Analgesic Drugs

Published on 27/02/2015 by admin

Filed under Anesthesiology

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 1 (1 votes)

This article have been viewed 2110 times

Analgesic Drugs

The ideal analgesic should relieve pain with a minimum of side-effects. When formulating a management plan, it is worth considering what contributes to pain perception and associated distress. The International Association for the Study of Pain (IASP) defines pain as ‘an unpleasant sensory and emotional experience associated with actual or potential tissue damage or described in terms of such damage’. It is clear from this definition that the degree of tissue damage and perception of pain are not necessarily correlated. Pain perception is a complex phenomenon, involving sensory, emotional and cognitive processes. Thus, while analgesic drugs can be effective in relieving both acute and chronic pain, other factors may also need to be addressed.

There is considerable evidence that patients continue to suffer pain, despite a wide range of available analgesic drugs. When devising a management plan for both acute and chronic settings, using ‘balanced’ or ‘multimodal analgesia’ may be helpful. These terms refer to the use of combinations of drugs acting by different mechanisms or at different sites within the pain pathway. Analgesic combinations may be additive or synergistic in their mode of action, with resultant dose-sparing effects and a reduction in side-effects. Effective and repeated assessment of patients is essential in determining optimal analgesic management.

OPIOIDS

Opioids are the most frequently used analgesics for the treatment of moderate to severe pain. Despite this, there are still major gaps in our knowledge of their clinical pharmacology, with the choice of drug and dose being largely empirical. Although they may be highly effective, control of dynamic (pain on movement) or incident (breakthrough) pain may be poor and side-effects may be a significant problem. The term opioid refers to all drugs, both synthetic and natural, that act on opioid receptors. Opiates are naturally occurring opioids derived from the opium poppy Papaver somniferum. The most widely used opioid is morphine, although a variety of different opioids are available. Incomplete cross-tolerance occurs between them, so an alternative should be tried if morphine is poorly tolerated. There is increasing evidence that in any one individual both analgesic response and side effect profile will be, at least partly, due to that person’s genetic make up. Differences in Single Nucleotide Polymorphisms (SNPs) almost certainly contribute to this variability in response between different opioids, with potential candidate genes including ABCB1 (encoding p-glycoprotein, a membrane transporter), STAT6 (signal transducer and activator of transcription 6) and beta-arrestin (intracellular protein involved in receptor internalization).

Dose conversion between different opioids is an inexact science, with equi-analgesic doses being based on relative potencies, often derived from studies that are not designed for dose equivalence calculation. Table 5.1 shows suggested equi-analgesic doses for some opioids.

Mechanism of Action

Opioid receptors belong to the G-protein coupled family of receptors with seven transmembrane domains, an extracellular N-terminal and intracellular C-terminal. Activation results in changes in enzyme activity such as adenylate cyclase or alterations in calcium and potassium ion channel permeability.

Opioid receptors were originally classified by pharmacological activity in animal preparations, and later by molecular sequence. The three main receptors were classified as μ (mu) or OP3, κ (kappa) or OP1 and δ (delta) or OP2. Another opioid-like receptor has been identified recently; it is termed the nociceptin orphanin FQ peptide receptor. Receptor nomenclature has changed several times in the last few years; the current International Union of Pharmacology (IUPHAR) classification is MOP (mu), KOP (kappa), DOP (delta) and NOP for the nociceptin orphanin FQ peptide receptor (Table 5.2).

Opioid receptors are distributed widely in both central and peripheral nervous systems. The different effects of currently available opioids are dependent on complex interactions at various receptors. There is a range of endogenous neuropeptide ligands active at these receptors (Table 5.2); they function as neurotransmitters, neuromodulators and neurohormones. The endogenous tetrapeptide endomorphins 1 and 2 are potent agonists acting specifically at the MOP receptor; they play a role in modulating inflammatory pain.

The analgesic action of morphine and most other opioids is related mainly to agonist activity at the MOP receptor. Unfortunately, many of the unwanted effects of opioids are also related to activity at this receptor. At a cellular level, MOP receptor activation has an overall inhibitory effect via: (i) inhibition of adenylate cyclase; (ii) increased opening of potassium channels (hyperpolarization of postsynaptic neurones, reduced synaptic transmission); and (iii) inhibition of calcium channels (decreases presynaptic neurotransmitter release).

Pharmacodynamic Effects of Opioids

The ubiquitous nature of opioid receptors implies that agents acting at them have wide-ranging effects, some of which may be problematic. Some opioids or their metabolites also have activity at other receptors, e.g. methadone acts at the N-methyl-D-aspartate (NMDA) receptor. These particular actions are discussed for individual agents below. The more general effects of opioids are described in this section.

Analgesic Action

Opioids with agonist activity mainly at the MOP receptor, and to a lesser extent at the KOP receptor, have analgesic effects. Analgesic effects have also been demonstrated for the spinal DOP receptor in certain situations.

