Prescribing drugs of dependence in general practice

Part C2 - The role of opioids in pain management - Chapter 1

Evidence-based guidance for opioids in acute and chronic pain

Download PDF

Last revised: 22 Jun 2020

As with any treatment, prescription of opioids should be based on:

  • a comprehensive medical assessment
  • a diagnosis
  • consideration of the likely risks and benefits of any medication, as well as alternative interventions
  • a management plan derived through shared decision making (SDM) and continual clinical monitoring.

Opioid treatment seeks to maximise outcomes for the health and social functioning of the patient while minimising risks. To minimise risks, opioids should be prescribed at the lowest effective dose for the shortest clinical timeframe.

Accountable prescribing also involves an assessment of patient risk stratification, provision of adequate therapeutic monitoring, dose limitations and compliance with national and state law.

As a general precaution, GPs should avoid prescribing opioids to patients with comorbid alcohol or substance use disorders (SUDs) or polydrug use. Patients who use two or more psychoactive drugs in combination (polydrug use) and those with a history of substance misuse may be more vulnerable to major harms. GPs should consider seeking specialist opinion in the management of these patients.

Key points

  • Most acute pain conditions presenting in general practice can be treated with non-opioid analgesia.
  • GPs should be familiar with common acute pain presentations where opioids are not recommended.
  • Patients with an acute pain in the context of existing chronic pain should be assessed with caution and usually by, or in conjunction with, their usual doctor or healthcare team.
  • GPs should prioritise non-opioid therapies for initial pain management.
  • Opioid medications should only be used for the treatment of acute pain when non-opioid pain medications and therapies have failed or are likely to fail to provide adequate pain relief.

When opioid medications are prescribed for treatment of acute pain, they are often prescribed in addition to paracetamol and non-steroidal anti-inflammatory drugs (NSAIDs)

Evidence statements

Grade

The efficacy of opioid therapy in acute pain is supported by strong evidence from randomised controlled trials (RCTs)7

 

Long-term opioid use often begins with mistreatment of acute pain8

When opioids are used for acute pain, clinicians should prescribe the lowest effective dose of immediate-release opioids and should prescribe no greater quantity than needed for the expected duration of pain severe enough to require opioids8

Strong recommendation, very low quality of evidence

Less than three days of opioid analgesia will often be sufficient for acute pain; more than seven days will rarely be needed8

Strong recommendation, very low quality of evidence

Evidence into practice - Acute pain

Background to managing acute pain in general practice

Acute pain is an unpleasant sensory and emotional experience usually related to surgery, injury or disease. It is associated with actual or potential tissue damage to non-neural tissue and is experienced due to activation of nociceptors. This is also known as nociceptive pain.

Acute pain includes inflammatory pain; that is, pain that occurs in response to tissue injury and the subsequent inflammatory response. Typically, inflammatory pain disappears after resolution of the initial tissue injury. However, in chronic disorders (eg rheumatoid arthritis) the pain may persist for as long as inflammation is active.

Effective management of acute pain requires tailoring treatment to the individual patient; awareness of the science behind contextual and placebo effects; competence with multimodal analgesia (ie the concurrent use of different classes of analgesics); and providing patient reassurance and education, including expected duration of pain episode and warning signs that would require immediate medical attention.

In managing acute pain, GPs should:

  • undertake a complete biopsychosocial assessment of the patient with pain
  • be familiar with the evidence for selected acute pain presentations in general practice and conditions where opioids are not routinely recommended
  • prescribe opioid medications only for the treatment of acute nociceptive pain when non-opioid pain medications and therapies have failed or are likely to fail
  • undertake a patient selection/exclusion process before commencing opioids.

If opioids are commenced for the pain of acute nociception, there is a need to give clear direction about the anticipated duration of therapy. Usually three days or less of opioid therapy will be sufficient for non-traumatic pain not related to major surgery. Typically, opioids should be weaned and ceased as the acute injury heals. Even in complex post-surgical cases this should be within 90 days.5

For a review of all acute pain issues, GPs are advised to review the evidence collated by ANZCA’s Faculty of Pain Medicine.

 

Minor analgesic for pain

GPs should prioritise non-opioid therapies for initial pain management or for the patient being assessed and managed in a general practice. GPs should be familiar with the evidence for selected acute pain presentations in general practice and conditions where opioids are not routinely recommended (Table 1).

In managing acute pain presentations:

  • paracetamol by itself is no longer first-line treatment for most mild to moderate acute pain because of lack of clinical effect9–11 and possible superiority of non-steroidal anti-inflammatory drugs (NSAIDs)12
  • ibuprofen and naproxen are appropriate first-line NSAIDs for mild to moderate acute pain (based on effectiveness,11 adverse effect profile,13 cost and over-the-counter [OTC] availability)
  • non-selective NSAIDs given in addition to paracetamol improve analgesia compared with either medicine given alone, in particular ibuprofen combined with paracetamol14,15
  • paracetamol or NSAIDs combined with codeine (at a dose above 60 mg) provide clinically important pain relief in the immediate term16
  • cyclooxygenase-2 (COX-2) selective NSAIDs are second-line medications for mild to moderate pain based on their similar effectiveness to non-selective NSAIDs.17
Table 1

Table 1

Acute pain conditions where opioid medications are not recommended 18,19

Table 1. Acute pain conditions where opioid medications are not recommended 18,19

Patient selection/exclusion process for opioid  therapy

Opioid medications should only be used for the treatment of acute pain when non-opioid pain medications and therapies have failed or are likely to fail to provide adequate pain relief, or other contraindications exist.

There are several patient groups in which caution is advised before commencing opioid therapy (refer to Table 2). GPs are advised to be familiar with prescribing precautions in these groups. When opioids are used for acute pain, clinicians should prescribe the lowest effective dose of immediate-release opioids and should prescribe no greater quantity than needed for the expected duration of pain severe enough to require opioids.8 The number of doses dispensed should be no more than the number needed (and should not be pro re nata [PRN]). This should be based on the expected duration of pain severe enough to justify prescribing opioids for that condition.

Usually three days or less of opioid therapy will be sufficient for non-traumatic pain not related to major surgery. Continuing requirements for opioid therapy after this time should prompt review. Long-term and problematic opioid use, including sharing with others, often has its origins in the acute pain setting. Clinical discipline is needed to restrict opioid use to time periods and conditions for which it is beneficial.

Table 2

Table 2

Patient groups that require caution when considering opioid therapy20

Table 2. Patient groups that require caution when considering opioid therapy20

Evidence for selected acute pain presentations in general practice

Pain in patients with an existing chronic pain condition

Patients with existing chronic pain sometimes present with acute pain, which is a specific area of pain management. Before initiating care, GPs are strongly advised to be familiar with issues involving:

  • acute exacerbations of existing chronic pain
  • opioid withdrawal presenting as acute pain
  • new painful presentation or diagnosis unrelated to chronic pain.
Acute musculoskeletal pain

For acute back pain, targeted reassurance can result in improved changes in psychological factors such as fear, worry, anxiety, catastrophising and healthcare utilisation.21 Given that most patients with acute or sub-acute low back pain improve over time regardless of treatment, clinicians and patients should select non-pharmacologic treatment with superficial heat (moderate-quality evidence), or massage (low-quality evidence).22

For acute pain resulting from strains, sprains or sports injuries (eg ankle sprain), oral and topical NSAIDs are effective analgesics.23–27

Renal colic

Renal colic is a common presentation in general practice. Similar analgesia may be achieved with intravenous (IV) paracetamol,28 non-selective NSAIDs and opioids.29 Hence IV paracetamol and NSAIDs should be considered as first-line treatments before opioid medication is considered.29–32

Compared to standard therapy, ureteral calculus expulsive therapy using alpha-blockers (eg tamsulosin) reduces the number of pain episodes, the need for analgesic medication and even hospitalisation.33

Biliary colic

Guidelines recommend relief of biliary colic pain may be achieved with NSAIDs.7 These have been found to be better than placebo or spasmolytics and equivalent to opioids.34 NSAIDs may also lower the rate of complications, in particular preventing progression to cholecystitis.34 Opioids are an alternative for patients who cannot take or fail to respond to NSAIDs.35,36

Dental pain

Guidelines recommend dental pain can be appropriately managed with non-opioid medications, or non-opioid combination analgesics.7,19 The combination of paracetamol and NSAIDs is more effective than paracetamol or NSAIDs alone.14 This combination may be a more effective analgesic, with fewer untoward effects, than many of the currently available opioid-containing formulations.37 This is particularly well documented for the combination of paracetamol and ibuprofen in the setting of wisdom tooth removal.15

Primary dysmenorrhoea

Simple analgesics such as paracetamol, aspirin and NSAIDs are effective in most women. These options interrupt the activity of cyclo-oxygenase pathways, thereby inhibiting prostaglandin production. Nonselective NSAIDs are more effective analgesics in dysmenorrhoea than placebo; however, they are associated with an increased rate of adverse effects.38 There appears to be no difference between NSAIDs and paracetamol with regard to efficacy and safety.38 Oral contraceptives are an option for women who wish to avoid pregnancy.

 
Herpes zoster–associated pain

Herpes zoster–associated pain may be severe; early and effective treatment is essential. Analgesia options during herpes zoster include:

  • antiviral agents (particularly famciclovir or valaciclovir) given within 72 hours of rash onset39
  • paracetamol in addition to an opioid such as oxycodone39–41 or tramadol
  • gabapentin – a single dose of 900 mg has been shown to reduce acute pain intensity by 66% (33% for placebo) and also reduced the area and severity of allodynia for up to six hours.42 This was also found with pregabalin (150 mg)43
  • topical lignocaine patches (5%) applied for 12 hours twice daily (on intact skin).44

For prevention of post-herpetic neuralgia, the early administration of antiviral therapy has not been shown to be effective.45 Early intervention with corticosteroids46 or antidepressants47 was also not effective.

 
Tension-type headache

Tension-type headache (TTH) is a common cause of acute pain. Psychological, physical and environmental factors are important in TTH and should be addressed during assessment and treatment. Oral paracetamol (1000 mg), ibuprofen (400 mg) and ketoprofen (25 mg) have similar efficacy.48However, a combination of paracetamol/aspirin and caffeine appears to be superior to paracetamol alone. There is limited low-quality evidence for physiotherapy or other manual interventions.49,50

 
Medication-overuse headache and rebound headaches

Medication-overuse headache (MOH) is one of the most common chronic headache disorders and a public health problem with a worldwide prevalence of 1–2%. It is a condition characterised by chronic headache and overuse of different headache medications. Withdrawal of the overused medication is recognised as the treatment of choice.

Rebound headaches can occur after taking an opioid and during the withdrawal stage. They are common with short-acting opioids (eg codeine).

In both MOH and rebound headaches, aim to use non-opioid and non-pharmacological treatments to wean and cease opioid usage, and manage persistent symptoms with the described strategies. Most patients experience withdrawal symptoms lasting 2–10 days after detoxification. The most common symptom is an initial worsening of the headache, accompanied by various degrees of nausea, vomiting, hypotension, tachycardia, sleep disturbances, restlessness, anxiety and nervousness. 

