Skip to Main Content
Book cover for Oxford Textbook of Palliative Medicine (5 edn) Oxford Textbook of Palliative Medicine (5 edn)

Contents

Book cover for Oxford Textbook of Palliative Medicine (5 edn) Oxford Textbook of Palliative Medicine (5 edn)
Disclaimer
Oxford University Press makes no representation, express or implied, that the drug dosages in this book are correct. Readers must therefore always … More Oxford University Press makes no representation, express or implied, that the drug dosages in this book are correct. Readers must therefore always check the product information and clinical procedures with the most up to date published product information and data sheets provided by the manufacturers and the most recent codes of conduct and safety regulations. The authors and the publishers do not accept responsibility or legal liability for any errors in the text or for the misuse or misapplication of material in this work. Except where otherwise stated, drug dosages and recommendations are for the non-pregnant adult who is not breastfeeding.

An understanding of the principles of clinical pharmacology is essential to enable clinicians to prescribe drugs safely and effectively. When prescribing any drug the intention is to gain a therapeutic effect (e.g. the use of opioids to reduce pain), while avoiding harm (e.g. opioid-induced drowsiness). All drugs have the potential to cause side effects, many of which are predictable from knowledge of clinical pharmacology.

Patients with serious or life-threatening illness present challenges to the safe and effective use of medications. Patient-related factors, such as age, gender, and co-morbidities, may influence the choice of drug, starting dose, and route of administration. Most patients receive multiple drugs and clinicians must always be mindful of the risk of drug–drug interactions. Given the risks of polypharmacy in those with advanced illness, options for concurrent non-drug therapy always should be considered. This is certainly important in the case of pain, for which a holistic approach, one that acknowledges that pharmacotherapy is only one strategy among many that may yield favourable outcomes, is often best.

Opioids and other drugs that have the potential for misuse or abuse, addiction, or drug diversion (see Chapter 9.5) are commonly used in palliative care, and these drugs are regulated to a greater or lesser extent in every country. Prescribers may need to make adjustments based on the wider social context implicated in these regulations, and impact of taking medications that may be inconsistently available or arduous to prescribe. In some countries, excessive regulation impedes patient access, particularly in community settings. Clinicians may need to consider whether therapy can be more effectively provided in different care settings (e.g. hospital versus community) or when using specific drugs or formulations, or rates of titration and monitoring. Inclusion of a drug, such as codeine or morphine, on the World Health Organization (WHO) Model List of Essential Medicines (WHO, 2013) increases the likelihood of availability, but does not mean unrestricted access in every country (Junger et al., 2013). Indeed, opioid drugs exemplify the international variation in access that characterizes many types of drugs; some provide their populations with a large number of different opioids and opioid formulations, whereas others fail to ensure access to any. The WHO continues to seek to improve international access to controlled drugs, in particular opioids.

With increasing health-care costs, groups of clinical, pharmacy, and finance professionals are working to promote cost-effective and safer prescribing. In the United Kingdom, efficacy and cost-effectiveness of drugs are assessed at a national level by the National Centre for Health and Care Excellence (NICE). Consideration of cost-effectiveness is now an integral part of the rationale for appropriate prescribing.

Clinical guidelines are recommendations, based on appraisal of the best available evidence. When evidence is limited or absent, expert opinion is necessary to guide and support best practice. For example, most of the evidence-based guidelines for opioid use in palliative care that have been developed by the European Association of Palliative Care are based on weak evidence (Caraceni et al., 2012). In addition to the development of guidelines by professional societies, national guidances (e.g. NICE (2012) or Scottish Intercollegiate Guidelines Network (2008)) have been developed to help inform local, regional, or hospital-specific practices in a manner that considers local variation in resources and practice. A working knowledge of both international guidelines and national guidances is essential.

Formularies are also a useful resource to guide the prescribing and dispensing of medicines. Many provide detailed information about formulations and doses of drugs. If a formulary is promoted or required in practice, it is important to consider the target audience and the context for any recommendations provided. In palliative care, the most widely recognized formulary is the Palliative Care Formulary (Palliativedrugs.com, 2012). This is regularly updated and includes advice about the many drugs that are used off-licence in the palliative care setting. Other generic formularies, such as the British National Formulary (BNF), include a much wider selection of medications (Joint Formulary Committee, 2014). The choice of drugs in local or regional formularies is often restricted due to budget constraints.

Evidence-based medicine (EBM) is centred on the judicious use of current best evidence about the risks and benefits of interventions to inform clinical decision-making. The principles of EBM are discussed in detail in Chapter 19.2. It is important to recognize that the evidence provided by clinical trials evaluates the efficacy of an intervention, such as an opioid, in an ideal/controlled setting. Strict inclusion and exclusion criteria are applied and an enriched or selected sample may be studied, with structured and often short-term follow-up. Conducting high-quality clinical trials in the palliative care setting is especially challenging (Kaasa et al., 2006) and the evidence used to inform prescribing in patients with advanced illness typically originates from studies of relatively healthier populations.

Effectiveness should be distinguished from efficacy. Effectiveness refers to the benefits and burdens of a drug in the wider context, at a population level and as part of everyday practice. Information about efficacy is needed to identify substances that have the potential for clinical benefit; information about effectiveness provides more relevant and actionable information about the likelihood and extent of the therapeutic effect in a given patient.

Cost-effectiveness refers to a comparison of effectiveness for a target indication against cost, that is, the ratio of effectiveness to cost. Clearly, it is more efficient to use the cheaper of two drugs that are equally effective and safe, and this information often informs local guidance.

From the clinical perspective, the expectation of benefit must be compared to the expectation of risk to determine whether treatment or a change in treatment is justified. Although it can be difficult to estimate the balance between the potential risks and benefits of a particular treatment in an individual case, it is necessary to do so before action is taken.

The likelihood of benefit or risk in the individual is informed by an understanding of likely outcomes in the population overall. Two broad measures that can be useful as part of this assessment are number needed to treat (NNT) and number needed to harm (NNH). The NNT estimates the number of patients that would need to be given a treatment for one of them to achieve a desired outcome (e.g. 50% pain relief). The NNH is calculated for adverse effects in a similar way. Although these measures have been criticized because they are derived from controlled clinical trials data, they nevertheless provide a useful point of comparison among drugs (Christensen and Kristiansen, 2006).

The cut-offs in pain relief or severity of an adverse effect used to calculate the NNT and NNH, respectively, are accepted by convention. In fact, perceived benefit or risk may be strongly influenced by individual variation or therapeutic context. For example, patients with severe pain may feel that a 30% or smaller reduction in pain relief is clinically meaningful. In addition, some patients may be willing to tolerate mild/moderate side effects to achieve a small improvement in pain control if this translates into improved function or quality of life. Knowledge of the patient’s drug history in terms of previous success or failures of treatment will also guide choice of drug in prescribing. Prescribers also must always check individual patient factors, such as renal function and known allergies, to further inform assessment of risk/harm and potential for drug interactions.

Information about side effects of a drug must continue to be gathered after the drug is licensed. This is particularly important when a drug is used in the palliative care context, which is usually characterized by patients with advanced illness and the use of multiple drugs adapted for off-licence indications. In these situations, the use of a drug may be associated with a side effect liability quite different than that expected based on clinical trials information.

Clinical pharmacology is broadly divided into pharmacokinetics (‘what the body does to the drug’) and pharmacodynamics (‘what the drug does to the body’). In palliative medicine, drugs are not often used to cure or modify underlying disease but are predominantly focused on improving symptoms. They often are administered with the intent to continue treatment until death. Knowledge of pharmacokinetic variation, between patients and across time as disease worsens, combined with an understanding of the basic modes of drug action, underpins the logical selection and use of the most appropriate treatment.

Pharmacokinetics encompasses the absorption, distribution, metabolism, and excretion of drugs (ADME). Detailed descriptions of each of these processes are available in pharmacology textbooks. To some extent, inter-individual variation in kinetics is genetically determined. Each process also is influenced by many other factors, however, and in the the palliative care setting, these also contribute to large inter-individual variation and the potential for significant changes across time (Box 9.1.1).

Box 9.1.1
Factors affecting pharmacokinetics

Age. Both pharmacokinetic and pharmacodynamic factors change at the extremes of age. Metabolism and volume of distribution are often reduced in the elderly leading to increased free drug concentrations in the plasma. Hepatic blood flow may have declined by 40–50% by age 75, with reduced clearance of opioids. Increased central nervous system sensitivity to opioid effects is also found in the elderly.

Hepatic disease has unpredictable effects. Although there may be little clinical consequence, severe hepatic failure with coexisting encephalopathy can lead to a marked increase in sensitivity to drug effects. Reduction in plasma protein concentration, which occurs with liver failure, will also have an effect on plasma concentrations of free unbound drug.

Renal failure has a significant impact on drug response. Some of this effect is due to changes in the concentrations of parent drug and metabolites. Some is related to pharmacodynamic changes apparent when drug effects compound the uraemic state. Drugs with active, renally-cleared metabolites, for example, morphine, tend to be more problematic because of metabolite accumulation.

Obesity results in a larger volume of distribution and prolonged elimination t½.

Hypothermia, hyperthermia, hypotension, and hypovolaemia may also result in variable absorption, distribution and metabolism of opioids.

At the cellular level, absorption occurs across lipid cell membranes and is a passive process along a concentration gradient. For most drugs, this process takes place in the small intestine. As long as the drug is in solution, has a degree of lipid solubility, and there is sufficient surface area and time for diffusion in the small bowel, then problems should not arise.

