20
PRACTICAL THERAPEUTICS Drugs 49 (I): 51-70, 1995 0012-6667/95/0001-0051/$20.00/0 © Ads International Limited. All rights reserved, Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management Mechanisms of Action and Rationale for Optimum Use Jeremy Cashman and Gregory McAnulty Department of Anaesthesia, St George's Hospital, London, England Contents Summary , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , 1, The Mechanism of Action of Nonsteroidal Anti-Inflammatory Drugs (NSAIDs) 1 ,1 Tissue Injury and the Arachidonic Acid Cascade ,"" 1 ,2 Inhibition of Prostaglandin Biosynthesis """"'" 1 ,3 Central Antinociceptive Mechanism of Action of NSAIDs , 2, Dose-Response Effects and Enantiomeric NSAIDs 2,1 Variability in Drug Response , 2,2 Stereoisomerism and NSAIDs , , 3, Adverse Responses to NSAIDs ",. 3.1 Adverse Gastrointestinal Effects 3.2 Adverse Renal Effects .. , , , , 3.3 Aspirin and Bronchospasm '" 4, Choosing an NSAID for Use in the Peri operative Period . 4.1 Rational Prescribing of NSAIDs ...... . 4.2 Pharmacokinetic-Pharmacodynamic Relationships. 5. Dosage Regimens . . 5.1 Oral and Rectal . , . 5.2 Parenteral . . . . . , 6. Preoperative Administration 7, Conclusions . . . , . . . . . . 51 52 52 55 57 58 58 59 60 61 62 63 63 63 63 65 65 65 66 66 Summary Nonsteroidal anti-inflammatory drugs (NSAIDs) are a group of agents with similar actions but diverse chemical structures. Aspirin (acetylsalicylic acid) and sodium salicylate were the first drugs of this type to be used clinically. However, over the past 3 decades there has been a dramatic increase in the number of NSAIDs available for the treatment of postoperative pain. Tissue injury, such as occurs with surgical intervention, is associated with the release of numerous inflammatory mediators including prostaglandins. Prosta- glandins derived from the arachidonic acid cascade are implicated in the produc- tion of inflammatory pain, and in sensitising nociceptors to the actions of other mediators. They are synthesised from arachidonic acid via the endoperoxide bio- synthesis pathway, the initial step of which is catalysed by the enzyme cyclo- oxygenase. Two forms ofthe cyclo-oxygenase enzyme (COX-I and COX-2) have been characterised. COX-l is important in circumstances where prostaglandins

Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

Embed Size (px)

Citation preview

Page 1: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

PRACTICAL THERAPEUTICS Drugs 49 (I): 51-70, 1995 0012-6667/95/0001-0051/$20.00/0

© Ads International Limited. All rights reserved,

Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management Mechanisms of Action and Rationale for Optimum Use

Jeremy Cashman and Gregory McAnulty Department of Anaesthesia, St George's Hospital, London, England

Contents Summary , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , 1, The Mechanism of Action of Nonsteroidal Anti-Inflammatory Drugs (NSAIDs)

1 ,1 Tissue Injury and the Arachidonic Acid Cascade ,"" 1 ,2 Inhibition of Prostaglandin Biosynthesis """"'" 1 ,3 Central Antinociceptive Mechanism of Action of NSAIDs ,

2, Dose-Response Effects and Enantiomeric NSAIDs 2,1 Variability in Drug Response , 2,2 Stereoisomerism and NSAIDs , ,

3, Adverse Responses to NSAIDs ",. 3.1 Adverse Gastrointestinal Effects 3.2 Adverse Renal Effects .. , , , , 3.3 Aspirin and Bronchospasm '"

4, Choosing an NSAID for Use in the Peri operative Period . 4.1 Rational Prescribing of NSAIDs ...... . 4.2 Pharmacokinetic-Pharmacodynamic Relationships.

5. Dosage Regimens . . 5.1 Oral and Rectal . , . 5.2 Parenteral . . . . . ,

6. Preoperative Administration 7, Conclusions . . . , . . . . . .

51 52 52 55 57 58 58 59 60 61 62 63 63 63 63 65 65 65 66 66

Summary Nonsteroidal anti-inflammatory drugs (NSAIDs) are a group of agents with similar actions but diverse chemical structures. Aspirin (acetylsalicylic acid) and sodium salicylate were the first drugs of this type to be used clinically. However, over the past 3 decades there has been a dramatic increase in the number of NSAIDs available for the treatment of postoperative pain.

