
Contents
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
Physical pain in patients with cancer Physical pain in patients with cancer
-
Assessment of pain Assessment of pain
-
Pain assessment tools Pain assessment tools
-
Visual analogue scales (VAS) Visual analogue scales (VAS)
-
Categorical verbal rating scales (VRS) Categorical verbal rating scales (VRS)
-
Categorical numerical rating scales (NRS) Categorical numerical rating scales (NRS)
-
-
Assessing pain in people with dementia at the end of life Assessing pain in people with dementia at the end of life
-
-
Pain classification Pain classification
-
Principles of pain management Principles of pain management
-
Classification of analgesics Classification of analgesics
-
Non-opioids Non-opioids
-
Opioids Opioids
-
Adjuvant analgesics Adjuvant analgesics
-
-
-
Breakthrough pain Breakthrough pain
-
Step 1: Non-opioid ± adjuvants Step 1: Non-opioid ± adjuvants
-
Step 2: Opioid for mild–moderate pain + non-opioid ± adjuvants Step 2: Opioid for mild–moderate pain + non-opioid ± adjuvants
-
Step 3: Strong opioid + non-opioid ± adjuvants Step 3: Strong opioid + non-opioid ± adjuvants
-
-
Step 1 analgesics Step 1 analgesics
-
Step 2 analgesics Step 2 analgesics
-
Codeine phosphate Codeine phosphate
-
Tramadol Tramadol
-
Adverse effects Adverse effects
-
-
-
Step 3 analgesics Step 3 analgesics
-
Starting morphine (Fig. ) Starting morphine (Fig. )
-
Opioid titration Opioid titration
-
Opioid adverse effects and toxicity Opioid adverse effects and toxicity
-
Adverse effects Adverse effects
-
Opioid toxicity Opioid toxicity
-
-
Changing route of administration of opioids Changing route of administration of opioids
-
Alternative opioids for moderate to severe pain Alternative opioids for moderate to severe pain
-
Indications for prescribing an initial opioid other than morphine Indications for prescribing an initial opioid other than morphine
-
Indications for changing to alternative opioids (opioid switching, rotation or substitution) Indications for changing to alternative opioids (opioid switching, rotation or substitution)
-
Choice of alternative opioid Choice of alternative opioid
-
Mechanism of opioid switching Mechanism of opioid switching
-
Suggested regime for opioid switching Suggested regime for opioid switching
-
-
Other opioids Other opioids
-
Diamorphine Diamorphine
-
-
Switching oral morphine to oral methadone Switching oral morphine to oral methadone
-
Method Adapted from Palliative Care Formulary Method Adapted from Palliative Care Formulary
-
Stop morphine Stop morphine
-
Subcutaneous methadone Subcutaneous methadone
-
Switching other opioids CSCI to methadone CSCI Switching other opioids CSCI to methadone CSCI
-
-
-
Topical opioids Topical opioids
-
Equivalent opioid doses Equivalent opioid doses
-
-
Opioid Equivalence for Transdermal Patches – Adult Use Opioid Equivalence for Transdermal Patches – Adult Use
-
Approximate Equivalent Doses of Opioid Analgesics for Adults Approximate Equivalent Doses of Opioid Analgesics for Adults
-
Intravenous use for pain emergencies Intravenous use for pain emergencies
-
‘Opioid-resistant’ cancer pain ‘Opioid-resistant’ cancer pain
-
Pseudo-resistant? Pseudo-resistant?
-
Semi-resistant? Semi-resistant?
-
Resistant Resistant
-
-
ALWAYS REVIEW PATIENT REGULARLY AFTER ANY OPIOID SWITCH AS CONVERSION RATIOS ARE APPROXIMATE AND CONSIDERABLE INTER-PATIENT VARIATION MAY OCCUR. ALWAYS REVIEW PATIENT REGULARLY AFTER ANY OPIOID SWITCH AS CONVERSION RATIOS ARE APPROXIMATE AND CONSIDERABLE INTER-PATIENT VARIATION MAY OCCUR.
-
-
Neuropathic pain Neuropathic pain
-
Pharmacological management of neuropathic pain Pharmacological management of neuropathic pain
-
Antidepressant drugs, Antidepressant drugs,
-
Tricyclic antidepressants (TCAs) Tricyclic antidepressants (TCAs)
-
-
-
Non-pharmacological management of neuropathic pain Non-pharmacological management of neuropathic pain
-
Management of specific problems Management of specific problems
-
Malignant bone pain Malignant bone pain
-
Radiotherapy Radiotherapy
-
Bisphosphonates, Bisphosphonates,
-
-
-
-
Anaesthetic procedures in palliative care Anaesthetic procedures in palliative care
-
Introduction Introduction
-
General principles of practice General principles of practice
-
Resuscitation equipment Resuscitation equipment
-
Aseptic technique Aseptic technique
-
Drug preparation Drug preparation
-
-
Intrathecal and epidural (neuraxial) techniques Intrathecal and epidural (neuraxial) techniques
-
Common indications for the use of neuraxial techniques Common indications for the use of neuraxial techniques
-
Possible contraindications to neuraxial techniques (risk–benefit decision): Possible contraindications to neuraxial techniques (risk–benefit decision):
-
Comparison of continuous versus bolus techniques (Table ) Comparison of continuous versus bolus techniques (Table )
-
-
Choice of drug: opioids, local anaesthetics, clonidine, ziconotide Choice of drug: opioids, local anaesthetics, clonidine, ziconotide
-
Opioids Opioids
-
-
Opioids: typical dosing schedule Opioids: typical dosing schedule
-
Side-effects of local anaesthetics Side-effects of local anaesthetics
-
Clonidine Clonidine
-
-
Guidelines for percutaneous intrathecal catheter insertion Guidelines for percutaneous intrathecal catheter insertion
-
Follow-up care Follow-up care
-
-
-
-
Chemical neurolysis for cancer pain Chemical neurolysis for cancer pain
-
Neurolytic agents Neurolytic agents
-
Coeliac plexus block Coeliac plexus block
-
Superior hypogastric block Superior hypogastric block
-
Ganglion impar block Ganglion impar block
-
Interpleural block Interpleural block
-
Saddle block Saddle block
-
-
Complementary therapies and other non-pharmacological pain interventions Complementary therapies and other non-pharmacological pain interventions
-
-
Pain and difficulties in communication Pain and difficulties in communication
-
Examples of unusual behaviours indicating pain Examples of unusual behaviours indicating pain
-
Verbal expression, e.g.: Verbal expression, e.g.:
-
Facial expression, e.g.: Facial expression, e.g.:
-
Behavioural expression, e.g.: Behavioural expression, e.g.:
-
Physical expression, e.g.: Physical expression, e.g.:
-
-
-
Further reading Further reading
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
-
Cite
Abstract
This chapter covers physical pain in patients with cancer, assessment of pain, pain classification, principles of pain management, breakthrough pain, step 1 analgesics, step 2 analgesics, and step 3 analgesics. The chapter also covers opioid equivalence for transdermal patches, approximate equivalent doses for opioid analgesics for adults, neuropathic pain, anaesthetic procedures in palliative care, chemical neurolysis for cancer pain, and pain and difficulties in communication.
I think the simplest and probably the best definition of pain is what the patient says hurts. I think that they may be expressing a very multi-faceted thing. They may have physical, psychological, family, social and spiritual things all wound up in this one whole experience. But I think we should believe people and once you believe somebody you can begin to understand, and perhaps tease out the various elements that are making up the pain.
Cicely Saunders
The Specificity Theory of pain, which was proposed by René Descartes in the sixteenth century, is one of the original pain theories. It states that pain intensity relates directly to the amount of associated tissue injury. Its unidimensional approach to pain implies that 100% of pain is treatable by analgesics.
Dr Henry Beecher recognized the limitations of this theory in the Second World War when he observed that only one out of three soldiers carried into a combat hospital complained of enough pain to require morphine.1 He noted that a mismatch between pain intensity and injury severity occurs in certain situations.
Melzack and Wall proposed the Gate Control Theory of pain in 1965, and suggested that a spinal cord mechanism existed that regulated the transmission of pain sensations between the periphery and the brain.2 The theory shifted attention away from the peripheral source of injury towards the spinal cord and brain. It provided the first physiological mechanism for psychological interventions to minimize pain (e.g. distraction or relaxation).
Following on from their work an increasingly multidimensional approach to pain was adopted, and clinicians now recognize that pain perception is governed by a multitude of factors (the Neuromatrix Theory of pain).3
Pain is an unpleasant sensory and emotional experience associated with actual or potential tissue damage, or described in terms of such damage.
International Association for the Study of Pain
Dame Cicely Saunders also recognized that other important factors can influence the pain experience. In parallel with the work of others, she developed the concept of ‘total pain’ from her understanding that the origins of pain may be:
Physical
Social
Psychological
Spiritual
The concept of total pain has become a central tenet of palliative care practice. It recognizes that cancer pain is often a complex, chronic pain with multiple, coexisting causes. Effective management of cancer pain requires a multidisciplinary approach that addresses the patient’s concerns and fears, as well as treating the physical aspects of pain. As a result, the provision of analgesics should be combined with the provision of emotional, social and spiritual supports.
One patient in particular, a lady whose name should be recorded, a Mrs. Hinson, there were days that I remember saying to her, ‘Tell me about your pain,‘ and without any more prompting from me, she went on to say, ‘Well doctor, it began in my back, but now it seems that all of me is wrong.‘ And she gave a description about her symptoms. And then she went on to say, ‘I could have cried for the pills and the injections, but I knew that I mustn’t, the world seemed to be against me, nobody seemed to understand how I felt. My husband and son were marvellous, but they were having to stay off work and lose their money, but it’s so wonderful to begin to feel safe again.’ So really, she’s talked about physical pain, she’s talked about emotional pain of feeling shut away and all the emotional burden that she couldn’t share. She was talking about social pain, financial in that case, but the impact on her family. And then the spiritual need, I think it is spiritual, of the security, the safety, to look at herself and who she was and just be herself. And then the concept of what I called total pain, and really started to lecture about, really comes from there.
Cicely Saunders
This chapter will focus on the management of physical pain caused by cancer, but the principles of therapy are generally applicable to the management of pain caused by other progressive, non-malignant conditions.
Physical pain in patients with cancer
Pain is a common and feared symptom of cancer (Table 6a.1). Many patients with non-malignant, life-threatening disease also suffer pain. Despite the availability of effective methods of controlling pain, significant numbers continue to receive inadequate pain relief.
The cancer itself | Bony/visceral/soft tissue involvement, nerve compression or infiltration, muscle spasm, ulceration, raised intracranial pressure, etc. |
Complications of the cancer | Pressure sores, constipation, post-herpetic neuralgia, candidiasis, lymphoedema, etc. |
Treatment of the cancer | Neuropathy caused by chemotherapy, mucositis caused by radiotherapy, post-operative pain, etc. |
Co-morbidities | Angina, diabetic neuropathy, arthritis, etc. |
The cancer itself | Bony/visceral/soft tissue involvement, nerve compression or infiltration, muscle spasm, ulceration, raised intracranial pressure, etc. |
Complications of the cancer | Pressure sores, constipation, post-herpetic neuralgia, candidiasis, lymphoedema, etc. |
Treatment of the cancer | Neuropathy caused by chemotherapy, mucositis caused by radiotherapy, post-operative pain, etc. |
Co-morbidities | Angina, diabetic neuropathy, arthritis, etc. |
Cancer pain may be acute or chronic, and chronic pain may be further subdivided:
Background pain is a persistent baseline pain. Background pain is managed by the regular administration of analgesics
Breakthrough pain occurs when pain ‘breaks through’ a regular pain-medicine schedule. The quality of breakthrough pain may feel very much like the background pain, except that it is more severe. Although it generally has the same aetiology as background pain, this is not always the case. Breakthrough pain is relieved with rescue or short-acting medications that are taken only at the time of a breakthrough pain episode (section) p. 232
One-quarter of patients do not experience pain
One-third of those with pain have a single pain
One-third have two pains
One-third have three or more pains
Induce depression
Exacerbate anxiety
Interfere with social performance and impair the quality of relationships
Negatively impact on physical capability
Prevent work and reduce income
Challenge existential beliefs
Constantly impact on the patient’s experience of pain
Assessment of pain
What we were aiming at as we started to work together in St Joseph’s, was a patient who was alert and themselves and free of pain. For the great majority of patients that isn’t that difficult with drugs that are available to anybody in a method that is simple. But it does include the very important careful analysis and assessment at the beginning. One long interview and careful examination can carry an awful lot of shorter ones.
Cicely Saunders
Failure to assess pain is a critical barrier to good pain management.
Pain is a subjective experience, and so patient self-report is the gold standard of pain assessment
Evaluation and treatment of pain are best achieved by a team approach, and the patient should be encouraged to take an active role in his/her own pain management
Many patients will have more than one type of pain, and each pain should be assessed separately
Patients should be re-assessed at regular intervals following initiation of treatment, and also at each report of new or altered pain
Seek to establish a relationship with the patient, and encourage the patient to do most of the talking
Begin with wide-angle open questions before clarifying and focussing on more specific ones
Watch the patient for clues regarding pain
Avoid jumping to conclusions
Description of the onset and duration of the pain
Description of the pain, e.g. location, quality, pattern, character
Rating of pain intensity (including a current pain rating, and ratings when pain is worst and least)
Description of aggravating and relieving factors
Description of associated symptoms and signs
Description of the effects of pain on functioning and quality of life
Description of current pain management regimen and assessment of effectiveness
Summary of the past history of pain management
Identification of the patient’s goals of treatment
Physical examination
Diagnostic testing, where appropriate
Pain assessment tools
I think there’s a pain somewhere in the room, but I could not positively say that I have got it.
Mrs Gradgrind (Hard Times, Charles Dickens)
Failure to assess pain is the most common cause of poor pain control. Pain scales can be useful tools in the systematic assessment of symptoms. Not only can they can encourage patient communication and facilitate the professional’s understanding of the patient’s experience, they can also be used to chart the trend of the patient’s response to therapy. However, tools should never be seen as substitutes for a complete pain assessment. Moreover, they may need to be tailored to the needs of specific populations, for example generic tools are often unsuitable for use with those people who are cognitively impaired.
The ideal qualities of a pain assessment tool are that:
It is easy to administer
It is valid and reliable
It shows sensitivity to treatment effect
It has multilingual validity
Pain assessment tools may be unidimensional or multidimensional:
Unidimensional tools provide information about the intensity of the pain experienced by the patient, and take the form of visual analogue scales, verbal rating scales and numeric rating scales
Multidimensional pain measuring tools provide information about additional aspects of the pain such as history, location, affective component and quality of the pain. Examples include the McGill Pain Questionnaire and the Brief Pain Inventory (Fig. 6a.1).
Brief pain inventory. Reproduced with permission from Doyle et al. (eds) (2004) The Oxford Textbook of Palliative Medicine (3rd edn). Oxford: Oxford University Press.
Visual analogue scales (VAS)
The VAS is a line with extremes marked as ‘no pain’ and ‘worst pain’. Patients are asked to mark the point in the line that best describes their pain.
No pain_________________________Worst pain
Categorical verbal rating scales (VRS)
These involve a sequence of words describing different levels of pain intensity e.g.:
None Mild Moderate Severe
Categorical numerical rating scales (NRS)
These use numbers or gradations that indicate the severity of the pain experience:
0___1___2___3___4___5___6___7___8___9___10
No pain Worst pain
Assessing pain in people with dementia at the end of life
Pain is under-reported and under-treated in cognitively impaired older people. A comprehensive review of the literature reveals that people dying with dementia represent the ‘disadvantaged dying’.4
As dementia progresses, and cognitive function and the ability to communicate verbally declines, it becomes increasingly more difficult to ascertain accurately wishes and needs and, consequently, people with dementia often have painful conditions that go unnoticed. This loss of language communication presents a major challenge in assessing pain in this group as self-reporting is required for most pain assessment tools.
In the absence of verbal self-report, nurses and carers are forced to rely increasingly on non-verbal and behavioural cues of physical and emotional pain. They need to use a combination of indicators to determine levels of pain, e.g. crying, facial grimacing, etc. Using observation skills rather than relying on verbal responses is essential, as is consideration that any sign of agitated behaviour may be emotional and not a result of pain.
Behaviour commonly associated with pain may be absent or difficult to interpret; e.g. extrapyramidal signs in people with dementia syndromes may mislead the healthcare professional, who may interpret the movements as an indication of pain.5 There is no agreed physiological or biological chemical marker of pain and in people who are unable to communicate there is no benchmark that can be used to allow a definitive diagnosis of pain.5
A number of pain assessment tools have been developed for use in people with dementia.6,7 However, it remains a complex area of clinical assessment practice. It is important to select a scale that matches the person’s abilities; even their pre-dementia educational level and pre-existing abilities influence their ability to use pain scales.5,6
Enlisting the assistance of a carer or family member who is familiar with the usual behaviours and responses of the person with dementia is also essential in identifying behaviour changes that may indicate pain or discomfort.
The Abbey Pain Scale is an assessment tool that is presented in a form suitable for clinical use and is both easy and quick to administer. It was developed with care-home residents with end- or late-stage dementia using a combination of quantitative and qualitative data collection and analysis processes.8
The Abbey Pain Scale has been shown to be most useful as an aid to intervention where the scale is used to assess changes prior to, and following the administration of analgesia (Fig. 6a.2).