A reduced rate of absorption may occur if there is delayed emptying of the stomach. This might arise as part of a pathological process or pharmacological agents that slow gastric motility, such as anticholinergic drugs or opioid analgesics.

Many drugs are now formulated as modified-release preparations, which need to remain in the small bowel for a specified period to achieve the expected absorption profile. In patients with either increased or decreased gastrointestinal transit time, there is a risk that the expected time–action relationship may not materialize. In either case, the prolonged duration or extent of therapeutic effects may be lost.

Absorption and bioavailability are not the same. The bioavailability of a drug is the percentage of administered drug that gains access unchanged to the systemic circulation. Bioavailability is of most clinical relevance after oral administration. Extensive first-pass hepatic metabolism results in relatively low bioavailability and/or large inter-individual variability for some drugs. For example, the bioavailability of oral morphine is just 35% on average and the range is 15–64%, whereas oxycodone has a bioavailability of 75% and range of 60–87%. The difference in bioavailability complicates the challenge of safe dose selection when changing between oral and parenteral routes and is one of the main reasons that dose titration is essential to identify an effective opioid dose.

Bioavailability can be altered by disease processes that affect hepatic function, or by exposure to drugs that either induce or inhibit enzymes of the cytochrome P450 (CYP450) system. In patients with chronic liver disease, for example, blood may be ‘shunted’ from portal to systemic vessels; this bypasses hepatic enzymes, reduces the first-pass effect, and increases bioavailability. These changes in hepatic function may have a profound effect on drug levels after oral administration but relatively little effect when the drug is given parenterally.

The volume of distribution (V  d) is a theoretical volume in which the total amount of drug would need to be uniformly distributed to achieve the blood concentration. For very lipophilic drugs which are taken up into fat stores or muscle, such as fentanyl, the volume may be many times body size.

The V  d is important as a determinant of half-life (t  ½) and is also of theoretical importance in the calculation of the loading dose of a drug where one is needed. Changes caused by disease, such as cachexia or renal failure, may shift a drug’s concentration–time relationships. Other related processes with potentially profound effects on drug kinetics or dynamics also may occur as a result of alteration in body composition or physicochemical environment. For example, all opioids are weak bases and dissociate into free-base and ionized fractions when dissolved in solution. The ionized form is active at the receptor site, whereas the free-base form is more lipid soluble. The relative proportions of ionized and unionized drug are dependent on pH and pK  a, and may change with the effects of disease.

All opioids also bind to plasma proteins, such as albumin and glycoproteins, in varying degrees. Opioid molecules which are unbound and unionized are capable of diffusing to the site of action, the proportion of which is known as the diffusible fraction. The concentration of the diffusible fraction and other factors such as lipid solubility determine the speed of onset of the drug. The diffusible fraction of a drug may change with hypoalbuminaemia associated with advanced illness.

A high lipid solubility facilitates diffusion across the blood–brain barrier into the brain and therefore is associated with a rapid onset of action. However this view is simplistic in that it is the ionized form that is active at opioid receptor. Speed of onset is therefore better represented as a complex function of both lipid solubility and percentage of the drug that ionized at physiological pH. Morphine has a high diffusible fraction but low lipid solubility which results in a slow onset of action. Alfentanil, however, has both a high diffusible fraction and a higher lipid solubility, which together explain the more rapid onset of action. Baseline values for both drugs may shift with varied disease-related factors that alter V  d, protein binding or the proportion of the ionized form of the drug.

Drug biotransformation takes place mainly in the liver and contributes both to the rate of elimination of a drug and its bioavailability. The rate at which metabolism proceeds usually determines the clearance; however, where removal is particularly rapid (high extraction ratio) the rate of delivery of drug to the liver rather than the rate of metabolism, may determine clearance (flow-dependent kinetics). For such drugs, if liver blood flow is markedly reduced, drug accumulation will result.

The biochemical processes of drug metabolism are complex. Two phases of metabolism are usually described. Phase I reactions involve oxidation, reduction, hydrolysis, hydration, dethioacetylation, and isomerization. Of these reactions, oxidation catalysed by members the CYP450 superfamily of enzymes are the most important and best characterized. Phase II reactions usually involves conjugation; this may take the form of glucuronidation, glycosylation, sulphation, methylation, acetylation, or conjugation with glutathione or certain amino acids. All of the reactions involve the production of products which are more water-soluble and amenable to excretion by the kidney. In some circumstances, phase II reactions may take place without a prior phase I reaction. When phase I reactions do occur, they may prepare the drug molecule for a phase II reaction by producing or uncovering a chemically reactive group, which then forms the substrate for a phase II reaction.

Most opioid metabolism, both phase I and II reactions, occurs in the liver. Hydrophilic metabolites are predominantly excreted renally, although a small amount may be excreted in the bile or unchanged in the urine. Opioid metabolites may be active and contribute to both the overall analgesic and side effect profile (Smith, 2011). Metabolism of individual opioids is shown in Table 9.1.1.

Table 9.1.1
Properties of commonly used opioids in palliative medicine
DrugpKaOral bioavailability (%)LipophilicityProtein binding (%)Volume of distributionMetabolic enzymesActive metabolitesExcreted unchanged in urine (%)Half-life

Morphine

7.9

15–64

+

30

3–5 L/kg

UGT2B7

M6G (A)

M3G (CNS+)

10

1.7–3 hours

Codeine

8.2

60–90

+

20

3–4 L/kg

UGT2B7

CYP2D6

Morphine (A)

0

2–4 hours

Diamorphine

7.6

++

70 L

Esterases

Morphine (A)

Minimal

2–3 minutes

Hydromorphone

8.2

50

+

<10

295 L

UGT2B7

UGT1A3

H3G (I)

H6G (A)

6

2–3 hours

Tramadol

9.4

70–90

+

20

2.6–2.9 L/kg

CYP3A4

CYP2D6

M1

90

6 hours

Buprenorphine

8.4

15

+++

96

430 L

CYP3A4

UGT1A1/1A3

Minimal

*Complicated by enterohepatic recirculation

Meperidine (pethidine)

8.5

++

60–80

3–5 L/kg

CYP3A4

CYP2B6

CYP2C19

Norpethidine (A, CNS+)

5

3–6 hours

Oxycodone

8.5

60–87

+

45

2–3 L/kg

CYP3A4

CYP2D6

Oxymorphone (A)

< 10

3–4 hours

Methadone

8.3

60–90

++

60–90

3–6 L/kg

CYP3A4

CYP2B6

(CYP2D6,2C9,2C19,1A2)

15–60

15–20 hours

(13–47)

Fentanyl

8.4

+++

90

3–8 L/kg

CYP3A4

< 7

2–7 hours

Alfentanil

6.5

++

90

0.4–1 L/kg

CYP3A4

Minimal

1–2 hours

DrugpKaOral bioavailability (%)LipophilicityProtein binding (%)Volume of distributionMetabolic enzymesActive metabolitesExcreted unchanged in urine (%)Half-life

Morphine

7.9

15–64

+

30

3–5 L/kg

UGT2B7

M6G (A)

M3G (CNS+)

10

1.7–3 hours

Codeine

8.2

60–90

+

20

3–4 L/kg

UGT2B7

CYP2D6

Morphine (A)

0

2–4 hours

Diamorphine

7.6

++

70 L

Esterases

Morphine (A)

Minimal

2–3 minutes

Hydromorphone

8.2

50

+

<10

295 L

UGT2B7

UGT1A3

H3G (I)

H6G (A)

6

2–3 hours

Tramadol

9.4

70–90

+

20

2.6–2.9 L/kg

CYP3A4

CYP2D6

M1

90

6 hours

Buprenorphine

8.4

15

+++

96

430 L

CYP3A4

UGT1A1/1A3

Minimal

*Complicated by enterohepatic recirculation

Meperidine (pethidine)

8.5

++

60–80

3–5 L/kg

CYP3A4

CYP2B6

CYP2C19

Norpethidine (A, CNS+)

5

3–6 hours

Oxycodone

8.5

60–87

+

45

2–3 L/kg

CYP3A4

CYP2D6

Oxymorphone (A)

< 10

3–4 hours

Methadone

8.3

60–90

++

60–90

3–6 L/kg

CYP3A4

CYP2B6

(CYP2D6,2C9,2C19,1A2)

15–60

15–20 hours

(13–47)

Fentanyl

8.4

+++

90

3–8 L/kg

CYP3A4

< 7

2–7 hours

Alfentanil

6.5

++

90

0.4–1 L/kg

CYP3A4

Minimal

1–2 hours

Enzymes: CYP, cytochrome P450, UGT, UDP-glucuronosyltransferase. Metabolites: M3G, morphine-3-glucuronide, M6G, morphine-6-glucuronide H3G, Hydromorphone-3-glucuronide, H6G, Hydromorphone-6-glucuronide, M1, O-desmethyl tramadol. Active metabolites: A, analgesically active, CNS+, CNS excitability.

