, and the inhalation of sub-
anaesthetic doses of 50% nitrous oxide in oxygen.
Palliative treatment of cancer pain
Great advances have been made in this field, and
most patients can be maintained virtually free of
pain by application of the principles of the
WHO ‘analgesic ladder’ (Table 7.3). It isessen-
tial to appreciate that pain is not an invariable
accompaniment to cancer, and about one-third
of cancer patients remain pain-free. A clear
understanding of the common pain syndromes
associated with neoplasms and their pathophys-
iological mechanisms, the psychological state of
the patient, and the indications and limitations
of the available therapeutic approaches, is vital
to effective management. Short-acting drugs, e.g.
pethidine (meperidine), are unsuitable because
they require frequent dosing, without additional
benefit.
tment
Several different types of pain may be associ-
ated with cancer (see Table 7.15), and these often
co-exist: some 80% of patients who are experi-
encing pain have two or more types and about
20% four or more types. The pain may be due to
the disease itself or to the debility it causes, and to
coexisting disease or treatment, and so a com-
bination of therapeutic approaches is often
required. Thus a multidisciplinary approach,
including specialist ‘Macmillan’ pharmacists, has
been adopted increasingly to form pain teams,
which take a holistic approach to the patient’s
needs. The clinical aspects of cancer and its
overall management are discussed in detail in
Chapter 10.
Types of pain
Because pain can have many causes, some of
which are not directly related to neoplastic
activity, especially careful assessment and diag-
nosis of the pain is essential in cancer patients.
Regular reassessment is required to take account
of disease progression and the occurrence of
unrelated intercurrent disease. Depending on its
aetiology, the pain in terminal disease may only
be partially responsive to opioid analgesics, so a
combination of analgesics with different phar-
macological actions is often necessary. Adjunc-
tive agents, radiotherapy or surgical approaches
may be appropriate in special circumstances.
Tumour infiltration of tissues
Visceral organs
Tumour invasion of the stomach, biliary tract,
intestine, uterus or bladder causes intense
contraction of the local smooth muscle with
increased pressure and local ischaemia. Visceral
pain is characteristically increasing in intensity,
diffuse, unlocalized and continuous. Opioid
drugs are the most effective in these circum-
stances, but treatment must be started on the
appropriate step of the WHO ‘analgesic ladder’
and must be given in adequate doses. A simple
analgesic, usually paracetamol (acetaminophen),
added to the opioid is often helpful. A regular
laxative, combining both a stimulant and a stool
softener (see Chapter 3), is also needed to
prevent constipation that would otherwise add
to the pain and complicate the picture.
Nerves
Infiltration or compression of local nerves by a
tumour may cause a variety of symptoms
according to the site of involvement. These
include hyperaesthesia (increased sensitivity),
dysaesthesia (painful sensation), neuralgia
(paroxysmal nerve pain), allodynia (pain elici-
tated by light touch). Motor disturbances and
sensory loss may also occur. The tumour may
cause persistent mechanical stimulation of high-
threshold nociceptors, i.e. those not readily stim-
ulated, and partial damage to axons and nerve
membranes, resulting in increased sensitivity to
sympathetic stimulation and pressure. Nerve
pain may respond to standard opioid analgesics,
at least partially, but unacceptable side-effects
often occur before adequate analgesia is
achieved, so a combined approach is often
required. Analgesic adjuvants, nerve blocks or
neurosurgical procedures (see above) may be
more successful, but the last two of these may
not always be achievable (see above), or desirable
in very frail patients, and may add their own
problems.
Some special pain situations 509
Bones
Bone pain is common in cancer and may be due
either to a primary tumour (e.g. multiple
myeloma) or to metastases (e.g. from breast, lung
or prostate cancer). Bone tumours stimulate local
pain receptors directly and induce the production
of PGs that may cause osteolysis (solution of the
mineral component of bone), sensitize free nerve
endings and augment pain perception. Inhibitors
of PG release, e.g. NSAIDs (see Chapter 12), are
thus the logical choice to treat bone pain.
Although NSAIDs exert their main analgesic
action at peripheral sites, there is also a central
component. It is worth maximizing the dosage of
individual agents and changing drugs if side-
effects are excessive or the response is poor,
because individuals vary considerably, both in
their response to different NSAIDs and in the
occurrence of side-effects. Bisphosphonates, e.g.
alendronic acid, disodium pamidronate, ibandronic
acid, sodium clodronate and zoledronic acid, can
also help by inhibiting osteolysis. Strontium
ranelate both inhibits osteolysis and stimu-
lates mineralization and may also be helpful
(unlicensed indication). However, cytotoxic
chemotherapy or single-dose local palliative
radiotherapy, if appropriate (see Chapter 10), may
have a dramatic, if temporary, analgesic effect.