Many of the principles of assessing and managing pain in people with dementia also apply to people with learning disabilities when providing end-of-life care. The Disability Distress Assessment Tool (DisDAT),9,10 developed to aid in the assessment of distress in people with learning disabilities, may also be of use in guiding practitioners in assessing distress in the dementia group.
Pain classification
Two main types of pain may be identified on the basis of the mechanism by which pain is produced: nociceptive and neuropathic. Different types of pain respond with varying degrees of effectiveness to different types of analgesics, and so it is important that clinicians determine what type of pain a patient is experiencing in order to prescribe the most appropriate drug. Many patients with cancer have mixed pain syndromes, i.e. a combination of nociceptive and neuropathic pain, and may need combination therapy.
Nociceptive pain may result from injury to somatic structures (somatic pain) or injury to visceral structures (visceral pain) (Fig. 6a.3.):
Somatic pain results from the stimulation of skin, muscle or bone receptors. The neural pathways involved are normal and intact and the pain is typically well localized, and may be felt in the superficial cutaneous areas (e.g. cellulitis) or deeper musculoskeletal areas (e.g. bone pain). It may be described as aching, stabbing, throbbing or pressure.
Visceral pain results from infiltration, compression or the distension of thoracic or abdominal viscera. It is often poorly localized and may be described as gnawing, cramping, aching or sharp. It may be referred to cutaneous sites (e.g. shoulder-tip pain from diaphragmatic irritation due to liver capsule distension), and the cutaneous site may be tender. Mechanism of action of referred pain is poorly understood.

Neuropathic pain is caused by injury to the peripheral and/or CNS (Fig. 6a.4). The underlying mechanisms of neuropathic pain are poorly understood, but broadly speaking, pain occurs because the injured nerves either react abnormally to stimuli or discharge spontaneously.
Typically, pain that arises from damage to the peripheral nervous system is termed ‘deafferentation’ pain, while pain that arises from injury to the spinal cord or brain is termed ‘central’ pain
Neuropathic pain is typically described as being different in character to nociceptive pain. Patients may describe having a dull ache with a ‘vice like’ quality, or burning, tingling, shooting or electric shock-like sensations. It may be associated with hyperalgesia and allodynia.

Allodynia | Pain caused by a stimulus which does not normally provoke pain |
Analgesia | Absence of pain in response to stimulation which would normally be painful |
Causalgia | A syndrome of sustained burning pain, allodynia and hyperpathia after a traumatic nerve lesion, often combined with vasomotor dysfunction and later trophic changes |
Central pain | Pain associated with a lesion in the central nervous system (brain and spinal cord) |
Dysaesthesia | An unpleasant abnormal sensation which can be either spontaneous or provoked |
Hyperaesthesia | An increased sensitivity to stimulation |
Hyperalgesia | An increased response to a stimulus that is normally painful |
Hyperpathia | A painful syndrome characterized by an increased reaction to a stimulus, especially a repetitive stimulus, and an increased threshold |
Neuralgia | Pain in the distribution of a nerve |
Neuropathy | A disturbance of function or pathological change in a nerve |
Neuropathic pain | Pain which is transmitted by a damaged nervous system, and which is usually only partially opioid-sensitive |
Nociceptor | A receptor preferentially sensitive to a noxious stimulus or to a stimulus which would become noxious if prolonged |
Nociceptive pain | Pain which is transmitted by an undamaged nervous system and is usually opioid-responsive |
Pain | An unpleasant sensory and emotional experience associated with actual or potential tissue damage or described in terms of such damage |
Pain threshold | The least experience of pain which a subject can recognize |
Pain tolerance level | The greatest level of pain which a subject is prepared to tolerate |
Allodynia | Pain caused by a stimulus which does not normally provoke pain |
Analgesia | Absence of pain in response to stimulation which would normally be painful |
Causalgia | A syndrome of sustained burning pain, allodynia and hyperpathia after a traumatic nerve lesion, often combined with vasomotor dysfunction and later trophic changes |
Central pain | Pain associated with a lesion in the central nervous system (brain and spinal cord) |
Dysaesthesia | An unpleasant abnormal sensation which can be either spontaneous or provoked |
Hyperaesthesia | An increased sensitivity to stimulation |
Hyperalgesia | An increased response to a stimulus that is normally painful |
Hyperpathia | A painful syndrome characterized by an increased reaction to a stimulus, especially a repetitive stimulus, and an increased threshold |
Neuralgia | Pain in the distribution of a nerve |
Neuropathy | A disturbance of function or pathological change in a nerve |
Neuropathic pain | Pain which is transmitted by a damaged nervous system, and which is usually only partially opioid-sensitive |
Nociceptor | A receptor preferentially sensitive to a noxious stimulus or to a stimulus which would become noxious if prolonged |
Nociceptive pain | Pain which is transmitted by an undamaged nervous system and is usually opioid-responsive |
Pain | An unpleasant sensory and emotional experience associated with actual or potential tissue damage or described in terms of such damage |
Pain threshold | The least experience of pain which a subject can recognize |
Pain tolerance level | The greatest level of pain which a subject is prepared to tolerate |
Principles of pain management
The right dose of an analgesic is the dose that relieves pain without causing unmanageable side-effects. Some individuals may simply require regular paracetamol, while others may need doses of oral morphine in excess of 1000mg/day.
More than twenty years ago, the World Health Organization (WHO) published Cancer Pain Relief,12 a book which set out the principles of cancer pain management including the use of the three-step analgesic ladder. The WHO method can be summarized in five phrases:
By the mouth
By the clock
By the ladder
For the individual
Attention to detail
The WHO ladder indicates that a structured, yet flexible, approach to the management of cancer pain is important. The steps of the ladder illustrate that the process of selecting analgesics should be dependent on an assessment of the intensity of pain experienced by the patient, rather than aetiology of the pain.
The oral route is the preferred route for analgesics, including morphine. For persistent pain, analgesics should be taken at regular time intervals and not ‘as needed’. Adjuvant drugs should be prescribed when required.
Each patient should be regularly re-assessed in order to determine response to treatment, and to ensure that he/she experiences maximum benefit with as few adverse effects as possible. Pain relief can be achieved for about 80% of patients by adopting the basic principles of ‘by the mouth, by the ladder and by the clock’.13
Classification of analgesics
The three classes of analgesics referred to in the WHO analgesic ladder are: non-opioids, opioids and adjuvants (co-analgesics) (Fig. 6a.5).

Non-opioids
Non-opioids are analgesic drugs that mediate their effect through receptors other than opioid receptors. Examples of non-opioids are aspirin, paracetamol and NSAIDs.
Opioids
Opioids are drugs that are agonists at opioid receptor sites. There are at least three types of opioid receptor (mu, kappa and delta), which are found in several areas of the brain and throughout the spinal cord. Stimulation of opioid receptors results in a variety of responses, including analgesia, respiratory depression, myosis, reduced gastrointestinal motility and euphoria. Differences between opioids relate, in part, to differences in receptor affinity.
Opioids for mild to moderate pain are so-called because they are conventionally used to treat pain of this intensity. They are sometimes called referred to as ‘weak’ opioids.
Opioids for moderate to severe pain are used to treat moderate to severe pain and are sometimes called ‘strong opioids’. They are available as immediate release and modified release preparations. Immediate–release (i/r) opioids typically reach peak plasma concentrations within one hour of ingestion and have a duration of action of 1–4h, depending on formulation. Modified-release (m/r) opioids typically reach their peak plasma concentrations 2–6h after ingestion, and the plasma levels are sustained over a period of 12–24h, depending on formulation.
Adjuvant analgesics
These are drugs which do not function primarily as analgesics but can act to relieve pain in specific circumstances. The choice of adjuvant drug is generally guided by the nature of the underlying pain (i.e. nociceptive or neuropathic). Examples of adjuvant analgesics are listed in Table 6a.3.
Adjuvant analgesic . | Use . |
---|---|
Antidepressant | Neuropathic pain |
Anticonvulsant | Neuropathic pain |
Antispasmodic | Pain due to colic |
Bisphosphonate | Pain due to bone metastases |
Corticosteroid | Pain due to oedema or nerve compression |
Muscle relaxant | Pain due to muscle spasm or cramp |
NMDA-receptor blocker | Neuropathic pain |
Adjuvant analgesic . | Use . |
---|---|
Antidepressant | Neuropathic pain |
Anticonvulsant | Neuropathic pain |
Antispasmodic | Pain due to colic |
Bisphosphonate | Pain due to bone metastases |
Corticosteroid | Pain due to oedema or nerve compression |
Muscle relaxant | Pain due to muscle spasm or cramp |
NMDA-receptor blocker | Neuropathic pain |
Breakthrough pain
Breakthrough pain is a transient increase in pain intensity over background pain. Usually, breakthrough pain is related to background pain; typically, it is of rapid onset, severe in intensity and self-limiting, with an average duration of 30 minutes.15
Three types of breakthrough pain are described:
Incident pain: pain is related to movement (either voluntary or involuntary)
Idiopathic/spontaneous pain: no identifiable cause; lasts longer than incident pain
End of dose failure: prior to scheduled dose of analgesia; gradual onset. Often not regarded as true breakthrough pain16
Breakthrough pain affects over 50% of patients with cancer, and also occurs in patients with non-malignant disease. It is a poor prognostic indicator17 and may lead to decreased functioning, anxiety and depression and longer stays in hospital.18,19 Every patient on an opioid should have access to breakthrough analgesia in order to ensure optimal pain control.
Management20
Non-pharmacological management: if possible, treat the underlying cause (e.g. surgery, chemotherapy, radiotherapy); avoid precipitating factors (if possible); physical therapy; education about limitations and exacerbating factors; and patient counselling to reduce anxiety
Optimize around-the-clock medication
Specific pharmacological interventions:
It is generally appropriate to use a short-acting opioid at a dose of one-sixth of the total 24-hour dose of opioid. However, it is recognized that the relationship between the 24-hour dose of opioid and the breakthrough dose is not fixed, and some patients may require individual titration of the breakthrough dose to control their incident pain.
Newer analgesics are available that offer more rapid onset and offset of analgesia than traditional short-acting opioids. These agents may offer certain advantages over traditional short-acting opioids because they have a more suitable pharmacokinetic profile. Examples include oral transmucosal fentanyl citrate (OTFC/Actiq) or buccal preparations and nasal sprays which are becoming available imminently.
Pharmacological management should be tailored to the type of breakthrough pain:
Incident pain: pre-emptive use of a short-acting opioid at an appropriate interval before activity (if predictable)
Idiopathic/spontaneous: use of a short-acting opioid when pain occurs
End of dose failure: alter the around-the-clock medication to increase the dose or shorten the dosing interval
Step 1: Non-opioid ± adjuvants
Patients with mild pain should be treated with a non-opioid analgesic, and if a specific indication exists, the use of the non-opioid analgesic may be combined with that of an adjuvant analgesic.
For example, a patient with mild neuropathic pain may be prescribed paracetamol 500mg–1g q6–8h regularly (max. daily dose 4g) in combination with amitriptyline 25–75mg o.d.
Step 2: Opioid for mild–moderate pain + non-opioid ± adjuvants
Patients who fail to achieve adequate relief after a trial of a non-opioid analgesic, or who present with moderate pain, should be treated with an opioid conventionally used for the treatment of mild to moderate pain. The opioid may be combined with a non-opioid in order to achieve an additive analgesic effect. Adjuvant drugs may also be used as required.
For example, a patient with moderate neuropathic pain may be prescribed codeine 30–60mg q6h in combination with paracetamol 1g q6h and amitriptyline 25–75mg o.d.
Some authors have questioned the usefulness of step 2 of the ladder because, in a systematic review, opioids for mild to moderate pain (either alone or in combination with non-opioids) were shown to be no better than full doses of NSAIDs alone.21 In practice, however, clinicians have found the range of choices afforded by the ladder to be helpful.
Combination preparations of opioids and non-opioids are available for use at step 2, and are some of the most widely used medicines in the community. e.g. co-codamol
Step 3: Strong opioid + non-opioid ± adjuvants
Patients who fail to achieve adequate relief despite appropriate prescription of step 2 drugs, or who present with severe pain should be treated with an opioid conventionally used for severe pain. Adjuvant drugs may be used as required.
Oral morphine, in either immediate-release (i/r) or modified-release (m/r) form, is the opioid of choice for treating moderate or severe cancer pain for reasons of its familiarity, availability and cost.
Step 1 analgesics
Aspirin, paracetamol and NSAIDs are all possible step 1 analgesics, although in practice aspirin is difficult to tolerate at analgesic doses due to its side-effects. The choice between paracetamol and the NSAIDs should be based on a risk/benefit analysis for each patient.
Paracetamol’s analgesic and antipyretic activities are similar to that of aspirin, but it lacks an anti-inflammatory effect. Its mode of action for its analgesic effect remains unclear, although serotonin modulation has been suggested. It has minimal toxicity in adults at recommended doses, but at higher doses can cause fatal hepatotoxicity and renal damage.
NSAIDs are a heterogenous group of drugs that are of use in the treatment of pain mediated by prostaglandins, which serve to sensitize nociceptors. They have conventionally been thought to have particular use in the management of bone pain, but their analgesic effect in this area has probably been somewhat over-estimated.
NSAIDs produce their analgesic effect through inhibition of one or both of the isoenzymes of cyclo-oxygenase: COX-1 or COX-2.
Non-specific COX inhibitors, such as aspirin, ibuprofen and diclofenac, inhibit both isoenzymes. But their use is associated with a number of adverse effects (notably GI and renal toxicity).
Newer NSAIDs inhibit COX-2 only, and are associated with reduced GI effects. However, they may still adversely affect renal function and they have recently been associated with an increased risk of adverse vascular events. Several epidemiological studies have implied that the increased risk of vascular events is a class effect, and is associated with the use of conventional NSAIDs also; this area is being closely studied
A dose–response relationship exists in terms of desired effects and both renal and gastrointestinal effects. Therefore, it is good practice initially to use the lowest effective dose of any NSAID, and to increase it only according to need
Patients appear to vary in their analgesic response to NSAIDs and it may be appropriate to try a drug from a different class if the first-choice drug is ineffective or not tolerated
. | No or low NSAID GI risk factors* . | NSAID GI risk factors* . |
---|---|---|
No CVD | Non-selective NSAID ± PPI | COX-2 inhibitor ± PPI, or non-selective NSAID + PPI |
CVD | Consider alternative analgesia first (e.g. paracetamol ±tramadol) then a non-selectiveNSAID ± PPI (cautiously) | Consider alternative analgesia first (e.g. paracetamol ±tramadol) then a non-selectiveNSAID ± PPI (cautiously) |
. | No or low NSAID GI risk factors* . | NSAID GI risk factors* . |
---|---|---|
No CVD | Non-selective NSAID ± PPI | COX-2 inhibitor ± PPI, or non-selective NSAID + PPI |
CVD | Consider alternative analgesia first (e.g. paracetamol ±tramadol) then a non-selectiveNSAID ± PPI (cautiously) | Consider alternative analgesia first (e.g. paracetamol ±tramadol) then a non-selectiveNSAID ± PPI (cautiously) |
Gastrointestinal (GI) risk factors include age >65yrs, a previous history of peptic ulcer disease, concurrent medication, e.g. aspirin, warfarin, selective serotonin-reuptake inhibitors (SSRIs).
Abbreviations: NSAID, non-steroidal anti-inflammatory drug; CVD, cardiovascular disease; PPI, proton pump inhibitor, COX-2, cyclo-oxygenase inhibitor-2.
Ibuprofen | 200–400mg t.d.s. p.o. |
Diclofenac | 50mg t.d.s. (can be used p.o., SC, or supps.) |
Celecoxib | up to 200mg b.d. p.o. |
Naproxen | 250–500mg b.d. (p.o. or per rectum) |
Ketorolac | 10–30mg SC every 4–6h p.r.n.; max. 90mg daily (max. 60mg for elderly); max. duration 2 days |
Parecoxib | 40mg, then 20–40mg every 6–12 h p.r.n.; max. |
80mg daily (max. 40mg for elderly) |
Ibuprofen | 200–400mg t.d.s. p.o. |
Diclofenac | 50mg t.d.s. (can be used p.o., SC, or supps.) |
Celecoxib | up to 200mg b.d. p.o. |
Naproxen | 250–500mg b.d. (p.o. or per rectum) |
Ketorolac | 10–30mg SC every 4–6h p.r.n.; max. 90mg daily (max. 60mg for elderly); max. duration 2 days |
Parecoxib | 40mg, then 20–40mg every 6–12 h p.r.n.; max. |
80mg daily (max. 40mg for elderly) |
Step 2 analgesics
Codeine phosphate
Codeine is metabolized by the hepatic cytochrome enzyme CYP2D6 to morphine. Codeine is a much weaker agonist at mu receptors than morphine, and the analgesic effect is highly dependent on the production of its active metabolite, morphine, by the CYP2D6 system.
Some 7% of Caucasians and 1–3% of Asian people are CYP2D6 poor metabolizers, and do not experience effective analgesia with codeine. CYP2D6 activity may also be inhibited by antipsychotic agents (haloperidol, levomepromazine and thioridazine) and antidepressants (amitriptyline, fluoxetine and paroxetine).