Source data from: Medicines and Healthcare Products Regulatory Agency (MHRA), available from <http://www.mhra.gov.uk>; Drugs.com, <http://www.drugs.com>, Copyright © 2000–2014 Drugs.com. All rights reserved.; Twycross R. WA (ed), Palliative Care Formulary 4+, Palliativedrugs.com Ltd, Copyright © 2012;
Rook EJ et al., Pharmacokinetics and pharmacokinetic variability of heroin and its metabolites: Review of the literature, Current Clinical Pharmacology, Volume 1, Issue 1, pp. 109–18, Copyright © 2006reference
; and
Ashley C et al. (ed), The Renal Drug Handbook, Third Edition, Radcliffe Publishing, Oxford, UK, Copyright ©2008.

The two major organs of elimination are the liver and kidneys, both of which are susceptible to pharmacological and pathophysiological sources of variability. Clearance is defined as the volume of blood which is completely cleared of the drug in a unit of time and reflects the efficiency of the elimination process. It is usually measured in mL/minute or L/hour. It is a major determinant of t  ½ and of the steady-state drug concentration.

This is perhaps the most well-known and commonly used pharmacokinetic parameter. The elimination half-life (t  ½) is a measure of the time taken for half the drug in the body to be removed and generally correlates closely with duration of action. After repeated dosing is initiated, or the dose of an existing regimen is changed, five to six half-lives are required to approach steady-state concentration, irrespective of the route of administration or dosing interval. Drugs with a long t  ½ accumulate for a relatively prolonged period of time, and as a result, the concentration may surpass the effective therapeutic range and build up to toxic levels. In the clinical setting, this accumulation has largely been a problem during methadone therapy. Methadone is exceptionally complex because its slow elimination phase is highly variable (beta t  ½, 15–60 hours) and preceded by a rapid distribution phase (t  ½, 2–3 hours); the overall half-life is relatively long (> 20 hours).

The aim of any dosing regimen in an individual patient is to achieve a concentration of drug in the blood that is high enough to give the intended effect without producing side effects. This concentration can never be completely steady as peaks will occur at the point of maximum drug absorption after administration, and troughs will occur immediately before each dose (Fig. 9.1.1). The degree of swing between peak and trough concentrations is determined by the drug’s elimination t  ½ and the frequency of drug administration.

 Steady-state plasma concentration of a short-acting drug showing peaks and troughs after each dose.
Fig. 9.1.1

Steady-state plasma concentration of a short-acting drug showing peaks and troughs after each dose.

The time taken for a drug to reach steady-state plasma concentration is dependent on the half-life. As noted, five to six half-lives are required to approach steady-state drug concentration if the same dose of drug is given at a constant time interval; four t  ½ yields approximately 95% of this concentration. This applies only to drug where elimination is governed by ‘first-order’ kinetics. Fortunately, this comprises the vast majority of drugs, including opioids. Phenytoin is a notable exception, which involves both ‘first-’ and ‘zero-order’ processes.

The t  ½ of morphine is 2–4 hours. Therefore, when morphine is administered every 4 hours, steady state will be 95% achieved after approximately 16 hours. Titration of the dose on a daily basis ensures that dose changes are occurring at steady state. In contrast, the methadone requires approximately 4–7 days, and occasionally much longer, to achieve steady state. Taking this into consideration a loading dose of methadone is often used followed by a period of cautious titration (after a number of days) to minimize the risk of toxicity.

Drugs produce their effects on the body by binding with receptors, modifying enzyme processes, or by direct chemical or physical actions. Opioids exert their influence by interacting with opioid receptors (primarily the mu opioid receptor).

Receptors are specialized proteins within the cell membrane which are integral for communication between the cell and the outside world. They are highly specific for certain ligands, such as specific hormones, cytokines and/or drugs.

Opioid receptors were originally classified by pharmacological activity and later by molecular sequencing (Pasternak, 2004). There are three types of classical opioid receptor: μ (mu) or MOR, κ (kappa) or KOR, and δ (delta) or DOR. Another opioid-like receptor has been identified named the nociceptin or orphanin FQ peptide receptor or NOR (Mollereau et al., 1994).

The three classical receptors are activated differentially by the endogenous opioids (encephalins, endorphins, and dynorphins) (Table 9.1.2). Exogenous opioids, such as morphine, act primarily at the MOR. Different opioids may show differential binding to sites on the MOR and may also bind to other opioid or non-opioid receptors (Pasternak and Pan, 2011).

Table 9.1.2
Classical opioid receptor genes, distribution, endogenous ligands, and function
ReceptorGeneExpression*Endogenous ligandFunction

Mu (µ)

MOR

OPRM1

Central nervous system:

Brain including cerebral cortex, thalamus, hypothalamus, striatum, amygdala, periaqueductal grey.

Spinal cord, pre- and postsynaptic neurons

Peripheral nervous system

Immune cells

Beta-endorphin

Encephalins

Endomorphins

Analgesia

Respiratory depression

Reduced GI motility

Miosis

Euphoria

Sedation

Physical dependence

Kappa (κ)

KOR

OPRK1

Central nervous system:

Brain including cerebral cortex, thalamus, hypothalamus, striatum, periaqueductal grey

Spinal cord

Peripheral nervous system

Dynorphins

Analgesia

Miosis

Dysphoria

Hallucinations

Sedation

Delta (δ)

DOR

OPRD1

Central nervous system:

Brain including cerebral cortex, striatum, olfactory bulb

Peripheral nervous system

Encephalins

Beta-endorphin

Analgesia

Respiratory depression

Reduced gastrointestinal motility

Tolerance

Mood regulation

ReceptorGeneExpression*Endogenous ligandFunction

Mu (µ)

MOR

OPRM1

Central nervous system:

Brain including cerebral cortex, thalamus, hypothalamus, striatum, amygdala, periaqueductal grey.

Spinal cord, pre- and postsynaptic neurons

Peripheral nervous system

Immune cells

Beta-endorphin

Encephalins

Endomorphins

Analgesia

Respiratory depression

Reduced GI motility

Miosis

Euphoria

Sedation

Physical dependence

Kappa (κ)

KOR

OPRK1

Central nervous system:

Brain including cerebral cortex, thalamus, hypothalamus, striatum, periaqueductal grey

Spinal cord

Peripheral nervous system

Dynorphins

Analgesia

Miosis

Dysphoria

Hallucinations

Sedation

Delta (δ)

DOR

OPRD1

Central nervous system:

Brain including cerebral cortex, striatum, olfactory bulb

Peripheral nervous system

Encephalins

Beta-endorphin

Analgesia

Respiratory depression

Reduced gastrointestinal motility

Tolerance

Mood regulation

Source: data from
Peckys D and Landwehrmeyer GB, Expression of mu, kappa, and delta opioid receptor messenger RNA in the human CNS: A 33P in situ hybridization study, Neuroscience, Volume 88, Issue 4, pp. 1093–135, Copyright © 1999reference
;
Stein C et al., Attacking pain at its source: new perspectives on opioids. National Medicine, Volume 9, Issue 8, pp. 1003–8, Copyright © 2003reference
; and
Borner C et al, Comparative analysis of mu-opioid receptor expression in immune and neuronal cells, Journal of Neuroimmunology, Volume 18, Issue 1–2, pp. 56–63, Copyright © 2007.reference

The opioid receptors belong to the superfamily of G protein-coupled receptors. Each consists of an extracellular N-terminus, seven transmembrane helices, three extra and intracellular loops, and an intracellular C-terminus. Each receptor type is coded for by a different gene (Meng et al., 1993; Knapp et al., 1994; Wang et al., 1994). The three receptors share a high degree of homology with most variation found in the extracellular loops and N-terminal domains (Minami and Satoh, 1995). The extracellular loops are particularly important as they determine ligand binding. Opioid receptors are widely, yet differentially, distributed in both central and peripheral nervous systems (Table 9.1.2) (Minami and Satoh, 1995).

The µ-opioid receptor is clinically the most important of the family is, as it is responsible for the inhibition of nociceptive pathways and is exploited by all exogenous opioids. Knockout studies in mice show that the MOR is essential for morphine-induced analgesia (Matthes et al., 1996). Many of the unwanted effects of opioids are also related to activity at this receptor (Table 9.1.2). The MOR is expressed on central and peripheral neurons, and the latter are up-regulated in response to inflammatory stimuli. Peripherally, MOR are found pre- and postsynaptically. For example, approximately 70% of MOR receptors in the dorsal horn are expressed on the primary afferent terminations (presynaptic), where they modulate afferent transmission (Stein et al., 2003). At a cellular level, µ-opioid receptor activation results in an overall inhibitory effect (Box 9.1.2).

Box 9.1.2
Downstream consequences of µ-opioid receptor activation

Inhibition of adenylyl cyclase

Increased opening of potassium channels (hyperpolarization of post-synaptic neurons, reduced synaptic transmission)

Inhibition of calcium channels (decreases pre-synaptic neurotransmitter release).

δ- and κ-opioid receptors also are involved in the modulation of pain. Knock-out studies in mice show that KOR may influence chemical visceral pain and thermal nociception (Simonin et al., 1998). Studies with selective opioid antagonists suggest that oxycodone analgesia depends on binding to the KOR receptor (Smith et al., 2001); other studies indicate that oxycodone is more like morphine and exerts its analgesic effects through MOR activation (Kalso et al., 1990; Chen et al., 1991; Yoburn et al., 1995). Pharmacological studies also suggest a role for KOR in mediating the dysphoric and sedative effects of opioids (Mark, 1990).

Combination opioid receptor knockout studies suggest that DOR plays a role in modulating mechanical and inflammatory pain (Martin et al., 2003). δ-opioid receptor knock-out mice also do not exhibit analgesic tolerance to morphine (Nitsche et al., 2002).