Opioids should be titrated against pain; if higher than necessary doses are given, respiratory depression and excessive sedation may result. If the pain is incompletely opioid-responsive, as may occur with neuropathic pain, then care must be taken with dose titration and a detailed reassessment of analgesic response is essential. Opioids exert their analgesic effect by:

Central Nervous System

In addition to analgesia there are several potential central nervous system (CNS) effects of opioids:

Sedation and sleep. Opioids interfere with rapid-eye-movement sleep with changes in the EEG including a progressive decrease in EEG frequency and production of delta waves. However, burst suppression is not seen, even with large doses. Opioid-related ventilatory depression is more common during sleep. Opioid-induced sedation may be used therapeutically, e.g. critical care setting. There is a dose-related reduction in minimum alveolar concentration (MAC) for volatile anaesthetics, though there is a floor to this effect. Opioids alone do not act as reliable anaesthetic agents.

Mood. Significant euphoria is uncommon when opioids are used to treat pain but it occurs frequently when they are used inappropriately. Dysphoria (possibly via a KOP receptor action) and hallucinations can occur. Commonly, the hallucinations are visual in nature and may only affect part of the visual field.

Miosis. This is mediated via a KOP receptor effect on the Edinger-Westphal nucleus of the oculomotor nerve.

Tolerance (i.e. requirement of increasing doses to achieve the same effect) is important clinically because a significant number of patients are receiving long-term opioid therapy for malignant and chronic non-malignant pain; it is also relevant in illicit drug use. Additionally, tolerance may develop much more acutely, e.g. when opioids are used for pain control before surgery or when given intrathecally. However, after initial dose titration, the majority of patients on long-term opioids are usually maintained on a stable dose.

At a cellular level, tolerance is caused by a progressive loss of active receptor sites combined with uncoupling of the receptor from the guanosine triphosphate (GTP)-binding subunit. There is also some evidence that the NMDA receptor may play a role in acute tolerance, via protein kinase C activity lifting the magnesium block at the NMDA site. There may therefore be a rationale for using ketamine in situations of acute tolerance, for example in the postoperative period. Interactions with the NOP receptor may also be important.

The incomplete cross-tolerance between different types of opioids may result from differential activity at opioid receptor subtypes. Withdrawal may occur if there is abrupt cessation of opioids or if an antagonist is given.

Opioid-induced hyperalgesia. This is a paradoxical response where an increase in opioid dose results in hyperalgesia. There is good basic science evidence of underlying mechanisms, but its importance in clinical practice is unclear. It may be part of the spectrum of opioid toxicity, or can occur in isolation. It has been shown to occur after systemic administration of potent short acting opioids such as remifentanil.

Addiction. This is defined as the compulsive use of opioids to the detriment of the patient in terms of physical, psychological or social function. Drug-seeking behaviour is not usually a problem if opioids are used appropriately for pain relief in both acute and chronic situations.

Respiratory

Opioids may cause respiratory depression, particularly in the elderly, neonates and when given without titrating effect to analgesic response. Tolerance does develop to this phenomenon, so it is less of a problem in chronic use. However, care must be taken if nociceptive input is reduced or removed, e.g. after a nerve block. Sensitivity to CO2 is reduced, even with small doses of MOP agonists. This is caused by depression of sensitivity of neurones on the ventral surface of the medulla.

Opioids, given at sufficient dose, are effective at suppressing the stress response to laryngoscopy and airway manipulation. They may reduce the plasma concentrations of catecholamines, cortisol and other stress hormones by inhibiting the pituitary-adrenal axis, reducing central sympathetic outflow and influencing central neuroendocrine responses. Opioids also suppress cough activity and mucociliary function. This may cause inadequate clearing of secretions and hypostatic pneumonia, especially if there is associated sedation and respiratory depression. This antitussive activity is, at least in part, mediated peripherally.

Cardiovascular

In normovolaemic patients, the majority of opioids have no significant cardiovascular depressant effect. However, if histamine is released, then there may be tachycardia, decrease in systemic vascular resistance and a reduction in arterial pressure. Bradycardia may occur in response to some opioids. There is no direct action on baroreceptors but a minimal reduction in preload and afterload may occur. There is no effect on cerebral autoregulation. However, if respiratory depression is present, then the resultant increase in PaCO2 may increase cerebral blood flow.

Opioids decrease central sympathetic outflow. Therefore, in patients who are relying on increased sympathetic tone to maintain cardiovascular stability, opioids may lead to haemodynamic compromise. This may be severe, particularly if potent opioids are given by rapid intravenous bolus.

Other Effects

Myoclonic jerks may occur if there is opioid toxicity and may be associated with sedation and hallucinations. Urinary retention and urgency may occur, probably related to a centrally mediated mechanism as these problems are much more common when neuraxial opioids are used. Pruritus is relatively common after neuraxial administration. The nose, face and torso are particularly affected and this may be reversed by a low dose of a MOP antagonist. Muscle rigidity is a recognized complication, particularly after intravenous bolus administration of potent phenylpiperidines. This may cause significant problems with ventilation because of chest wall rigidity and decreased respiratory compliance. It may be minimized by co-administration of opioids with intravenous anaesthetic agents and benzodiazepines, reversed by naloxone or prevented by neuromuscular blocking agents. Thermoregulation is impaired to a similar extent as that seen with volatile agents. With long-term use, depression of the immune system may occur. Endocrine problems include impaired adrenal and sexual function, and infertility.