 
Migraine

Patients who experience mild migraine-related headache and disability may be effectively treated with simple analgesics, either alone or in combination with an antiemetic. Paracetamol,51 aspirin,52 ibuprofen53 and diclofenac54 have all shown benefit. Appropriate antiemetics include (parenteral) metoclopramide55 and prochlorperazine.56,57

Triptans or IV ketorolac58 may be particularly effective in the presence of severe pain and disability where simple analgesia has failed to provide adequate relief in the past.59

Opioids are of limited benefit in the treatment of migraine and are not recommended.60 Opioid use for migraine is associated with more severe headache-related disability, symptomology, comorbidities (depression, anxiety and cardiovascular disease and events), and greater healthcare utilisation.61 However, opioids may be considered as a last resort when other migraine treatments are contraindicated.62

 
Cluster headaches

Cluster headache is a rare primary headache disorder, presenting predominantly in males,63–65 with recurrent acute episodes of brief severe unilateral periorbital pain. Guidelines for the treatment of cluster headache attacks propose first-line treatments to be high-flow oxygen (eg 100% oxygen at 6–12 L/min)66–69 and tryptans (eg sumatriptan 6 mg subcutaneously, zolmitriptan 5 mg and 10 mg intranasally).70,71

 
Temporomandibular dysfunction

Headache or facial pain attributed to the temporomandibular joint is often associated with tension headache72 and cervical spine dysfunction.73 Teeth grinding (bruxism) is a common contributor. The most effective therapies include interocclusal appliance therapy.74 There is limited evidence for the successful pharmacological management of temporomandibular dysfunction (TMD) pain.75 If a medication is going to be used, the best evidence exists for naproxen (1000 mg/day), which is more effective than celecoxib (200 mg/day) and placebo.76

 
 

Key points

  • Chronic non-cancer pain (CNCP) is a collection of clinical conditions that may have involvement of single or multiple pathophysiological mechanisms leading to persistent pain.
  • Outside of end-of-life care, the research on long-term opioid therapy for chronic pain remains limited and insufficient to determine long-term benefits. Available evidence suggests dose-dependent risk of serious harms.
  • Guidelines identify only a selective place for opioids in the management of some chronic pain conditions; opioid use is not routine and is not first-line or second-line therapy. Opioids for CNCP should be reserved for selected patients with moderate or severe pain that significantly affects function or quality of life and that has not responded to other therapies.

Evidence into practice – Chronic pain

Chronic pain has been historically defined as continuous or recurrent pain that persists for an extended period (generally more than three months). However, the biological mechanisms for chronic pain are quite different from those of acute nociception, and should not be considered as ‘unhealed’ acute pain. CNCP is a collection of clinical conditions with involvement of single or multiple pathophysiological mechanisms leading to persistent pain. It is also an individual, multifactorial experience influenced by culture, previous pain events, beliefs, expectations, mood and resilience.

Due to methodological weaknesses of chronic pain studies, interpretation and translation of evidence into practice is difficult. There is limited evidence to determine long-term benefits (outside of end-of-life care); however, there is evidence of risk of harm that increases with dose. While guidelines suggest opioids in the management of some chronic pain conditions, they are not recommended for routine or first-line use. In managing CNCP, GPs should:

  • undertake a complete biopsychosocial assessment of the patient with pain
  • optimise non-drug therapies, and optimise non-opioid therapies as the primary interventions of care.

Opioids for CNCP should be reserved for selected patients with moderate or severe pain that significantly affects function or quality of life and which has not responded to other therapies. If primary interventions fail or are suboptimal, opioid therapies may be considered. GPs should share the decision-making process with the patient, and if opioid therapy is considered, there should be:

  • a patient selection/exclusion process before a therapeutic opioid trial
  • formal care planning based on specific goals and risks
  • an opioid trial, which is undertaken to determine a patient’s response to opioid therapy. This trial includes the selection of an appropriate opioid, formal measures of analgesia and functionality, a trial of dose reduction, and a drug cessation plan if the trial fails
  • an ongoing assessment and evaluation by the accountable prescriber if the trial shows opioid benefit
  • opioid tapering and cessation if suboptimal results or aberrant behaviour occurs.

Long-term use should be uncommon, undertaken with caution and based on consideration of the likely risks and benefits of opioids. Intermittent use is preferable.

GPs should also be aware of chronic pain conditions where there are known clinical complexities involving opioids. These complex clinical areas include the exacerbation or new acute pain in patients on long-term opioid therapy, managing opioids after a non-fatal overdose, and managing the inherited patient.

Some patients on long-term treatment with opioids in CNCP may represent de facto maintenance treatment for iatrogenic opioid dependence. GPs should aim to taper patients taking >100 mg oral morphine equivalent (OME) per day.


Assessment of patients with chronic non-cancer pain

Key point

Management of CNCP should be based on a comprehensive biopsychosocial assessment, a diagnosis, and thoughtful consideration of the likely risks and benefits of any intervention or medication.

The foundation of care for CNCP is a comprehensive biopsychosocial assessment, and a complete documentation of the patient’s pain condition, general medical condition and psychosocial history, psychiatric status, and substance use history.77,78 A biopsychosocial assessment completes a more complex understanding of the patient’s pain perspective. Factors do not always neatly fit into one category; for example, depression may have physical and social aspects as well as being a psychological factor influencing pain.

A comprehensive pain assessment includes a:

  • general assessment and pain specific history (that explores the pain type, severity, functional impact, context, and the patient’s meaning of pain, expectations and fears)
  • physical examination (assessing for signs of tissue damage or disease that might indicate nociceptive and/or neuropathic mechanisms of pain)
  • psychological assessment (the pain experience is affected by mood, stress, coping skills, fear avoidance, and catastrophising).
This will inform the selection of treatment options most likely to be effective. The assessment may need to be repeated reasonably frequently, particularly while establishing a diagnosis and appropriate pain management.


Non-drug therapies for chronic pain

Key point

The basis for good pain management is a strong continuous therapeutic relationship.

Evidence statements

Grade

Non-drug therapy and non-opioid pharmacologic therapy are preferred for chronic pain8

Strong recommendation, low quality of evidence

Before any therapies are undertaken, GPs are advised to consider the science and evidence of contextual and placebo effects in pain management. The manner in which a clinician explains and delivers therapy has an impact on outcome.79 Practitioners who master not only the disease and treatment, but also cultivate a therapeutic relationship may be more effective at pain management.80

The basis for pain management is a strong continuous therapeutic doctor–patient relationship.

Management should optimise general health, including addressing comorbid physical and mental health conditions. GPs should provide adequate patient education about the causes of chronic pain. Further information regarding neuroscience education can be found here.

GPs should engage in a frank discussion and negotiation about different pain strategies and the goals of treatment. These goals should go beyond pain relief alone (eg improvement in physical, social, emotional and mental functioning, including an increase in activity) and there should be an agreement that if the goals are not met, then the treatment will be discontinued.

Non-drug and non-opioid pharmacological therapies are preferred options for management of CNCP.8 Multimodal interventions are effective.81 Exercise,82–86 selective psychological interventions,87,88 social interventions, and nonopioid therapies are all advocated.

Most patients with chronic pain are physically deconditioned from inactivity.89 Movement and exercise therapies, regardless of their form, are recommended in the management of patients with chronic pain.86 Physiotherapists and exercise physiologists with an interest in chronic pain are advocated. Further information on the effectiveness of movement and exercise interventions can be found here.

Psychological approaches aim to increase self-management, behavioural change, and cognitive change rather than directly eliminate the locus of pain. Psychologist intervention is advocated for assessment and management of selected presentations. Further information on the effectiveness of psychological interventions can be found here.

Box 1. Handbook of non-drug interventions

The RACGP’s Handbook of non-drug interventions (HANDI) provides multiple non-drug therapies for various pain conditions such as osteoarthritis, back pain and foot pain.
For an explanation of exercise and reconditioning, refer to ‘Graded exercise therapy for chronic fatigue syndrome’.

Non-opioid medication for chronic pain

Effective management requires knowledge of the frameworks for pain, different treatment approaches, and an individualised multifaceted intervention plan for the patient5,78,86,89–93 depending on specific contributors of pain.

Where analgesics are considered appropriate, the optimal approach to manage chronic pain is without opioids, prioritising use of minor analgesics and analgesic adjuvants (eg antidepressants, anticonvulsants) instead.5,8 ‘Adjuvants’ refers to medications that are co-administered with analgesics to enhance pain relief via other pathways. The evidence for effectiveness of adjuvants varies for pains with different nociceptive, neuropathic and nociplastic origins. A brief overview of the evidence can be found here.

In countries where they are available, cannabinoids may have a place in relieving chronic neuropathic pain.94,95

However, the evidence for effectiveness is considered weak and their role is being debated in Australia.96,97

GPs should prescribe one analgesic/adjuvant drug at a time and monitor individual responses. While it is accepted practice to run a clinical trial over several weeks, many of the agents will elicit an analgesic response within the first two weeks of treatment (if achieved, the response tends to last).98–100 If functionally meaningful benefit does not ensue, stop the drug and try an alternative. Because of individual variation in pain, patients may respond to one drug within a class, but not to others (eg a patient may respond to naproxen, but not ibuprofen). Therapy failure due to inadequate relief or side effects should not stop doctors from trialling alternative medications from the same class (eg NSAIDs, anti-epileptics). Combination adjuvants (anticonvulsants and antidepressants) are common but inadequately studied.

For patients who achieve clinically meaningful analgesia, use the lowest individualised effective dose to minimise adverse effects. Reassess regularly (eg every two weeks), and trial medication withdrawal to determine that the response is due to the drug and not natural history. It is recognised that patients treated with a placebo can also improve over time.100


Patient selection/exclusion process before an opioid trial

Key point

Long-term opioid therapy is dependent on an appropriate patient selection process, considered care planning, and an ‘opioid trial’ to determine responsiveness to opioid treatment.

Evidence statements

Grade

GPs should review the patient’s history of controlled substance prescriptions using state prescription drug monitoring program (PDMP) data to determine whether the patient is receiving opioid dosages or dangerous combinations that put him or her at high risk for overdose

Strong recommendation, very low quality of evidence

Before starting opioid therapy, GPs should evaluate risk factors for opioid-related harms8

Strong recommendation, very low quality of evidence

If alternatives to opioid treatment fail, have limited benefit or are inappropriate, a supervised opioid trial may be initiated. Opioids for CNCP should be reserved for select patients with moderate or severe pain that significantly affects function or quality of life and that has not responded to other therapies.

Prescription of opioids, as with any treatment, should be based on a comprehensive medical assessment; a diagnosis; thoughtful consideration of the likely risks and benefits of any medication, as well as alternative interventions; and a management plan derived through SDM and continual clinical monitoring.

In selecting or excluding patients before starting opioid therapy, GPs should:

  • be aware of the limitations of opioids in managing some pain syndromes (eg functional visceral pain, such as irritable bowel syndrome and bladder pain syndrome, is not sensitive to opioids)
  • be aware of patient groups or contexts requiring additional caution or exclusion. Additional advice needs to be considered when patient selection for opioid therapy involves pregnancy, workers’ compensation injuries, patients who drive, patients with sleep apnoea and disordered breathing, patients aged 65 or older, patients with renal disease, patients with hepatic disease, culturally and linguistically diverse patients, and patients with mental health conditions
  • evaluate risk factors for opioid-related harms in individual patients, including review of the patient’s history of controlled substance prescriptions using the Prescription Shopping Programme (PSP) or state PDMP data to determine whether the patient is receiving opioid dosages or dangerous combinations (especially benzodiazepines101) that put him or her at high risk for overdose
  • avoid prescribing opioids to patients with polydrug use, comorbid alcohol or substance use disorders. GPs should consider tapering benzodiazepines and seeking specialist opinion or a specialised pain management facility90 in the management of these patients.