G-protein-coupled receptors, including MOR, KOR and DOR, have been shown to form different configurations, including homo- and hetero-dimers and oligomers, with unique internalization and activation pathways. Dimerization modulates receptor pharmacology and this process could present targets for novel interventions (Milligan, 2005). The development of MOR-DOR heteromers exemplifies this potential. There is evidence that there is an increased abundance of MOR–DOR heteromers in chronic pain and/or chronic exposure to morphine, and that ligand binding to DOR in the context of MOR-DOR is not associated with the development of opioid tolerance, in contrast to ligand binding to DOR alone. MOR-DOR may represent a new pharmacological target with the potential induce analgesia without tolerance (Costantino et al., 2012).

Genetic polymorphism caused by alternative splicing of mRNA has been shown to give rise to various human MOR receptor subtypes. Much of the variation is found in the intracellular C-terminal domain and includes the creation of potential phosphorylation sites (Pasternak and Pan, 2011). These receptor subtypes have differential expression patterns and demonstrate activation profiles that vary among the various MOR agonists; they are likely to explain some of the clinical variation in opioid response (Pasternak, 2004). Genetic polymorphism also may be involved individual variation in pain responses. For example, the minor T allele of OPRM1 rs563649 is associated with higher expression levels of the MOR-1K isoform and also with high pain sensitivity (Shabalina et al., 2009).

Other cellular adaptations are likely to be involved in the varied responses to chronic opioid exposure, such as variation in the development of tolerance (Ferguson, 2001; Bailey and Connor, 2005). Chronic morphine exposure leads to little change in MOR expression but does seem to produce changes in the non-neuronal population of glial cells, with increased activation provoking central sensitization (Watkins et al., 2005).

Although it is widely accepted that clinically relevant pharmacological tolerance to opioid analgesic effects is not an issue for the majority of patients with cancer-related pain, it is difficult to assess analgesic tolerance clinically (Chang et al., 2007). There does not appear to be a simple correlation between exposure to opioids and induction of analgesic tolerance. A process of adaptation occurs, which is likely to depend on diverse factors, but this process cannot be fully explained on the basis of current knowledge of cellular mechanisms.

Based on their interactions with receptors, opioid compounds can be divided into agonist, agonist–antagonist, and antagonist classes (Table 9.1.3). Most opioids used in the clinical setting are full agonist drugs. Opioid antagonists, such as naloxone and naltrexone, bind to MOR and produce no agonist activity. Agonist–antagonist drugs include a group of mixed agonist–antagonists, which are agonists at one or more opioid receptor subtypes and antagonists at others, and partial agonists. The mixed agonist–antagonists, such as pentazocine, are seldom used in the management of patients with advanced illness. Buprenorphine is a partial agonist that is being used more as an analgesic with the advent of a transdermal delivery system. Buprenorphine is believed to have a ceiling effect at doses of 8–16 mg/day (Walsh and Eissenberg, 2003). However as the recommended analgesic doses are much lower than the ceiling dose (equivalent to up to 3–4 mg/day, or 2 × 70-microgram/hour patches), buprenorphine typically is used with doses in the linear part of the dose-response curve and clinically performs as a full agonist during the management of pain. At the higher doses used to treat heroin addiction, the partial agonist effect may be encountered (Greenwald et al., 2003).

Table 9.1.3
Table of opioid receptor agonists and antagonists
Receptor effectDescriptionExamples

Agonists

An agonist is a drug that has affinity for and binds to cell receptors to induce changes in the cell that stimulate physiological activity

The agonist opioid drugs have no clinically relevant ceiling effect to analgesia

Morphine

Diamorphine

Oxycodone

Pethidine

Hydromorphone

Methadone

Fentanyl

Tramadol

Partial agonist

A partial agonist has low intrinsic activity (efficacy) so that its dose–response curve exhibits a ceiling effect at less than the maximum effect produced by a full agonist

Buprenorphine

Antagonist

Antagonist drugs have no intrinsic pharmacological action but can interfere with the action of an agonist

Competitive antagonists bind to the same receptor and compete for receptor sites, whereas non-competitive antagonists block the effects of the agonist in some other way

Naloxone

Naltrexone

Mixed agonist–antagonist

The mixed agonist–antagonist drugs produce agonist effects at one receptor and antagonist effects at another

Pentazocine

Butorphanol

Nalbuphine

Receptor effectDescriptionExamples

Agonists

An agonist is a drug that has affinity for and binds to cell receptors to induce changes in the cell that stimulate physiological activity

The agonist opioid drugs have no clinically relevant ceiling effect to analgesia

Morphine

Diamorphine

Oxycodone

Pethidine

Hydromorphone

Methadone

Fentanyl

Tramadol

Partial agonist

A partial agonist has low intrinsic activity (efficacy) so that its dose–response curve exhibits a ceiling effect at less than the maximum effect produced by a full agonist

Buprenorphine

Antagonist

Antagonist drugs have no intrinsic pharmacological action but can interfere with the action of an agonist

Competitive antagonists bind to the same receptor and compete for receptor sites, whereas non-competitive antagonists block the effects of the agonist in some other way

Naloxone

Naltrexone

Mixed agonist–antagonist

The mixed agonist–antagonist drugs produce agonist effects at one receptor and antagonist effects at another

Pentazocine

Butorphanol

Nalbuphine

Efficacy is defined by the maximal response induced by administration of the active agent. In practice, this is determined by the degree of analgesia produced following dose escalation through a range limited by the development of adverse effects. Potency, in contrast, reflects the dose–response relationship and is typically defined by the intensity of a specified effect, such as analgesia, associated with a specific dose. Potency is influenced by pharmacokinetic factors (i.e. how much of the drug enters the body’s systemic circulation and then reaches the receptors) and by affinity to drug receptors.

Clinically, the utility of potency measurements is created by comparing drugs using relative potency ratios, or the ratio of doses required to produce the same analgesic effect. The relative potency of each of the commonly used opioids is based upon a comparison with 10 mg of oral morphine. Data from single-and repeated-dose studies in patients with acute or chronic pain have been used to develop ‘equianalgesic’ or dose conversion tables. The term ‘equianalgesic’ is however misleading as there is wide inter-individual differences in response to different opioids and such tables should be used only as a guide when switching between opioids (Riley et al., 2006). In particular, care should be exercised when switching from one opioid to another as part of the management of opioid toxicity. In this situation, conservative conversions should be used followed by individual titration. Opioid switching is discussed in detail in Chapter 9.4.

The clinical utility of an opioid therapy is determined by a favourable balance between analgesic efficacy and side effects. Many variables may influence whether a dose exists that yields this balance. These include intensity of pain; prior opioid exposure in terms of drug, duration, and dose (and the degree of cross-tolerance that this confers); age; route of administration; level of consciousness and metabolic abnormalities; and genetic polymorphism in the expression of relevant enzymes or receptors (Droney et al., 2012).

The clinical use of combinations of different opioids is increasing with the aims to (a) improve analgesia, (b) reduce side effects, and (c) limit the development of opioid tolerance. The rationale behind this practice is to utilize the inherent differences in the pharmacodynamic and pharmacokinetic properties of this group of drugs to maximize potential benefit and minimize adverse effects (Fallon and Laird, 2011). Hypotheses of the pharmacodynamic mechanisms include splice variation in opioid receptors, the receptor activation versus endocytosis (RAVE) theory, and the formation of opioid receptor homo- and heterodimers which results in changes to G-protein signalling cascades (Davis et al., 2005).

In animal studies, there is some evidence of analgesic synergism between methadone and other µ-agonist opioids (Bolan et al., 2002). In addition two small retrospective case series of patients with uncontrolled cancer-related pain have reported that low dose methadone in combination with existing opioid improved analgesia (McKenna and Nicholson, 2011; Haughey et al., 2012).

A combination product of oxycodone with morphine in a fixed-dose ratio 3:2 is currently being evaluated in clinical trials (MoxDuo®). It has been trialled in phase II and phase III studies for the management of acute postoperative pain and results suggest that there may be a difference in side effect profile when compared with the individual opioids (Webster et al., 2010; Richards et al., 2011; Webster, 2012). There are currently no trials published in the chronic pain or cancer pain setting.

Further research into the use of opioid combinations in warranted. The potential benefits from this strategy must be weighed against other factors, such as poor patient compliance, confusion over dosing, and prescriber dosing errors, along with potentially unanticipated increased side effects.

Some drugs exert effects by affecting enzyme processes, rather than by binding to receptors. Many act by inhibition of enzyme actions. For example, non-steroidal anti-inflammatory drugs block the effect of the enzyme cyclooxygenase and thereby interfere with the synthesis of prostaglandins and exert anti-inflammatory activity.

Other drugs produce intended effects through a direct chemical or physical action. Antacids are an example of drugs with a direct chemical action; they are bases which neutralize gastric acid. Drugs with a physical mode of action include the bulk laxatives, such as ispaghula husk.

Patients with palliative care needs may already be receiving drugs for a variety of conditions. Some may still be beneficial but others may no longer contribute to improving prognosis or symptoms. Rationalization of the therapeutic regimen always should be considered. If further drugs to relieve symptoms are added, this adds to the potential for drug interaction. Interaction is adverse if it causes therapeutic failure or toxicity from any one drug. Remembering all the possible drug interactions is virtually impossible, but knowledge of the underlying mechanisms of drug interaction can put the prescriber on guard together with frequent consultation with prescribing information is important.