Opioid Structure

The structures of opioid analgesics are diverse, although for most opioids it is usually the laevorotatory (laevo) stereoisomer that is the active compound. The structures of some of the common agents are shown in Figures 5.1 and 5.2. Agents in current use include phenanthrenes (e.g. morphine – Fig. 5.1), phenylpiperidines (e.g. meperidine (pethidine), fentanyl – Fig. 5.2) and diphenylpropylamines (e.g. methadone, dextropropoxyphene). Structural modification affects agonist activity and alters physicochemical properties such as lipid solubility. A tertiary nitrogen is necessary for activity separated from a quaternary carbon by an ethylene chain. Chemical modifications that produce a quaternary nitrogen significantly reduce potency as a result of decreased CNS penetration. If the methyl group on the nitrogen is changed, antagonism of analgesia may be produced.

Other important positions for activity and metabolism, as seen on the morphine molecule (Fig. 5.1), include the C-3 phenol group (the distance of this from the nitrogen affects activity) and the C-6 alcohol group. Potency may be increased by hydroxylation of the C-3 phenol; oxidation of C-6 (e.g. hydromorphone); double acetylation at C-3 and C-6 (e.g. diamorphine); hydroxylation of C-14 and reducing the double bond at C-7/8. Further additions at the C-3 OH group reduce activity. A short-chain alkyl substitution is found in mixed agonist-antagonists, hydroxylation or bromination of C-14 produces full antagonists and removal or substitution of the methyl group reduces agonist activity.

Pharmacokinetics and Physicochemical Properties

Knowledge of the specific physicochemical properties and pharmacokinetics of individual agents is important in determining the optimal route of drug delivery in order to achieve an effective receptor site concentration for an appropriate duration of action. All opioids are weak bases. The relative proportion of free and ionized fractions is dependent on plasma pH and the pKa of the particular opioid. The amount of opioid diffusing to the site of action (diffusible fraction) is dependent on lipid solubility, concentration gradient and degree of binding. Plasma concentrations of albumin and αl-acid glycoprotein as well as tissue binding determine the availability of the unbound, unionized fraction. This diffusible fraction moves into tissue sites in the brain and elsewhere; the amount reaching receptors is dependent not only on lipophilicity but also on the amount of non-specific tissue binding, e.g. CNS lipids.

The ionized, protonated form is active at the receptor site. This has important implications for speed and duration of activity. For example, morphine is relatively hydrophilic and penetrates the blood–brain barrier slowly. However, a large mass of any given dose eventually reaches the receptor site because of low levels of nonspecific tissue binding. This effect-site equilibration time (t½keo) is measured by assessing the effect of opioids on the EEG. The offset time may also be prolonged, resulting in a longer duration of action than would be expected from the plasma half-life. Most opioids have a very steep dose-response curve. Therefore, if the dose is near the minimum effective analgesic concentration (MEAC), very small fluctuations in plasma or effect-site concentrations may lead to large changes in the level of analgesia.

Opioids tend to have a large volume of distribution (VD) because of their high lipid solubility. A consequence of this can be that redistribution, particularly after a bolus dose or short infusion, can have significant effects on plasma concentrations. In addition, first-pass effects in the lung may remove significant amounts of drug from the circulation, reducing the initial peak plasma concentration. However, the drug re-enters the plasma several minutes later. Plasma concentrations of opioids such as fentanyl, sufentanil and meperidine (pethidine) are affected by this; the effect is negligible for remifentanil. Other lipophilic amines such as lidocaine and propranolol are affected similarly and may reduce pulmonary uptake of co-administered opioids.

After prolonged infusion, significant sequestration in fat stores and other body tissues occurs for highly lipid-soluble opioids. This is reflected in the ‘context-sensitive t½’ i.e. the time taken for the plasma concentration to reduce by 50% after an infusion designed to maintain constant plasma concentrations has been stopped (see Chapter 1). The context-sensitive t½ is increased after prolonged infusion for most opioids apart from remifentanil. For example, the elimination t½ for fentanyl after bolus administration is 3–5 h, but increases to 7–12 h after prolonged infusion.

Most opioid metabolism occurs in the liver (phase I and II reactions) with the hydrophilic metabolites predominantly excreted renally, although a small amount may be excreted in the bile or unchanged in the urine. As a result, hepatic blood flow is one of the major determinants of plasma clearance. Metabolism of individual drugs is shown in Table 5.3. Enterohepatic recirculation may occur when water-soluble metabolites excreted in the gut may be metabolized by gut flora to the parent opioid and then reabsorbed. Lipid-soluble opioids may diffuse into the stomach, become ionized because of the low pH and then be reabsorbed in the small intestine; this results in a secondary peak in plasma concentration.

A summary of physicochemical and pharmacokinetic properties of some opioids is shown in Table 5.4. Metabolism (including production of active metabolites), distribution between different tissues and elimination all interact within individual subjects to produce clinically important actions at receptor sites.