Care planning for an opioid trial

Key points

  • Long-term opioid therapy is dependent on an appropriate patient selection process, considered care planning, and an ‘opioid trial’ to determine responsiveness to opioid treatment.
  • A treatment plan is discussed including a plan to discontinue opioids if there is no objective functional improvement.

The aim of an opioid trial is to discover the individual’s responsiveness to opioid therapy in terms of decreased pain, increased function and improved quality of life. Before starting opioid therapy for chronic pain, GPs should:

  • ensure patients are aware of known risks and realistic benefits of opioid therapy, and of their responsibilities for managing therapy (including risks to other individuals if opioids are intentionally or unintentionally shared) • establish treatment goals with all patients, including realistic goals for pain and function, and consider how opioid therapy will be discontinued if goals are not met or benefits do not outweigh risks8,92
  • consider a UDT to assess for prescribed medications as well as other controlled prescription drugs and illicit drugs.

Care plans should incorporate strategies to mitigate risk including having only one prescriber for ongoing analgesics;5,91 offering naloxone when factors that increase risk for opioid overdose are present (eg history of overdose or substance use disorder, higher opioid dosages or concurrent benzodiazepine use); and sharing care for high-risk patients with clinical specialists.8

While screening for opioid risk, treatment agreements89 and urine testing5,78,92,93,102 are often advocated, they have not been shown to reduce overall rates of opioid prescribing, misuse, or overdose.5,103 Patient treatment agreements do serve an administrative role, clarify expectations and behaviour standards, and have educational purposes.104,105 GPs should use their discretion when considering candidates for a urine drug screen (UDS) and treatment agreements.

It is important to have an ‘exit strategy’ if the trial fails to achieve agreed outcomes. The strategy should preserve the therapeutic relationship while also managing possible misuse of opioids.

Box 2. Naloxone

Discussion about take-home naloxone (THN) as part of an overdose response plan is an effective brief intervention. The use of naloxone fits within both harm reduction strategies and patient-centred care. Naloxone is safe, effective, inexpensive, and relatively easy to administer via intramuscular (IM) injection.106–108

Naloxone injection is dual listed as a Schedule 4 medicine subsided by the Pharmaceutical Benefits Scheme (PBS), and as a Schedule 3 medicine available from a pharmacist. It is beneficial for GPs or pharmacists to provide patients with brief instructions on how to prevent, recognise and respond to overdose including how to store, carry and administer naloxone. It may be appropriate to involve the patient’s family and/or other potential overdose witnesses relevant to the patient, as these are the people most likely to be responding to an overdose. GPs should also advise patients that naloxone can be obtained over-the-counter (OTC) from local pharmacies by a third party to protect co-residents (eg children) of the patient from intentional or unintentional overdose.

Prescribers are encouraged to provide a prescription for THN for patients at high-risk of overdose (eg high opioid dose, complex care, or recently released from a controlled environment). For further resources on naloxone therapy, refer to the Cope Australia website.

Box 3. Care planning tool

All patients prescribed longer-term opioids for CNCP should have a care plan, and one principal prescribing doctor nominated. The patient should be managed primarily by the one GP. When the principal GP is absent, another GP should be designated to manage the patient in accordance with the care plan. 

A simple tool for care planning in chronic pain is available at the NSW Pain Management Network website.

Care planning for high-risk patients

For GPs with a specific interest in this area, guidance for planning and use of care plans in high-risk patients has been developed by the NSW Department of Health for both opioid dependent patients and patients with severe comorbidity.

Undertaking an opioid trial

Key point

An opioid trial is undertaken to discover the individual’s responsiveness to opioid therapy in terms of decreased pain, increased function and improved quality of life.

Opioids for CNCP should be reserved for select patients with moderate or severe pain that significantly affects function or quality of life and that has not responded to other therapies. An opioid trial should occur in conjunction with formal measures of analgesia and functionality. Validated brief assessment tools that measure pain and function (eg Pain, Enjoyment, General activity [PEG]109 in Appendix E) or other validated assessment tools may be helpful and time effective.

When trialling opioids for chronic pain, opioid selection should be based on evidence of safety and benefit for the pain type and patient factors. The opioid should be in addition to appropriate non-drug therapy and non-opioid drug therapies.

Unless clinically contraindicated, consider selecting an effective opioid with less potential for harm such as oral tapentadol, transdermal buprenorphine or oral tramadol.8,78 Start opioids at a low dosage, increase gradually (refer to individual opioids) and monitor opioid effectiveness using the 5As of chronic pain management and the PEG tool until optimal dose is attained.8,78,101

Patients likely to benefit from opioids in the long term will demonstrate a favourable response within 2–4 weeks of initiating therapy. It is accepted practice to run a clinical trial over 8–12 weeks to allow for normal variations of life. An important part of any trial is to reduce opioid doses to establish minimal needs.

Patients who do not experience clinically meaningful pain relief early in treatment (ie within three months) are unlikely to experience pain relief with longer-term use.110 Suboptimal therapeutic response may be due to failure to recognise psychosocial aspects of the patient’s pain.

If the initial choice of opioid is ineffective, or if adverse effects are unacceptable, opioid rotation may be tried. In clinical practice, opioid rotation must be performed with consideration of individual patient characteristics, comorbidities (eg concurrent psychiatric, pulmonary, renal, or hepatic illness), and concurrent medications.111 Reduce the starting dose of the new opioid by 50% of the calculated equianalgesic dose of the first opioid. This reduction in dose may result in improved tolerability while incomplete cross-tolerance may result in equivalent efficacy.

To minimise risks, opioids should be prescribed at the lowest effective dose for the shortest clinical timeframe. Caution should be used when increasing dose from low to moderate (ie above 40–50 mg per day OME).8 While there is no international consensus on a maximum (ceiling) dose,5,112 a ceiling of 100 mg per day OME is recommended for Australian GP prescribing.5,8,92 Above this dose, specialist review is advocated.92

 

Ongoing therapy, assessment and monitoring

Key points

  • Long-term opioid therapy requires ongoing structured monitoring and review of benefits and harms.
  • GPs should taper and discontinue opioids in the absence of functional improvement, when planned care fails, or aberrant behaviours become apparent.

Long-term opioid use should be uncommon, undertaken with caution and based on thoughtful consideration of the likely risks and benefits of opioids. If opioids provide benefit (pain relief and improved functioning), opioid analgesia can be an acceptable long-term therapeutic option.

GPs should continually evaluate treatment efficacy and harms, and monitor for known comorbidities. Patients should be re-evaluated at least every three months, and within four weeks of any dose escalation.8

Although the evidence for use of instruments to assess patient-reported safety, efficacy, or misuse of current opioid therapy for chronic pain is lacking,113 it is advised that patients are evaluated using the 5As of chronic pain management.8,114 Only continue opioid therapy if there is documented clinically meaningful improvement in pain and function that outweighs risks to patient safety.5,8,78,101

While baseline UDTs are recommended,5,78,92,93,102 prescribers can use discretion for ongoing testing according to patient risk. Consider UDTs at initiation and then at least annually to assess for prescribed medications as well as other controlled prescription drugs and illicit drugs.8,92 When using UDTs be aware of benefits and limitations, appropriate test ordering and interpretation, and have a plan to use results.92

In all states except NSW, GPs should also seek state regulatory authorisation for any patient who has been prescribed Schedule 8 (S8) opioids for two months or longer.115 In NSW, only certain oral opioids (buprenorphine, hydromorphone and methadone) require an authority.

Pharmaceutical Benefits Scheme (PBS) regulations dictate an independent opioid prescribing review at 12 months of therapy. The RACGP recommends a structured review using a checklist for this purpose.

Long-term opioid prescriptions should be at the lowest effective dose, and regular attempts at reduction should be scheduled.103 Intermittent use has also been advocated.103 Proper management of opioids in well-selected patients with no history of SUD can lead to long-term (≥26 weeks) pain relief for about 25% of patients.116,117 Continued professional monitoring of health outcomes is required.

Where continued opioid therapy is inappropriate due to failure of benefits to outweigh harms, or if there is evidence of problematic use (eg rapid dose escalation), work with patients to taper and discontinue opioids.8,90,101 This involves preserving the therapeutic relationship, optimising non-opioid therapies, and working with patients to taper opioids to lower dosages or to discontinue opioids.5,90 Patients have the right to respectful care that promotes their dignity, privacy and safety; abruptly discontinuing opioid treatment without offering a feasible plan for tapering treatment could be considered a failure of duty of care.

GPs should regularly assess whether the patient satisfies individual state health legal definitions of drug dependence and seek advice from the state regulatory authorities if issues arise. All states and territories run drug and alcohol advisory services, which offer information, advice, referral, intake, assessment and support 24 hours a day. They offer services for individuals, their family and friends, GPs, and other health professionals.

Chronic post-surgical pain

Key points

  • The prevention of the progression of acute to chronic pain should be the aim of all practitioners whenever they treat an acutely painful condition.
  • Both surgical and patient factors influence the development of chronic post-surgical pain (CPSP).
  • CPSP may have biomedical components – GPs need to understand the role of different analgesics and adjuvants in different pain processes.

Management of chronic post-surgical pain

CPSP is defined as pain developing and persisting beyond the time expected for the normal healing process (ie persisting for at least two months). CPSP can affect 15–60% of all surgical patients.118 The incidence of CPSP varies with the type of operation and it is more common where nerve trauma is inevitable (eg amputation) or where the surgical field is richly innervated (eg chest wall).119–121 For example, following:

  • inguinal hernia repair or caesarean section, 10% of patients report ongoing pain and disabilities six months after surgery
  • mastectomy or lumpectomy, 20–30% of women report chronic pain
  • amputation, thoracotomy or coronary bypass surgery, 30–50% of patients report persistent pain.122

Depending on the type of surgery, CPSP is often neuropathic pain (on average one-third of cases; range from 6% to 54%). Pain involving such a neuropathic component is usually more severe than nociceptive pain and often affects the quality of life more adversely.123

Nociceptive and neuropathic processes in the periphery (including nerve injury) and neuroplastic processes (leading to peripheral and central sensitisation) are all implicated in the transition from acute to chronic pain. The relative degree of ongoing inflammation or intraoperative nerve injury resulting in peripheral and central sensitisation may explain the variation in risk and, to an extent, the characteristics of CPSP for different operations. In addition to the type of surgery, the development of CPSP is influenced by patient factors including:

  • psychological factors such as anxiety, depression, stress, fear of surgery, hypervigilance and pain catastrophising124,125
  • demographic factors, such as younger age for adults and female gender
  • genetic and epigenetic factors, which influence both the sensitivity of individuals to analgesics and their risk of CPSP.126,127

All practitioners should aim to prevent the progression of acute to chronic pain. Treatments that address nociceptive and neuropathic pain should be considered, including opioids where appropriate.

Pregabalin and gabapentin may have a role in preventing CPSP due to their effect on neuropathic pain. However, considerable uncertainty exists regarding efficacy with limited and contradictory evidence.7

Chronic visceral pain

Visceral pain refers to pain in the trunk and abdominal areas of the body that includes the heart, lungs, abdominal and pelvic organs. Examples of chronic visceral pain include chronic chest pain, chronic pancreatitis, and chronic pelvic pain.