An adverse drug reaction can be defined as an unwanted or harmful reaction experienced following administration of a drug, or combination of drugs, under normal conditions of use that is suspected of being related to the drug. For example opioids act via the µ-opioid receptor to slow gut transit and cause constipation. Opioid-related side effects are summarized in Box 9.1.3 and can be transient or persistent. Chapter 9.4 describes in more detail the principles of opioid switching which can reduce individual adverse drug reactions.

Box 9.1.3
Opioid side effects listed by system
Gastrointestinal system

Nausea

Constipation

Dry mouth

Vomiting

Ileus

Nervous system

Somnolence

Confusion

Myoclonus

Abnormal dreams

Hallucinations

Hyperalgesia

Genitourinary system

Urinary retention

Respiratory system

Cough decreased

Respiratory depression

Skin

Hyperhidrosis

Pruritus

Endocrine

Hypogonadism

Immunosuppression

Opioid analgesics are one of the drugs most frequently associated with adverse drug events. A study of 3695 inpatient adverse drug reactions found that 16% were attributable to opioids (Davies et al., 2009). Risk of opioid drug reactions increases in older patients, in those with underlying cardiac or respiratory disease, and when co-prescribed with other sedative medications such as benzodiazepines (Bernard and Bruera, 2000).

Pharmacokinetic interaction arises through alterations in the rate and extent of absorption and changes in metabolism (both pre-systemic and elimination), distribution, and renal excretion. The clinical impact of theoretical interactions can be difficult to predict.

Drugs such as metoclopramide and anticholinergics, which alter the rate of gastric emptying, may affect the speed of absorption of other agents. Some drugs bind others in the gastrointestinal tract and affect their bioavailability. For example, care is necessary if antacid preparations, iron salts, or cholestyramine are used concurrently with certain drugs.

Drug interactions resulting from changes in the rate of metabolism by the liver will result both in changes of bioavailability for those drugs with a significant first-pass effect, and decreased clearance. Steady-state concentrations of drug may be profoundly affected.

A number of drugs (particularly phenobarbital, carbamazepine, phenytoin and rifampicin) are capable of inducing the cytochrome P450 and glucuronidase enzymes in the liver. There are a myriad of substrates for this interaction, including methadone, warfarin, corticosteroids, and anticonvulsant drugs. Increased pre-systemic metabolism may result in the need to increased doses to achieve therapeutic levels. Conversely some drugs may inhibit CYP enzymes, individually or as an entire superfamily (Table 9.1.4). Certain foodstuffs may also induce or inhibit hepatic enzyme systems; for example, grapefruit contains furanocoumarins which inhibit CYP3A (Hanley et al., 2011).

Table 9.1.4
Potential for drug interactions involving the cytochrome P450 enzyme system
Inhibitors −CYP1A2CYP2B6CYP2C8CYP2C9CYP2C19CYP2D6CYP2E1CYP3A4

Amiodarone

Clopidogrel

Amiodarone

Amiodarone

Celecoxib

Amiodarone

Alcohol (acute use)

Amiodarone

Ciprofloxacin

Paroxetine

Fluconazole

Fluconazole

Esomeprazole

Celecoxib

Disulfiram

Bicalutamide

Diclofenac

Sertraline

Ibuprofen

Ibuprofen

Fluconazole

Duloxetine

Clarithromycin

Fluvoxamine

Omeprazole

Metronidazole

Fluoxetine

Fluoxetine

Diclofenac

Pantoprazole

Miconazole

Lansoprazole

Haloperidol

Diltiazem

Quinine

Omeprazole

Modafinil

Levomepromazine

Erythromycin

Trimethoprim

Pantoprazole

Omeprazole

Methadone

Fluconazole (high dose)

Quinine

Rabeprazole

Paroxetine

Grapefruit juice

Sertraline

Quinine

Haloperidol

Sertraline

Imatinib

Itraconazole

Verapamil

Inhibitors −CYP1A2CYP2B6CYP2C8CYP2C9CYP2C19CYP2D6CYP2E1CYP3A4

Amiodarone

Clopidogrel

Amiodarone

Amiodarone

Celecoxib

Amiodarone

Alcohol (acute use)

Amiodarone

Ciprofloxacin

Paroxetine

Fluconazole

Fluconazole

Esomeprazole

Celecoxib

Disulfiram

Bicalutamide

Diclofenac

Sertraline

Ibuprofen

Ibuprofen

Fluconazole

Duloxetine

Clarithromycin

Fluvoxamine

Omeprazole

Metronidazole

Fluoxetine

Fluoxetine

Diclofenac

Pantoprazole

Miconazole

Lansoprazole

Haloperidol

Diltiazem

Quinine

Omeprazole

Modafinil

Levomepromazine

Erythromycin

Trimethoprim

Pantoprazole

Omeprazole

Methadone

Fluconazole (high dose)

Quinine

Rabeprazole

Paroxetine

Grapefruit juice

Sertraline

Quinine

Haloperidol

Sertraline

Imatinib

Itraconazole

Verapamil

SubstratesCYP1A2CYP2B6CYP2C8CYP2C9CYP2C19CYP2D6CYP2E1CYP3A4

Amitriptyline

Diclofenac

Diclofenac

Amitriptyline

Amitriptyline

Amitriptyline

Domperidone

Alfentanil

Metronidazole

Domperidone

Ketamine

Ibuprofen

Celecoxib

Citalopram

Codeine

Paracetamol

Amitriptyline

Midazolam

Duloxetine

Methadone

Naproxen

Diclofenac

Clopidogrel

Duloxetine

Theophylline

Carbamazepine

Mirtazapine

Flutamide

Omeprazole

Fluoxetine

Diazepam

Fluoxetine

Citalopram

Modafinil

Haloperidol

Repaglinide

Gliclazide

Diclofenac

Haloperidol

Clonazepam

Omeprazole

Methadone

Rosiglitazone

Glimepiride

Esomeprazole

Methadone

Dexamethasone

Ondansetron

Mirtazapine

Tamoxifen

Glipizide

Ibuprofen

Methylphenidate

Diazepam

Oxycodone

Naproxen

Ibuprofen

Lansoprazole

Metoclopramide

Domperidone

Pantoprazole

Olanzapine

Ketamine

Methadone

Mirtazapine

Esomeprazole

Quinine

Ondansetron

Methadone

Naproxen

Omeprazole

Etoricoxib

Rabeprazole

Paracetamol

Metronidazole

Omeprazole

Ondansetron

Exemestane

Reboxetine

Ropinirole

Naproxen

Pantoprazole

Oxycodone

Fentanyl

Risperidone

Theophylline

Omeprazole

Phenobarbital

Paroxetine

Finasteride

Sertraline

Warfarin

Tamoxifen

Rabeprazole

Promethazine

Granisetron

Simvastatin

Warfarin

Sertraline

Risperidone

Haloperidol

Tamoxifen

Warfarin

Sertraline

Ketamine

Trazodone

Tamoxifen

Medroxyprogesterone

Venlafaxine

Tramadol

Methadone

Zopiclone

Trazodone

Methylphenidate

Venlafaxine

SubstratesCYP1A2CYP2B6CYP2C8CYP2C9CYP2C19CYP2D6CYP2E1CYP3A4

Amitriptyline

Diclofenac

Diclofenac

Amitriptyline

Amitriptyline

Amitriptyline

Domperidone

Alfentanil

Metronidazole

Domperidone

Ketamine

Ibuprofen

Celecoxib

Citalopram

Codeine

Paracetamol

Amitriptyline

Midazolam

Duloxetine

Methadone

Naproxen

Diclofenac

Clopidogrel

Duloxetine

Theophylline

Carbamazepine

Mirtazapine

Flutamide

Omeprazole

Fluoxetine

Diazepam

Fluoxetine

Citalopram

Modafinil

Haloperidol

Repaglinide

Gliclazide

Diclofenac

Haloperidol

Clonazepam

Omeprazole

Methadone

Rosiglitazone

Glimepiride

Esomeprazole

Methadone

Dexamethasone

Ondansetron

Mirtazapine

Tamoxifen

Glipizide

Ibuprofen

Methylphenidate

Diazepam

Oxycodone

Naproxen

Ibuprofen

Lansoprazole

Metoclopramide

Domperidone

Pantoprazole

Olanzapine

Ketamine

Methadone

Mirtazapine

Esomeprazole

Quinine

Ondansetron

Methadone

Naproxen

Omeprazole

Etoricoxib

Rabeprazole

Paracetamol

Metronidazole

Omeprazole

Ondansetron

Exemestane

Reboxetine

Ropinirole

Naproxen

Pantoprazole

Oxycodone

Fentanyl

Risperidone

Theophylline

Omeprazole

Phenobarbital

Paroxetine

Finasteride

Sertraline

Warfarin

Tamoxifen

Rabeprazole

Promethazine

Granisetron

Simvastatin

Warfarin

Sertraline

Risperidone

Haloperidol

Tamoxifen

Warfarin

Sertraline

Ketamine

Trazodone

Tamoxifen

Medroxyprogesterone

Venlafaxine

Tramadol

Methadone

Zopiclone

Trazodone

Methylphenidate

Venlafaxine

Inducers +CYP1A2CYP2B6CYP2C8CYP2C9CYP2C19CYP2D6CYP2E1CYP3A4

Carbamazepine

Carbamazepine

Carbamazepine

Carbamazepine

Carbamazepine

Alcohol (chronic use)