Codeine is available in tablet and syrup formulations. Doses of 30–60mg p.o. q4h are generally prescribed, to a maximum dose of 240mg in 24 hours. It is also available in compound preparations, where it is combined with a non-opioid. The compound preparations may contain either low-dose (8mg) or high-dose (30mg) codeine, and so it is important to stipulate the dose of codeine required in the preparation, e.g. ‘paracetamol 500mg/codeine 30mg’. Studies have shown that there is no additional analgesic benefit from preparations that contain only 8mg of codeine combined with paracetamol compared with paracetamol alone.
Tramadol
Tramadol displays weak opioid activity and it is also a noradrenaline and selective serotonin-reuptake inhibitor (SSRI). It is metabolized by both CYP2D6 to the active metabolite, and additionally by CYP3A4. Like codeine, CYP2D6 inhibitors can reduce its analgesic effect. CYP3A4 inducers (e.g. carbamazepine) can also reduce analgesia.
There is some disagreement as to which step of the ladder tramadol should be prescribed. But, traditionally, it has proved suitable as a step 2 analgesic because, at therapeutic doses, its analgesic effect is similar to that of an opioid for mild to moderate pain in combination with a non-opioid.
It is not classed as a ‘controlled drug’ and this has some practical prescribing advantages. One of the limitations of tramadol in cancer pain is the extent to which the dose can be titrated. It is recommended that, except in special clinical circumstances, doses of 400mg a day should not be exceeded. At doses above this, the risk of convulsions may be increased.
Adverse effects
Adverse effects include nausea, vomiting, dizziness and drowsiness. Hallucinations and confusion, as well as rare cases of drug dependence and withdrawal, have also been reported. Seizures may occur with use, and appear to be most likely in people taking drugs that lower the seizure threshold, or who have a history of seizures
Adverse effects may be ameliorated by slow titration
Tramadol has the potential for serious drug interactions, and may cause serotonin syndrome (particularly when combined with other serotonergic drugs, such as most antidepressants)
Caps: 50mg; sol. tabs 50mg; sachets effervescent powder 50mg
Tabs: m/r (12h) 75mg, 100mg, 150mg, 200mg
Caps: m/r (12h) 50mg, 100mg, 150mg, 200mg
Tabs: m/r (24h) 150mg, 200mg, 300mg, 400mg
Inj.: 100mg/2mL
Starting dose: 50mg q.d.s. p.o. or 100mg b.d. p.o. (12h m/r)
Step 3 analgesics
Opioids are safe, effective and appropriate drugs for the management of cancer pain, provided that:
Appropriate starting doses and titration are observed
The properties and relative potencies of different opioids are understood
Opioid-related adverse effects are monitored and managed
Prescribers are aware that some types of pain are poorly responsive to opioids and require other types of analgesics (adjuvant analgesics)
Morphine is the strong analgesic of choice unless there is a clinical reason to use an alternative opioid ( p.245).
Morphine appears to have no clinically relevant ceiling effect to analgesia. Doses of oral morphine may vary significantly between individuals to achieve the same end-point of pain relief
The liver is the principal site of morphine metabolism
Morphine undergoes a significant first-pass metabolism, which results in the production of two main metabolites: morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G)
M6G is believed to be a more potent analgesic than morphine itself, while the pharmacology of M3G remains unclear
Morphine is well tolerated in patients with mild-to-moderate hepatic failure. In those with more severe hepatic failure it may be necessary to begin with a reduced dose of morphine and titrate to effect
The products of morphine metabolism are eliminated by the kidney; as a result, patients with renal impairment are at an increased risk of accumulating metabolites and developing adverse effects ( p.707)
Morphine can be administered via oral, rectal, SC, IV, topical, epidural and intrathecal routes. Oral is the preferred route
If patients are unable to take morphine orally, the preferred alternative route is SC. There is generally no indication for IM morphine for chronic cancer pain because SC administration is simpler and less painful
The relative potency of morphine varies according to route of administration (p. 244)
Oral Morphine is available in two formulations: immediate-release (i/r; also known as ‘normal release’) and modified-release (m/r). Peak plasma concentrations of i/r preparations usually occur within the first hour after oral administration, and the analgesic effect lasts for about 4h. In contrast, m/r preparations produce a delayed peak plasma concentration after 2–6h, and analgesia lasts for 12 or 24h.
Morphine metabolism can be induced or inhibited by a variety of medications:
Carbamazepine, phenobarbital, phenytoin and rifampicin all accelerate the clearance of morphine
Phenothiazines, tricyclic antidepressants and cimetidine all interfere with morphine metabolism and increase the effect of morphine
Lorazepam and other benzodiazepines that undergo glucuronidation competitively inhibit morphine metabolism
Morphine i/r (4h) | |
Oramorph | 10mg/5mL; 20mg/1mL |
Sevredol | 10, 20, 50mg scored tabs |
Morphine suppositories | 10mg, 15mg, 20mg, 30mg (Equianalgesic dose by oral and rectal routes) |
Morphine m/r (12h) | |
MST and Zomorph can be used interchangeably. | |
MST Continus | 5, 10, 15, 30, 60, 100, 200mg tabs |
Morphgesic SR | 20, 30, 60, 100, 200mg susp. |
10, 30, 60, 100mg tabs | |
Zomorph | 10, 30, 60, 100, 200mg caps (can be opened and administered via an NG/PEG tube, or sprinkled on food) |
Morphine m/r (24h) | |
MXL | 30, 60, 90, 120, 150, 200mg caps |
Morphine i/r (4h) | |
Oramorph | 10mg/5mL; 20mg/1mL |
Sevredol | 10, 20, 50mg scored tabs |
Morphine suppositories | 10mg, 15mg, 20mg, 30mg (Equianalgesic dose by oral and rectal routes) |
Morphine m/r (12h) | |
MST and Zomorph can be used interchangeably. | |
MST Continus | 5, 10, 15, 30, 60, 100, 200mg tabs |
Morphgesic SR | 20, 30, 60, 100, 200mg susp. |
10, 30, 60, 100mg tabs | |
Zomorph | 10, 30, 60, 100, 200mg caps (can be opened and administered via an NG/PEG tube, or sprinkled on food) |
Morphine m/r (24h) | |
MXL | 30, 60, 90, 120, 150, 200mg caps |
Starting morphine (Fig. 6a.6)
The starting dose of morphine depends on whether or not the patient has used opioids before and his/her previous dose.
An i/r morphine regime gives greatest flexibility for initial dose titration, but it may be appropriate to start some patients directly on m/r morphine, e.g.:
Patients who are already taking step 2 opioids
Patients with less severe pain
Patients who have difficulties with compliance
Outpatients
Adult, not pain-controlled on full-dose step 2 analgesics: i/r preparation 5–10mg 4h
Elderly, cachectic or not taking step 2 analgesics: i/r preparation5mg 4h
Very elderly and frail: Oramorph 2.5mg 4h
Adult, not pain-controlled on full-dose step 2 opioids: 15–30mg morphine m/r 12h
Elderly, cachectic or not taking step 2 analgesics: m/r 10-15mg morphine 12h
Very elderly and frail: 5-10mg 12h morphine m/r

Flowchart adapted from Cancer Care Alliance of Teeside, Durham and North Yorkshire Network Supportive and Palliative Care Guidelines 2006.
Opioid titration
Titration is a systematic process of incremental dose adjustment based on the patient’s needs and responses
The goal of titration is to use the smallest dose that provides satisfactory pain relief with the fewest side-effects
The need for dose adjustment is based on reported severity of pain and frequency of need for breakthrough doses: ’titrating to effect’.
Titration may be upward (a.k.a. ‘dose escalation’) or downward (tapering), depending on the patient’s needs and responses.
Dose adjustments are calculated based on a percentage of the total dose of breakthrough analgesia required in previous 24-hour period (most commonly an increase or decrease of 30 – 50%)
The increment percentage tends to decrease a little as the dose increases, e.g. 5–10–15–20–30–40–60–80–100–130–160–200mg
Opioid adverse effects and toxicity
Adverse effects
The management of opioid-related adverse effects is a fundamental aspect of opioid therapy, and clinicians should adopt a proactive approach, wherever possible. The use of opioid analgesia should not be affected by unfounded fears about pharmacological tolerance, dependence or respiratory depression. In the palliative care setting, pharmacological tolerance and psychological dependence rarely occur. There is often significant inter-individual variation in sensitivity to opioid-related adverse effects, and this may be explained, in part, by genetic variability. Other factors influencing the development of adverse effects are:
The presence of renal and hepatic impairment
The use of polypharmacy
The patient’s age
The patient’s cognitive function
The extent of the patient’s disease
The dose and type of opioid
The route of administration of opioid
Many of the side-effects caused by opioids are non-specific, so it is important as a first step to determine whether the symptoms are related to opioid use or to underlying disease.
Common management strategies for various opioid-related side-effects are listed in Table 6a.6, and are mainly based on a dose reduction of systemic opioid ± use of a specific therapy to treat the adverse effect.
Opioid-related nausea and vomiting | Although these symptoms are commonly present during initiation of opioid therapy, they are frequently self-limiting and usually require only a short course of anti-emetic therapy. |
If symptoms persist, metoclopramide 10-20mg p.o. t.d.s. is generally used as first-line anti-emetic. Haloperidol 1.5–3mg o.n. initially is also useful. | |
Opioid-related constipation | Prescription of regular, prophylactic laxatives is recommended. Studies comparing laxatives, especially in palliative care patients, are lacking. Various laxatives have been investigated, but findings remain inconclusive and choice of laxative is often dependent on local practice. A combination of a stimulant laxative (senna) and a stool softener (docusate) may be used. |
Methylnaltrexone has been developed specifically for opioid-induced constipation, see p. 319 | |
If problem persists, laxatives should be titrated upwards, and enemas may be used. The addition of metoclopramide for its pro-kinetic effects may be beneficial. | |
If problem is severe, consider prescription of alternative opioid. Fentanyl and buprenorphine may be less constipating. | |
Consider non-pharmacological approaches, such as fluid intake, dietary fibre and mobilization. However, in the palliative care population these options may not be appropriate. | |
Opioid-related drowsiness | This symptom is commonly present in the first days of opioid therapy, and is usually self-limiting. Patients should be advised to expect some sedation during the first days of treatment, and not to drive. If persistent, consider treatment of contributing factors: • Sleep hygiene • Review of other centrally acting drugs e.g. benzodiazepines • Correction of metabolic disturbances (e.g. hypercalcaemia) If pain-free, consider a reduction of opioid dose. If pain present, consider addition of a psychostimulant, e.g. methylphenidate |
Opioid-related delirium | Opioid-related delirium is often associated with the combined effect of the opioid with other contributing factors, e.g. infection, electrolyte disturbance, CNS metastases or organ failure. Treat contributory causes where appropriate and possible. |
If pain-free, consider a reduction of opioid dose. If pain is present, consider opioid switch or use of adjuvants. | |
Discontinue other centrally acting agents. | |
Use neuroleptics, such as haloperidol. | |
Opioid-related xerostomia | Meticulous mouth care. |
Consider stopping anticholinergic drugs where possible. | |
Pilocarpine 2% eye drops by mouth or 5mg by mouth 3 times daily may be of use. | |
Opioid-related pruritus | More common with spinal than with systemic opioids • Use Alternative opioid • If unsuccessful, treat opioid-induced pruritus with 5-HT3 antagonists such as ondansetron |
Opioid-related sweating | Exclude other causes of sweating |
Consider alternative opioid | |
Consider use of antimuscarinic drugs | |
Rarely: | Hyperalgesia and allodynia have been reported with high-dose opioids. The symptoms may be associated with signs of toxicity. Characteristically, the patient reports that an increase in the opioid dose leads to worsening pain. Substitution of an alternative opioid often resolves the symptoms. Alternatively, reduction of dose and the addition of an alternative co-analgesic may be useful. |
Opioid-related nausea and vomiting | Although these symptoms are commonly present during initiation of opioid therapy, they are frequently self-limiting and usually require only a short course of anti-emetic therapy. |
If symptoms persist, metoclopramide 10-20mg p.o. t.d.s. is generally used as first-line anti-emetic. Haloperidol 1.5–3mg o.n. initially is also useful. | |
Opioid-related constipation | Prescription of regular, prophylactic laxatives is recommended. Studies comparing laxatives, especially in palliative care patients, are lacking. Various laxatives have been investigated, but findings remain inconclusive and choice of laxative is often dependent on local practice. A combination of a stimulant laxative (senna) and a stool softener (docusate) may be used. |
Methylnaltrexone has been developed specifically for opioid-induced constipation, see p. 319 | |
If problem persists, laxatives should be titrated upwards, and enemas may be used. The addition of metoclopramide for its pro-kinetic effects may be beneficial. | |
If problem is severe, consider prescription of alternative opioid. Fentanyl and buprenorphine may be less constipating. | |
Consider non-pharmacological approaches, such as fluid intake, dietary fibre and mobilization. However, in the palliative care population these options may not be appropriate. | |
Opioid-related drowsiness | This symptom is commonly present in the first days of opioid therapy, and is usually self-limiting. Patients should be advised to expect some sedation during the first days of treatment, and not to drive. If persistent, consider treatment of contributing factors: • Sleep hygiene • Review of other centrally acting drugs e.g. benzodiazepines • Correction of metabolic disturbances (e.g. hypercalcaemia) If pain-free, consider a reduction of opioid dose. If pain present, consider addition of a psychostimulant, e.g. methylphenidate |
Opioid-related delirium | Opioid-related delirium is often associated with the combined effect of the opioid with other contributing factors, e.g. infection, electrolyte disturbance, CNS metastases or organ failure. Treat contributory causes where appropriate and possible. |
If pain-free, consider a reduction of opioid dose. If pain is present, consider opioid switch or use of adjuvants. | |
Discontinue other centrally acting agents. | |
Use neuroleptics, such as haloperidol. | |
Opioid-related xerostomia | Meticulous mouth care. |
Consider stopping anticholinergic drugs where possible. | |
Pilocarpine 2% eye drops by mouth or 5mg by mouth 3 times daily may be of use. | |
Opioid-related pruritus | More common with spinal than with systemic opioids • Use Alternative opioid • If unsuccessful, treat opioid-induced pruritus with 5-HT3 antagonists such as ondansetron |
Opioid-related sweating | Exclude other causes of sweating |
Consider alternative opioid | |
Consider use of antimuscarinic drugs | |
Rarely: | Hyperalgesia and allodynia have been reported with high-dose opioids. The symptoms may be associated with signs of toxicity. Characteristically, the patient reports that an increase in the opioid dose leads to worsening pain. Substitution of an alternative opioid often resolves the symptoms. Alternatively, reduction of dose and the addition of an alternative co-analgesic may be useful. |
Additional strategies for the management of opioid-related side-effects include:
Opioid switching (section p. 248)
Use of adjuvant analgesics that act in a synergistic manner to reduce opioid requirements ( p. 269)
Use of nerve blocks or alternative routes of administration, such as epidural ( p. 282)
Use of anticancer therapies to reduce tumour burden p. 279.
Opioid toxicity
Opioid toxicity may be precipitated by:
Excessive increase in opioid dose or inappropriate use of opioids for the management of an opioid-insensitive pain
Dehydration or renal impairment
Infection
Other change in disease status, e.g. hepatic function, weight loss
Reduction in analgesic requirements because pain is relieved by other methods
Drug interactions, e.g. the co-administration of amitriptyline increases the bioavailability of morphine
Drowsiness
Hallucinations (most commonly visual)
Confusion
Vomiting
Myoclonus
Pinpoint pupils
Respiratory depression (if severe)
Treat the underlying cause where appropriate (e.g. correct dehydration)
Reduce opioid dose
Prescribe haloperidol for management of delirium
Prescribe benzodiazepine (e.g. clonazepam, midazolam) for symptomatic management of myoclonus
Consider switch to alternative opioid if dose reduction fails to resolve toxicity or results in uncontrolled pain
Pain acts as a physiological antagonist to the central depressant effect of opioids, and strong opioids, when used appropriately, do not cause respiratory depression in patients with pain. Clinicians should take care to distinguish between true respiratory depression attributable to opioids and the slow and irregular breathing that may accompany the terminal phase.
If respiratory depression does occur, reduction of the opioid dose is usually all that is required immediately. Subcutaneous infusions of opioids should be temporarily stopped to allow plasma levels to decrease, before restarting at a lower dose.
Naloxone is rarely used in palliative care practice and is only indicated if significant respiratory depression is present that is attributable to opioids; acute opioid withdrawal symptoms and pain can be severe in patients who have been on long-term opioids.
Respiratory rate <8 breaths/min, or
<10–12 breaths/min, difficult to rouse and clinically cyanosed, or
<10–12 breaths/min, difficult to rouse and SaO2 <90% on pulse oximeter
Stop opioid
Secure IV access
Dilute 0.4mg naloxone in 10mL sodium chloride 0.9%
Give 0.5mL (=20mcg naloxone) every 2min IV until satisfactory respiratory status
Review renal function, pain and analgesic requirements
NB. Naloxone has a half-life of 5–20min. As the half-life of most opioids is longer than this, it is important to continue to assess the patient and give naloxone at further intervals if necessary
Changing route of administration of opioids
In the palliative care setting the IM route is normally avoided because of issues of painful injections, cachexia and reduced muscle mass. The SC route is often used, and is appropriate for those patients who are unwilling or unable to take oral medications for such reasons as:
Reduced level of consciousness
Dysphagia
Nausea/vomiting
Tablet burden
Malabsorption
SC administration is not an alternative to ineffective oral treatment. In such cases, the clinician should reassess the patient, determine the reason for their poor response to therapy, and develop a targeted plan to deal with the problem.