Visceral pain is diffused and can be difficult to localise.128 It may be accompanied by nausea, vomiting and changes in vital signs. Treatment goals are both causal and symptomatic and involve holistic strategies including patient education.129

Fibromyalgia

Fibromyalgia is a common and potentially disabling condition affecting 2% of the population in developed countries, predominantly young to middle-aged women.130 Fibromyalgia has a varied and fluctuating clinical spectrum. Cardinal features include widespread musculoskeletal pain and tenderness; poor quality, unrefreshing sleep; significant levels of fatigue; cognitive disturbances, particularly problems with concentration and memory; and high distress levels.131

Fibromyalgia pain is considered to be due to central sensitisation.132 Exercise remains the strongest therapeutic recommendation.133 Specific guideline recommendations should be consulted prior to therapies.134–137

Complex regional pain syndromes

Complex regional pain syndrome (CRPS) is a debilitating, painful condition in a limb associated with sensory, motor, autonomic, skin and bone abnormalities. Pain is typically the leading symptom, but it is often associated with limb dysfunction and psychological distress.138 There is usually a history of precipitating event or injury; however, the severity of the clinical pain presentation is often disproportionate to the severity of the inciting event.

Exact mechanisms for the pathogenesis of CRPS are not understood.139–143 A combination of elements including inflammation, dysfunction within sympathetic and somatosensory nervous system, and cortical (not psychological) factors are thought to contribute to the generation and perpetuation of symptoms.138 There is no evidence that paracetamol assists in CRPS, and insufficient evidence regarding the efficacy of NSAIDs and opioids. Although there is no evidence supporting the long-term effectiveness of anticonvulsants for CRPS, these agents may be useful in providing pain relief in the earlier stage of the disease.144

GPs should seek specialist advice with presentations with CRPS.

Key point

GPs should use caution in patients presenting with acute exacerbations of chronic pain.

Acute exacerbation of existing chronic pain

If patients experience an acute exacerbation of CNCP, the aetiology of the pain must be identified rather than just treating it as an acute nociceptive event. The aetiology may be multifactorial, and it is advised that the patient’s mental health status and social situation should be formally assessed to determine if additional resources may be appropriate. It may also be appropriate to check the PDMP and patient-controlled electronic medical record (if present) for history of opioid prescriptions.

The treatment for the chronic pain patient may be significantly different from the acute episode. Exacerbations of pain should be managed with non-opioid therapy: physical therapies and NSAIDs should be used for periodic flareups of mild to moderate inflammatory or non-neuropathic pain.19,89 Because of potential risks and adverse effects, clinicians are encouraged to avoid prescribing increased dosage or additional opioids.19

New painful condition in a patient with chronic pain

As with acute exacerbation of chronic pain, in any new pain presentation it is important to identify the source of pain rather than just treating for an acute nociceptive event, avoid prescribing increased dosage or additional opioids,19 use non-opioid therapy as appropriate,89 and assess the patient’s mental health status and social situation to determine if additional resources are required.

If a new presentation does dictate opioid treatment, consider monitoring in an appropriate care setting. Dosing opioids for acute pain in a patient already taking opioids is problematic. The patient may require a higher dose to achieve the same analgesic effect. The higher dose puts the patient at greater risk for an adverse event. Note that patients already taking opioids do not require a longer than normal course of treatment for acute pain. Once the acute event has passed, reduce opioids to maintenance levels.

Opioid withdrawal presenting as acute pain

Consider opioid withdrawal when evaluating opioid-tolerant patients who present with acute pain complaints or gastrointestinal symptoms. Opioid withdrawal can occur when patients have stopped, lost or overused their medications. Patients are often reluctant to share this information with their clinician.

Opioid withdrawal generally presents with anxiety 12 hours after the last dose, and becomes physically detectable within 24 hours after the last use of short-acting opioids and 48 hours after last use of long-acting opioids.

Unless the patient is otherwise medically unstable, withdrawal is not life threatening. However, it may be very distressing and necessitate reassurance and comfort measures. All states and territories run an alcohol and drug information service, which offers information, advice, referral, intake, assessment and support 24 hours a day.

Managing opioid therapy after non-fatal overdose

Patients who have had a presentation or admission for opioid accidental or non-accidental overdose are at significant risk for another overdose and further harms.145

At two years, the cumulative incidence of repeated overdose was:145

  • 17% (95% confidence interval [CI]: 14%, 20%) for patients receiving high dosages of opioids after the index overdose
  • 15% (CI: 10%, 21%) for those receiving moderate dosages
  • 9% (CI: 6%, 14%) for those receiving low dosages
  • 8% (CI: 6%, 11%) for those receiving no opioids.

Opioid discontinuation after overdose is associated with lower risk for repeated overdose.145

Non-fatal opioid overdose is an opportunity to identify and treat substance use disorders, pain and mental health issues.

All patients admitted or presenting to hospital emergency departments (EDs) with non-fatal opioid overdose must have a full pain and psychiatric evaluation prior to discharge, and consideration of opioid cessation or provision of naloxone for peer or family member administration. A clear plan for opioid safety after discharge and communication with usual treating practitioners in the community is essential.

Naloxone distribution programs are firmly rooted in the principles of harm reduction. Naloxone is safe, effective, inexpensive, and relatively easy to administer via intramuscular (IM) injection.106

The inherited patient – Continuation of long-term opioid management plans initiated by other healthcare providers

Key points

  • Patients with chronic pain and/or SUDs have the same entitlement to respectful and appropriate care as other patients.
  • Referral according to clinical handover standards should be arranged by the original authorised prescriber.
  • Clinical handover in primary care takes time. Adequate prescriptions from the original prescriber should cover this handover period.
  • All new patients ‘inherited’ by the practice should receive a comprehensive re-evaluation and renewed care planning.
  • Patient care strategies must be defensible, rational, and compassionate. Doctors should prescribe opioids according to their best clinical judgement, with individual risk benefit analysis for each patient.
  • Before opioid prescriptions are written, relevant state authority must be obtained.
  • In the event of an SUD being detected, the management plan should also include remedial programs or referral to appropriate drug misuse agencies.
  • Patients who satisfy practice appropriateness criteria and are accepted should be prescribed ongoing medication under the continued care of a single doctor.

Overview

Patients often arrive from other practices or institutions requesting continuation of their long-term opioid management programs. These situations can produce a therapeutic dilemma for the new practitioner. Patients’ histories and management regimes may be quite complex, and some practices and institutions have prescribing practices that are variable or may not be evidence based or safe.

Patient requests for ongoing pain management may be quite legitimate, but, unfortunately, this type of presentation is also common for drug-seeking behaviour for opioids. In both situations, the patient may be physically dependent on opioids. When these patients are deprived of opioid medication, they may experience acute withdrawal.

In the inherited-patient situation, caution is advised while acknowledging the patient’s right to respectful care that promotes their dignity, privacy and safety. Patients with legitimate clinical indications for long-term opioid therapy have expressed concern regarding stigmatisation and lack of access to ongoing care. It is important GPs independently make a thorough clinical assessment of each patient’s opioid use, and develop an individual management treatment plan consistent with clinical guidelines while safeguarding against abuse and diversion. Treatment should seek to maximise health outcomes across a range of domains (eg patient pain acceptance, functionality, risk).

In the event of an SUD being detected, this plan should also include remedial programs or referral to appropriate drug misuse agencies. Practice policies should be in place to assist the management of the inherited patient using drugs of dependence.


Comprehensive clinical assessment of the inherited patient

The assessment of an inherited patient with an existing long-term opioid treatment plan includes:

  • a review of the patient’s general health conditions, including any SUD (eg alcohol, benzodiazepines, opioids)
  • a full social and psychological assessment
  • confirmation of the pain diagnosis, which may require a formal in-depth review of the pain diagnosis including radiology, response to therapeutic interventions, and current functionality
  • establishing the medical necessity and the appropriateness of prescribing regimen
    • assess the risk–benefit ratio – some existing plans may have evidence-based multimodal therapy; others may have quite high-risk or inappropriate regimes with mixed and multiple opioids, multiple psychoactive drugs or drugs that are contraindicated or not consistent with regulations
    • confirm that opioid drugs and doses are correct (ie calculate the patient’s daily morphine equivalent dose for all opioids prescribed to determine whether these dosages are safe or at increased risk)
    • determine pain and function with this level of opioids and other medications and ask ‘Is this patient at risk if I maintain the same prescription?’
  • deciding whether the regimen represents rational accountable prescribing, is outside the scope of practice or the doctor, or needs optimisation
  • assessing aberrant behaviours using the 5As approach
    • tolerance and physical dependence are normal consequences of sustained use of opioid analgesics and are not the same as addiction
    • patients treated for ‘legitimate’ pain may become physically dependent on the supervised therapy of opioid medications, but this does not constitute a substance use disorder.
  • contacting prescription shopping services or real-time prescription monitoring services.

Prescribers should verify patient-reported treatment plans and any identified concerns about misuse or dependency with the previous prescriber and pharmacist directly.

Box 4.

Triaging the inherited patient An assessment of an inherited patient will triage patients into one of three groups:

  • Patients who have reasonable function and are being managed on a course of therapy that is both reasonable and appropriate for the diagnosis
  • Patients who have been managed in a fashion that is not totally consistent with the new clinician’s experience and resources, and may reflect a clinical picture that can be optimised
  • Patients whose course of therapy is (for a variety of reasons), not evidence based, or presents significant risk to the patient and others, and so is not something the new clinician feels able to support

Assessment of risk with the inherited patient

Even when a case for ongoing care is established, the general risk category of the patient should be considered and GPs are not obliged to take full responsibility for care of all patients. For each patient, the risk category is dependent on patient factors, practitioner factors and resources.

Box 5.

Risk categories

Patients may be placed into one of three risk categories:

  • Group I (low risk) includes patients who the clinician feels able to confidently manage on their own
  • Group II (medium risk) includes patients who the clinician feels able to co-manage with speciality support
  • Group III (high risk) includes patients who the clinician feels are best referred to speciality clinics

Higher-risk situations also include those where:

  • staff safety may be impacted
  • care is outside the expertise of the practitioner
  • long-term health prospects of patients are being compromised by lack of access to state or territory facilities
  • patients have serious mental illness or are taking antipsychotic medication
  • patients have past family or personal history of substance misuse
  • polydrug use is present
  • patients have been recently discharged from a correctional services facility.

For more information about deciding when to seek advice or consider referral to a psychiatrist or pain/addiction specialist refer to Prescribing drugs of dependence in general practice, Part A.

Care planning for the inherited patient
Patients who satisfy practice appropriateness criteria and are accepted should be prescribed ongoing medication under the continued care of a single doctor. This doctor needs to:
  • communicate empathically but honestly with patients about opioid use (including realistic expectations about the likely or potential outcomes of their treatment)
  • develop a defensible, rational, and compassionate patient care strategy
  • obtain relevant permits to prescribe opioids from the state or territory authorities.

Patients need to be informed about the purpose, importance, benefits and risks of their medicines. This enables them to engage in SDM, which has been shown to build trust, prevent harm and reduce surprise and distress if complications or adverse events occur.

Not all SDM will result in agreement. It is challenging when there is pharmacologic or other therapeutic disagreement with a patient. But there is a difference between patient-centred care and patient-controlled care. Acknowledging patient beliefs, expectations and preferences does not involve crossing professional boundaries, ignoring laws or continuing therapy that is considered detrimental to the patient’s health.