Carbamazepine

Phenobarbital

Modafinil

Phenobarbital

Phenobarbital

Phenobarbital

Phenobarbital

Dexamethasone

Rifampicin

Phenobarbital

Rifampicin

Rifampicin

Modafinil

Tobacco

Rifampicin

Phenobarbital

Phenytoin

Rifampicin

St John’s wort

Inducers +CYP1A2CYP2B6CYP2C8CYP2C9CYP2C19CYP2D6CYP2E1CYP3A4

Carbamazepine

Carbamazepine

Carbamazepine

Carbamazepine

Carbamazepine

Alcohol (chronic use)

Carbamazepine

Phenobarbital

Modafinil

Phenobarbital

Phenobarbital

Phenobarbital

Phenobarbital

Dexamethasone

Rifampicin

Phenobarbital

Rifampicin

Rifampicin

Modafinil

Tobacco

Rifampicin

Phenobarbital

Phenytoin

Rifampicin

St John’s wort

Adapted with permission from
Andrew Dickman, Drugs in Palliative Care, Second Edition, Oxford University Press, Oxford UK, Copyright © 2012 Andrew Dickman by permission of Oxford University press.reference

The most important drug interactions in the kidney involve competition between agents for active tubular secretion. Active tubular secretion is used by organic acids, and the most frequent interactions are caused by the loop diuretics and some non-steroidal anti-inflammatory drugs. Although renal excretion of some drugs is pH dependent, in general this has minor implications in normal therapeutics. There are a few exceptions, however; for example, methadone’s renal clearance is considerably enhanced by concurrent use of urinary acidifiers such as acetazolamide (Bellward et al., 1977).

Some drug–drug interactions occur at a receptor level. For example, buprenorphine is a partial agonist and morphine is a full agonist, and morphine-induced analgesia may be reversed or limited by competition at the receptor level if buprenorphine is added.

The preferred route of administration for many drugs including opioids is oral. Oral formulations of opioids include immediate-release (IR) syrups, tablets or capsules, and modified-release (MR) tablets or capsules. MR formulations slowly release the drug into the gut, allowing treatment once or twice daily depending on the formulation (e.g. morphine, oxycodone). Care must be taken in prescribing, such that the correct formulation is dispensed, and when more than one formulation is given (e.g. IR and MR) the patient understands how and when to use each preparation.

Opioid drugs can also be given by other routes, including, transdermal, transmucosal (sublingual, buccal, nasal, rectal), and parenteral by injection either intravenously or more commonly subcutaneously. Fentanyl and buprenorphine products have been formulated for the transmucosal and transdermal routes. In addition specialist pain services may use epidural or intrathecal opioids in the palliative setting where appropriate. Prescriptions of these drugs should specify the exact formulation, as formulations differ in systemic availability and may not be interchangeable.

IR preparations are absorbed in the stomach or proximal small bowel, so that absorption is complete within a few hours on ingestion. For example, IR morphine and oxycodone reach peak effects within 1 hour. Modified- or sustained-release formulations allow a drug to be released over 12–24 hours, resulting in a smoother concentration profile of the drug in the blood, extended duration of action, and reduction in tablet burden for the patient.

Drugs that are absorbed through the buccal, nasal, or rectal mucosa avoid first-pass metabolism in the liver by uptake into veins that drain directly into the systemic circulation. This results in higher bioavailability and often a faster onset of action when compared to the oral route. IR transmucosal fentanyl products may be useful in treatment of breakthrough/incident pain. The rapid speed of onset of action (approximately 10 minutes) and short duration of action (≥ 1 hour) may sometimes be more suited to the temporal characteristics of breakthrough/incident pain than conventional IR opioid (Twycross et al., 2012; Davies et al., 2013). There does not, however, appear to be a meaningful relationship between background opioid dose and the effective dose of transmucosal fentanyl, and therefore, titration is essential (Zeppetella, 2011).

Some lipid-soluble drugs are well absorbed through the skin, and their transdermal delivery via ‘patches’ allows controlled release over many hours or days. Opioid examples include fentanyl and buprenorphine. Different formulations have been developed and are not interchangeable and preparations may last for 3–7 days depending on drug/formulation. Clinical trials suggest good patient satisfaction with this mode of delivery. Care must be taken, given the wide variability in drug absorption, especially in cachexic or pyrexial patients (Heiskanen et al., 2009).

The preferred parenteral route of administration in palliative patients is subcutaneous and opioids may be given as stat injections or as a continuous subcutaneous infusion. When continuous subcutaneous infusions are used and multiple drugs combined, care must be taken to ensure drug interactions are avoided, as precipitation of one drug in solution will clearly limit therapeutic effect (e.g. cyclizine and oxycodone are not compatible) (Dickman et al., 2002).

Combination products are attractive and may aid compliance by reducing tablet burden. To be effective, the frequency of administration of the two drugs should be the same. Combination products to not allow for titration of one drug without the other, and this may be a concern with some combinations. For example, concerns have been raised regarding combinations of opioid analgesics with paracetamol because of the need to limit titration of the paracetamol and risk of liver damage in overdose.

Compliance or adherence is the extent to which a patient follows a prescribed drug regimen. It is important that decisions regarding treatment are jointly made by the prescriber and patient. Allowing adequate time to explain principles and goals of therapy, expected benefit and possible side effects, and plans for review and follow-up is essential. In addition, exploration of patient (and family) concerns regarding addiction, tolerance, side effects, or fear that treatment implies the final stages of life are essential when using opioids for patients with advanced illness (NICE, 2012). The proactive management or prevention of side effects, for example, provision of laxatives for opioid-induced constipation, can improve compliance. In general, more complex regimens with high frequency of administration and/or multiple drugs reduce compliance.

Pharmacogenomics is the study of how genetic variation influences response to drugs. It is the cornerstone of personalized medicine, which aims to tailor treatment to the individual to maximize efficacy and minimize adverse reactions. Two techniques have been used to study pharmacogenomics: the candidate gene approach and genome-wide association. The candidate gene approach targets single nucleotide polymorphisms (SNPs) in genes already known to be important in pharmacokinetic (e.g. drug metabolizing enzymes, drug transporters) and pharmacodynamic (e.g. receptors, ion channels, enzymes) pathways. Genome-wide association studies cast a much wider net examining millions of SNPs across the entire genome at a time and may therefore provide new biological insights into mechanisms (Wilke et al., 2008).

In recent years, the field of pharmacogenomics has exploded to provide a wealth of information to inform personalized prescribing across the medical specialties. In oncology, response to certain chemotherapy agents can now be predicted, for example, variation in UGT1A1 is associated with severe neutropenia from irinotecan (Innocenti et al., 2004). In cardiology, response to warfarin, statins, and clopidogrel have all been associated with genetic factors (Johnson and Cavallari, 2013). In HIV medicine, screening programmes have been used to reduce the risk of hypersensitivity reactions to abacavir by testing for HLA B*5701, which is associated with the condition (Mallal et al., 2008). Work continues on how this knowledge may best be translated into clinical practice (Johnson et al., 2012).

Study of the CYP enzyme 2D6 (CYP2D6) gene has provided perhaps the best examples of how pharmacokinetics and ultimately opioid response is linked to genetic variation. CYP2D6 is involved in the metabolism of several opioids including codeine, tramadol, and oxycodone. Over 70 CYP2D6 alleles have been described which directly affect the final protein; these include SNPs, deletions, insertions, and copy number variation (Leandro-Garcia et al., 2009). The sum functional effect of this variation has been classified into four main phenotypes: poor, intermediate, extensive, and ultrarapid metabolizers.

Codeine is partially (10%) metabolized to morphine by CYP2D6 (Lotsch, 2005). Approximately 10% of Caucasians are poor metabolizers and experience little analgesia from codeine (Sindrup et al., 1990; Persson et al., 1995). Conversely 3% of Caucasians are ultrarapid metabolizers and have a higher incidence of codeine-related adverse reactions (Kirchheiner et al., 2007). There have been case reports of fatal neonatal opioid toxicity in children breastfed by mothers who are ultrarapid metabolizers following ingestion of codeine (Madadi et al., 2007). The CYP2D6 phenotype has also been suggested to affect response to tramadol and oxycodone by altering ratios of the parent opioid to the more active metabolites (Samer et al., 2003, 2010b), although the clinical relevance of this is debated (Gronlund et al., 2010; Samer et al., 2010a, 2010b; Andreassen et al., 2012).

Pharmacodynamic candidate gene studies in palliative care patients suggest that opioid receptor SNPs, for example, OPRM1 A118G, influence patients’ requirements for opioids (Klepstad et al., 2004; Campa et al., 2008; Walter and Lotsch, 2009). Individual pain susceptibility also influences analgesic response, and therefore, many more candidate genes from pain signalling and modulatory pathways, for example, COMT (Rakvag et al., 2005; Rakvag et al., 2008), also may be important in opioid responsiveness.

Pain experience and opioid response are complex traits and therefore influenced by a myriad of gene–gene and gene–environment interactions. Recently, genetic association studies have begun to explore interactions between variants from more than one gene. This has thus far been limited to two candidate SNPs at a time (Reyes-Gibby et al., 2007; Campa et al., 2008). The concept of gene–gene/environment interactions or epistasis provides a huge challenge for the future of opioid pharmacogenetics, both practical and analytical. Further work needs to be done to unpick the complexities behind opioid response to be able to develop a useful predictive tool to inform clinical practice.