Factors affecting pharmacokinetics include:

image Age. Systemic dose is often calculated on body weight, although there is little evidence to support this in adult clinical practice. Age is often more important because of both pharmacokinetic and pharmacodynamic factors. 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 years, with reduced clearance of opioids. Increased CNS sensitivity to opioid effects is also found in the elderly.

image Hepatic disease has unpredictable effects, although there may be little clinical difference unless there is coexisting encephalopathy. Reductions in plasma protein concentrations also have effects on plasma concentrations of free unbound drug.

image Renal failure may have significant effects for opioids with renally excreted active metabolites such as morphine, diamorphine and meperidine.

image Obesity will result in a larger VD and prolonged elimination t½. This may be a particular problem if infusions are being used.

image Hypothermia, hypotension and hypovolaemia may also result in variable absorption and altered distribution and metabolism.

Routes of Administration

Opioids given parenterally have 100% bioavailability (see Chapter 1), although peak plasma concentrations may be affected by site of administration and haemodynamic status. Opioids may be given by many routes; variations between specific agents are discussed below. It is unclear how much cross-tolerance exists for different routes of administration, e.g. intravenous versus epidural.

The choice of route is dependent on the clinical situation and several factors may need to be considered:

image If there is delayed gastrointestinal transit time, the biological half-life may be prolonged with orally administered agents. Similarly, if there is rapid GI transit time or reduced area for absorption, then there may be reduced absorption, particularly of long acting agents.

image Intrathecal administration is associated with fewer supraspinal effects, although both urinary retention and pruritus may be more common. Highly lipid-soluble opioids (e.g. fentanyl) do not spread readily in cerebrospinal fluid (CSF). It is claimed that they are less likely than water-soluble opioids (e.g. morphine) to cause late respiratory depression due to rostral spread.

image Dural penetration from epidural administration is dependent on molecular size and lipophilicity For example, only 3–5% of morphine crosses into the CSF, with a peak concentration after 60–240 min; fentanyl peaks at approximately 20 min.

MOP Agonists

Morphine is the standard opioid against which other agents are compared. Other MOP agonists have a similar pharmacodynamic profile but differ in relative potency, pharmacokinetics and biotransformation to other active metabolites.

Phenylpiperidine opioids (Fig. 5.2) are potent MOP receptor agonists with moderate (alfentanil) to high (sufentanil) lipid solubility and good diffusion through membranes. Both potency and time to reach the effect site vary considerably. In contrast to morphine, these agents do not cause histamine release. All except remifentanil may cause postoperative respiratory depression as a result of secondary peaks in plasma concentrations. This may be caused by release from body stores if large doses have been infused intra-operatively. Fentanyl, alfentanil and sufentanil are metabolized mainly in the liver to inactive metabolites. Very little is excreted unchanged in the urine (Table 5.3).

Morphine

Morphine is a relatively hydrophilic phenanthrene derivative. It may be given orally, rectally, topically, parenterally and via the neuraxial route. The standard parenteral dose for adults is 10 mg, although many factors affect this and the dose should be titrated to effect. Its oral bioavailability is dependent on first-pass hepatic metabolism and may be unpredictable (35–75%). Oral morphine is available either as immediate-release liquid, simple tablet or as a modified-release preparation. Single-dose studies of morphine bioavailability indicate that the relative potency of oral:intramuscular morphine is 1:6 although, with repeated regular administration, this ratio becomes approximately 1:3. The dose of short-acting morphine for breakthrough pain should be approximately one-sixth of the total daily dose. Morphine has a plasma half-life of approximately 3 h and a duration of analgesia of 4–6 h.

Morphine is metabolized, at least in part, by microsomal UDP glucuronyl transferases (UDPGT) found in the liver, kidney and intestines. Several of these metabolites may have clinically significant effects (see below). Although morphine conjugation occurs in the liver, there is evidence that extrahepatic sites may also be important, e.g. kidney, gastrointestinal tract. The site of conjugation on the molecule also varies, leading to a variety of metabolites (Table 5.3). After glucuronidation, metabolites are excreted in urine or bile, dependent on molecular weight and polarity; more than 90% of morphine metabolites are excreted in the urine. The main metabolite in humans is morphine-3-glucuronide (60–80%) and this may have an excitatory effect via CNS actions not related to opioid receptor activation. Morphine-6-glucuronide (M-6-G) is active at the MOP receptor, producing analgesia and other MOP-related effects. It is significantly more potent than morphine. Therefore, M-6-G produces significant clinical effects despite only 10% of morphine being metabolized in this way. As it is renally excreted, it may accumulate in patients with impaired renal function, causing respiratory depression. Current evidence indicates that accumulation of morphine metabolites, especially M-6-G, becomes significant when creatinine clearance declines to 50 mL min–1 or less.

Meperidine (Pethidine)

Meperidine (pethidine) is available as parenteral and oral preparations. There is no evidence that this opioid provides any advantage over morphine, e.g. treatment of colic-type pain. It is fairly short acting in terms of analgesia, and if repeated doses are given, the metabolite normeperidine can accumulate (t½ ~ 15 h). This is a CNS stimulant and can cause seizures, especially if there is renal dysfunction. Its clearance is significantly reduced in hepatic disease. Chronic use may result in enzyme induction and an increase in normeperidine plasma concentrations. Its metabolism is decreased by the oral contraceptive pill.