In this situation, it is necessary to maintain professional discipline and clinical honesty. If a patient refuses the advice of a GP, they should be advised about the implications of deciding not to receive the healthcare offered. The patient should be given sufficient time to consider and clarify any information in order to make an informed decision, taking into account the context of the clinical situation.146

Where GPs suspect an opioid use disorder, they should be honest with the patient while reassuring them that effective treatments are available. Scripting sentences and techniques for GPs are available to assist in politely refusing to provide opioid prescriptions. Appropriate management includes the initiation of remedial programs or referral to appropriate drug misuse centres. In some states such as Victoria, South Australia (and possibly New South Wales in 2018) GPs are permitted to prescribe opioid replacement therapy (ORT). Advice from state-based drug and alcohol services is available at all times.

GPs should not evict patients from their practice because of manipulative behaviour or therapy refusal.

 

Using legislation to assist with challenging patients

Clinicians should not see legislation covering the prescription of controlled substances as a barrier to appropriate prescribing. Instead, legislation acts as protection for prescribers as well as for patients.

When patients make demands for opioids, practitioners can strongly state their duty to act within state, territory and national legislative frameworks, and to manage their prescribing practices within the laws and clinical and professional standards.

Practitioners should determine who is the patient’s authorised prescriber. If the prescriber is not in the state or territory where the consultation is being made, new authorisation will need to be obtained. Time for the necessary processes will need to allowed.

 

Using opioid rotation and rationalisation

When opioids are less effective than expected or a new patient presents with ‘irrational pharmacotherapy’, then opioid rotation, consolidation (eg using one type, longer acting) or taper (in some cases, to the point of discontinuation) should be considered.

Opioid rotation may help to:

  • improve analgesia
  • lower the dose of the prescribed medication
  • manage loss of analgesic efficacy or the presence of toxicity.147

Note that clinically, patients often show incomplete cross-tolerance when rotated from one analgesic to another.

 

Use urine drug testing

A baseline UDT should be performed at the initial visit, with a request to include detection of oxycodone and other drugs not usually recognised by immunoassay.

Further testing is used as clinically indicated. Unexpected results from such tests should be interpreted within their limitations: fentanyl, buprenorphine, synthetic drugs, anabolic steroids, and usually oxycodone are not routinely detected and must be requested as additional tests (at extra cost to the patient).

Note that drug misusers may adopt a variety of methods, such as switching urine samples, to influence results.148

 

All state and territory health authorities have 24-hour telephone access to assist with drug and alcohol queries.

GPs can get urgent advice and support for patients on opioid therapy, and seek information on referral options or advice on issues on care coordination between multiple providers.
 

Key points

  • Long-term treatment with opioids in CNCP may represent de facto maintenance treatment for iatrogenic opioid dependence.
  • GPs should follow an ‘exit strategy’ for dealing with failure to achieve agreed outcomes of opioid treatment.
  • GPs can effectively wean their patients from opioids if there is no benefit, or if risks outweigh benefits.
  • Lack of improvement, intolerable side effects and abnormal behaviour are signs of opioid trial failure and indicate the need to taper and/or discontinue opioids.
  • GPs should be familiar with opioid reduction and withdrawal processes.

Background to opioid tapering

It is not unusual for opioid medication to become ineffective over time, or for adverse effects to develop. Where opioid trials fail to achieve agreed outcomes or harms outweigh benefits, GPs should follow their ‘exit strategy’ to taper and/or cease opioids while preserving the therapeutic relationship.

In other cases, long-term treatment with opioids in CNCP may represent de facto maintenance treatment for iatrogenic opioid dependence. Tapering should be attempted for all patients who have been on long-term opioid therapy of greater than 100 mg OME per day. Reduction in opioid dose may reduce adverse effects including cognitive impairment and the likelihood of non-fatal or fatal unintentional overdose.

In all cases, patients have the right to respectful care that promotes their dignity, privacy and safety. Ending a relationship with a patient or abruptly discontinuing opioid treatment without offering a feasible plan for tapering or addiction treatment could be considered an abrogation of the accountable prescriber’s duty. Such approaches rarely solve the problem.

Patients can experience significant improvement in pain severity, functioning, and mood when their opioid therapy is tapered to a lower, safer dose, or ceased. At present there is very little research available on comparative effectiveness of interventions for opioid withdrawal in CNCP.150

GPs should regularly assess whether the patient satisfies the legal definition of drug dependence. If the patient has an opioid use disorder, opioid therapy for pain should be discontinued and the addiction professionally, sympathetically and appropriately treated. Seek authority from the state regulatory authorities when treating these patients.

Box 6.

Educational videos about opioid cessation

Brainman pain management resources, developed by the Hunter Integrated Pain Service at John Hunter Hospital and Hunter Medicare Local,
Understanding pain: Brainman stops his opioids

Managing opioid tapering or discontinuation

Before attempting opioid discontinuation, GPs should be familiar with opioid reduction and withdrawal processes and patient experiences.5,90 Some patients will experience stable or improved pain after an opioid taper.151–153 However, short-term withdrawal can lead to transitory increased pain and hyperalgesia.154–156 For patients with long-term opioid treatment for CNCP:

  • there is no published comparison of speed of tapers157
  • there is no evidence to support switching to buprenorphine or methadone for tapering157
  • key predictors of opioid tapering dropout or relapse are depression, high pain scores and high opioid doses157 (there is not yet any research to support addressing these factors through pharmacologic and psychological interventions). These patients are also those at higher risk of opioid-related adverse events and tapering of the dose should be strongly considered
  • the use of α2-adrenergic agonists (eg clonidine) can reduce symptoms of withdrawal via sympathetic activity.

Where there is no evidence of substance use disorder

If weaning is required after a shorter period of opioid therapy, such as after failure to achieve the goals of an opioid trial, or after a negotiated treatment phase for acute pain, then a faster rate of weaning is generally appropriate. One option is a stepwise reduction of the daily opioid dose each week by 10–25% of the starting dose.5

If weaning is required in response to significant adverse effects or opioid misuse, then daily stepwise reduction may be more appropriate. Alternatively, immediate opioid cessation and pharmacological treatment of withdrawal symptoms can be considered.

Otherwise, a decrease of 10% of the original dose every 5–7 days until 30% of the original dose is reached, followed by a weekly decrease by 10% of the remaining dose, rarely precipitates withdrawal symptoms and facilitates adherence.157


Where there are complex patient comorbidities

Discontinuing opioid therapy is often hindered by patients’ psychiatric comorbidities and poor coping skills, as well as the lack of formal guidelines for the prescribers.

If a previous attempt at opioid weaning has proven unsuccessful, then the rate of tapering can be slowed. This can be achieved by reducing the size of the dose reduction each month and/or by increasing the time spent at each dose level (eg two or three months between reductions).


Where there is evidence of substance use disorder

Importantly, the legislative requirements for prescribing S8 drugs vary depending on the person’s dependence, but all are consistent for patients with respect to SUD: S8 medications (most opioids, alprazolam and flunitrazepam) cannot be prescribed without a permit or an appropriate approval from the relevant state or territory health department’s pharmaceutical services unit.

In some cases, it may become apparent during weaning that the primary problem is opioid dependency rather than pain. For patients with opioid use disorder, GPs should offer or arrange evidence-based treatment (usually medication-assisted treatment with buprenorphine-naloxone or methadone in combination with behavioural therapies). Referral to an addiction specialist is advised.5,8

Monitoring the taper

The goal of tapering is to improve or maintain patient wellbeing while opioids are being withdrawn. Schedule frequent visits and ask about and emphasise the benefits of taper (eg improved pain, mood, alertness) at each appointment. Referral for counselling or other support during the taper is recommended, especially if there are significant behavioural issues. If a patient is not successfully reducing their dose, or there is an escalation in dose beyond prescription, involve other practitioners.

Managing withdrawal symptoms

Symptoms of withdrawal, such as nausea, diarrhoea, muscle pain and myoclonus can be managed with clonidine 0.1–0.2 mg orally every six hours, or simple supportive therapy. Symptoms of mild opioid withdrawal may persist for six months after opioids have been discontinued. However, do not treat withdrawal symptoms with opioids or benzodiazepines after discontinuing opioids.

Rapid recurrence of tolerance can occur from months to years after prior chronic use.
 