Opioids have different analgesic potencies when administered by different routes. Doses of opioids need to be adjusted, therefore, when their route of administration is altered in order to avoid under- or over-dosing. For example, intrathecal and epidural morphine are approximately 100 and 10 times, respectively, more potent than oral morphine.
The average relative potency ratio of oral morphine to subcutaneous morphine is between 1:2 and 1:3 (i.e. 20–30mg of morphine by mouth is equianalgesic to 10mg by SC injection).22
Alternative opioids for moderate to severe pain
A number of alternative strong opioid analgesics are available which have their place in palliative care practice (Table 6a.7):
Morphine and similar drugs | Morphine Diamorphine Hydromorphone Oxycodone |
Fentanyl and similar drugs | Fentanyl Alfentanil Sufentanil |
Methadone | Methadone |
Mixed agonist–antagonist | Buprenorphine |
Not recommended | Pethidine |
Morphine and similar drugs | Morphine Diamorphine Hydromorphone Oxycodone |
Fentanyl and similar drugs | Fentanyl Alfentanil Sufentanil |
Methadone | Methadone |
Mixed agonist–antagonist | Buprenorphine |
Not recommended | Pethidine |
Indications for prescribing an initial opioid other than morphine
Patient choice
Past history of unacceptable adverse effects associated with prior use of morphine
Patient unwilling or unable to take oral morphine regularly, and wishes to take a less invasive form of opioid than morphine administered via a syringe driver—a transdermal opioid such as fentanyl may be appropriate
Subacute/partial intestinal obstruction—a less constipating opioid such as fentanyl may be appropriate
Renal failure
Indications for changing to alternative opioids (opioid switching, rotation or substitution)
Patients who experience poor analgesia or significant side-effects may benefit from a change in their opioids, called ‘opioid switching’. Opioid switching involves discontinuation of the previously used opioid and initiation of the new one at the equianalgesic dose
This approach of opioid switching is based on observations that inter-individual response may vary from opioid to opioid, and that switching to another opioid may lead to better opioid responsiveness with fewer side-effects.23 Switching between opioids complicates pain management and this is a disadvantage for non-specialists (for whom it is not recommended without expert advice).
Choice of alternative opioid
The decision to use alternative opioids in place of morphine is best made by a palliative care specialist. Rigorous evidence favouring the use of one opioid over another is often lacking, but it is generally accepted that individual opioids have characteristics that may result in clinicians preferentially using a particular opioid in certain clinical situations (Table 6a.8).
Diamorphine | More soluble than morphine, and often used as the opioid in a syringe driver for SC infusion |
Fentanyl | Of use in those patients experiencing inadequate pain relief or unacceptable toxicity with another opioid |
Of use in those patients with renal impairment | |
Associated with less constipation than other opioids, and so may be used in patients for whom constipation is a refractory problem | |
OTFC preparations of use in treatment of incident pain | |
Transdermal preparation of use for patients who are unwilling or unable to take oral opioids | |
Buprenorphine | Of use in those patients experiencing inadequate pain relief or unacceptable toxicity with another opioid |
Transdermal preparation of use for patients who are unwilling or unable to take oral opioids | |
Of use in renal impairment | |
Methadone | Of use in patients with refractory pain that has a neuropathic component |
Of use in renal impairment | |
Of use where cost is an economic consideration | |
Oxycodone | Of use in those patients experiencing inadequate pain relief or unacceptable toxicity with another opioid |
Hydromorphone | Of use in those patients experiencing inadequate pain relief or unacceptable toxicity with another opioid |
May be of use in renal impairment |
Diamorphine | More soluble than morphine, and often used as the opioid in a syringe driver for SC infusion |
Fentanyl | Of use in those patients experiencing inadequate pain relief or unacceptable toxicity with another opioid |
Of use in those patients with renal impairment | |
Associated with less constipation than other opioids, and so may be used in patients for whom constipation is a refractory problem | |
OTFC preparations of use in treatment of incident pain | |
Transdermal preparation of use for patients who are unwilling or unable to take oral opioids | |
Buprenorphine | Of use in those patients experiencing inadequate pain relief or unacceptable toxicity with another opioid |
Transdermal preparation of use for patients who are unwilling or unable to take oral opioids | |
Of use in renal impairment | |
Methadone | Of use in patients with refractory pain that has a neuropathic component |
Of use in renal impairment | |
Of use where cost is an economic consideration | |
Oxycodone | Of use in those patients experiencing inadequate pain relief or unacceptable toxicity with another opioid |
Hydromorphone | Of use in those patients experiencing inadequate pain relief or unacceptable toxicity with another opioid |
May be of use in renal impairment |
A number of factors influence choice of an appropriate opioid:
Individual patient factors (e.g. preference, compliance, renal and other organ function, co-morbidities, out-patient vs in-patient setting, stable vs uncontrolled pain)
Drug profile
Possible/desirable routes of administration
Comparative analgesic effects
Comparative adverse effect profile
Other potential therapeutic effects
Availability
Mechanism of opioid switching
The equianalgesic doses of different opioids drugs are only approximations. A number of factors can affect their accuracy:
Individual patient variation (differences in absorption, metabolism, excretion)
The sequence of opioid switching
Equianalgesic doses can vary according to the dose of opioid (e.g. at higher doses more conservative estimates of opioid conversion are recommended)
Some estimations are derived from studies of single doses rather than continued therapy, and may therefore require adjustment in clinical practice
Although guidelines and conversion tables should be consulted, each drug must be titrated against pain and side-effects for each individual patient
Suggested regime for opioid switching
Calculate the equianalgesic dose of the new drug according to guidelines or tables p. 261–266
Reduce the calculated dose of the new opioid by about 30% to account for incomplete cross-tolerance and inter-individual variability in potency
Further adjust the dose, if necessary, according to prior pain control (e.g. smaller reductions may be indicated in patients with uncontrolled pain)
Reassess and titrate new opioid against pain and side-effects
Other opioids
Diamorphine
Diamorphine is more soluble in water than morphine, and is used as an injectable strong opioid in a syringe driver for subcutaneous infusion.
NB It is unusual to give diamorphine IV in a hospice setting. Subcutaneous administration is most commonly used (onset of action is 10–20 minutes) unless the pain is very severe and immediate relief is needed.
Conversion ratio of oral morphine: parenteral diamorphine is 3mg: 1mg
To convert from oral morphine to SC diamorphine, divide the total dose of oral morphine by three, e.g.:
10mg 4h morphine
≅60mg oral morphine in 24h
≅20mg diamorphine by CSCI over 24h
Inj.: 5mg, 10mg, 30mg, 100mg, 500mg
Oxycodone is a strong opioid analgesic similar to morphine.
May be given by p.o., p.r., SC, IM and IV routes
It is available in 4-h i/r, and 12-h m/r oral preparations, and is used in a similar way to morphine (remember to prescribe a laxative
It is a useful, alternative opioid in selected patients who develop side-effects with morphine
Although a recent meta-analysis failed to find any clinically significant difference in adverse effect profile between morphine and oxycodone,24 inter-individual variation means that some individuals who are intolerant of morphine may benefit from an opioid switch to oxycodone
Oxycodone is metabolized to noroxycodone, oxymorphone and their glucuronides. However, the analgesic activity and profile of the metabolites is not yet known, and caution is advised when using oxycodone in patients with renal impairment
Oxycodone is metabolized by CYP2D6, but the extent to which its analgesic effect is dependent upon this system is unclear. There have been reports of reduced analgesic effect when used in combination with CYP2D6 inhibitors
The conversion ratio of morphine to oxycodone is approximately 2:1.
If a patient is already on morphine, then the total 24-hour dose of morphine should be divided by two for the equivalent 24-hour dose of oxycodone; e.g.: morphine 60mg/24h = morphine m/r 30mg b.d. = oxycodone 30mg/24h = oxycodone m/r 15mg b.d.
Caps: i/r 5mg, 10mg, 20mg (OxyNorm)
Tabs: m/r (12h) 5mg, 10mg, 20mg, 40mg, 80mg (OxyContin)
Liquid: 5mg/5mL, 10mg/1mL
Injection: 10mg/1mL, 1mL and 2mL amps
Hydromorphone is a strong opioid analgesic very similar to morphine.
Hydromorphone can be given by p.o., SC, IM, IV and spinal routes
It is used in a similar way to morphine (remember to prescribe a laxative)
Similarly to morphine, when administered orally it undergoes an extensive first-pass metabolism, resulting in the production of three major metabolites: hydromorphone-3-glucuronide (H-3-G), dihydromorphine and dihydroisomorphine.
Hydromorphone and its metabolites are eliminated renally
It is used widely in North America as an alternative to diamorphine, which is not available. It is available in 4h i/r and 12h m/r preparations, but the injection is not routinely available in the UK
Hydromorphone and morphine generally have the same adverse effects, although there may also be individual variation in adverse effect profile between patients.
When converting from oral morphine to oral hydromorphone, the manufacturers recommend a ratio of 7.5:1 (i.e. morphine 10mg hydromorphone 1.3mg)
Caps: 1.3mg, 2.6mg (Palladone)
Caps: m/r (12h) 2mg, 4mg, 8mg, 16mg, 24mg (Palladone SR)
Caps may be opened and sprinkled on food
Inj.: 10mg/1mL, 20mg/1mL, 50mg/1mL
Injections available in UK as a special order from Martindale. See BNF for contact details.
Fentanyl is a semi-synthetic opioid with a high degree of lipid solubility. It is 75–100 times as potent as morphine
It can be given by IV, IM, SC, transdermal and spinal routes
It is metabolized in the liver to mainly inactive metabolites, so it is of use in patients with renal failure
Although both fentanyl and morphine act at the mu receptor, patients switched from long-term morphine to fentanyl may experience withdrawal symptoms despite good pain relief because fentanyl is a highly selective mu agonist.25 Symptoms may be treated using rescue doses of immediate release morphine until they resolve after a few days
Fentanyl shares the side-effect profile of morphine and other opioids, but may cause less constipation, nausea and vomiting. Nevertheless, laxatives are generally still required.26 In one large trial, although constipation occurred less frequently with fentanyl than with morphine, it was still the most frequent adverse effect reported with each drug (52% and 65% of adverse events reported, respectively).28 Local skin erythema or pruritus has been reported with use of transdermal fentanyl (<5%)
The transdermal route is of obvious benefit to patients with dysphagia, vomiting or malabsorption. There are two types of transdermal preparations: gel-reservoir and matrix
Analgesia is delayed for at least 12 hours after application of the first patch, and steady state may not be achieved for 48h. Transdermal fentanyl has a prolonged duration of action; each patch lasts about 72 hours and plasma concentrations are halved about 17 hours after removal
As a result, transdermal fentanyl should only be used in those patients with stable opioid requirements
Up to 25% of patients need a patch change every 48h due to pharmacokinetic variability
Oral i/r morphine is commonly used as the breakthrough analgesic, though oral transmucosal fentanyl citrate may also be used p. 253
Used fentanyl patches should be disposed of safely, as a substantial amount of fentanyl can remain in patches (even after 3 days in situ). In a group of patients studied, 28–84% of fentanyl remained in used patches.27 Patches should be folded with the sticky sides together, wrapped and disposed of either by returning to the pharmacist, or placing in the rubbish well out of reach of children and others
Clinicians should be aware that increased body heat (e.g. fever, humid climate) and direct heat (e.g. from an electric blanket or heat pad) may increase the rate of absorption of fentanyl
Sweating may decrease drug absorption because it prevents the patch from sticking to the skin
Transdermal fentanyl should be used only in patients with stable pain. It should not be used for titration in patients with uncontrolled pain.
The starting dose of transdermal fentanyl is calculated on the basis of the oral morphine equivalent dose and individual patient factors. Consult tables and adjust appropriately p. 261.
From i/r opioid: apply patch when convenient and use oral i/r opioid only as required
From twice-daily m/r opioid: apply patch at the same time as the last dose of m/r oral opioid
(From once-daily m/r opioid: apply the patch 12h after the last dose of m/r opioid)
Breakthrough/rescue doses of i/r opioids may be needed whilst transdermal absorption is established.
It takes up to 72 hours for a steady state of fentanyl to be achieved. Titration, in 25mcg/hour steps, if required, should take place no more frequently than every 3 days.
The dose of oral m/r opioid is calculated by consulting dose conversion tables. The dose should be adjusted appropriately for individual factors.
Remove patch and give the first dose of oral m/r opioid approx. 8h later.
If a patient appears well pain-controlled the fentanyl patch should be continued as normal, and breakthrough doses given as required.
If the patient has uncontrolled pain and death appears to be imminent, it is often not appropriate to stop the fentanyl patch and switch to an alternative opioid administered via syringe driver (because of the adverse effects associated with the inaccurate estimations of dose equivalencies in the terminal phase). In these circumstances, the fentanyl patch may be continued and supplementary regular analgesia may be given by a second opioid infusion via syringe driver.
Note: Care must be taken to use the total 24-h dose of opioid as the basis for calculations of breakthrough doses, i.e. fentanyl + SC opioid
Fentanyl lozenges (on a stick) are rapidly absorbed through the buccal mucosa, leading to onset of pain relief within 5–10 minutes. The maximum effect is reached within 20–40 minutes with a duration of action of 1–3h. Bioavailability is about 50%.
Incident pain
The optimal dose of OTFC cannot be predicted from, and is not directly related to, the daily dose of regular opioid being taken for background pain. Therefore, the effective dose has to be found by titration. There is no conversion ratio between fentanyl lozenges and other opioid formulations (including fentanyl patches).
The lozenge should be placed in the mouth and sucked, constantly moving it from one cheek to the other
It should not be chewed
Water can be used to moisten the mouth beforehand
The lozenge should be consumed within 15min
Partially consumed lozenges should be dissolved under hot running water, and the handle disposed out of reach of children
Initial dose is 200mcg, regardless of the dose of the regular opioid. This dose is probably approx. equivalent to morphine 2mg IV
A second lozenge of the same strength can be used if pain is not relieved after 15 minutes
No more than two lozenges should be used to treat any individual pain episode
Continue with this dose for a further 2 or 3 episodes of breakthrough pain, allowing the second lozenge when necessary
If pain is still not controlled, increase to the next higher dose lozenge
Continue to titrate in this manner until a dose is found that provides adequate analgesia with minimum adverse effects
No more than four doses per day should be used (consider increasing regular strong opioid dose for background pain)
Administration of fentanyl by subcutaneous infusion will allow therapeutic blood levels to be achieved more rapidly, and also allows for the use of fentanyl in patients with uncontrolled pain
Calculate dose as equivalent to transdermal patch, e.g. 25mcg/h = 600mcg/24h; for convenience (and considering the widely variable absorption from a patch) use 500mcg/24h CSCI as equivalent to a 25mcg/h patch
Large volumes are needed for high doses: consider substituting alfentanil (see Alfentanil)
Compatible with most commonly used drugs in palliative care
Fentanyl has been used topically to manage painful skin ulcers.
Patches: 12, 25, 50, 75, 100mcg/h (Durogesic DTrans); starting dose: (25mcg/h) patch every 3 days
Inj.: 50mcg/1mL, 100mcg/2mL, 500mcg/10mL; starting dose: 500mcg/24h CSCI
Lozenge with applicator: 200, 400, 600, 800, 1200, 1600mcg (Actiq); starting dose: 200mcg lozenge regardless of regular opioid dose
Alfentanil is a selective mu-receptor opioid agonist, similar to fentanyl. However, it has a more rapid onset of action and a shorter duration of action. It is mainly metabolized in the liver to inactive compounds.
Its short-lasting effect means it has been used for incident pain e.g. dressing changes, but it is generally not used for other types of breakthrough pain.
Alfentanil has been given by CSCI in a syringe driver, appears to mix with most other commonly used drugs in palliative care and should be diluted with water. Compared to fentanyl, an equianalgesic dose can be used in a much smaller volume, making CSCI of large doses possible.
Injection: 1mg/2mL, 5mg/10mL, 5mg/1mL
Starting dose: 500mcg/24h CSCI (equivalent to diamorphine 5mg CSCI)
Sufentanil is a synthetic opioid very similar to fentanyl, but with a more rapid onset and shorter half life. It can be used as an alternative to alfentanil if the fentanyl dose necessitates too large a volume for the portable syringe driver in use. It has also been used sublingually. The clinically derived sufentanil to fentanyl relative potency is approximately 20:1.
Buprenorphine is a potent semi-synthetic opioid with mixed agonist/antagonist properties. No ceiling dose has been shown for the analgesic effect of buprenorphine in clinical studies, and so it can be used clinically as though it were an agonist at the mu-opioid receptor. The highest reported dose of transdermal buprenorphine in the literature is 140mcg/h.29
It may be given by the epidural, IM, IV, SC and SL routes, but the transdermal route is most commonly used in palliative care practice
It is available in two forms of transdermal delivery systems. Both delivery systems contain the active drug incorporated into a polymer matrix, which is also the adhesive layer
The time to reach steady-state plasma concentrations is approximately 24–48 hours, but additional analgesia may be required for the first 24–48 hours of use
Buprenorphine concentrations decrease to about one-half in 12 hours following patch removal. It has an apparent half-life of about 26 hours
Buprenorphine is metabolized in the liver, principally to an inactive metabolite, norbuprenorphine
It may be used in mild-to-moderate liver failure, and in renal failure( p. 707)
The adverse effect profile of buprenorphine is similar to that of other strong opioids
Transdermal buprenorphine should be used only in those patients with stable pain. It should not be used for titration in patients with uncontrolled pain.