Ashley, C. and Currie, A. (2008).

The Renal Drug Handbook
(3rd ed.). Oxford: Radcliffe Publishing.

Bernard, S.A. and Bruera, E.  

2000
.
Drug interactions in palliative care.
 
Journal of Clinical Oncology
, 18, 1780–1799.

Borner, C., Stumm, R., Hollt, V., and Kraus, J. (

2007
).
Comparative analysis of mu-opioid receptor expression in immune and neuronal cells.
 
Journal of Neuroimmunology
, 188(1–2), 56–63.

Caraceni, A., Hanks, G., Kaasa, S., et al. (

2012
).
Use of opioid analgesics in the treatment of cancer pain: evidence-based recommendations from the EAPC.
 
The Lancet Oncology
, 13, e58–e68.

Dickman A. (2012).

Drugs in Palliative Care
(2nd ed.). Oxford: Oxford University Press.

Dickman A., Littlewood, C., and Varga J. (2002).

The Syringe Driver: Continuous Subcutaneous Infusions in Palliative Care
. Oxford: Oxford University Press.

Droney, J., Riley, J., and Ross, J. (

2012
).
Opioid genetics in the context of opioid switching.
 
Current Opinion in Supportive and Palliative Care
, 6, 10–16.

Fallon, M. T. and Laird, B. J. (

2011
).
A systematic review of combination step III opioid therapy in cancer pain: an EPCRC opioid guideline project.
 
Palliative Medicine
, 25, 597–603.

Joint Formulary Committee (2014).

British National Formulary
(68th ed.). London: BMJ Group and Pharmaceutical Press.

Junger, S., Brearley, S., Payne, S., et al. (

2013
).
Consensus building on access to controlled medicines: a four-stage Delphi consensus procedure.
 
Journal of Pain and Symptom Management
, 46(6), 897–910.

Minami, M. and Satoh, M. (

1995
).
Molecular biology of the opioid receptors: structures, functions and distributions.
 
Neuroscience Research
, 23, 121–145.

National Institute of Health and Clinical Excellence (2012). Opioids in Palliative Care: Safe and Effective Prescribing of Strong Opioids for Pain in Palliative Care of adults CG140. London: NICE.

Palliativedrugs.com (2012). Palliative Care Formulary 4+. [Online] Available at: http://www.palliativedrugs.com>.

Pasternak, G.W. (

2004
).
Multiple opiate receptors: deja vu all over again.
 
Neuropharmacology
, 47(Suppl. 1), 312–323.

Peckys, D. and Landwehrmeyer, G.B. (

1999
).
Expression of mu, kappa, and delta opioid receptor messenger RNA in the human CNS: a 33P in situ hybridization study.
 
Neuroscience
, 88(4), 1093–1135.

Rook, E.J., Huitema, A.D., Van Den Brink, W., Van Ree, J.M., and Beijnen, J.H. (

2006
).
Pharmacokinetics and pharmacokinetic variability of heroin and its metabolites: review of the literature.
 
Current Clinical Pharmacology
, 1(1), 109–118.

Shabalina, S.A., Zaykin, D.V., Gris, P., et al. (

2009
).
Expansion of the human mu-opioid receptor gene architecture: novel functional variants.
 
Human Molecular Genetics
, 18(6), 1037–1051.

Smith, H.S. (

2011
).
The metabolism of opioid agents and the clinical impact of their active metabolites.
 
Clinical Journal of Pain
, 27, 824–838.

Stein, C., Schafer, M., and Machelska, H. (

2003
).
Attacking pain at its source: new perspectives on opioids.
 
Nature Medicine
, 9(8), 1003–1008.

Walter, C. and Lotsch, J. (

2009
).
Meta-analysis of the relevance of the OPRM1 118A>G genetic variant for pain treatment.
 
Pain
, 146, 270–275.

Zeppetella, G. (

2011
).
Opioids for the management of breakthrough cancer pain in adults: a systematic review undertaken as part of an EPCRC opioid guidelines project.
 
Palliative Medicine
, 25, 516–524.

Andreassen, T.N., Eftedal, I., Klepstad, P., et al. (

2012
).
Do CYP2D6 genotypes reflect oxycodone requirements for cancer patients treated for cancer pain? A cross-sectional multicentre study.
 
European Journal of Clinical Pharmacology
, 68, 55–64.

Bailey, C.P. and Connor, M. (

2005
).
Opioids: cellular mechanisms of tolerance and physical dependence.
 
Current Opinion in Pharmacology
, 5, 60–68.

Bellward, G.D., Warren, P.M., Howald, W., Axelson, J.E., and Abbott, F.S. (

1977
).
Methadone maintenance: effect of urinary pH on renal clearance in chronic high and low doses.
 
Clinical Pharmacology & Therapeutics
, 22, 92–99.

Bolan, E.A., Tallarida, R.J., and Pasternak, G.W. (

2002
).
Synergy between mu opioid ligands: evidence for functional interactions among mu opioid receptor subtypes.
 
Journal of Pharmacology and Experimental Therapeutics
, 303, 557–562.

Campa, D., Gioia, A., Tomei, A., Poli, P., and Barale, R. (

2008
).
Association of ABCB1/MDR1 and OPRM1 gene polymorphisms with morphine pain relief.
 
Clinical Pharmacology & Therapeutics
, 83, 559–566.

Chang, G., Chen, L., and Mao, J. (

2007
).
Opioid tolerance and hyperalgesia.
 
Medical Clinics of North America
, 91, 199–211.

Chen, Z., Irvine, R., Somogyi, A., and Bochner, F. (

1991
).
Mu receptor binding of some commonly used opioids and their metabolites.
 
Life Sciences
, 48, 2165–2171.

Christensen, P.M. and Kristiansen, I.S. (

2006
).
Number-needed-to-treat (NNT)—needs treatment with care.
 
Basic & Clinical Pharmacology & Toxicology
, 99, 12–16.

Costantino, C.M., Gomes, I., Stockton, S.D., Lim, M.P., and Devi, L.A. (

2012
).
Opioid receptor heteromers in analgesia.
 
Expert Reviews in Molecular Medicine
, 14, e9.

Davies, A., Buchanan, A., Zeppetella, G., et al. (

2013
).
Breakthrough cancer pain: an observational study of 1000 European oncology patients.
 
Journal of Pain and Symptom Management
, 46(5), 619–628.

Davies, E.C., Green, C.F., Taylor, S., Williamson, P.R., Mottram, D.R., and Pirmohamed, M. (

2009
).
Adverse drug reactions in hospital in-patients: a prospective analysis of 3695 patient-episodes.
 
PLoS One
, 4, e4439.

Davis, M.P., Legrand, S.B., and Lagman, R. (

2005
).
Look before leaping: combined opioids may not be the rave.
 
Supportive Care in Cancer
, 13, 769–774.

Ferguson, S.S. (

2001
).
Evolving concepts in G protein-coupled receptor endocytosis: the role in receptor desensitization and signaling.
 
Pharmacological Reviews
, 53, 1–24.

Greenwald, M.K., Johanson, C.E., Moody, D.E., et al. (

2003
).
Effects of buprenorphine maintenance dose on mu-opioid receptor availability, plasma concentrations, and antagonist blockade in heroin-dependent volunteers.
 
Neuropsychopharmacology
, 28, 2000–2009.

Gronlund, J., Saari, T.I., Hagelberg, N.M., Neuvonen, P.J., Olkkola, K.T., and Laine, K. (

2010
).
Exposure to oral oxycodone is increased by concomitant inhibition of CYP2D6 and 3A4 pathways, but not by inhibition of CYP2D6 alone.
 
British Journal of Clinical Pharmacology
, 70, 78–87.

Hanley, M.J., Cancalon, P., Widmer, W.W., and Greenblatt, D.J. (

2011
).
The effect of grapefruit juice on drug disposition.
 
Expert Opinion on Drug Metabolism & Toxicology
, 7, 267–286.

Haughey, C., Watson, M., and White, C. (

2012
).
Use of methadone as a coanalgesic: response to McKenna and Nicholson.
 
Journal of Pain and Symptom Management
, 43, e5–e6.

Heiskanen, T., Matzke, S., Haakana, S., Gergov, M., Vuori, E., and Kalso, E. (

2009
).
Transdermal fentanyl in cachectic cancer patients.
 
Pain
, 144, 218–222.

Innocenti, F., Undevia, S. D., Iyer, L., et al. (

2004
).
Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan.
 
Journal of Clinical Oncology
, 22, 1382–1388.

Johnson, J.A., Burkley, B.M., Langaee, T.Y., et al. (

2012
).
Implementing personalized medicine: development of a cost-effective customized pharmacogenetics genotyping array.
 
Clinical Pharmacology & Therapeutics
, 92, 437–439.

Johnson, J.A. and Cavallari, L.H. (

2013
).
Pharmacogenetics and cardiovascular disease--implications for personalized medicine.
 
Pharmacological Reviews
, 65, 987–1009.

Kaasa, S., Hjermstad, M.J., and Loge, J.H.-V. (

2006
).
Methodological and structural challenges in palliative care research: how have we fared in the last decades?
 
Palliative Medicine
, 20, 727–734.

Kalso, E., Vainio, A., Mattila, M.J., Rosenberg, P.H., and Seppala, T. (

1990
).
Morphine and oxycodone in the management of cancer pain: plasma levels determined by chemical and radioreceptor assays.
 