  1. Cohen M, Quintner J, Buchanan D. Is chronic pain a disease? Pain Med 2013;14(9):1284–88.
  2. Upshur CC, Luckmann RS, Savageau JA. Primary care provider concerns about management of chronic pain in community clinic populations. J Gen Intern Med 2006;21(6):652–55.
  3. Henderson JV, Harrison CM, Britt HC, Bayram CF, Miller GC. Prevalence, causes, severity, impact, and management of chronic pain in Australian general practice patients. Pain Med 2013;14(9):1346–61.
  4. O’Rorke JE, Chen I, Genao I, Panda M, Cykert S. Physicians’ comfort in caring for patients with chronic nonmalignant pain. Am J Med Sci 2007;333(2):93–100.
  5. Australian and New Zealand College of Anaesthetists. Recommendations regarding the use of opioid analgesics in patients with chronic non-cancer pain. Melbourne: ANZCA, 2015 PM1-2010.pdf [Accessed 19 July 2017].
  6. Guyatt GH, Oxman AD, Vist GE, et al. GRADE: An emerging consensus on rating quality of evidence and strength of recommendations. BMJ 2008;336(7650):924–26.
  7. Schug S, Palmer G, Scott D, et al. Acute pain management: Scientific evidence. 4th edn. Melbourne: ANZCA, 2015 Documents/APMSE4_2015_Final [Accessed 19 July 2017].
  8. Dowell D, Haegerich TM, Chou R. CDC guideline for prescribing opioids for chronic pain – United States, 2016. JAMA 2016;315(15):1624–45.
  9. Saragiotto BT, Machado GC, Ferreira ML, Pinheiro MB, Abdel Shaheed C, Maher CG. Paracetamol for low back pain. Cochrane Database Syst Rev 2016(6):CD012230.
  10. Williams CM, Maher CG, Latimer J, et al. Efficacy of paracetamol for acute low-back pain: A double-blind, randomised controlled trial. Lancet 2014;384(9954):1586–96.
  11. Moore RA, Derry S, Aldington D, Wiffen PJ. Single dose oral analgesics for acute postoperative pain in adults – An overview of Cochrane reviews. Cochrane Database Syst Rev 2015(9):CD008659.
  12. Moore RA, Derry S, Wiffen PJ, Straube S, Aldington DJ. Overview review: Comparative efficacy of oral ibuprofen and paracetamol (acetaminophen) across acute and chronic pain conditions. Eur J Pain 2015;19(9):1213–23.
  13. Moore RA, Derry S, Aldington D, Wiffen PJ. Adverse events associated with single dose oral analgesics for acute postoperative pain in adults – An overview of Cochrane reviews. Cochrane Database Syst Rev 2015(10):CD011407.
  14. Ong CK, Seymour RA, Lirk P, Merry AF. Combining paracetamol (acetaminophen) with nonsteroidal antiinflammatory drugs: A qualitative systematic review of analgesic efficacy for acute postoperative pain. Anesth Analg 2010;110(4):1170–79.
  15. Bailey E, Worthington HV, van Wijk A, Yates JM, Coulthard P, Afzal Z. Ibuprofen and/or paracetamol (acetaminophen) for pain relief after surgical removal of lower wisdom teeth. Cochrane Database Syst Rev 2013;12:CD004624.
  16. Shaheed CA, Maher CG, McLachlan AJ. Investigating the efficacy and safety of over-the-counter codeine containing combination analgesics for pain and codeine based antitussives. Canberra: Therapeutic Goods Association, 2016 [Accessed 19 July 2017].
  17. Blondell RD, Azadfard M, Wisniewski AM. Pharmacologic therapy for acute pain. Am Fam Physician 2013;87(11):766–72.
  18. Bendtsen L, Evers S, Linde M, et al. EFNS guideline on the treatment of tension-type headache – Report of an EFNS task force. Eur J Neurol 2010;17(11):1318–25.
  19. Thorson D, Biewen P, Bonte B, et al. Acute pain assessment and opioid prescribing protocol. Bloomington, MN: Institute for Clinical Systems Improvement, 2014 Opioids.pdf [Accessed 1 September 2017].
  20. Australian and New Zealand College of Anaesthetists. Guidelines on acute pain management. Melbourne: ANZCA, 2013 Documents/ps41-2013-guidelines-on-acute-painmanagement [Accessed 19 July 2017].
  21. Traeger AC, Hubscher M, Henschke N, Moseley GL, Lee H, McAuley JH. Effect of primary care-based education on reassurance in patients with acute low back pain: Systematic review and meta-analysis. JAMA Intern Med 2015;175(5):733–43.
  22. Qaseem A, Wilt TJ, McLean RM, Forciea MA, Clinical Guidelines Committee of the American College of Physicians. Noninvasive treatments for acute, subacute, and chronic low back pain: A clinical practice guideline from the American College of Physicians. Ann Intern Med 2017;166(7):514–30.
  23. Chung JW, Zeng Y, Wong TK. Drug therapy for the treatment of chronic nonspecific low back pain: Systematic review and meta-analysis. Pain Physician 2013;16(6):E685–704.
  24. van den Bekerom MP, Sjer A, Somford MP, Bulstra GH, Struijs PA, Kerkhoffs GM. Non-steroidal anti-inflammatory drugs (NSAIDs) for treating acute ankle sprains in adults: Benefits outweigh adverse events. Knee Surg Sports Traumatol Arthrosc 2015;23(8):2390–99.
  25. Massey T, Derry S, Moore RA, McQuay HJ. Topical NSAIDs for acute pain in adults. Cochrane Database Syst Rev 2010(6):CD007402.
  26. Predel HG, Giannetti B, Seigfried B, Novellini R, Menke G. A randomized, double-blind, placebo-controlled multicentre study to evaluate the efficacy and safety of diclofenac 4%
  27. spray gel in the treatment of acute uncomplicated ankle sprain. J Int Med Res 2013;41(4):1187–202.
  28. Predel HG, Hamelsky S, Gold M, Giannetti B. Efficacy and safety of diclofenac diethylamine 2.32% gel in acute ankle sprain. Med Sci Sports Exerc 2012;44(9):1629–36.
  29. Serinken M, Eken C, Turkcuer I, Elicabuk H, Uyanik E, Schultz CH. Intravenous paracetamol versus morphine for renal colic in the emergency department: A randomised double-blind controlled trial. Emerg Med J 2012;29(11):902–95.
  30. Holdgate A, Pollock T. Nonsteroidal anti-inflammatory drugs (NSAIDs) versus opioids for acute renal colic. Cochrane Database Syst Rev 2005(2):CD004137.
  31. Afshar K, Jafari S, Marks AJ, Eftekhari A, MacNeily AE. Nonsteroidal anti-inflammatory drugs (NSAIDs) and nonopioids for acute renal colic. Cochrane Database Syst Rev 2015(6):CD006027.
  32. Turk C, Petrik A, Sarica K, et al. EAU guidelines on diagnosis and conservative management of urolithiasis. Eur Urol 2016;69(3):468–74.
  33. Sin B, Koop K, Liu M, Yeh JY, Thandi P. Intravenous acetaminophen for renal colic in the emergency department: Where do we stand? Am J Ther 2017;24(1):e12–e19.
  34. Campschroer T, Zhu Y, Duijvesz D, Grobbee DE, Lock MT. Alpha-blockers as medical expulsive therapy for ureteral stones. Cochrane Database Syst Rev 2014;4:CD008509.
  35. Colli A, Conte D, Valle SD, Sciola V, Fraquelli M. Metaanalysis: Nonsteroidal anti-inflammatory drugs in biliary colic. Aliment Pharmacol Ther 2012;35(12):1370–78.
  36. National Institute for Health and Clinical Excellence. Gallstone disease: Diagnosis and initial management. NICE guidelines CG188. London: NICE, 2014. Available at www. [Accessed 21 July 2017].
  37. Zakko SF. Uncomplicated gallstone disease in adults. UpToDate, 2016 uncomplicated-gallstone-disease-in-adults?topicKey=GAST %2F654&elapsedTimeMs=5&source=machineLearning&se archTerm=biliary+colic&selectedTitle=1%7E150&view=print &displayedView=full&anchor=H25 [Accessed 21 July 2017].
  38. Moore PA, Hersh EV. Combining ibuprofen and acetaminophen for acute pain management after thirdmolar extractions: Translating clinical research to dental practice. J Am Dent Assoc 2013;144(8):898–908.
  39. Marjoribanks J, Proctor M, Farquhar C, Derks RS.
  40. Nonsteroidal anti-inflammatory drugs for dysmenorrhoea. Cochrane Database Syst Rev 2010(1):CD001751.
  41. Cunningham A, Breuer J, Dwyer D, et al. The prevention and management of herpes zoster. Med J Aust 2008;188(3):171–76.
  42. Dworkin RH, Johnson RW, Breuer J, et al.
  43. Recommendations for the management of herpes zoster. Clin Infect Dis 2007;44(Suppl 1):S1–26.
  44. Dwyer DE, Cunningham AL. 10: Herpes simplex and varicella-zoster virus infections. Med J Aust 2002;177(5):267–73.
  45. Berry JD, Petersen KL. A single dose of gabapentin reduces acute pain and allodynia in patients with herpes zoster. Neurology 2005;65(3):444–47.
  46. Jensen-Dahm C, Rowbotham MC, Reda H, Petersen KL.
  47. Effect of a single dose of pregabalin on herpes zoster pain. Trials 2011;12:55.
  48. Lin PL, Fan SZ, Huang CH, et al. Analgesic effect of lidocaine patch 5% in the treatment of acute herpes zoster: A double-blind and vehicle-controlled study. Reg Anesth Pain Med 2008;33(4):320–25.
  49. Chen N, Li Q, Yang J, Zhou M, Zhou D, He L. Antiviral treatment for preventing postherpetic neuralgia. Cochrane Database Syst Rev 2014(2):CD006866.
  50. Chen N, Yang M, He L, Zhang D, Zhou M, Zhu C.
  51. Corticosteroids for preventing postherpetic neuralgia. Cochrane Database Syst Rev 2010(12):CD005582.
  52. Saarto T, Wiffen PJ. Antidepressants for neuropathic pain: A Cochrane review. J Neurol Neurosurg Psychiatry 2010;81(12):1372–73.
  53. Moore RA, Derry S, Wiffen PJ, Straube S, Bendtsen L. Evidence for efficacy of acute treatment of episodic tensiontype headache: Methodological critique of randomised trials for oral treatments. Pain 2014;155(11):2220–28.
  54. Chaibi A, Russell MB. Manual therapies for cervicogenic headache: A systematic review. J Headache Pain 2012;13(5):351–59.
  55. Luedtke K, Allers A, Schulte LH, May A. Efficacy of interventions used by physiotherapists for patients with headache and migraine-systematic review and metaanalysis. Cephalalgia 2016;36(5):474–92.
  56. Derry S, Moore RA. Paracetamol (acetaminophen) with or without an antiemetic for acute migraine headaches in adults. Cochrane Database Syst Rev 2013;4:CD008040.
  57. Kirthi V, Derry S, Moore RA. Aspirin with or without an antiemetic for acute migraine headaches in adults. Cochrane Database Syst Rev 2013;4:CD008041.
  58. Rabbie R, Derry S, Moore RA. Ibuprofen with or without an antiemetic for acute migraine headaches in adults. Cochrane Database Syst Rev 2013;4:CD008039.
  59. Derry S, Rabbie R, Moore RA. Diclofenac with or without an antiemetic for acute migraine headaches in adults. Cochrane Database Syst Rev 2013;4:CD008783.
  60. Colman I, Brown MD, Innes GD, Grafstein E, Roberts TE, Rowe BH. Parenteral metoclopramide for acute migraine: Meta-analysis of randomised controlled trials. BMJ 2004;329(7479):1369–73.
  61. Friedman BW, Esses D, Solorzano C, et al. A randomized controlled trial of prochlorperazine versus metoclopramide for treatment of acute migraine. Ann Emerg Med 2008;52(4):399–406.
  62. Coppola M, Yealy DM, Leibold RA. Randomized, placebo-controlled evaluation of prochlorperazine versus metoclopramide for emergency department treatment of migraine headache. Ann Emerg Med 1995;26(5):541–46.
  63. Taggart E, Doran S, Kokotillo A, Campbell S, Villa-Roel C, Rowe BH. Ketorolac in the treatment of acute migraine: A systematic review. Headache 2013;53(2):277–87.
  64. Thorlund K, Mills EJ, Wu P, et al. Comparative efficacy of triptans for the abortive treatment of migraine: A multiple treatment comparison meta-analysis. Cephalalgia 2014;34(4):258-67.
  65. Tepper SJ. Opioids should not be used in migraine. Headache 2012;52(Suppl 1):30–34.