The use of buprenorphine in association with other opioids, as for example during opioid switching or for the management of breakthrough pain, has been of concern because of a possible partial agonist effect, which might reduce analgesia or induce withdrawal symptoms. However, recent data suggest that no interference between buprenorphine and other opioid mu-agonists exists within the usual clinical dose ranges30
A study evaluated the safety and effectiveness of IV morphine in patients with advanced cancer who were receiving transdermal buprenorphine (mean transdermal buprenorphine dose of 44.5 mg/hour, mean IV-morphine dose of 6.1 mg). It found that morphine provided a strong and rapid analgesic effect, and interference between the two opioids was not noted31
Buprenorphine may have a wider safety profile with regards to respiratory depression compared to other opioids—because of its partial agonist activity it has a demonstrated ceiling effect on respiratory depression
However, in the event of respiratory depression occurring, higher doses of naloxone may be required compared with other opioids. The effects of naloxone may be delayed, and it may need to be given in repeated doses because of the long duration of activity of buprenorphine
It has been suggested that buprenorphine’s potential for abuse is limited compared to other opioids—because of the fact that the subjective and physiological effects experienced by patients show a ceiling effect. This is due to its partial agonist activity
Transtec: 35mcg/h, 52.5mcg/h and 70mcg/h patches, which release the drug over a 3-4 day period
BuTrans: 5mcg/h, 10mcg/h and 20mcg/h patches, which release the drug over a 7-day period
Tabs (sublingual): 200mcg
The absorption of buprenorphine from the sublingual mucosa is rapid, occurring within 5 minutes. However, the oral bioavailability of buprenorphine is very low (approximately 10%), and additional swallowing of buprenorphine contributes little to overall absorption
Sublingual buprenorphine is often not tolerated by patients due to its adverse effects profile, which includes dizziness, nausea, vomiting, drowsiness and lightheadedness
As a result, alternative oral opioids, such as i/r morphine, are generally used to provide breakthrough pain relief for patients using transdermal buprenorphine
Methadone is a synthetic opioid agonist at the mu- and delta-opioid receptors, and is also an NMDA-receptor antagonist. Its NMDA-receptor antagonism has led to the clinical impression that methadone has a particular place in the management of neuropathic pain.
It may be given by the p.o., p.r., IV and SC routes
Oral administration is followed by rapid gastrointestinal absorption with measurable plasma levels at 30 minutes.
Tissue accumulation of methadone with chronic administration results in the potential for toxicity. Also, there is considerable inter-individual variation in methadone pharmacokinetics, which means that dose conversion and titration is difficult to predict accurately
Methadone is extensively metabolized in the liver by the cytochrome P450 CYP3A3/4 isoenzyme to inactive metabolites. It is then excreted mainly by the faecal route, and so does not accumulate in renal failure
Methadone should only be used in the palliative care setting by specialists experienced in its use. Even among experienced physicians, occasional serious toxicity can occur during the administration of methadone.37 Contrary to what would be expected, toxicity appears to occur more frequently in patients previously exposed to high doses of opioids than in patients receiving low doses
All the typical opioid side-effects can be expected. Methadone also has an antidiuretic effect.
Methadone has the potential for numerous and complex drug interactions.32 Its metabolism is increased by a number of other drugs, and their use in combination with methadone may then precipitate pain flare and opioid withdrawal symptoms. Other interactions that inhibit metabolism can lead to overdose and toxicity (Table 6a.9).
Decrease methadone levels . | Increase methadone levels . |
---|---|
Phenytoin | Fluconazole |
Phenobarbital | SSRIs |
Carbamazepine (not sodiumvalproate or gabapentin) | |
Rifampicin |
Decrease methadone levels . | Increase methadone levels . |
---|---|
Phenytoin | Fluconazole |
Phenobarbital | SSRIs |
Carbamazepine (not sodiumvalproate or gabapentin) | |
Rifampicin |
Methadone is rarely used as a first-line analgesic in the UK since it has a long half life and is difficult to titrate safely. Equianalgesic ratios of morphine (and other opioids) to methadone are dose-dependent. In single dose studies these ratios may vary from 1:1 at low doses of an oral opioid to as high as 20:1 for those patients receiving oral morphine in excess of 300mg per day. Opioid rotation to methadone is difficult because of the wide variability of equianalgesic ratios, but there have been several guidelines published.37 (The appropriate conversion ratios should be used when switching from other strong opioids.)
Switching oral morphine to oral methadone
The switch to methadone is successful (i.e. improved pain relief and/or reduced toxicity) in about 75% of patients.
Method Adapted from Palliative Care Formulary37
Stop morphine
If switching from immediate release morphine (e.g. oramorph):
give the first dose of methadone (after stopping morphine) ≥2h(if pain present) or 4h (if pain free) after the last dose of morphine.
If switching from modified release morphine:
give the first dose of methadone (after stopping morphine) ≥6hours (if pain present) or 12h (if pain free) after the last dose of a 12h preparation (e.g. MST), or ≥12h (if pain present) or 24h (if pain free) after the last dose of a 24h preparation (e.g. MXL).
give a single loading dose of oral methadone 1/10 of the previous total 24h oral morphine dose, up to a maximum of 30mg.
give oral methadone (1/3 of the loading dose i.e. 1/30 of the previous total 24h oral morphine dose) every 3 hours if needed, up to a maximum of 30mg per dose.
Example 1: Morphine 300mg/24h p.o. = loading dose of methadone 30mg p.o., with 10mg 3hourly p.r.n.
Example 2: Morphine 1200mg/24h p.o. = loading dose of methadone 120mg p.o., with 40mg 3hourly p.r.n.; however, both are limited to the maximum of 30mg.
on day 6, the amount of methadone taken over the previous 2 days is noted and divided by 4 to give a regular 12hourly dose, with 1/6–1/10 of the 24h dose given every 3h p.r.n.
Example: Methadone 80mg p.o. in previous 48h=Methadone 20mg every 12h and 5mg p.o. every 3h p.r.n.
If ≥2 doses/day of p.r.n. methadone continue to be needed, the dose of regular methadone should be increased once a week, guided by p.r.n. use.
Converting oral opioid (other than morphine) to oral methadone
Calculate the morphine equivalent daily dose and then follow the guidelines for morphine
Converting from oral methadone to SC methadone
give½–¾ of the 24h oral dose subcutaneously over 24h
For patients in severe pain and who need more analgesia in < 3h:
Give the previously used opioid every hour p.r.n. (50–100% of the p.r.n. dose used before switching). (If neurotoxicity was apparent with the preswitch dose, use an appropriate dose of an alternative strong opioid)
If there has been a rapid escalation of the preswitch opioid dose, calculate the dose of methadone using the pre-escalation dose of the opioid. If a patient becomes over-sedated, reduce the dose generally by 33–50% (some centres monitor the level of consciousness and respirations every 4h for 24h). If a patient develops opioid abstinence symptoms, give p.r.n. doses of the previous opioid until symptoms settle.
Subcutaneous methadone
Although SC methadone has been used, there may be problems with skin reactions, partly because methadone in solution is acidic. If it is necessary to use SC methadone, dilute it as much as possible.
Switching other opioids CSCI to methadone CSCI
Direct CSCI switching between other opioids and methadone is not recommended.
Mixtures: 1mg/1mL, 10mg/1mL
Tabs: 5mg
Linctus: 2mg/5mL (for cough)
Injection 25mg/1mL, 50mg/1mL
Pethidine is a synthetic opioid and has similar agonist effects to morphine. However, it has a short duration of action, and, when given regularly, active metabolites accumulate and can cause convulsions. Although accumulation of metabolites is more likely in patients with renal disease, toxicity has been observed in patients with normal renal function and, as a result, its use is contraindicated in the management of chronic cancer pain.
Topical opioids
Although opioid receptors are not evident in normal tissue, they are found in inflamed tissue. As a result, there has been some interest in the efficacy of topical opioids for the management of ulcers and pressure sores. The use of topical opioids is associated with the potential advantage of a reduction or avoidance of the adverse effect profile associated with the use of systemic opioids.
Most studies describing the topical effects of morphine have mixed the opioid in Intrasite gel, although there are anecdotal reports, in which morphine (or diamorphine) has been mixed with flamazine or metronidazole gel.
A randomized, double-blind, placebo-controlled, crossover pilot study reported an analgesic effect when morphine was applied topically to painful ulcers.38 However, patient numbers were small and, therefore, the data should be interpreted with caution. Treatment was generally well tolerated by patients, and although local reactions were described during the study, these were mild and possibly not related to morphine. No systemic adverse effects were reported.
Equivalent opioid doses
The following tables show the approximately equivalent oral, transdermal and subcutaneous opioid doses.
Opioid Equivalence for Transdermal Patches – Adult Use
Transdermal patches are not suitable for patients who require rapid titration of strong opioid medication for severe pain and should be restricted to those diagnosed with stable pain.
24 hourly Oral Morphine Dose (mg) . | Fentanyl Patch Strength (mcg/hr) . | 4 Hourly Oral Morphine (mg) (also breakthrough medication dose) . |
---|---|---|
<90 | 25 | <15 |
90–134 | 37 | 15–20 |
135–189 | 50 | 25–30 |
190–224 | 62 | 35 |
225–314 | 75 | 40–50 |
315–404 | 100 | 55–65 |
405–494 | 125 | 70–80 |
495–584 | 150 | 85–95 |
585–674 | 175 | 100–110 |
675–764 | 200 | 115–125 |
765–854 | 225 | 130–140 |
855–944 | 250 | 145–155 |
945–1034 | 275 | 160–170 |
1035–1124 | 300 | 175–185 |
24 hourly Oral Morphine Dose (mg) . | Fentanyl Patch Strength (mcg/hr) . | 4 Hourly Oral Morphine (mg) (also breakthrough medication dose) . |
---|---|---|
<90 | 25 | <15 |
90–134 | 37 | 15–20 |
135–189 | 50 | 25–30 |
190–224 | 62 | 35 |
225–314 | 75 | 40–50 |
315–404 | 100 | 55–65 |
405–494 | 125 | 70–80 |
495–584 | 150 | 85–95 |
585–674 | 175 | 100–110 |
675–764 | 200 | 115–125 |
765–854 | 225 | 130–140 |
855–944 | 250 | 145–155 |
945–1034 | 275 | 160–170 |
1035–1124 | 300 | 175–185 |
Please note conversion factors may change depending on direction of conversionNOTE: Transdermal Fentanyl Patch has 72 hour duration of action i.e. changed every 3 days
Buprenorphine Matrix Patch/Patches micrograms/hr . | 24 hr Oral Morphine Dose (mg) . |
---|---|
35 | 50–97 |
52.5 | 76–145 |
70 | 101–193 |
Buprenorphine Matrix Patch/Patches micrograms/hr . | 24 hr Oral Morphine Dose (mg) . |
---|---|
35 | 50–97 |
52.5 | 76–145 |
70 | 101–193 |
NOTE: Transtec Patch is replaced every 3–4 days
. | 5 micrograms/hr . | 10 micrograms/hr . | 20 micrograms/hr . |
---|---|---|---|
Oral Tramadol | ≤ 50mg/day | 50-100mg/day | 100–150mg/day |
Oral Codeine | ~30–60mg/day | ~60–120mg/day | ~120–180mg/day |
Oral Dihydrocodeine | ~60mg/day | ~60–120mg/day | ~120–180mg/day |
. | 5 micrograms/hr . | 10 micrograms/hr . | 20 micrograms/hr . |
---|---|---|---|
Oral Tramadol | ≤ 50mg/day | 50-100mg/day | 100–150mg/day |
Oral Codeine | ~30–60mg/day | ~60–120mg/day | ~120–180mg/day |
Oral Dihydrocodeine | ~60mg/day | ~60–120mg/day | ~120–180mg/day |
NOTE: Butrans Patch is replaced every 7 days
Approximate Equivalent Doses of Opioid Analgesics for Adults
Caution should be used when converting opioids in opposite directions as potency ratios may be different
Conversion factors for guidance when converting from one opioid to another . |
---|
Oral Morphine to Subcutaneous (SC) Diamorphine – Divide by 3 Eg 30 mg Oral Morphine = 10 mg SC Diamorphine |
Oral Morphine to Oral Oxycodone – Divide by 2 Eg 30 mg Oral Morphine = 15 mg Oral Oxycodone |
Oral Morphine to Subcutaneous Morphine – Divide by 2 Eg 30 mg Oral Morphine = 15 mg SC Morphine |
Oral Morphine to Oral Hydromorphone – Divide by 7.5 Eg 30 mg Oral Morphine = 4 mg Oral Hydromorphone |
Oral Oxycodone to SC Oxycodone – Divide by 2 (Suggested safe practice) Eg 10 mg Oral Oxycodone = 5 mg SC Oxycodone |
Oral Hydromorphone to SC Hydromorphone – Divide by 2 Eg 4 mg Oral Hydromorphone = 2 mg SC Hydromorphone |
SC Diamorphine to SC Oxycodone – Treat as equivalent up to doses of 60 mg/24 hrs Caution should be used when converting higher doses (Suggested safe practice) Eg 10 mg SC Diamorphine = 10 mg SC Oxycodone |
SC Diamorphine to SC Alfentanil – Divide by 10 Eg 10 mg SC Diamorphine = 1 mg SC Alfentanil |
SC Diamorphine to SC Morphine – ratio is between 1:1.5 and 1:2 – Multiply by 1.5 Eg 10 mg SC Diamorphine = 15 mg SC Morphine |
Oral Tramadol to Oral Morphine– Divide by 10 (Suggested safe practice)* Eg 100 mg Oral Tramadol = 10 mg Oral Morphine |
Oral Codeine / Dihydrocodeine to Oral Morphine – Divide by 10 Eg 240 mg Oral Codeine / Dihydrocodeine = 24 mg Oral Morphine |
Conversion factors for guidance when converting from one opioid to another . |
---|
Oral Morphine to Subcutaneous (SC) Diamorphine – Divide by 3 Eg 30 mg Oral Morphine = 10 mg SC Diamorphine |
Oral Morphine to Oral Oxycodone – Divide by 2 Eg 30 mg Oral Morphine = 15 mg Oral Oxycodone |
Oral Morphine to Subcutaneous Morphine – Divide by 2 Eg 30 mg Oral Morphine = 15 mg SC Morphine |
Oral Morphine to Oral Hydromorphone – Divide by 7.5 Eg 30 mg Oral Morphine = 4 mg Oral Hydromorphone |
Oral Oxycodone to SC Oxycodone – Divide by 2 (Suggested safe practice) Eg 10 mg Oral Oxycodone = 5 mg SC Oxycodone |
Oral Hydromorphone to SC Hydromorphone – Divide by 2 Eg 4 mg Oral Hydromorphone = 2 mg SC Hydromorphone |
SC Diamorphine to SC Oxycodone – Treat as equivalent up to doses of 60 mg/24 hrs Caution should be used when converting higher doses (Suggested safe practice) Eg 10 mg SC Diamorphine = 10 mg SC Oxycodone |
SC Diamorphine to SC Alfentanil – Divide by 10 Eg 10 mg SC Diamorphine = 1 mg SC Alfentanil |
SC Diamorphine to SC Morphine – ratio is between 1:1.5 and 1:2 – Multiply by 1.5 Eg 10 mg SC Diamorphine = 15 mg SC Morphine |
Oral Tramadol to Oral Morphine– Divide by 10 (Suggested safe practice)* Eg 100 mg Oral Tramadol = 10 mg Oral Morphine |
Oral Codeine / Dihydrocodeine to Oral Morphine – Divide by 10 Eg 240 mg Oral Codeine / Dihydrocodeine = 24 mg Oral Morphine |
Limited evidence suggests that, when converting from Oral Tramadol to Oral Morphine, the dose should be divided by 5. However this may result in too high a dose for some patients hence locally agreed suggested safe practice is to divide by 10 and ensure appropriate breakthrough dose is available.