Meperidine has other significant effects related to activity at non-opioid receptors. For example, its atropine-like action may cause a tachycardia, in addition to direct myocardial depression at high doses. It was used originally as a bronchodilator. It can also reduce shivering related to hypothermia or epidurals, although the mechanism for this is not fully understood. Meperidine also has a local anaesthetic-like membrane stabilizing action.

Fentanyl

Fentanyl is available in a variety of preparations for parenteral, transdermal and transmucosal (including buccal) administration. Due to high first-pass metabolism (~ 70%) it is not given orally. It is approx. 80–100 times more potent than morphine in the acute setting, although it is approx. 30–40 times as potent when given chronically e.g. slow-release transdermal patches. With transdermal administration, the patch and underlying dermis act as a reservoir and plasma concentration does not reach steady state until approx 15 h after initial application. Plasma concentration also declines slowly after removal (t½ ~ 15–20 h).

Fentanyl is very lipophilic with a relatively short duration of action. There are several new buccal/transmucosal preparations developed for rapid onset breakthrough pain. These aim to have a very rapid onset in ~ 10 minutes, although this may not be the case in clinical practice. Fentanyl has a large VD with rapid peripheral tissue uptake limiting initial hepatic metabolism. This may result in significant variability in plasma concentrations and secondary plasma peaks. It binds to αl-acid glycoprotein and albumin; 40% of the protein-bound fraction is taken up by erythrocytes. The lung may be important in exerting a first-pass effect on fentanyl (up to 75% of the dose), thus buffering the plasma from high peak drug concentrations.

Remifentanil

Remifentanil is available for parenteral use as a lyophilized white crystalline powder containing glycine. It should not be administered epidurally or spinally. After being made up in solution, it is stable for 24 h. This MOP receptor agonist has a similar potency to fentanyl and is ~ 20 times more potent than alfentanil. It differs from other opioids in that it has an ester linkage, resulting in degradation by tissue and non-specific plasma esterases. This process is non-saturable and clearance is significantly greater than hepatic blood flow. Plasma cholinesterase deficiency does not affect clearance. Hepatic and renal dysfunction have no effect on clearance also, although increased opioid sensitivity in hepatic disease may result in a lower dosage requirement. Other situations requiring a reduction in dose include haemorrhage or shock. Hydrolysis produces a carboxylic acid metabolite with limited action at the MOP receptor (~ 1000 times < remifentanil). It is not thought to be clinically significant, even in renal dysfunction.

Remifentanil has a rapid blood–brain equilibration time of just over 1 min, with a short context-sensitive half-time of 3–5 min which is unaffected by duration of infusion. This makes it ideally suited for infusion during anaesthesia and in the critical care setting. It may be titrated rapidly to achieve the desired effect. The high clearance and low VD imply that the offset of effect is caused by metabolism rather then redistribution. Hypothermia, such as may occur in cardiac surgery, may reduce clearance by up to 20%.

There is some evidence to suggest that acute opioid tolerance and hyperalgesia may occur after remifentanil infusions. If high doses are used without neuromuscular blockade, muscle rigidity may be a problem. It is unlikely to be a problem when using a concentration of 100 μg mL –1 or less and an infusion rate of 0.2–0.5 μg kg–1 min–1. Bradycardia has also been reported.

Methadone

Methadone is a diphenylpropylamine. It has very good oral bioavailability (~ 85%) with an oral to parenteral ratio of 1:2. Its plasma half-life can be very variable (3–50 h, average 24 h) but its duration of action is relatively short. With repeated dosing, problems with accumulation can occur because of this discrepancy between half-life and analgesic effect. Careful monitoring is therefore required when converting patients to long-term methadone. Also, there is incomplete cross-tolerance with morphine. The racemic mixture in common use has agonist actions at the MOP receptor (mainly the laevo-isomer) as well as antagonist activity at the NMDA receptor (dextro-isomer). Given the importance of this receptor in central sensitization in a variety of pain states, there may be cases where methadone offers particular advantages over and above other opioids, e.g. neuropathic pain.

Plasma concentrations of methadone can be reduced by carbamazepine and its metabolism is accelerated by phenytoin.

Codeine

Codeine is a constituent of opium. Up to 10% of a dose of codeine is metabolized by the hepatic microsomal enzyme CYP2D6 to morphine, which contributes significantly to its analgesic effect. The rest is metabolized in the liver to norcodeine and then conjugated to produce glucuronide conjugates of codeine, norcodeine and morphine. Codeine is considerably less potent than morphine. Around 8% of Western Europeans are deficient in the CYP2D6 enzyme due to genetic polymorphism and such individuals may not experience adequate analgesia with codeine. Similarly, with “super-metabolizers”, there may be problems with opioid toxicity. It can cause significant histamine release and its intravenous administration should be avoided. It has marked antitussive effects and also causes significant constipation. It is often combined with paracetamol.