  66. Buse DC, Pearlman SH, Reed ML, Serrano D, Ng-Mak DS, Lipton RB. Opioid use and dependence among persons with migraine: Results of the AMPP study. Headache 2012;52(1):18–36.
  67. Finocchi C, Viani E. Opioids can be useful in the treatment of headache. Neurol Sci 2013;34(Suppl 1):S119–24.
  68. Broner SW, Sun-Edelstein C, Lay CL. Cluster headache in women. Curr Pain Headache Rep 2007;11(2):127–30.
  69. Finkel AG. Epidemiology of cluster headache. Curr Pain Headache Rep 2003;7(2):144–49.
  70. Fischera M, Marziniak M, Gralow I, Evers S. The incidence and prevalence of cluster headache: A meta-analysis of population-based studies. Cephalalgia 2008;28(6):614–18.
  71. Bennett MH, French C, Schnabel A, Wasiak J, Kranke P. Normobaric and hyperbaric oxygen therapy for migraine and cluster headache. Cochrane Database Syst Rev 2008(3):CD005219.
  72. Cohen AS, Burns B, Goadsby PJ. High-flow oxygen for treatment of cluster headache: A randomized trial. JAMA 2009;302(22):2451–57.
  73. Robbins MS, Starling AJ, Pringsheim TM, Becker WJ,
  74. Schwedt TJ. Treatment of cluster headache: American Headache Society evidence-based guidelines. Headache 2016;56(7):1093–106.
  75. Bennett MH, French C, Schnabel A, Wasiak J, Kranke P, Weibel S. Normobaric and hyperbaric oxygen therapy for the treatment and prevention of migraine and cluster headache. Cochrane Database Syst Rev 2015(12):CD005219.
  76. Law S, Derry S, Moore RA. Triptans for acute cluster headache. Cochrane Database Syst Rev 2013;7:CD008042.
  77. Francis GJ, Becker WJ, Pringsheim TM. Acute and preventive pharmacologic treatment of cluster headache. Neurology 2010;75(5):463–73.
  78. van der Meer HA, Speksnijder CM, Engelbert R,
  79. Lobbezoo F, Nijhuis-van der Sanden MW, Visscher CM. The association between headaches and temporomandibular disorders is confounded by bruxism and somatic complaints. Clin J Pain 2016. doi: 10.1097/ AJP.0000000000000470.
  80. Costa YM, Conti PC, de Faria FA, Bonjardim LR.
  81. Temporomandibular disorders and painful comorbidities: Clinical association and underlying mechanisms. Oral Surg Oral Med Oral Pathol Oral Radiol 2017;123(3):288–97.
  82. Schiffman E, Ohrbach R, List T, et al. Diagnostic criteria for headache attributed to temporomandibular disorders. Cephalalgia 2012;32(9):683–92.
  83. Mujakperuo HR, Watson M, Morrison R, Macfarlane TV.
  84. Pharmacological interventions for pain in patients with temporomandibular disorders. Cochrane Database Syst Rev 2010(10):CD004715.
  85. Ta LE, Dionne RA. Treatment of painful temporomandibular joints with a cyclooxygenase-2 inhibitor: A randomized placebo-controlled comparison of celecoxib to naproxen. Pain 2004;111(1-2):13–21.
  86. Kelley JM, Kraft-Todd G, Schapira L, Kossowsky J, Riess H. The influence of the patient-clinician relationship on healthcare outcomes: A systematic review and metaanalysis of randomized controlled trials. PLoS One 2014;9(4):e94207.
  87. National Opioid Use Guideline Group. Canadian guideline for safe and effective use of opioids for chronic non-cancer pain. Part B: Recommendations for practice. Ontario:
  88. NOUGG, 2010. Available at http://nationalpaincentre. [Accessed 21 July 2017].
  89. Klinger R, Colloca L, Bingel U, Flor H. Placebo analgesia: Clinical applications. Pain 2014;155(6):1055–58.
  90. Miller FG, Kaptchuk TJ. The power of context: Reconceptualizing the placebo effect. J R Soc Med 2008;101(5):222–25.
  91. Lee C, Crawford C, Swann S, Active Self-Care Therapies for Pain Working Group. Multimodal, integrative therapies for the self-management of chronic pain symptoms. Pain Med 2014;15(Suppl 1):S76–85.
  92. Hayden JA, van Tulder MW, Malmivaara A, Koes BW. Exercise therapy for treatment of non-specific low back pain. Cochrane Database Syst Rev 2005(3):CD000335.
  93. Fransen M, McConnell S, Harmer AR, Van der Esch M, Simic M, Bennell KL. Exercise for osteoarthritis of the knee: A Cochrane systematic review. Br J Sports Med 2015;49(24):1554–57.
  94. Fransen M, McConnell S, Hernandez-Molina G,
  95. Reichenbach S. Exercise for osteoarthritis of the hip. Cochrane Database Syst Rev 2014;4:CD007912.
  96. Busch AJ, Barber KA, Overend TJ, Peloso PM, Schachter CL. Exercise for treating fibromyalgia syndrome. Cochrane Database Syst Rev 2007(4):CD003786.
  97. Scottish Intercollegiate Guidelines Network. Management of chronic pain (SIGN 136). Edinburgh: SIGN, 2013 [Accessed 1 September 2017].
  98. Williams AC, Eccleston C, Morley S. Psychological therapies for the management of chronic pain (excluding headache) in adults. Cochrane Database Syst Rev 2012;11:CD007407.
  99. Eccleston C, Hearn L, Williams AC. Psychological therapies for the management of chronic neuropathic pain in adults. Cochrane Database Syst Rev 2015;10:CD011259.
  100. Hooten W, Timming R, Belgrade M, et al. Assessment and management of chronic pain. Bloomington, MN: Institute for Clinical Systems Improvement, 2013 c26a36d83686607ad89ee835daa3c9db3f4c.pdf [Accessed 1 September 2017].
  101. Kahan M, Mailis-Gagnon A, Wilson L, Srivastava A, National
  102. Opioid Use Guideline Group. Canadian guideline for safe and effective use of opioids for chronic noncancer pain: Clinical summary for family physicians. Part 1: General population. Can Fam Physician 2011;57(11):1257–66, e407-18.
  103. The Royal Australasian College of Physicians. Prescription opioid policy: Improving management of chronic nonmalignant pain and prevention of problems associated with prescription opioid use. Sydney: RACP, 2009.
  104. Manchikanti L, Abdi S, Atluri S, et al. American Society of Interventional Pain Physicians (ASIPP) guidelines for responsible opioid prescribing in chronic non-cancer pain: Part 2: Guidance. Pain Physician 2012;15(3 Suppl):S67–116.
  105. National Center for Injury Prevention and Control. Common elements in guidelines for prescribing opioids for chronic pain. NCIPC, 2014 drugoverdose/pdf/common_elements_in_guidelines_for_ prescribing_opioids-a.pdf [Accessed 21 July 2017].
  106. Moulin D, Boulanger A, Clark AJ, et al. Pharmacological management of chronic neuropathic pain: Revised consensus statement from the Canadian Pain Society. Pain Res Manag 2014;19(6):328–35.
  107. Whiting PF, Wolff RF, Deshpande S, et al. Cannabinoids for medical use: A systematic review and meta-analysis. JAMA. 2015;313(24):2456-73.
  108. Finnerup NB, Attal N, Haroutounian S, et al. Pharmacotherapy for neuropathic pain in adults: A systematic review and meta-analysis. Lancet Neurol 2015;14(2):162–73.
  109. Australian and New Zealand College of Anaesthetists. Statement on ‘medicinal cannabis’ with particular reference to its use in the management of patients with chronic noncancer pain (PM10). Melbourne: ANZCA, 2015 [Accessed 21 July 2017].
  110. Stacey BR, Barrett JA, Whalen E, Phillips KF, Rowbotham MC. Pregabalin for postherpetic neuralgia: Placebocontrolled trial of fixed and flexible dosing regimens on allodynia and time to onset of pain relief. J Pain 2008;9(11):1006–17.
  111. Wang F, Ruberg SJ, Gaynor PJ, Heinloth AN, Arnold LM. Early improvement in pain predicts pain response at endpoint in patients with fibromyalgia. J Pain 2011;12(10):1088–94.
  112. Therapeutics Initiative. Benefits and harms of drugs for ‘neuropathic’ pain. Vancouver: University of British Columbia, 2015 [Accessed 21 July 2017].
  113. Hughes MA, Biggs JJ, Theise MS, Graziano K, Robbins RB, Effiong AC. Recommended opioid prescribing practices for use in chronic non-malignant pain: A systematic review of treatment guidelines. J Manag Care Med 2011;14(3):52.
  114. National Opioid Use Guideline Group. Canadian guideline for safe and effective use of opioids for chronic non-cancer pain. Ontario: NOUGG, 2010. Available at http://nationalpaincentre. [Accessed 21 July 2017].
  115. Deyo RA, Von Korff M, Duhrkoop D. Opioids for low back pain. BMJ 2015;350:g6380.
  116. Chou R, Fanciullo GJ, Fine PG, et al. Clinical guidelines for the use of chronic opioid therapy in chronic noncancer pain. J Pain 2009;10(2):113–30.
  117. Fagan MJ, Chen JT, Diaz JA, Reinert SE, Stein MD. Do internal medicine residents find pain medication agreements useful? Clin J Pain 2008;24(1):35–38.
  118. Bazazi AR, Zaller ND, Fu JJ, Rich JD. Preventing opiate overdose deaths: Examining objections to takehome naloxone. J Health Care Poor Underserved 2010;21(4):1108–13.
  119. McDonald R, Strang J. Are take-home naloxone programmes effective? Systematic review utilizing application of the Bradford Hill criteria. Addiction 2016;111(7):1177–87.
  120. Strang J, McDonald R, Alqurshi A, Royall P, Taylor D, Forbes B. Naloxone without the needle – Systematic review of candidate routes for non-injectable naloxone for opioid overdose reversal. Drug Alcohol Depend 2016;163:16–23.
  121. Krebs EE, Lorenz KA, Bair MJ, et al. Development and initial validation of the PEG, a three-item scale assessing pain intensity and interference. J Gen Intern Med 2009;24(6):733–38.
  122. Boudreau D, Von Korff M, Rutter CM, et al. Trends in long-term opioid therapy for chronic non-cancer pain. Pharmacoepidemiol Drug Saf 2009;18(12):1166–75.
  123. Smith HS, Peppin JF. Toward a systematic approach to opioid rotation. J Pain Res 2014;7:589–608.
  124. The British Pain Society. Opioids for persistent pain: Good practice. London: The British Pain Society, 2010.
  125. Becker WC, Fraenkel L, Edelman EJ, et al. Instruments to assess patient-reported safety, efficacy, or misuse of current opioid therapy for chronic pain: A systematic review. Pain 2013;154(6):905–16.
  126. Gourlay DL, Heit HA, Almahrezi A. Universal precautions in pain medicine: A rational approach to the treatment of chronic pain. Pain Med 2005;6(2):107–12.
  127. Jammal W, Gown G. Opioid prescribing pitfalls: Medicolegal and regulatory issues. Aust Prescr 2015;38:198–203.
  128. Noble M, Treadwell JR, Tregear SJ, et al. Long-term opioid management for chronic noncancer pain. Cochrane Database Syst Rev 2010(1):CD006605.
  129. Häuser W, Bock F, Engeser P, Tölle T, Willweber-Strumpf A, Petzke F. Long-term opioid use in non-cancer pain. Dtsch Ärztebl Int 2014;111(43):732–40.
  130. Tawfic Q, Kumar K, Pirani Z, Armstrong K. Prevention of chronic post-surgical pain: The importance of early identification of risk factors. J Anesth 2017;31(3):424–31.
  131. Wylde V, Hewlett S, Learmonth ID, Dieppe P. Persistent pain after joint replacement: Prevalence, sensory qualities, and postoperative determinants. Pain 2011;152(3):566–72.
  132. Chan MT, Wan AC, Gin T, Leslie K, Myles PS. Chronic postsurgical pain after nitrous oxide anesthesia. Pain 2011;152(11):2514–20.
  133. Macrae WA. Chronic post-surgical pain: 10 years on. Br J Anaesth 2008;101(1):77–86.
  134. Kehlet H, Jensen TS, Woolf CJ. Persistent postsurgical pain: Risk factors and prevention. Lancet 2006;367(9522):1618–25.
  135. Treede RD, Rief W, Barke A, et al. A classification of chronic pain for ICD-11. Pain 2015;156(6):1003–7.
  136. Theunissen M, Peters ML, Bruce J, Gramke HF, Marcus MA. Preoperative anxiety and catastrophizing: A systematic review and meta-analysis of the association with chronic postsurgical pain. Clin J Pain 2012;28(9):819–41.