Oral medication . | ||||||
---|---|---|---|---|---|---|
Opioid . | Step 2: mild-to-moderate pain . | Step 3: moderate-to-severe pain . | ||||
Codeine | 30–60mg q.d.s. | Not applicable | ||||
Morphine i/r | 2.5–5mg 4h | 10mg 4h | 20mg 4h | 30mg 4h | 40mg 4h | 60mg 4h |
Morphine m/r | 5–15mg b.d. | 30mg b.d. | 60mg b.d. | 90mg b.d. | 120mg b.d. | 180mg b.d. |
Oxycodone i/r | 1.5–3mg 4–6h | 5mg 4h | 10mg 4h | 15mg 4h | 20mg 4h | 30mg 4h |
Oxycodone m/r | 5mg b.d. | 10–20mg b.d. | 30mg b.d. | 40–50mg b.d. | 60mg b.d. | 90mg b.d. |
Hydromorphone i/r | Not applicable | 1.3mg 4h | 2.6mg 4h | 3.9mg 4h | 5.2mg 4h | 6.5mg 4h |
Hydromorphone m/r | Not applicable | 4mg b.d. | 8mg b.d. | 12mg b.d. | 16mg b.d. | 20mg b.d. |
Tramadol i/r | Not applicable | 50mg 4h | Not applicable | |||
Tramadol m/r | 50mg b.d. | 100–150mg b.d. | Not applicable | |||
Approximate 24horal morphine | 30mg | 60mg | 120mg | 180mg | 240mg | 360mg |
Oral medication . | ||||||
---|---|---|---|---|---|---|
Opioid . | Step 2: mild-to-moderate pain . | Step 3: moderate-to-severe pain . | ||||
Codeine | 30–60mg q.d.s. | Not applicable | ||||
Morphine i/r | 2.5–5mg 4h | 10mg 4h | 20mg 4h | 30mg 4h | 40mg 4h | 60mg 4h |
Morphine m/r | 5–15mg b.d. | 30mg b.d. | 60mg b.d. | 90mg b.d. | 120mg b.d. | 180mg b.d. |
Oxycodone i/r | 1.5–3mg 4–6h | 5mg 4h | 10mg 4h | 15mg 4h | 20mg 4h | 30mg 4h |
Oxycodone m/r | 5mg b.d. | 10–20mg b.d. | 30mg b.d. | 40–50mg b.d. | 60mg b.d. | 90mg b.d. |
Hydromorphone i/r | Not applicable | 1.3mg 4h | 2.6mg 4h | 3.9mg 4h | 5.2mg 4h | 6.5mg 4h |
Hydromorphone m/r | Not applicable | 4mg b.d. | 8mg b.d. | 12mg b.d. | 16mg b.d. | 20mg b.d. |
Tramadol i/r | Not applicable | 50mg 4h | Not applicable | |||
Tramadol m/r | 50mg b.d. | 100–150mg b.d. | Not applicable | |||
Approximate 24horal morphine | 30mg | 60mg | 120mg | 180mg | 240mg | 360mg |
In all opioid conversions there is uncertainty. It is safer to err on the side of underestimating analgesic requirements when converting between opioids but ensure that the patient has ready access to appropriate breakthrough analgesia. If in doubt about a conversion seek specialist advice if you are unfamiliar with the particular opioid.
Subcutaneous medication . | ||||||
---|---|---|---|---|---|---|
Opioid . | Step 2: mild-to-moderate pain . | Step 3: moderate-to-severe pain . | ||||
Alfentanil CSCI (24h) | 1mg 24h | 2mg 24h | 4mg 24h | 6mg 24h | 8mg 24h | 12mg 24h |
Diamorphine SC (4h p.r.n.) | 2.5mg 4h | 2.5–5mg 4h | 5–7.5mg 4h | 10mg 4h | 15mg 4h | 20mg 4h |
Diamorphine CSCI (24h) | 10mg 24h | 20mg 24h | 40mg 24h | 60mg 24h | 80mg 24h | 120mg 24h |
Morphine SC (4h p.r.n.) | 2.5–5mg 4h | 5mg 4h | 10mg 4h | 15mg 4h | 20mg 4h | 30mg 4h |
Morphine CSCI (24h) | 15mg 24h | 30mg 24h | 60mg 24h | 90mg 24h | 120mg 24h | 180mg 24h |
Oxycodone SC (4h p.r.n.) | 2.5mg 4h | 2.5mg 4h | 5mg 4h | 7.5mg 4h | 10mg 4h | 15mg 4h |
* Oxycodone CSCI (24h) | 7.5mg 24h | 15mg 24h | 30mg 24h | 50mg 24h | 60mg 24h | 90mg 24h |
Approximate 24 horal morphine | 30mg | 60mg | 120mg | 180mg | 240mg | 360mg |
Subcutaneous medication . | ||||||
---|---|---|---|---|---|---|
Opioid . | Step 2: mild-to-moderate pain . | Step 3: moderate-to-severe pain . | ||||
Alfentanil CSCI (24h) | 1mg 24h | 2mg 24h | 4mg 24h | 6mg 24h | 8mg 24h | 12mg 24h |
Diamorphine SC (4h p.r.n.) | 2.5mg 4h | 2.5–5mg 4h | 5–7.5mg 4h | 10mg 4h | 15mg 4h | 20mg 4h |
Diamorphine CSCI (24h) | 10mg 24h | 20mg 24h | 40mg 24h | 60mg 24h | 80mg 24h | 120mg 24h |
Morphine SC (4h p.r.n.) | 2.5–5mg 4h | 5mg 4h | 10mg 4h | 15mg 4h | 20mg 4h | 30mg 4h |
Morphine CSCI (24h) | 15mg 24h | 30mg 24h | 60mg 24h | 90mg 24h | 120mg 24h | 180mg 24h |
Oxycodone SC (4h p.r.n.) | 2.5mg 4h | 2.5mg 4h | 5mg 4h | 7.5mg 4h | 10mg 4h | 15mg 4h |
* Oxycodone CSCI (24h) | 7.5mg 24h | 15mg 24h | 30mg 24h | 50mg 24h | 60mg 24h | 90mg 24h |
Approximate 24 horal morphine | 30mg | 60mg | 120mg | 180mg | 240mg | 360mg |
Nb. In all opioid conversions there is uncertainty. It is safer to err on the side of underestimating analgesic requirement when converting between opioids but ensure that the patient has ready access to appropriate breakthrough analgesia. If in doubt about a conversion always seek specialist advice if you are unfamiliar with the particular opioid.
The manufacturer of oxycodone states clearly that the conversion factor between oxycodone sc and diamorphine sc is 1:1. While most clinicians are comfortable with this ratio when converting from oxycodone sc to diamorphine sc, when converting from diamorphine sc to oxycodone sc some would advocate a 2:1 ratio with provision for breakthrough medication. As in every dose conversion the clinician must assess each individual patient and clinical situation.
Intravenous use for pain emergencies
Pain is a more terrible lord of mankind than even death itself.
Albert Schweitzer (1875–1965)
Various protocols have been described:
Monitor respiratory rate and conscious level regularly
Draw up morphine diluted to 10mL with water
If opioid naive dilute 5mg of morphine in 10mL of water
If patient is on regular opioid use equivalent 4h dose based on previous opioid use in last 24h ( see ‘Breakthrough doses’, p. 232) diluted in 10mL water
Give appropriate morphine dilution IV at 1mL/minute (total over 10 minutes)
Stop if pain <5/10 or toxicity develops
Repeat the above after a further 10–20 minutes if required
Calculate the total dose of morphine administered and multiply by 6
Start maintenance infusion with CSCI, or give regular oral morphine equivalent dose over 24h
‘Opioid-resistant’ cancer pain
Pseudo-resistant?
Underdosing
Poor alimentary absorption of opioid (rare, except where there is an ileostomy)
Poor alimentary intake because of vomiting
Ignoring psychological aspects of patient care
Semi-resistant?
Bone pain
Raised intracranial pressure
Neuropathic pain
Resistant
Patients who have chronic unremitting pain from a deteriorating condition are particularly at risk of spiritual pain. Referral for psychological/spiritual support is important, and/or that of a complementary therapist.
Using Hay’s seven-model assessment,39 (spiritual pain) is broken down into:
Spiritual suffering—interpersonal or intra-psychic anguish
Inner resource deficiency—diminished spiritual capacity
Belief system problem—lack or loss of personal meaning system
Religious request—a specifically expressed religious need
ALWAYS REVIEW PATIENT REGULARLY AFTER ANY OPIOID SWITCH AS CONVERSION RATIOS ARE APPROXIMATE AND CONSIDERABLE INTER-PATIENT VARIATION MAY OCCUR.
When changing from one opioid to another because of toxicity, dose reduction of 30-50% may be necessary.
Use caution in the elderly and in patients with renal or significant hepatic impairment. Consider reduced doses. In severe renal impairment or dialysis patients, alfentanil may be the preferred opioid. Contact the Palliative Care Team for advice.
Breakthrough doses for each opioid are calculated as approximately ⅙th daily dose (ie 4 hourly dose) but lower doses may be used if effective. eg For a patient on Oral Morphine 60 mg over 24 hours, breakthrough dose is 10 mg orally but a lower dose may be effective in some patients.
When converting opioids at higher doses, caution should be used. Lower doses (30-50% reduction) may be necessary due to incomplete cross tolerance.
For further advice contact Palliative Care Team
Neuropathic pain
The medical treatment of neuropathic pain is unsatisfactory, and pain can prove difficult to control in a significant number of patients with this condition.
The early trials of analgesics for neuropathic pain considered neuropathic pain as a uniform entity, but more recent trials have assessed various pain syndromes. It has emerged from this data that analgesics may vary in efficacy according to the type of pain syndrome. Most of the trials on neuropathic pain have studied patients who have post-herpetic neuralgia and painful polyneuropathy. Fewer trials have looked at neuropathic pain due to cancer infiltration, and, as a result, much of the data on the efficacy of different drugs is extrapolated from data collected from patients with non-malignant pain.
The NNT (number of patients needed to treat to obtain one responder to the active drug) is commonly used as an indication of the overall efficacy of individual drugs. It is a relatively crude measure however, as it can be hampered by methodological variability across RCTs and a lack of consideration for other important outcomes (e.g. quality of life).
Table 6a.13 lists the NNT of analgesics on the basis of data listed in the European Federation of Neurological Societies (EFNS) guidelines. The data was taken from class I/II studies on painful polyneuropathy with 95% confidence intervals (CI). Unless otherwise specified, the NNT used was for 50% pain relief.
Drug . | NNT . |
---|---|
Amitriptyline, clomipramine, imipramine (TCA—balanced monoamine uptake) | 2.1 (CI 1.8–2.6)* |
Desipramine (TCA—mainly noradrenaline reuptake inhibition) | 2.5 (CI 1.9–3.6)* |
Venlafaxine (SNRI) | 4.6 (CI 2.9–10.6) |
Duloxetine (SNRI) | 5.2 (CI 3.7–8.5) |
Oxcarbazepine (antiepileptic)† | 5.9 (CI 3.2–42.2) |
Lamotrigine (antiepileptic)† | 4.0 (CI 2.1–42) |
Gabapentin (antiepileptic)† | 3.9 (CI 3.2–5.1) |
Oxycodone (opioid) | 2.6 (CI 1.9–4.1) |
Tramadol | 3.4 (CI 2.3–6.4) |
Drug . | NNT . |
---|---|
Amitriptyline, clomipramine, imipramine (TCA—balanced monoamine uptake) | 2.1 (CI 1.8–2.6)* |
Desipramine (TCA—mainly noradrenaline reuptake inhibition) | 2.5 (CI 1.9–3.6)* |
Venlafaxine (SNRI) | 4.6 (CI 2.9–10.6) |
Duloxetine (SNRI) | 5.2 (CI 3.7–8.5) |
Oxcarbazepine (antiepileptic)† | 5.9 (CI 3.2–42.2) |
Lamotrigine (antiepileptic)† | 4.0 (CI 2.1–42) |
Gabapentin (antiepileptic)† | 3.9 (CI 3.2–5.1) |
Oxycodone (opioid) | 2.6 (CI 1.9–4.1) |
Tramadol | 3.4 (CI 2.3–6.4) |
Most of the data stems from relatively small crossover trials, which mayoverestimate efficacy
Data from patients with painful diabetic polyneuropathy
Abbreviations: NNT, number needed to treat; TCA, tricyclic antidepressant; SNRI, serotonin and noradrenaline reuptake inhibitor; CI, confidence interval
First-line: TCAs, gabapentin, pregabalin, opioids and tramadol
(Use of amitriptyline and nortriptyline is limited by their adverse effect profiles)
Second-line: duloxetine and venlafaxine
Venlafaxine lacks the sedative and antimuscarinic effects of the TCAs, but should not be used if there is a high risk of cardiac arrhythmias
First-line: TCAs, gabapentin, pregabalin, opioids and tramadol
(Topical lidocaine may also be considered as a first-line drug in elderly patients with allodynia and small areas of pain)
Second-line: capsaicin, valproate
First-line: oxcarbazepine, carbamazepine
First-line (spinal cord injury): gabapentin, pregabalin and opioids
First-line (post-stroke pain): amitriptyline
Second-line: Lamotrigine
Pharmacological management of neuropathic pain
The mechanism of action of antidepressant drugs in the management of neuropathic pain is unclear. Analgesia is often achieved at a lower dosage and faster (usually within a few days) than the onset of any antidepressant effect (which can take up to six weeks). The pain-relieving properties of antidepressants are independent of any effect on mood.
Two main groups of antidepressants are commonly used:
The older tricyclic antidepressants (TCAs)
The newer selective serotonin-reuptake inhibitors (SSRIs) and serotonin–noradrenaline reuptake inhibitors (SNRIs)
Tricyclic antidepressants (TCAs)
TCAs are often listed as first-line drugs for the treatment of neuropathic pain. In terms of NNT, they appear to have a similar efficacy regardless of the underlying condition: diabetes mellitus, post-herpetic neuralgia,traumatic nerve injury or stroke.
TCAs may be subdivided into two categories based on their chemical structure—tertiary and secondary amines:
The secondary amines (e.g. desipramine and nortriptyline) appear to be as effective as the tertiary agents (e.g. imipramine and amitriptyline) as analgesics in the treatment of neuropathic pain, but they produce markedly fewer adverse effects (Table 6a.14)
Adverse effects from TCAs can be significant, and may lead to up to 20% of patients withdrawing from treatment. TCAs are contraindicated in those patients with glaucoma and taking MAOIs
Amitriptyline can increase the bioavailability of morphine, thus leading to toxicity
Anticholinergic effects | Dry mouth, constipation, blurred vision, urinary retention, dizziness, tachycardia, memory impairment, delirium |
Alpha-1-adrenergic effects | Orthostatic hypotension/syncope |
Cardiac conduction delays/heart block | Arrhythmias, Q–T prolongation |
Other side-effects | Sedation, weight gain, excessive perspiration, sexual dysfunction |
Anticholinergic effects | Dry mouth, constipation, blurred vision, urinary retention, dizziness, tachycardia, memory impairment, delirium |
Alpha-1-adrenergic effects | Orthostatic hypotension/syncope |
Cardiac conduction delays/heart block | Arrhythmias, Q–T prolongation |
Other side-effects | Sedation, weight gain, excessive perspiration, sexual dysfunction |
Tabs: 10mg, 25mg, 50mg
Syrup: 25mg/5mL, 50mg/5mL
Start with amitriptyline 10–25mg nocte
Slowly titrate, as tolerated
Some patients do not see a benefit until after 4–6 weeks of treatment, and effective dosages are highly variable, but the average dosage is 75mg/day
severity of pain and the patient’s prognosis will dictate how long to persevere with antidepressants
many patients do not tolerate amitriptyline especially in higher doses, therefore consider changing to a secondary amine
SSRIs and SNRIs are two newer classes of antidepressants that act by selectively inhibiting the presynaptic reuptake of serotonin and noradrenaline.
SNRIs (e.g. duloxetine and venlafaxine) have the advantage that they are generally safer to use than TCAs.
The most common adverse effects associated with SNRIs are nausea, dizziness, drowsiness, constipation, increased sweating and dry mouth.
Duloxetine is contraindicated in patients with glaucoma and those taking MAOIs.
SSRIs (e.g. citalopram and paroxetine) are believed to be less efficacious in the treatment of neuropathic pain. Generally, they are not regarded as first or second-line agents.
As with epilepsy, neuronal hyperexcitability is thought to be of importance in the pathogenesis of neuropathic pain. Anticonvulsants act through a variety of mechanisms to depress synaptic transmission, elevate the threshold for the repetitive firing of nociceptive neurones and reduce discharges from the dorsal root ganglion cells.
Duloxetine does not cause cardiac conduction abnormalities whereas venlafaxine should not be used in established heart disease with a risk of ventricular arrhythmias.
Gabapentin/pregabalin:
Gabapentin is associated with diarrhoea and pregabalin with constipation
Gabapentin and pregabalin bind to voltage-gated calcium channels in the dorsal horn, resulting in a decrease in the release of excitatory neurotransmitters such as glutamate and substance P
Pregabalin is an analogue of gabapentin; it has the same mechanism of action but with a higher affinity for the presynaptic calcium channel
Both drugs have a similar adverse effects profile, which includes dizziness, drowsiness, peripheral oedema and dry mouth
Gabapentin should be titrated slowly according to individual tolerance, and is administered t.i.d.
Pregabalin has a short onset of action, and is administered b.d.
Doses should be adjusted in patients with renal impairment
Carbamazepine is associated with frequent and severe adverse reactions, including sedation, dizziness, gait abnormalities, hyponatraemia, hepatitis and aplastic anaemia. Oxcarbazepine shows little risk for crossed cutaneous allergy. Slow dose titration is recommended.
Lamotrigine is generally well tolerated and has shown efficacy in the treatment of diabetic peripheral polyneuropathy. Adverse effects include dizziness, nausea, headache and fatigue. It may induce potentially severe allergic skin reactions. Slow dose titration is recommended.
Sodium valproate, has been used for many years as an anticonvulsant. RCTs have only recently been carried out with this drug in the study of peripheral neuropathic pain; results are conflicting and firm conclusions as to the place of valproate in the management of neuropathic pain are lacking.
Caps: 100mg, 300mg, 400mg
Tabs: 600mg, 800mg
Starting dose: Day 1, 300mg nocte; day 2, 300mg b.d.; day 3, 300mg t.d.s. p.o.