Tissue injury, such as occurs with surgical intervention, is associated with the release of numerous inflammatory mediators including prostaglandins. Prosta­glandins derived from the arachidonic acid cascade are implicated in the produc­tion of inflammatory pain, and in sensitising nociceptors to the actions of other mediators. They are synthesised from arachidonic acid via the endoperoxide bio­synthesis pathway, the initial step of which is catalysed by the enzyme cyclo­oxygenase. Two forms ofthe cyclo-oxygenase enzyme (COX-I and COX-2) have been characterised. COX-l is important in circumstances where prostaglandins

Page 2: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

52 Cashman & McAnulty

have a protective effect such as gastric mucus production and renal blood flow maintenance. NSAIDs inhibit the synthesis of prostaglandins at 1 or more points in the endoperoxide pathway. Three mechanisms of inhibition of the biosynthetic enzymes have been proposed: (i) rapid, reversible competitive inhibition; (ii) irreversible, time-dependent inhibition; and (iii) rapid, reversible noncompetitive (free radical trapping) inhibition. In addition, there is evidence that NSAIDs have a central anti nociceptive mechanism of action that augments the peripheral effect. This may involve inhibition of central nervous system prostaglandins or inhibi­tion of excitatory amino acids or bradykinins.

There is considerable variability in the pain relief obtained from NSAIDs. Such variability in drug response may be explained in terms of differences be­tween agents with respect to either pharmacodynamic actions or pharmacokinetic parameters or a combination of both. Stereoisomerism, where preparations exist as racemic mixtures and where only 1 enantiomer is active, may also be important. However, chiral inversion from inactive to active enantiomer may occur and may be rapid or slow.

NSAIDs have numerous adverse effects. Gastrointestinal disturbances includ­ing ulceration are the commonest adverse responses to NSAIDs and carry the greatest risk of death. Also significant are renal impairment and an increased risk of postoperative haemorrhage. Asthma and allergic reactions are uncommon.

The choice of NSAID should be made on a rational basis. For short term perioperative use it is advisable to favour drugs with good safety profiles, which are available in a range of formulations. It is important to review therapy regularly, changing to an alternative NSAID if there is poor response to treatment. NSAIDs should not be used in patients with known contraindications to their use. Disparity between clinical effect and plasma concentration of some NSAIDs may be due to a complex, time-dependent concentration-effect relationship. Dosage of NSAIDs should be tailored to individual patient response, based on clinical as­sessment. Oral mandatory dosage regimens and intravenous (or even intramus­cular) infusions may result in steady state therapeutic plasma concentrations of NSAIDs and therefore provide consistent analgesia. Finally, there seems to be little benefit from preoperative administration of NSAIDs, indeed haemorrhagic complications may be more likely.

Sodium salicylate was first used for the treat­ment of rheumatic fever in 1875. In 1899, aspirin

(acetylsalicylic acid) was introduced as a result of its antipyretic, anti-inflammatory and analgesic ef­fects. Within a comparatively short period of time a number of other drugs with diverse chemical structures, which shared some, if not all, of the ef­fects (as well as the adverse effects) of aspirin, were discovered. As a result of the similarity of thera­

peutic action of these drugs they have tended to be

regarded as a single group known as the aspirin­

like drugs. More commonly these agents are re-

ferred to as the nonsteroidal anti-inflammatory drugs (NSAIDs).

1. The Mechanism of Action of Nonsteroidal Anti-Inflammatory Drugs (NSAIDs)

1.1 Tissue Injury and the Arachidonic Acid Cascade

Tissue damage is associated with release of a host of inflammatory mediators including hista­mine, serotonin (5-hydroxytryptamine), bradyki-

© Adis International Limited, All rights reserved. Drugs 49 (1) 1995

Page 3: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

NSAIDs in Peri surgical Pain Management

nin, platelet activating factor, interleukin-l, pros­taglandins, thromboxanes and leukotrienes.[l] The importance of these mediators in the inflammatory response can be ascertained by eliminating activity of each mediator in turn, either by using enzyme inhibitors or by blocking the pharmacological ef­fect of the mediator using specific antagonists.[l] The discovery that NSAIDs selectively inhibit the biosynthesis of prostaglandins led to the theory that prostaglandins are important inflammatory mediators)2,3] Furthermore, the analgesic effect of drugs that inhibit prostaglandin biosynthesis may be explained if prostaglandins are responsible for inflammatory pain.l4]