Opioids are generally not very effective in relieving bone pain and can sometimes be reduced in dose or withdrawn totally after the introduction of an NSAID, though many patients use the combined therapy.
Morphine in palliative care
Optimal use follows the guidelines given on pp.
470 and 490. Oral morphine is the first-line
opioid in palliative care. Initially, the oral dose of
plain tablets or liquid is given 4-hourly, with
rescue medication as required. Doubling the
bedtime dose usually enables the patient to sleep
throughout the night. With a 4-h duration of
action, morphine takes about 24 h to reach steady
state, so patients need re-evaluation daily. Once
stabilized, the change is made to modified-release
tablets (not to be crushed), usually formulated
for 12-hourly dosing. If pain increases, the dose
should be increased but the dose interval
retained.
If oral dosing is not possible or not tolerated,
rectal (dose equals oral dose) or SC routes
(potency relative to oral is 2, as a bolus or contin-
uous infusion) can be used. The choice may
depend on the availability of the desired dosage
form (see Table 7.4) and its suitability, e.g. the IM
route gives more pain with morphine and should
not be used. The IV route is uncommon in
palliative care because pharmacological tolerance
develops, leading to escalating doses.
A change to diamorphine (heroin) or hydromor-
phone (in North America or elsewhere that
heroin is not licensed) may be needed if high
doses are required, because morphine is not very
soluble and only small volumes can be injected
by the SC route. Also, if the SC route is unsuit-
able due to generalized oedema, coagulation
problems, poor peripheral circulation or local
adverse drug reactions, the IV route may be used
(potencies relative to oral morphine: diamorphine
(heroin), about 3; hydromorphone, about 15).
About 20% of patients fail to respond to these
measures. These will need spinal opioids, with
local anaesthetics and other adjuncts (see above).
Patients with liver failure
These patients often present a therapeutic
dilemma because most of the commonly used
analgesics are contra-indicated. In particular, the
effect of liver failure on the pharmacokinetics
and pharmacodynamics of the analgesic has to
be considered (see Chapters 2 and 3). Most
opioids are significantly metabolized by the liver
and will accumulate in liver disease if dosing
intervals are not adjusted. Further, the oral
bioavailability of opioids may be increased
owing to reduced first-pass metabolism.
Patients with liver failure are particularly sensi-
tive to the effects of opioids, as to other seda-
tives, because they are metabolized hepatically
and relatively small doses can precipitate
encephalopathy (see Chapter 3). Those with
cirrhosis are liable to develop oesophageal
varices (see Chapter 3), and NSAIDs should be
used cautiously as the gastrointestinal irritation
caused may precipitate catastrophic bleeding.
Chronic administration of large doses of hepato-
toxic drugs, e.g. paracetamol (acetaminophen),
tment
can further exacerbate the liver failure. Fortu-
nately, the severity of liver failure usually encountered in cancer patients does not require large dosage changes.
In practice, an estimate of the dosage interval
required to prevent accumulation occurring is
obtained by giving a cautious dose of opioid, the
subsequent dose being withheld until the pain
reappears. It is particularly important in this
situation to use drugs in which the analgesic
half-life is similar to the plasma half-life, e.g.
morphine (plus its active metabolite, morphine-6-
glucuronide), because the risk of accumulation is
then minimized, as the loss of analgesic effect
should correlate with drug clearance.
Patients with renal failure
Choosing an appropriate analgesic in this group
of patients does not usually present a problem
(but see Chapter 14). Some analgesics however
(e.g. the NSAIDs) can cause nephropathy, so
taking a medication history is important, to
determine whether the renal failure may initially
have been due to analgesic over-dosage or
misuse. NSAIDs may also be contra-indicated
because they may exacerbate fluid retention and
precipitate decompensation in developing heart
failure (see Chapter 4).
Opioid analgesics may present a significant
problem in renal failure. Morphine and its
glucuronide metabolites accumulate, requiring
an increased dosing interval or dose reduction.
Fentanyl is said to be safer in renal impairment
and can be given intravenously. If injections are
not tolerated, then transdermal patches can be
used. However, it takes about 24 h after applica-
tion of the first patch to reach an adequate
fentanyl plasma concentration. Further, the long
half-life of fentanyl (about 17 h) creates problems
if the drug accumulates, and replacement with
an alternative must be started at a low dose
concurrently with the removal of a patch, and
increased gradually thereafter. Alfentanil is being
used increasingly in palliative care for patients in
renal failure.
Pethidine (meperidine) should be avoided
because the toxic metabolite norpethidine accu-
mulates in renal failure and can lead to seizures.
Bạn đang đọc truyện trên: AzTruyen.Top