Usual maintenance dose: 0.9–1.2g/24h. Maximum recommended dose 1.8g/24h, but doses up to 2.4g/24h (and even higher) have been used
Tabs: 200mg, 500mg
Syrup: 200mg/5mL
Suppositories: available as special orders
Starting dose: 200mg t.d.s. p.o. or 500mg nocte p.o.; increase 200mg/day at 3-day intervals
Usual maintenance dose: 1–2g/24h. Max. 2.5g/24h in divided doses
Tabs:100mg, 200mg, 400mg
Liquid: 100mg/5mL
Supps: 125mg, 250mg
Starting dose: 100mg b.d. p.o.; increase from initial dose by increments of 200mg every week.
Usual maintenance dose: 0.8–1.2g/24h in two divided doses. Max. 1.6 –2g/24h
Equivalent rectal dosage: 125mg ≅ 100mg p.o.
Carbamazepine levels are increased (hence risk of toxicity) by dextropropoxyphene (co-proxamol), clarithromycin, erythromycin, fluoxetine, fluvoxamine
Caps: 25mg, 50mg, 75mg, 100mg, 150mg, 200mg, 300mg
Starting dose: 75mg b.d. (but some clinicians recommend lower starting doses); increase after 3 days to 150mg b.d.; increase after seven days to 300mg b.d.
Opioid agonists may relieve neuropathic pain by binding to the mu-opioid receptors in the CNS and thus dampening neuronal excitability. In addition, opioids may be helpful because many patients with cancer who are experiencing neuropathic pain have a nociceptive component to their pain.
If the pain seems to be resistant to first-line opioid (or opioid toxicity is a problem):
An alternative opioid analgesic may be tried for better effect
Medication can be given to counteract side-effects (e.g. psychostimulants for drowsiness)
The pain may be morphine-resistant
Methadone may be considered to be different from the other opioids with respect to neuropathic pain. It can either be tried as an alternative to a first-line opioid, or introduced later, when other options have failed ( see Methadone, p. 258).
The combination of morphine and gabapentin has been shown to be of use in the management of post-herpetic neuralgia. The study demonstrated synergistic effects, with better analgesia at lower doses of each drug than either given as a single agent.43
Topical agents work locally, directly at the site of application, with minimal systemic effects. Lidocaine like other local anaesthetics, seems to act through the inhibition of voltage-gated sodium channels.
Lidocaine patches appear to be safe with very low systemic absorption and only local adverse effects (mild skin reactions). Up to four patches a day for a maximum of 12 hours may be used to cover the painful area. Titration is not necessary.
Capsaicin is thought to elevate the pain threshold by reducing the amount of substance P available to act as a neurotransmitter. The application of the cream can itself cause stinging, which can be relieved by the use of Emla cream applied prior to the capsaicin. Capsaicin is a derivative of chilli pepper and must be applied with glove; it is given 3–4 times a day. Although the pain may increase to start with, perseverance may provide relief.
Cream: 0.075% 45g (Axsain)
Starting dose: apply topically 3–4 times daily
The NMDA receptor is thought to be involved in the development of the ‘wind-up’ phenomenon of neuropathic pain.
NMDA receptors are widely spread throughout the CNS, particularly in the spinal cord. Stimulation of pain fibres in the periphery cause the release of excitatory amino acids such as glutamate and aspartate, which in turn activate the NMDA receptor complex. A phenomenon known as ‘wind-up’ then occurs producing a magnified pain response which, clinically, is associated with the features of neuropathic pain such as allodynia (pain produced by a stimulus that is not normally painful, e.g. light touch) and hyperalgesia (an exaggerated and prolonged pain response to a mildly painful stimulus).
Ketamine and methadone are NMDA antagonists, and this may explain their efficacy in the management of neuropathic pain.
Ketamine is a dissociative anaesthetic which has strong analgesic properties. Its analgesic effect may be partly due to NMDA receptor blockade, i.e. receptor antagonism. Clinical reports indicate that, when added to opioids, low subanaesthetic ketamine doses may give improved analgesia with tolerable adverse effects. However, evidence for the effectiveness of this practice is limited, and insufficient data is available to state definitively that ketamine improves the effectiveness of opioid treatment in cancer pain.44 High-quality RCTs comprising greater numbers of patients and standardized, clinically relevant routes of ketamine administration, are needed.
The use of ketamine has been restricted by its adverse effects profile, which includes sedation, nausea, disagreeable psychological disturbances and hallucinations
It has been used by p.o., SC, IV, epidural and intrathecal routes, and in a very wide range of doses
Drugs that interact with CYP34A have the potential to affect ketamine metabolism (e.g. azole antifungals, macrolide antibacterials, HIV protease inhibitors and ciclosporin)
Clinicians with limited experience in using ketamine should seek expert consultation to develop an appropriate treatment plan
There are no studies comparing various titration or dosing schedules, or routes of administration. Ketamine is effective orally, but it is difficult to assess the potency ratio in view of the wide dose ranges used. Ketamine undergoes first-pass hepatic meta-bolism to an active metabolite, and one study suggests it may be more potent given orally than parenterally. In general, equivalent daily doses should initially be used when changing route.
Ketamine 10mg SC stat. may be given if indicated for severe pain
Start infusion of ketamine 50–100mg/24h CSCI (1–2.5mg/kg per 24h)
Increase ketamine dose by 50–100mg increments as indicated to maximum 500mg/24h CSCI
The use of short-term ‘burst therapy’ has also been reported:47
Starting dose 100mg/24h
Increase after 24h to 300mg/24h if 100mg/24h ineffective
Increase after further 24h to 500mg/24h if 300mg/24h ineffective
Stop 3 days after last dose escalation
Haloperidol or midazolam may be concurrently prescribed(either orally or parenterally) if the patient experiences dysphoria or hallucinations. Some palliative care practitioners recommend that patients at high risk of experiencing these adverse effects be given these medications prophylactically.
Although corticosteroids are commonly used to treat neuropathic pain, RCTs have not been used to evaluate their effectiveness. They may act by reducing inflammatory sensitization of nerves, or by reducing the pressure effects of oedema.
Dexamethasone has the least mineralocorticoid effect, and can be used once a day because of its long duration of effect.
A high initial dose may be used to achieve rapid results (dexamethasone 8mg/day will work in 1–3 days); the dose should then be rapidly reduced to the minimum that maintains benefit.
Long-term corticosteroids may be best avoided, although they can sometimes buy useful time whilst allowing other methods (e.g. radiotherapy or antidepressants) time to work (Table 6a.15):
Hydrocortisone has a high mineralocorticoid effect
Dexamethasone has a relatively high equivalent corticosteroid dose per tablet and less mineralocorticoid effects than either prednisolone or methylprednisolone, with consequently less problems with fluid retention
Prednisolone causes less proximal myopathy than dexamethasone.48 If steroids are not helpful for pain within five days, consider stopping them
Steroid . | Administration . | Equivalent dose (mg) . |
---|---|---|
Dexamethasone | oral/SC | 2 |
Prednisolone | oral/rectal | 15 |
Hydrocortisone | oral/IM/IV | 60 |
Methylprednisolone | oral/IM/IV | 12 |
Steroid . | Administration . | Equivalent dose (mg) . |
---|---|---|
Dexamethasone | oral/SC | 2 |
Prednisolone | oral/rectal | 15 |
Hydrocortisone | oral/IM/IV | 60 |
Methylprednisolone | oral/IM/IV | 12 |
Non-pharmacological management of neuropathic pain
Anaesthetic procedures:
spinal (epidural and intrathecal)
nerve blocks
cordotomy
Disease-modifying therapy e.g. RT CT
TENS
Acupuncture
Management of specific problems
Malignant bone pain
Cancer commonly metastasizes to bone. Although some bone metastases are painless, many frequently cause significant and debilitating pain. The management of patients with metastatic bone pain involves a multidisciplinary approach, and includes analgesia, radiotherapy, surgery, chemotherapy, hormone treatment, radioisotopes and bisphosphonates. Analgesia is generally the first option in most patients. Radiotherapy or surgery can be used for localized metastatic disease, and hemibody radiotherapy may be suitable for patients with disease extending to one region of the body. In patients with widespread painful bone involvement, the use of radioisotopes presents another treatment option.
Radiotherapy49
Radiotherapy has been shown to be effective in decreasing metastatic bone pain and in causing tumour shrinkage or growth inhibition
Up to 60% of patients will have some pain relief and about one-third of them will have complete pain relief following radiotherapy
A ‘pain flare’ is described in around 5% of patients, with the pain worsening in the first few days after treatment before it then settles
There is no difference in the efficacy between single-fraction radiotherapy and multifraction radiotherapy. However, the re-treatment rate and pathological fracture rate are higher after single-dose radiotherapy
Bisphosphonates form part of the standard therapy for hypercalcaemia and for the prevention of skeletal events in some cancers. There is evidence to support the effectiveness of bisphosphonates in providing some pain relief for bone metastases
There is insufficient evidence to recommend bisphosphonates for immediate effect as first-line therapy, to define the most effective bisphosphonates or their relative effectiveness for different primary neoplasms
Bisphosphonates should be considered where analgesics and/or radiotherapy are inadequate for the management of painful bone metastases
Patients may experience a ‘flu-like’ reaction post-treatment
Doses may need to be altered in renal impairment
Analgesic effect should be expected within 14 days. Disodium pamidronate, sodium clodronate and ibandronic acid need to be given every 3–4 weeks, but zoledronic acid has a longer duration of action (4–6 weeks)
It is unclear for how long bisphosphonates should be continued
The role of oral bisphosphonates has yet to be clarified in patients with metastatic bone disease
Inj.: 15mg, 30mg, 90mg (dry powder for reconstitution)
Dose: 90mg IV in 500mL sodium chloride 0.9% over 4h.
Inj.: 300mg/5mL, 300mg/10mL
Dose: 1.5g IV in 500mL sodium chloride 0.9% over 4h
p.o. 800mg or 520mg b.d.
Inj.: 4mg
Dose: 4mg IV in 100mL sodium chloride 0.9% over 30mins
Inj.: 6mg
Dose: 6mg IV in 500mL sodium chloride 0.9% over 1–2h.
p.o. 50mg daily
Doses adjusted according to renal fucntion. Calcium and Vitamin D supplements may be needed.
Strontium-89 and samarium-153 are radioisotopes that are approved in the USA and Europe for the palliation of pain from metastatic bone cancer. Radioisotopes are effective in providing pain relief with response rates of between 40% and 95%. Pain relief starts 1–4 weeks after the initiation of treatment, continues for up to 18 months and is also associated with a reduction in analgesic use in many patients
Thrombocytopenia and neutropenia are the most common toxic effects, but they are generally mild and reversible
Repeat doses are effective in providing pain relief in many patients
The development and clinical assessment of radioisotopes has focused mainly on their use in prostate cancer. Data also exists for their use in lung and breast cancer. Tumours with little or no osteoblastic reaction, for example renal carcinoma and myeloma, tend to show poor response to radioisotopes
External beam hemi-body irradiation is an alternative for multiple-site bone pain
The pain of bone metastases may respond to local infiltration or intra-lesional injection with depot corticosteroid ± local anaesthetic. Consideration should be given to the prophylactic pinning of osteolytic metastases in long bones. Vertebroplasty, in which injection of acrylic cement is administered percutaneously, into unstable fractures of the vertebrae may be worth considering if the patient is relatively well. Spinal or epidural anaesthetic blocks may also be needed
(Table 6a.16)
Pain . | Possible co-analgesics . |
---|---|
Headache due to cerebral oedema | Dexamethasone |
Painful wounds | Metronidazole |
Intestinal colic | Hyoscine butylbromide or hydrobromide |
Gastric mucosa | Lansoprazole |
Gastric distension | Asilone + domperidone |
Skeletal muscle spasm | Baclofen/diazepam |
Cardiac pain | Nitrates/nifedipine |
Oesphageal spasm | Nitrates/nifedipine |
Pain . | Possible co-analgesics . |
---|---|
Headache due to cerebral oedema | Dexamethasone |
Painful wounds | Metronidazole |
Intestinal colic | Hyoscine butylbromide or hydrobromide |
Gastric mucosa | Lansoprazole |
Gastric distension | Asilone + domperidone |
Skeletal muscle spasm | Baclofen/diazepam |
Cardiac pain | Nitrates/nifedipine |
Oesphageal spasm | Nitrates/nifedipine |
Patients with a history of substance abuse should not be denied effective analgesia for genuine pain. But management is more complex in these patients because:
Previous users of opioids can have high opioid tolerance and may need higher doses for effective pain relief
Concurrent use of alcohol and other CNS depressants can have additive effects and place the patient at risk
There may be a greater risk of dependence in such patients
Involve pain management or drug and alcohol specialists when possible.
Anaesthetic procedures in palliative care
Introduction
The majority of patients with cancer can have their pain needs met by following the WHO analgesic ladder. For the minority of those patients who do not obtain satisfactory pain relief with the WHO three-step ladder, a ‘fourth step’ (that is, interventional pain management) can be useful in helping to control pain, maintain psychomotor performance and improve quality of life.53,54 Patients are usually considered for interventional techniques when:
Conventional oral or parenteral therapies are proving unsuccessful, or where side-effects are intolerable
A specific nerve block is likely to provide good analgesia, with minimal or acceptable side-effects
Expertise and support is available
As with all interventions, patient selection is vital for success. General selection criteria include:
Patient competent/consented
Patient compliant
Absence of systemic infection
Absence of specific allergy
Absence of significant coagulopathy
Adequate support for post-procedural care and maintenance
A clear pathway for the management of complications after the patient is discharged from the primary unit
Unlike the general population receiving anaesthetic input, palliative care patients are often debilitated, have limited mobility and have a limited lifespan. Therefore, it is imperative that all that can be done to alleviate their pain is performed with the least inconvenience and with minimal compromise. Although many spinal procedures can easily be carried out at the bedside, the more involved interventions, such as chemical neurolysis, necessitate the use of radiological guidance, usually in a hospital setting. With this in mind, patients should, therefore, be selected for these procedures according to what is considered appropriate and acceptable to their level of disability.
General principles of practice
Resuscitation equipment
Infrequently, systemic toxicity can occur from the administration of local anaesthetic or neurolytic agents. If significant hypotension occurs due to spinal sympathetic blockade, boluses of ephedrine 3mg (i.e. 30mg/mL ampoule diluted to 10mL with sterile water, giving 3mg/mL) should be administered IV at 3–4 minute intervals according to response.
Aseptic technique
All equipment should be sterile and preferably disposable
Cleaning fluids should be disposed of prior to drawing up the local anaesthetic to avoid error
Both sterile gloves and a mask should be worn during the procedures
Drug preparation
Drugs and drug mixtures for intrathecal use should be preservative-free and prepared under sterile conditions. If combinations of drugs are used, care should be taken to ensure their stability and compatibility.55
Intrathecal and epidural (neuraxial) techniques
Direct delivery of opioids to the spinal cord via epidural and intrathecal techniques has become increasingly popular in recent decades,56,57,58 and has proven an effective and reversible way to provide profound analgesia with reduced systemic side-effects. Intrathecal opioids bind to the mu- and kappa-opioid receptors in the substantia gelatinosa of the spinal cord. This is achieved to a lesser extent with epidural opioids, which exert a simultaneous systemic and intrathecal effect (10% and 90%, respectively). See Fig. 6a.7.