Pharmacology and Toxicology
, 67, 322–328.

Kirchheiner, J., Schmidt, H., Tzvetkov, M., et al. (

2007
).
Pharmacokinetics of codeine and its metabolite morphine in ultra-rapid metabolizers due to CYP2D6 duplication.
 
Pharmacogenomics Journal
, 7, 257–265.

Klepstad, P., Rakvag, T.T., Kaasa, S., et al. (

2004
).
The 118 A > G polymorphism in the human mu-opioid receptor gene may increase morphine requirements in patients with pain caused by malignant disease.
 
Acta Anaesthesiologica Scandinavica
, 48, 1232–1239.

Knapp, R.J., Malatynska, E., Fang, L., et al. (

1994
).
Identification of a human delta opioid receptor: Cloning and expression.
 
Life Sciences
, 54, L463–L469.

Leandro-Garcia, L.J., Leskela, S., Montero-Conde, C., et al. (

2009
).
Determination of CYP2D6 gene copy number by multiplex polymerase chain reaction analysis.
 
Analytical Biochemistry
, 389, 74–76.

Lotsch, J. (

2005
).
Opioid metabolites.
 
Journal of Pain and Symptom Management
, 29, S10–S24.

Madadi, P., Koren, G., Cairns, J., et al. (

2007
).
Safety of codeine during breastfeeding: fatal morphine poisoning in the breastfed neonate of a mother prescribed codeine.
 
Canadian Family Physician
, 53, 33–35.

Mallal, S., Phillips, E., Carosi, G., et al. (

2008
).
HLA-B*5701 screening for hypersensitivity to abacavir.
 
The New England Journal of Medicine
, 358, 568–579.

Mark, J. (

1990
).
Kappa-opioid receptors and analgesia.
 
Trends in Pharmacological Sciences
, 11, 70–76.

Martin, M., Matifas, A., Maldonado, R., and Kieffer, B. L. (

2003
).
Acute antinociceptive responses in single and combinatorial opioid receptor knockout mice: distinct mu, delta and kappa tones.
 
European Journal of Neuroscience
, 17, 701–708.

Matthes, H.W., Maldonado, R., Simonin, F., et al. (

1996
).
Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the mu-opioid-receptor gene.
 
Nature
, 383, 819–823.

McKenna, M. and Nicholson, A.B. (

2011
).
Use of methadone as a coanalgesic.
 
Journal of Pain and Symptom Management
, 42, e4–e6.

Meng, F., Xie, G.X., Thompson, R.C., et al. (

1993
).
Cloning and pharmacological characterization of a rat kappa opioid receptor.
 
Proceedings of the National Academy of Sciences of the United States of America
, 90, 9954–9958.

Milligan, G. (

2005
).
Opioid receptors and their interacting proteins.
 
NeuroMolecular Medicine
, 7, 51–59.

Mollereau, C., Parmentier, M., Mailleux, P., et al. (

1994
).
ORL1, a novel member of the opioid receptor family. Cloning, functional expression and localization.
 
FEBS Letters
, 341, 33–38.

Nitsche, J.F., Schuller, A.G., King, M.A., Zengh, M., Pasternak, G.W., and Pintar, J.E. (

2002
).
Genetic dissociation of opiate tolerance and physical dependence in delta-opioid receptor-1 and preproenkephalin knock-out mice.
 
Journal of Neuroscience
, 22, 10906–10913.

Pasternak, G. and Pan, Y.X. (

2011
).
Mu opioid receptors in pain management.
 
Acta Anaesthesiologica Taiwanica
, 49, 21–25.

Persson, K., Sjostrom, S., Sigurdardottir, I., Molnar, V., Hammarlund-Udenaes, M., and Rane, A. (

1995
).
Patient-controlled analgesia (PCA) with codeine for postoperative pain relief in ten extensive metabolisers and one poor metaboliser of dextromethorphan.
 
British Journal of Clinical Pharmacology
, 39, 182–186.

Rakvag, T.T., Klepstad, P., Baar, C., et al. (

2005
).
The Val158Met polymorphism of the human catechol-O-methyltransferase (COMT) gene may influence morphine requirements in cancer pain patients.
 
Pain
, 116, 73–78.

Rakvag, T.T., Ross, J.R., Sato, H., Skorpen, F., Kaasa, S., and Klepstad, P. (

2008
).
Genetic variation in the catechol-O-methyltransferase (COMT) gene and morphine requirements in cancer patients with pain.
 
Molecular Pain
, 4, 64.

Reyes-Gibby, C.C., Shete, S., Rakvag, T., et al. (

2007
).
Exploring joint effects of genes and the clinical efficacy of morphine for cancer pain: OPRM1 and COMT gene.
 
Pain
, 130, 25–30.

Richards, P., Riff, D., Kelen, R., Stern, W., and MoxDuo Study Group. (

2011
).
Analgesic and adverse effects of a fixed-ratio morphine-oxycodone combination (MoxDuo) in the treatment of postoperative pain.
 
Journal of Opioid Management
, 7, 217–228.

Riley, J., Ross, J.R., Rutter, D., et al. (

2006
).
No pain relief from morphine? Individual variation in sensitivity to morphine and the need to switch to an alternative opioid in cancer patients.
 
Supportive Care in Cancer
, 14, 56–64.

Samer, C.F., Daali, Y., Wagner, M., et al. (

2010
a).
The effects of CYP2D6 and CYP3A activities on the pharmacokinetics of immediate release oxycodone.
 
British Journal of Pharmacology
, 160, 907–918.

Samer, C.F., Daali, Y., Wagner, M., et al. (

2010
b).
Genetic polymorphisms and drug interactions modulating CYP2D6 and CYP3A activities have a major effect on oxycodone analgesic efficacy and safety.
 
British Journal of Pharmacology
, 160, 919–930.

Scottish Intercollegiate Guidelines Network (

2008
).
Control of Pain in Adults with Cancer
. Edinburgh: Scottish Intercollegiate Guidelines Network.

Simonin, F., Valverde, O., Smadja, C., et al. (

1998
).
Disruption of the kappa-opioid receptor gene in mice enhances sensitivity to chemical visceral pain, impairs pharmacological actions of the selective kappa-agonist U-50,488H and attenuates morphine withdrawal.
 
EMBO Journal
, 17, 886–897.

Sindrup, S.H., Brosen, K., Bjerring, P., et al. (

1990
).
Codeine increases pain thresholds to copper vapor laser stimuli in extensive but not poor metabolizers of sparteine.
 
Clinical Pharmacology & Therapeutics
, 48, 686–693.

Smith, M., Ross, F., Nielsen, C., and Saini, K. (

2001
).
Oxycodone has a distinctly different pharmacology from morphine.
 
European Journal of Pain
, 15(suppl A), 135–136.

Stamer, U.M., Lehnen, K., Hothker, F., et al. (

2003
).
Impact of CYP2D6 genotype on postoperative tramadol analgesia.
 
Pain
, 105, 231–238.

Twycross, R., Prommer, E.E., Mihalyo, M., and Wilcock, A. (

2012
).
Fentanyl (transmucosal).
 
Journal of Pain and Symptom Management
, 44, 131–149.

Walsh, S.L. and Eissenberg, T. (

2003
).
The clinical pharmacology of buprenorphine: extrapolating from the laboratory to the clinic.
 
Drug and Alcohol Dependence
, 70, S13–27.

Wang, J.B., Johnson, P.S., Persico, A.M., Hawkins, A.L., Griffin, C.A., and Uhl, G.R. (

1994
).
Human mu opiate receptor. cDNA and genomic clones, pharmacologic characterization and chromosomal assignment.
 
FEBS Letters
, 338, 217–222.

Watkins, L.R., Hutchinson, M.R., Johnston, I.N., and Maier, S.F. (

2005
).
Glia: novel counter-regulators of opioid analgesia.
 
Trends in Neurosciences
, 28, 661–669.

Webster, L. (

2012
).
Efficacy and safety of dual-opioid therapy in acute pain.
 
Pain in Medicine
, 13(Suppl. 1), S12–20.

Webster, L., Richards, P., Stern, W., Kelen, R., and MoxDuo Study Group (

2010
).
A double-blind, placebo-controlled study of dual-opioid treatment with the combination of morphine plus oxycodone in patients with acute postoperative pain.
 
Journal of Opioid Management
, 6, 329–340.

Wilke, R.A., Mareedu, R.K., and Moore, J.H. (

2008
).
The pathway less traveled: moving from candidate genes to candidate pathways in the analysis of genome-wide data from large scale pharmacogenetic association studies.
 
Current Pharmacogenomics and Personalized Medicine
, 6, 150–159.

World Health Organization (2013). WHO Model List of Essential Medicines (18th ed.). [Online] Available at: <http://www.who.int/medicines/publications/essentialmedicines/en/>.

Yoburn, B., Shah, S., Chan, K., Duttaroy, A., and Davis, T. (

1995
).
Supersensitivity to opioid analgesics following chronic opioid antagonist treatment: relationship to receptor selectivity.
 
Pharmacology, Biochemistry and Behavior
, 51, 535–539.

Close
This Feature Is Available To Subscribers Only

Sign In or Create an Account

Close

This PDF is available to Subscribers Only

View Article Abstract & Purchase Options

For full access to this pdf, sign in to an existing account, or purchase an annual subscription.

Close