Partial Agonists

These agents have high affinity for the MOP receptor but limited efficacy (see Chapter 1). A ceiling effect is seen in the dose-response curve at less than the maximal analgesic effect of full MOP agonists. If given with a full MOP agonist, there may be a reduction in the maximal analgesic effect.

Opioid Antagonists

Naloxone is a short-acting opioid antagonist that is relatively selective for the MOP receptor. It is structurally similar to morphine, with some modifications resulting in antagonist activity including an OH group at C-14. It can reverse opioid-induced respiratory depression but repeated administration may be required because of its short duration of action; it can be given by continuous infusion. However, sudden and complete reversal of the analgesic effects of opioids may be accompanied by major cardiovascular and sympathetic responses. Naloxone has a very low oral bioavailability (~ 3%), allowing its use in combination with oxycodone (see below) to reduce gastrointestinal effects.

Naltrexone is a long-acting opioid antagonist used in the management of opioid dependence. It is available only in oral formulation.

New Developments in Opioid Pharmacology

There is considerable interest in developing opioid analgesics with an improved side effect profile, and there are some promising recent developments in this area. It has been recognized that by using agents which act on more than one type of opioid receptor there may be beneficial effects (see Fig. 5.3) Some recently developed agents include:

image Oxycodone/Naloxone (Targinact®). This drug combination utilizes a fixed ratio of oxycodone and naloxone (2:1). The oral naloxone has a greater affinity for opioid receptors in the gut than oxycodone and therefore preferentially binds to these receptors. As a result, gastrointestinal side effects may be reduced, without any effect on the central analgesic effect of oxycodone.

image Tapentadol. The analgesic effect of this agent is due to actions at several receptors, including the MOP receptor. It is much more potent at the MOP receptor than tramadol and also has a strong action in inhibiting noradrenaline reuptake. Preliminary evidence is that it is equianalgesic to oxycodone, but has reduced gastrointestinal side effects.

image MoxDuo® is a novel opioid combination of morphine and oxycodone in a 3:2 ratio, with promising evidence of at least 50% decrease in clinically significant side effects such as nausea, vomiting, and dizziness when compared to either opioid alone. This seems to be achieved by the need to use lower doses of both morphine and oxycodone to reach an equivalent analgesic effect.

PARACETAMOL

Paracetamol (acetaminophen) was first used in 1893 and is the only remaining p-aminophenol available in clinical practice. It is the active metabolite of the earlier, more toxic drugs acetanilide and phenacetin. Its structure is shown in Figure 5.4. Paracetamol is an effective analgesic and antipyretic but has no anti-inflammatory activity. In recommended doses, it is safe and has remarkably few side-effects.

Pharmacokinetics

Paracetamol is absorbed rapidly from the small intestine after oral administration; peak plasma concentrations are reached after 30–60 min. It may also be given rectally and intravenously (either as paracetamol or the prodrug proparacetamol). It has good oral bioavailability (70–90%); rectal absorption is more variable (bioavailability ~ 50–80%) with a longer time to reach peak plasma concentration. The plasma half-life is approx. 2–3 h.

Paracetamol is metabolized by hepatic microsomal enzymes mainly to the glucuronide, sulphate and cysteine conjugates. None of these metabolites is pharmacologically active. A minimal amount of the metabolite N-acetyl-p-amino-benzoquinone imine is normally produced by cytochrome P450-mediated hydroxylation. This reactive toxic metabolite is rendered harmless by conjugation with liver glutathione, then excreted renally as mercapturic derivatives. With larger doses of paracetamol, the rate of formation of the reactive metabolite exceeds that of glutathione conjugation, and the reactive metabolite combines with hepatocellular macromolecules, resulting in cell death and potentially fatal hepatic failure. The formation of this metabolite is increased by drugs inducing cytochrome P450 enzymes, such as barbiturates or carbamazepine.

Overdose and Hepatic Toxicity

In overdose, there is the potential for the toxic metabolite described above to cause centrilobular hepatocellular necrosis, occasionally with acute renal tubular necrosis. The threshold dose in adults is ~ 10–15 g. Accidental overdosage can occur if combined preparations such as co-codamol are used together with paracetamol. Doses of more than 150 mg kg–1 taken within 24 h may result in severe liver damage, hypoglycaemia and acute tubular necrosis. Individuals taking enzyme-inducing agents are more likely to develop hepatotoxicity.

Early signs include nausea and vomiting, followed by right subcostal pain and tenderness. Hepatic damage is maximal 3–4 days after ingestion, and may lead to liver failure and death. Treatment consists of gastric emptying and the specific antidotes methionine and acetylcysteine. The former offers effective protection up to 10–12 h after ingestion. Acetylcysteine is effective within 24 h and perhaps beyond. The plasma paracetamol concentration related to time from ingestion indicates the risk of liver damage. Acetylcysteine is given if the plasma paracetamol concentration is > 200 mg L–1 at 4 h and 6.25 mg L–1 at 24 h after ingestion.