Common indications for the use of neuraxial techniques
Unacceptable side-effects despite successful analgesia with systemic opioids
Unsuccessful analgesia despite escalating doses and use of sequential opioids
Intolerable neuropathic pain which may be amenable to spinal adjuvants
Sympathetically mediated pain amenable to sympathetic blockade
Incident pain which may benefit from numbness (local anaesthetic)
Possible contraindications to neuraxial techniques (risk–benefit decision):
Platelet count <20×109/L with clinical symptoms of poor clotting
Full anticoagulation or INR >1.5
Active infection with concurrent septicaemia
Concurrent chemotherapy likely to cause neutropenia
Occlusion of epidural space by tumour at the site of catheter tip placement (epidural catheters only)
Allergy or unmanageable side-effects from the anticipated treatment
Psychosocial issues that make technique untenable
Inadequate professional support in resolving problems
Inadequately investigated symptoms
Factors . | Intrathecal . | Epidural . |
---|---|---|
Infection rate | Same as epidural | Same as intrathecal |
Pain relief | Better for long-term | Good only for short-term |
Dose | Lower (10–20% of epidural dose) | Higher |
Pump refills | Less frequent | More frequent (higher volumes) |
Side-effects | Fewer | More |
Technical difficulty | Easier to place, less likely to become displaced | Potentially more difficult, and catheter may migrate |
Long-term complications | Less (approx. 5%) | More (approx. 55%) |
Catheter occlusion and fibrosis | Minimal | Higher frequency (leading to loss of analgesia/pain on injection) |
Epidural metastases | Less affected | More affected (may compromise drug delivery) |
Overall advantage | Effective analgesia with fewer complications |
Factors . | Intrathecal . | Epidural . |
---|---|---|
Infection rate | Same as epidural | Same as intrathecal |
Pain relief | Better for long-term | Good only for short-term |
Dose | Lower (10–20% of epidural dose) | Higher |
Pump refills | Less frequent | More frequent (higher volumes) |
Side-effects | Fewer | More |
Technical difficulty | Easier to place, less likely to become displaced | Potentially more difficult, and catheter may migrate |
Long-term complications | Less (approx. 5%) | More (approx. 55%) |
Catheter occlusion and fibrosis | Minimal | Higher frequency (leading to loss of analgesia/pain on injection) |
Epidural metastases | Less affected | More affected (may compromise drug delivery) |
Overall advantage | Effective analgesia with fewer complications |
Catheters may be externalized through the skin at the puncture site or may be tunnelled subcutaneously away from the spine. Alternatively, a totally implantable pump system may be employed if the patient has a life expectancy of several months.60
Comparison of continuous versus bolus techniques (Table 6a.18)
Factor . | Continuous . | Intermittent bolus . |
---|---|---|
Dose escalation | Higher | Lower |
Analgesic quality | Better | Fair |
Local anaesthetic combinations | Minimal motor or haemodynamic complications | Higher risk of motor or haemodynamic complications (caution recommended for intraspinal administration) |
Factor . | Continuous . | Intermittent bolus . |
---|---|---|
Dose escalation | Higher | Lower |
Analgesic quality | Better | Fair |
Local anaesthetic combinations | Minimal motor or haemodynamic complications | Higher risk of motor or haemodynamic complications (caution recommended for intraspinal administration) |
Ideally, the continuous infusion technique should be used, with a familiar delivery system, e.g. standard Graseby syringe pump delivering the infusion solution over 24 hours. The use of intrathecal bolus administration during pain crises has been described for use in a selected population of patients,61 but this is not considered routine practice in many institutions.58
Mechanical problems (catheter kinking, disconnection, dislodgement or pump failure) |
Skin breakdown at insertion site |
Infection: local, catheter, epidural abscess, meningitis, systemic infections |
CSF leak, causing headache or loss of analgesia |
CSF seroma |
Haematoma |
Catheter displacement, occlusion or migration |
Nerve damage (rare but possible): procedure-related, from an inflammatory mass at catheter tip, drug-induced neurotoxicity |
Mechanical problems (catheter kinking, disconnection, dislodgement or pump failure) |
Skin breakdown at insertion site |
Infection: local, catheter, epidural abscess, meningitis, systemic infections |
CSF leak, causing headache or loss of analgesia |
CSF seroma |
Haematoma |
Catheter displacement, occlusion or migration |
Nerve damage (rare but possible): procedure-related, from an inflammatory mass at catheter tip, drug-induced neurotoxicity |
Minor sedation: opioid dose excessive |
Urinary retention: commonest in males during first 24 hours |
Persistent nausea |
Pruritis |
Respiratory depression: may be severe and insidious if opioid naϊve |
Hyperalgesia: at higher doses |
Myoclonus: at higher doses, indicating toxicity |
Constipation |
Minor sedation: opioid dose excessive |
Urinary retention: commonest in males during first 24 hours |
Persistent nausea |
Pruritis |
Respiratory depression: may be severe and insidious if opioid naϊve |
Hyperalgesia: at higher doses |
Myoclonus: at higher doses, indicating toxicity |
Constipation |
Sedation: reduce dose of opioid
Urinary retention: usually requires once-only catheterization
Nausea: regular anti-emetic
Respiratory depression (RR less than 8/min or excessive drowsiness) see p. 244:
naloxone 100-400mcg
stop intrathecal infusion
reduce infusion dose
Hyperalgesia:
reduce opioid dose
consider addition of adjuvant agent
Myoclonus:
reduce opioid dose
check renal function
encourage rehydration
consider low-dose benzodiazepine therapy (myoclonus may not be opioid dose-dependent)
Constipation: regular laxatives from the outset of therapy
Choice of drug: opioids, local anaesthetics, clonidine, ziconotide
Opioids
Morphine is one of the least lipid-soluble opioids available, and when given in the spinal space, it has the slowest rate of uptake into the surrounding vasculature, which gives it a longer and primarily spinal site of action.57 Intrathecal morphine is regarded as 100 times more potent than a systemically given dose. Data from postoperative pain studies suggest that morphine is twice as potent as diamorphine by the intrathecal route. Intrathecal morphine should be preservative-free.
Diamorphine can be administered intrathecally, with a potency ratio of 1:100 (intrathecal: systemic). Diamorphine should be reconstituted in sodium chloride 0.9%. Alternatives to morphine and diamorphine are used less frequently.
Hydromorphone is an effective and affordable option for the morphine-intolerant patient. The potency of intrathecal hydromorphone is 5 times that of morphine. Of the lipophilic drugs, both fentanyl and sufentanil are used. Greater lipid solubility may be an advantage to achieve a rapid onset of action, but rapid systemic absorption means a shorter duration of action with a less dose-sparing effect when compared to systemic administration. There is no published data on dose equivalences.
Opioids: typical dosing schedule
If patient is opioid-naive, start with intrathecal diamorphine 0.5–1mg/24h or epidural diamorphine 2.5–5mg/24h
If the patient is established on a systemic opioid, the following regimen is recommended for minimizing the withdrawal phenomenon during route conversion:
give half of the systemic opioid by the established route
convert the remaining half dose numerically to diamorphine, and then divide by 100 to get the intrathecal dose
add 2mL of 0.5% bupivacaine to this diamorphine dose
add sodium chloride 0.9% up to a total of 10mL
infuse this solution over 24h
after 24h, reduce the systemic dose by 50% and titrate the intrathecal dose upwards (usually in increments of 10–30%)
attempt to discontinue the systemic opioid by day 2 (may not always be possible)
Local anaesthetics
Local anaesthetic agents are sodium channel blockers and are unique in their ability to block nerve impulses conducted proximally (pain relief) and impulses conducted distally (motor blockade). The conduction blockade produced is both painless and reversible. Table 6a.19 shows the most frequently used agents.
. | Lidocaine . | Bupivacaine . | Ropivacaine . |
---|---|---|---|
Onset | Rapid | Slower | Similar to bupivacaine |
Duration | Short (hours) | 2–3 times longer than lidocaine | Slightly longer than bupivacaine |
Typical dosage | 2mL 2% over 24h | 2mL 0.5% over 24h | 2mL 0.75% over 24h |
Advantages | Rapid onset and off-set | Synergism with opioids | May preferentially block sensory nerves |
Synergism with opioids | Two-thirds as potent as bupivacaine | ||
Synergism with opioids |
. | Lidocaine . | Bupivacaine . | Ropivacaine . |
---|---|---|---|
Onset | Rapid | Slower | Similar to bupivacaine |
Duration | Short (hours) | 2–3 times longer than lidocaine | Slightly longer than bupivacaine |
Typical dosage | 2mL 2% over 24h | 2mL 0.5% over 24h | 2mL 0.75% over 24h |
Advantages | Rapid onset and off-set | Synergism with opioids | May preferentially block sensory nerves |
Synergism with opioids | Two-thirds as potent as bupivacaine | ||
Synergism with opioids |
Side-effects of local anaesthetics
Postural or overt hypotension—sympathetic block
Numbness—sensory block
Leg weakness—motor block
Altered proprioception in low doses, even when motor weakness is not apparent
Urinary retention
‘Total spinal’—this potentially catastrophic event can occur if a large dose of local anaesthetic is delivered erroneously to the subarachnoid space. A profound drop in blood pressure is accompanied by motor paralysis of the lower limbs, spreading to the upper limbs and respiratory muscles and ultimately involving the brain. Both cardiovascular and ventilatory support are required until the local anaesthetic effects wear off
Clonidine
Clonidine is an α2-adrenergic agonist and appears to act at the level of the spinal cord. It acts synergistically with opioids, but is also a powerful analgesic when used alone in the management of neuropathic pain.55 It is a lipophilic compound and its spinal effect may be more pronounced with intrathecal rather than epidural administration. The dose of clonidine is often limited by the appearance of side-effects, such as sedation, hypotension and bradycardia. Nausea, pruritis and urinary retention have also been reported. Starting doses range from 10 to 20mcg/24h and should be titrated for analgesic effect and minimal side-effects. Doses above 150 mcg/24h should not be necessary.
Ziconotide
Ziconotide is thought to produce its analgesic effects through the blockade of specific N-type calcium channels found at the presynaptic terminals in the dorsal horn.65 Recognized side-effects of ziconotide include dizziness, gait imbalance, nausea, nystagmus, confusion and urinary retention. These side-effects usually occur after prolonged use, and may be severe. The administration of intrathecal ziconotide has been reported to be useful in the treatment of refractory pain in patients with cancer and AIDS.55,66 However, clinical experience is limited and longer term data is awaited. The British Pain Society guidelines57 suggest a starting dose of not more than 0.5mcg/24h, with increases of not more than 0.5mcg/24h no more often than once weekly. Ziconotide is incompatible with morphine.59
Other analgesics have been tried as intrathecal agents, including midazolam, ketamine octreotide, calcium channel blockers and neostigmine. As yet, there is no convincing body of evidence to support their use.57
Guidelines for percutaneous intrathecal catheter insertion
Patient consent
Nurse assistant present
Adequate space for aseptic trolley
Patient in sitting or lateral position, with head and knees curled toward the abdomen
Skin asepsis (e.g. chlorhexidine)
Local anaesthetic infiltration of skin at chosen level of insertion
18G Tuohy needle inserted into spinal space, piercing the dura, until CSF is free-flowing
Aseptic epidural catheter inserted to 15cm, and free-flow of CSF confirmed
Catheter secured and dressing applied to insertion site (one which allows daily visual inspection)
An antibacterial filter should be connected and may remain attached for up to a month
Connection made to infusion pump or syringe driver
Follow-up care
All staff involved in aftercare of the patient should be familiar with the possible side-effects and complications listed, and should be vigilant for signs of infection or problems developing
The catheter insertion site should be inspected on a daily basis
A pain chart should be kept initially until the analgesia is considered sufficient
If the patient is being discharged to the community, there should be liaison with the key members of the primary care team (GP, district nurse and community specialist palliative care nurse) prior to discharge and written guidelines should accompany the patient home
Patients should not be discharged home until dose escalation has been stabilized
Chemical neurolysis for cancer pain
Neurolysis of nerves by chemical means is indicated for patients with limited life expectancy.67 The use of neurolytic techniques has diminished over recent years due to advancements in spinal analgesia and increased life expectancies for cancer patients. However, neurolysis should be considered when:
The pain is severe, intractable and has failed to respond to other measures
The nociceptive pathway is readily identified and related to aperipheral nerve pathway or sympathetic chain
A trial block of local anaesthetic has been successful
The effects of the local anaesthetic block is acceptable to the patient
The goals of neurolysis include reduction in pain and reduction in the need for other pharmacotherapy. Neurolysis rarely is permanent and pain returns as a consequence of regrowth of neural structures or disease progression in the treated area. When used centrally, there is a risk of motor paralysis and sphincter weakness, which are generally unacceptable to most patients. For that reason, patients should be fully consented by the practitioner prior to the procedure.
Neurolytic agents
Alcohol and phenol are the two most commonly used agents.
Alcohol is hypobaric, hence the patient should be able to tolerate a position that allows the alcohol solution to float upwards to the affected nerve root.
Alcohol is usually associated with:
A burning sensation upon injection along the distribution of the nerve, followed by warm numbness
An increase in pain relief over a few days, maximal by one week
Phenol is hyperbaric, so the patient should be able to tolerate a position that allows the phenol to sink down to the nerve roots.
Phenol is characterized as follows:
Following injection, an initial local anaesthetic effect allows relief of pain which is then maintained by the neurolysis which may take 3–7 days to take full effect.
The density and duration of block is felt to be less than that of alcohol
Visceral cancer pain is often produced through a combination of visceral afferent stimulation as well as somatic and neuropathic elements. The sympathetic chain carries much nociceptive information, and blockade of the sympathetic chain may improve both visceral- and sympathetic-mediated pain.
Visceral cancer pain can often be alleviated by a combination of oral pharmacological therapy and neurolytic blockade of the sympathetic axis. Neurolytic techniques have a narrow risk–benefit ratio and, therefore, they should only be performed by experienced pain clinicians in appropriate surroundings56,68
Palliative physicians should be aware of the potential of these blocks and know when it is appropriate to refer patients for neurolytic intervention
Coeliac plexus block
The coeliac plexus is responsible for transmission of nociceptive information from the entire abdominal contents, excluding the descending colon and pelvic structures. It has been successfully used to combat pain from pancreatic cancer and other upper abdominal viscera. It is a relatively safe and simple technique, performed under computed tomography (CT) guidance. Patients referred for this procedure should be able to tolerate lying on an X-ray table, and should have no coagulopathy or local infection ( See Figs 6a.8 a and b).

a&b (a) Positioning for coeliac plexus block; (b) Deep anatomy showing placement of needles for coeliac placement block.
Possible complications include:
Orthostatic hypotension which may persist for days
Backache at the site of needle insertion: if backache and hypotension persist, observe serial haematocrit measurements to rule out retroperitoneal haematoma
Diarrhoea
Abdominal aortic dissection
Paraplegia and motor paralysis: rare
Superior hypogastric block
The superior hypogastric sympathetic ganglion transmits nociceptive information from the pelvis, excluding the distal Fallopian tubes and ovaries. Superior hypogastric blocks have been successfully used to manage pain of pelvic origin, other than ovarian pain. Neurological complications have not been reported with this block.
Ganglion impar block
This ganglion marks the end of the sympathetic chains and is situated at the sacrococcygeal junction. Visceral pain in the perineal area has been successfully treated with neurolytic blockade of this ganglion. These patients often present with a vague, poorly localized perineal pain, accompanied by a burning sensation or urgency, and are often difficult to treat using oral pharmacology alone. No complications have been reported with this block.
Interpleural block
Insertion of an epidural catheter into the pleural space and infusion of local anaesthetic or phenol has been described in the management of visceral pain associated, with oesophageal cancer69 and rib invasion by bone metastases (Fig. 6a.9). It is a relatively simple technique with few complications, which include:
Pneumothorax (avoid bilateral blocks)
Phrenic nerve palsy
Trauma to local structures caused by the needle, catheter or as a consequence of the phenol injection

Saddle block
This is a modified low spinal technique where phenol is injected into the CSF in the lumbar area, with the intention of causing chemical neurolysis of the low sacral nerve roots that serve the perineum and perianal area. It is useful for patients complaining of pain or excoriation in the ‘saddle’ area, but carries the potential risk of sphincter compromise (<10%).
Complementary therapies and other non-pharmacological pain interventions
A range of techniques and expertise exists which complement the pharmacological and interventional approaches that have dominated this module thus far (Table 6a.20).
Complementary therapies . | Other non-pharmacological interventions . |
---|---|
Acupuncture | Positioning |
Reflexology | Catheterization |
Aromatherapy | Reassurance |
Art therapy | Good communication |
Music therapy | Diversional therapy |
Touch therapy | TENS |
Splinting of a fractured limb | |
Psychological support |
Complementary therapies . | Other non-pharmacological interventions . |
---|---|
Acupuncture | Positioning |
Reflexology | Catheterization |
Aromatherapy | Reassurance |
Art therapy | Good communication |
Music therapy | Diversional therapy |
Touch therapy | TENS |
Splinting of a fractured limb | |
Psychological support |
( See also Chapter 11.)
These techniques are not just an adjunct to medication but point to the centrality of holistic patient-centred care. Not all approaches will be appropriate for every patient, but for some traditional medicine has little to offer either.
Pain and difficulties in communication
High prevalence of pain in the elderly population is a recognized reality. Almost half of those who die from cancer are over 75 years old. One study showed that 40–80% of elderly people in institutions are in pain. There is evidence that many patients suffering from some form of dementia receive no pain relief at all, despite the presence of a concomit-ant, potentially painful illness.
The reason for this lies in the difficulty in assessing those with communication difficulties. In addition, the elderly often minimize their pain, making it even more difficult to evaluate. The only indication of pain may be a patient’s unusual behaviour that returns to normal with adequate analgesia
Various attempts have been made to evaluate pain in such circumstances. The DOLOPLUS scale,70 developed in 1993, is based on observations of a patient’s behaviour in ten different situations that could be associated with pain. Pain is classified into somatic, psychomotor and psychosocial aspects and scores are allocated. A collective score level confirms the presence of pain.
Examples of unusual behaviours indicating pain71
Verbal expression, e.g.:
Crying when touched
Shouting
Becoming very quiet
Swearing
Grunting
Talking without making sense
Facial expression, e.g.:
Grimacing/wincing
Closing eyes
Worried expression
Behavioural expression, e.g.:
Jumping on touch
Hand pointing to body area
Increasing confusion
Rocking/shaking
Not eating
Staying in bed/chair
Withdrawn/no expression
Grumpy mood
Physical expression, e.g.:
Cold
Pale
Clammy
Change in colour
Change in vital signs if acute pain (e.g. BP, pulse)
Further reading
Book
Articles
Month: | Total Views: |
---|---|
October 2022 | 3 |
December 2022 | 3 |
January 2023 | 2 |
February 2023 | 4 |
March 2023 | 5 |
April 2023 | 1 |
May 2023 | 2 |
June 2023 | 2 |
July 2023 | 2 |
August 2023 | 3 |
September 2023 | 2 |
October 2023 | 3 |
November 2023 | 2 |
December 2023 | 2 |
January 2024 | 1 |
February 2024 | 1 |
March 2024 | 4 |
April 2024 | 2 |
May 2024 | 3 |
June 2024 | 5 |
July 2024 | 1 |
August 2024 | 13 |
September 2024 | 6 |
November 2024 | 1 |
January 2025 | 1 |