  137. Hinrichs-Rocker A, Schulz K, Jarvinen I, Lefering R, Simanski C, Neugebauer EA. Psychosocial predictors and correlates for chronic post-surgical pain (CPSP) – A systematic review. Eur J Pain 2009;13(7):719–30.
  138. Buchheit T, Van de Ven T, Shaw A. Epigenetics and the transition from acute to chronic pain. Pain Med 2012;13(11):1474–90.
  139. Mauck M, Van de Ven T, Shaw AD. Epigenetics of chronic pain after thoracic surgery. Curr Opin Anaesthesiol 2014;27(1):1–5.
  140. Wesselmann U, Baranowski AP, Borjesson M, et al.
  141. Emerging therapies and novel approaches to visceral pain. Drug Discov Today Ther Strateg 2009;6(3):89–95.
  142. Olesen AE, Farmer AD, Olesen SS, Aziz Q, Drewes AM. Management of chronic visceral pain. Pain Manag 2016;6(5):469–86.
  143. Queiroz LP. Worldwide epidemiology of fibromyalgia. Curr Pain Headache Rep 2013;17(8):356.
  144. Hauser W, Zimmer C, Felde E, Kollner V. What are the key symptoms of fibromyalgia? Results of a survey of the German Fibromyalgia Association. Schmerz 2008;22(2):176–83.
  145. Clauw DJ, Arnold LM, McCarberg BH, FibroCollaborative. The science of fibromyalgia. Mayo Clin Proc 2011;86(9):907–11.
  146. Macfarlane GJ, Kronisch C, Dean LE, et al. EULAR revised recommendations for the management of fibromyalgia. Ann Rheum Dis 2017;76(2):318–28.
  147. Angel Garcia D, Martinez Nicolas I, Saturno Hernandez PJ. Clinical approach to fibromyalgia: Synthesis of evidencebased recommendations, a systematic review. Reumatol Clin 2016;12(2):65–71.
  148. Clauw DJ. Fibromyalgia: A clinical review. JAMA 2014;311(15):1547–55.
  149. Fitzcharles MA, Ste-Marie PA, Goldenberg DL, et al. 2012 Canadian guidelines for the diagnosis and management of fibromyalgia syndrome: Executive summary. Pain Res Manag 2013;18(3):119–26.
  150. Hauser W, Thieme K, Turk DC. Guidelines on the management of fibromyalgia syndrome – A systematic review. Eur J Pain 2010;14(1):5–10.
  151. Goebel A, Barker C, Turner-Stokes L, et al. Complex regional pain syndrome in adults: UK guidelines for diagnosis, referral and management in primary and secondary care. London: RCP, 2012.
  152. Casale R, Atzeni F, Sarzi-Puttini P. The therapeutic approach to complex regional pain syndrome: Light and shade. Clin Exp Rheumatol 2015;33(1 Suppl 88):S126–39.
  153. Birklein F, O’Neill D, Schlereth T. Complex regional pain syndrome: An optimistic perspective. Neurology 2015;84(1):89–96.
  154. Borchers AT, Gershwin ME. Complex regional pain syndrome: A comprehensive and critical review. Autoimmun Rev 2014;13(3):242–65.
  155. Lohnberg JA, Altmaier EM. A review of psychosocial factors in complex regional pain syndrome. J Clin Psychol Med Settings 2013;20(2):247–54.
  156. Cossins L, Okell RW, Cameron H, Simpson B, Poole HM, Goebel A. Treatment of complex regional pain syndrome in adults: A systematic review of randomized controlled trials published from June 2000 to February 2012. Eur J Pain 2013;17(2):158–73.
  157. Goh EL, Chidambaram S, Ma D. Complex regional pain syndrome: A recent update. Burns Trauma 2017;5:2.
  158. Larochelle MR, Liebschutz JM, Zhang F, Ross-Degnan D, Wharam JF. Opioid prescribing after nonfatal overdose and association with repeated overdose: A cohort study. Ann Intern Med 2016;164(1):1–9.
  159. Zanini C, Sarzi-Puttini P, Atzeni F, Di Franco M, Rubinelli S. Building bridges between doctors and patients: The design and pilot evaluation of a training session in argumentation for chronic pain experts. BMC Med Educ 2015;15:89.
  160. Gammaitoni AR, Fine P, Alvarez N, McPherson ML, Bergmark S. Clinical application of opioid equianalgesic data. Clin J Pain 2003;19(5):286–97.
  161. NSW Therapeutic Advisory Group Inc. Preventing and managing problems with opioid prescribing for chronic noncancer pain. Sydney: NSW TAG, 2015. Available at www. practical-guidance/pain-guidance-july-2015.pdf [Accessed 26 July 2017].
  162. Busse JW, Craigie S, Juurlink DN, et al. Guideline for opioid therapy and chronic noncancer pain. CMAJ 2017;189(18):E659–E66.
  163. Windmill J, Fisher E, Eccleston C, et al. Interventions for the reduction of prescribed opioid use in chronic non-cancer pain. Cochrane Database Syst Rev 2013(9):CD010323.
  164. Nilsen HK, Stiles TC, Landro NI, Fors EA, Kaasa S, Borchgrevink PC. Patients with problematic opioid use can be weaned from codeine without pain escalation. Acta Anaesthesiol Scand 2010;54(5):571–79.
  165. Baron MJ, McDonald PW. Significant pain reduction in chronic pain patients after detoxification from high-dose opioids. J Opioid Manag 2006;2(5):277–82.
  166. Crisostomo RA, Schmidt JE, Hooten WM, Kerkvliet JL, Townsend CO, Bruce BK. Withdrawal of analgesic medication for chronic low-back pain patients: Improvement in outcomes of multidisciplinary rehabilitation regardless of surgical history. Am J Phys Med Rehabil 2008;87(7):527–36.
  167. Younger J, Barelka P, Carroll I, et al. Reduced cold pain tolerance in chronic pain patients following opioid detoxification. Pain Med 2008;9(8):1158–63.
  168. Hooten WM, Mantilla CB, Sandroni P, Townsend CO. Associations between heat pain perception and opioid dose among patients with chronic pain undergoing opioid tapering. Pain Med 2010;11(11):1587–98.
  169. Wang H, Akbar M, Weinsheimer N, Gantz S, Schiltenwolf M. Longitudinal observation of changes in pain sensitivity during opioid tapering in patients with chronic low-back pain. Pain Med 2011;12(12):1720–26.
  170. Berna C, Kulich RJ, Rathmell JP. Tapering long-term opioid therapy in chronic noncancer pain: Evidence and recommendations for everyday practice. Mayo Clin Proc 2015;90(6):828–42.
  171. International Association for the Study of Pain. Classification of chronic pain. 2nd edn. Washington DC: IASP, 2011 aspx?ItemNumber=1673 [Accessed 26 July 2017].
  172. Katz J, Weinrib A, Fashler SR, et al. The Toronto General Hospital Transitional Pain Service: Development and implementation of a multidisciplinary program to prevent chronic postsurgical pain. J Pain Res 2015;8:695–702.
  173. Denk F, McMahon SB, Tracey I. Pain vulnerability: A neurobiological perspective. Nat Neurosci 2014;17(2):192– 200.
  174. Eisenberger NI. The neural bases of social pain: Evidence for shared representations with physical pain. Psychosom Med 2012;74(2):126–35.
  175. Kosek E, Cohen M, Baron R, et al. Do we need a third mechanistic descriptor for chronic pain states? Pain 2016;157:1382–86.
  176. Merskey H, Bogduk N. International Association for the Study of Pain Task Force on Taxonomy. Classification of chronic pain: Descriptions of chronic pain syndromes and definitions of pain terms. 2nd edn. Seattle: IASP Press, 1994; p. 222.
  177. Smart KM, Blake C, Staines A, Thacker M, Doody C. Mechanisms-based classifications of musculoskeletal pain: Part 3 of 3: Symptoms and signs of nociceptive pain in patients with low back (+/– leg) pain. Man Ther 2012;17(4):352–57.
  178. Costigan M, Scholz J, Woolf CJ. Neuropathic pain: A maladaptive response of the nervous system to damage. Annu Rev Neurosci 2009;32:1–32.
  179. Freynhagen R, Baron R, Gockel U, Tolle TR. painDETECT: A new screening questionnaire to identify neuropathic components in patients with back pain. Curr Med Res Opin 2006;22(10):1911–20.
  180. O’Connor AB, Dworkin RH. Treatment of neuropathic pain: An overview of recent guidelines. Am J Med 2009;122(10 Suppl):S22–32.
  181. Arendt-Nielsen L, Nie H, Laursen MB, et al. Sensitization in patients with painful knee osteoarthritis. Pain 2010; 149(3):573–81.
  182. Kosek E, Ordeberg G. Lack of pressure pain modulation by heterotopic noxious conditioning stimulation in patients with painful osteoarthritis before, but not following, surgical pain relief. Pain 2000;88(1):69–78.
  183. Aranda-Villalobos P, Fernandez-de-Las-Penas C, NavarroEspigares JL, et al. Normalization of widespread pressure pain hypersensitivity after total hip replacement in patients with hip osteoarthritis is associated with clinical and functional improvements. Arthritis Rheum 2013;65(5):1262–70.
  184. Graven-Nielsen T, Wodehouse T, Langford RM, ArendtNielsen L, Kidd BL. Normalization of widespread hyperesthesia and facilitated spatial summation of deep-tissue pain in knee osteoarthritis patients after knee replacement. Arthritis Rheum 2012;64(9):2907–16.
  185. Kosek E, Ordeberg G. Abnormalities of somatosensory perception in patients with painful osteoarthritis normalize following successful treatment. Eur J Pain 2000;4(3):229–38.
  186. Rosenquist EWK. Evaluation of chronic pain in adults. UpToDate 2016 evaluation-of-chronic-pain-in-adults?source=search_ result&search=pain%20assessment&selectedTitle=1~150 - H15544430 [Accessed 3 March 2017].
  187. Kirsh KL, Jass C, Bennett DS, Hagen JE, Passik SD. Initial development of a survey tool to detect issues of chemical coping in chronic pain patients. Palliat Support Care 2007;5(3):219–26.
  188. Flor H. Psychological pain interventions and neurophysiology: Implications for a mechanism-based approach. Am Psychol 2014;69(2):188–96.
  189. Nicholas MK, Linton SJ, Watson PJ, Main CJ, Decade of the Flags Working Group. Early identification and management of psychological risk factors (‘yellow flags’) in patients with low back pain: A reappraisal. Phys Ther 2011;91(5):737–53.
  190. Ip HY, Abrishami A, Peng PW, Wong J, Chung F. Predictors of postoperative pain and analgesic consumption: A qualitative systematic review. Anesthesiology 2009;111(3):657–77.
  191. Leeuw M, Goossens ME, Linton SJ, Crombez G, Boersma K, Vlaeyen JW. The fear-avoidance model of musculoskeletal pain: Current state of scientific evidence. J Behav Med 2007;30(1):77–94.
  192. Hjermstad MJ, Fayers PM, Haugen DF, et al. Studies comparing numerical rating scales, verbal rating scales, and visual analogue scales for assessment of pain intensity in adults: A systematic literature review. J Pain Symptom Manage 2011;41(6):1073–93.
  193. Chen L, Vo T, Seefeld L, et al. Lack of correlation between opioid dose adjustment and pain score change in a group of chronic pain patients. J Pain 2013;14(4):384–92.
  194. Chou R, Turner JA, Devine EB, et al. The effectiveness and risks of long-term opioid therapy for chronic pain: A systematic review for a National Institutes of Health Pathways to Prevention Workshop. Ann Intern Med 2015;162(4):276–86.
  195. Sehgal N, Manchikanti L, Smith HS. Prescription opioid abuse in chronic pain: A review of opioid abuse predictors and strategies to curb opioid abuse. Pain Physician 2012;15(3 Suppl):ES67–92.
  196. Gordon A, Cone EJ, DePriest AZ, Axford-Gatley RA, Passik SD. Prescribing opioids for chronic noncancer pain in primary care: Risk assessment. Postgrad Med 2014;126(5):159–66.
  197. Di Blasi Z, Harkness E, Ernst E, Georgiou A, Kleijnen J. Influence of context effects
This event attracts CPD points and can be self recorded

Did you know you can now log your CPD with a click of a button?

Create Quick log

Advertising