NON-STEROIDAL ANTI-INFLAMMATORY DRUGS

The analgesic, anti-inflammatory and antipyretic effects of salicylates, derived from the bark of the willow tree, were described as early as 1763. Acetylsalicylic acid (aspirin) was first produced in 1853 (Fig. 5.4). More recently, many other NSAIDs have been developed with actions similar to aspirin. Perioperative analgesia using NSAIDs is free from many of the adverse effects of opioids, such as respiratory depression, sedation, nausea and vomiting and gastrointestinal stasis. NSAIDs have been shown to be effective analgesics in acute and chronic conditions, although significant contraindications and adverse effects limit their use.

Mechanism of Action

The mechanism of action of aspirin was discovered in the 1970s. It was shown to irreversibly inhibit the production of prostanoids (i.e. prostaglandins and thromboxanes) from arachidonic acid released from phospholipids in cell membranes (Fig. 5.5). The basal rate of prostaglandin production is low and regulated by tissue stimuli or trauma that activate phospholipases to release arachidonic acid. Prostaglandins are then produced by the enzyme prostaglandin endoperoxide synthase which has both cyclo-oxygenase and hydroperoxidase sites. At least two subtypes of cyclo-oxygenase enzyme have been identified in humans: COX-1 and COX-2. The prostanoids produced by COX-1 are functionally active in many areas, including the gastrointestinal tract, kidney, lung and cardiovascular systems. By contrast, the functional COX-2 enzyme is normally found less widely, e.g. brain, spinal cord, renal cortex, tracheal epithelium and vascular endothelium. However, COX-2 mRNA is widely distributed. In response to specific stimuli, especially those associated with inflammation, the expression of COX-2 isoenzyme is induced or upregulated, leading to increased local production of prostaglandins. A range of specific prostanoid receptors (e.g. EP1-4) are involved in peripheral sensitization associated with inflammation.

NSAIDs also have central effects as cyclo-oxygenases are widely distributed in both the peripheral and central nervous systems. NSAIDs may also have other mechanisms of action independent of any effect on prostaglandins, including effects on basic cellular and neuronal processes. NSAIDs are ‘non-selective’; they inhibit both COX-1 and COX-2.

Pharmacokinetics

All NSAIDs are rapidly absorbed. They are weak acids and are therefore mainly unionized in the stomach where absorption can occur. When given orally, most absorption occurs in the small intestine because the absorptive area of the microvilli of the small intestine is much more extensive. Most have pKa values lower than 5 and are therefore 99% ionized at a pH value greater than 7. Most are almost insoluble in water at body pH, although the sodium salt (diclofenac sodium, naproxen sodium) is more soluble. Ketorolac trometamol is the most soluble and can be given intravenously as a bolus and intramuscularly with less chance of significant irritation.

Most NSAIDs are highly protein bound (90–99%), with low volumes of distribution (approx. 0.1– 0.2 L kg–1). The unbound fraction is active. NSAIDs may potentiate the effects of other highly protein-bound drugs by displacing them from protein-binding sites (e.g. oral anticoagulants, oral hypoglycaemics, sulphonamides, anticonvulsants).

NSAIDs are mostly oxidized or hydroxylated and then conjugated and excreted in the urine. A few have active metabolites. For example, nabumetone is metabolized to 6-methoxy-2-naphthyl acetic acid, which is more active than the parent drug.

The interaction between NSAID and cyclo-oxygenase enzyme is often complex and plasma half-life may not reflect pharmacodynamic half-life. Diclofenac has a terminal half-life of 1–2 h. It is conjugated to glucuronides and sulphates, with 65% being excreted in the urine and 35% in the bile. The metabolites are less active than the parent compound. Ketorolac trometamol has a terminal half-life of 5 h and more than 90% is excreted renally. Naproxen has a terminal half-life of 12–15 h and is excreted almost entirely through the kidney as the conjugate. Tenoxicam is cleared mainly through the urine as the inactive hydroxypyridyl metabolite, although approximately 30% is via biliary excretion as the glucuronide.

Pharmacodynamics

NSAIDs are very effective analgesics, although their use is limited by adverse effects due to their general effect on prostanoid synthesis and the ubiquitous nature of prostanoids. Generally, the risk and severity of NSAID-associated side-effects is increased in the elderly population or those with other significant co-morbidity.

COX-2-SPECIFIC INHIBITORS

These drugs are anti-inflammatory analgesics that reduce prostaglandin synthesis by specifically inhibiting COX-2 with little or no effect on COX-1 (relative specificity varies between drugs). They were developed as an alternative to traditional NSAIDs with the aim of avoiding COX-1-mediated side-effects, primarily gastric ulceration and platelet effects.

Mechanism of Action

The mechanism of action is similar to that of NSAIDs (Fig. 5.4). Both COX-1 and COX-2 enzymes have very similar active sites and catalytic properties, although COX-2 has a larger potential binding site because of a secondary internal pocket. This has allowed design of drugs to target predominantly COX-2. COX-2 is induced at sites of inflammation and trauma, producing prostaglandins, and these drugs, inhibit this process. However, COX-2 is an important constitutive enzyme in the CNS, including the spinal cord, and inhibition at this site is thought to be an important mechanism also.

Pharmacodynamics

Gastrointestinal

One of the commonest side-effects of NSAIDs is gastrointestinal toxicity. Approximately 1 in 1200 patients receiving chronic NSAID treatment (> 2 months) die from related gastroduodenal complications. COX-1 isoenzyme is the predominant cyclo-oxygenase found in the gastric mucosa. The prostanoids produced here help to protect the gastric mucosa by reducing acid secretion, stimulating mucus secretion, increasing production of mucosal phospholipids and bicarbonate and regulating mucosal blood flow. Specific COX-2 inhibitors have less effect on these processes.

Short- to medium-term treatment with specific COX-2 inhibitors (up to 3 months) is associated with a significant reduction in the incidence of gastroduodenal ulceration. However, this effect is reduced during prolonged treatment and in patients taking low-dose aspirin. It is likely that the degree of COX-2 specificity is related to the efficacy of gastric protection.

Cardiovascular

Some large long-term studies of COX-2 inhibitors have found an increased risk of cardiovascular events (e.g. myocardial infarction, stroke) compared with traditional NSAIDs and placebo. This has led to the recommendation that they should not be used in patients with ischaemic heart or cerebrovascular disease and that their use in others should be guided by individual risk assessments for each patient. Some drugs have been withdrawn. A similar phenomenon has been reported when some of these drugs have been used after coronary artery bypass surgery. An increased risk of myocardial events may not be confined to COX-2 inhibitors – there is some evidence for increased risk with general NSAIDs.

The mechanism of this adverse outcome is under investigation. However, it may be that COX-2 specificity itself is the cause. COX-2 is present in vessel endothelium where it produces prostacyclins which inhibit platelet function and cause vasodilatation. Inhibition of COX-2 at this site may increase the likelihood of thrombus formation and occlusion, and therefore myocardial infarction and stroke. NSAIDs inhibit COX-2 in addition but they also inhibit COX-1 which causes significant impairment of platelet function. Therefore, the combined effect of NSAIDs is such that the risk of adverse cardiovascular and cerebrovascular events is not increased; in fact, the incidence may be decreased (certainly true for low-dose aspirin).

KETAMINE

Ketamine (2-chlorophenyl-2-methylaminocyclohexanone hydrochloride) is an anaesthetic agent that has analgesic actions at low doses. It is structurally similar to phencyclidine. It is available in parenteral formulation in a variety of concentrations (10–100 mg mL–1). It is normally presented as a racemic mixture, although the S(+) isomer is available in some countries and may have an improved therapeutic index. This isomer may be administered orally but bioavailability is relatively low and unpredictable. If given by the epidural route, the preservative-free formulation must be used.

Pharmacodynamics

Low-dose ketamine has analgesic effects in both acute and chronic pain. It appears to have an opioid-sparing effect in the postoperative setting, and there is evidence from systematic reviews of its efficacy for neuropathic pain. Its use may be limited by side-effects, although a Cochrane review of its use in the perioperative period did not find this to be a major problem. It is unclear what the optimal route of administration is. Given parenterally either intravenously or subcutaneously, bolus doses of 0.25–0.5 mg kg–1 or infusion rates of 0.125–0.25 mg kg–1 h–1 can provide analgesia.

Psychotomimetic effects can often limit use. Hallucinations and nightmares may be troublesome. In large doses, or in susceptible individuals, excess sedation can occur. Ketamine can cause hypertension, increased heart rate and cardiac output, and increased intracranial pressure.

FURTHER READING

Bell, R.F., Dahl, J.B., Moore, R.A., Kalso, E.A., Perioperative ketamine for acute postoperative pain. [Art. No.:CD004603]. Cochrane Database Syst. Rev. 2006;2006(1); doi:10.1002/14651858, CD004603.pub2

Christo, P.J. Opioid effectiveness and side effects in chronic pain. Anesthesiol. Clin. North America. 2003;21:699–713.

Colvin, L.A., Fallon, M.T. Opioid-induced hyperalgesia – a clinical challenge. Br. J. Anaesth. 2010;104:125–127.

Dietis, N., Guerrini, R., Calo, G., et al. Simultaneous targeting of multiple opioid receptors: a strategy to improve side-effect profile. Br. J. Anaesth. 2009;103:38–49.

Hinz, B., Brune, K. Paracetamol and cyclooxygenase inhibition: is there a cause for concern? Ann. Rheum. Dis. 2012;71(1):20–25.

Hocking, G., Cousins, M.J. Ketamine in chronic pain management: an evidence-based review. Anesth. Analg. 2003;97:1730–1739.

Mather, L.E. Trends in the pharmacology of opioids: implications for the pharmacotherapy of pain. Eur. J. Pain. 2001;5(Suppl. A):49–57.

McDonald, J., Lambert, D.G. Opioid receptors. Continuing Education in Anaesthesia, Critical Care and Pain. 2005;5:22–25.

Yaksh, T.L. Pharmacology and mechanisms of opioid analgesic activity. Acta Anaesthesiol. Scand. 1997;41:94–111.