Von Euler[5] coined the term prostaglandin to describe the extract from semen that contracted uterine smooth muscle. Although prostaglandins are derived from arachidonic acid and other poly­unsaturated fatty acids, the 20-carbon polyunsatu­rated essential fatty acid (arachidonic acid) is the major source in mammalian tissues. Prostaglan­dins derived from arachidonic acid contain 2 dou­ble bonds [e.g. prostaglandin E2 (PGE2), throm­boxane and prostacyclinJ. Analogous compounds synthesised from eicosatrienoic (linoleic) and eicosapentaenoic acid contain 1 less or 1 more dou­ble bond in the side chains (e.g. PGEl and PGE3, respectively). The various groups of prostaglan­dins, thromboxanes, hydroxy acids and leuko­trienes which retain the 20-carbon unsaturated fatty acid backbone are collectively known as eicosanoids. The 3 fatty acid precursors (eicosa­tetraenoic acid, eicosatrienoic acid and eicosa­pentaenoic acid) are derived directly or indirectly from dietary fat and are esterified into cell mem­branes. Their release, usually as a result of trauma, is the major stimulus for eicosanoid production as prostaglandins cannot be stored and are released as soon as they are synthesised.

A number of enzymes catalyse the conversion of arachidonic acid to prostaglandins, thrombox­anes, leukotrienes and hydroxy acids. The produc­tion of prostaglandins depends on the release of arachidonic acid from cell membrane phos­pholipids. Phospholipase activation liberates ara-

© Adis International Limited. All rights reserved.

53

chidonic acid which in turn acts as a substrate for cyclo-oxygenase and hence, because tissue dam­age is associated with inflammation, prostaglandin production accompanies the inflammatory re­sponse. Phospholipase A2 is activated in response to mechanical, chemical and immunological stim­ulation. The enzyme cyclo-oxygenase, which is a membrane-bound haemo- and glycoprotein with a molecular weight of7lkD, was isolated in 1976J6] Cyclo-oxygenase catalysed peroxidation of ara­chidonic acid results in a cyclic structure whereas peroxidation catalysed by lipo-oxygenase pro­duces straight chain hydroxy-peroxy acids (HPETEs) which can then be converted to hydroxy acids (HETEs). The cyclo-oxygenase and lipo­oxygenase pathways represent the major routes for the oxidative metabolism of arachidonic acid (fig. 1).[1]

Prostanoids do not generally activate nociceptors directly, but sensitise them to mechan­ical stimuli and chemical mediators of nociception such as bradykininJ4,7] However, the evidence clearly indicates that stable prostaglandins of the E series are involved in the hyperalgesia seen in acute inflammation.l81 PGE2 is the predominant eicosanoid in such inflammatory conditions, act­ing synergistically with other mediators to produce inflammatory pain.l l ,9] PGE2 has no direct pain producing activity,[lO] but it does sensitise recep­tors on afferent nerve endings to the actions of bra­dykinin and histamineV]

Many other cyclo-oxygenase products have been detected in inflammatory lesions, but usually they are present at only a fraction of the concentra­tion of PGE2. Prostacyclin is the most important of these in terms of inflammatory signs[l] and may even be present in comparable concentrations to PGE2JlO] Higgs et al.[ll] proposed that pro­stacyclin was a more potent vasodilator than PGE2 by several orders of magnitude. More recently however, Vane and Botting,D] have suggested that their potency is similar. Nevertheless, prostacyclin is a more potent hyperalgesic agent in both rats and dogsJ4,l2] In both these animal models the hyper­algesia produced by prostacyclin is immediate and

Drugs 49 (1) 1995

Page 4: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

54

Vasoconstrictionfvasodilatation Pain Inflammation Bronchial constriction Gastric mucus production

Fig. 1. The endoperoxide biosynthesis pathway.

of short duration. That produced by PGE2 is slow in onset and of long duration.!4,JO)

Higgs et aU8) report that carrageenin-induced hyperalgesia in rat paw preparations is likely to be mediated by release of a short-acting product of cyclo-oxygenase since the response is interrupted by indomethacin. This observation has prompted the suggestion that, because aspirin-like drugs would be ineffective against the prolonged activity of PGE2, the endogenous mediator of hyperalgesia is probably prostacyclin.!8l The lipo-oxygenase

© Adis International Limited. All rights reserved.

Platelet disaggregation Vasodilatation

Prostaglandins

Pain Inflammation Fever

Cashman & McAnulty

Platelet aggregation VasoconstrICtion

Bronchial constrictionfdilatation Blood flow

pathway results in the production of other arachid­onic acid metabolites which may also have algo­genic properties. Leukotriene B4 is a potent chemo­tactic factor for polymorphs, which in turn lowers the firing threshold of pain fibres and thus stimu­late nociceptors directly. HETE is also highly che­motactic (fig. 1).

It is now known that the cyclo-oxygenase en­zyme is encoded by 2 genes.!l3l Two forms of the cyclo-oxygenase enzyme (COX-l and COX-2) have been characterised.

Drugs 49 (1) 1995

Page 5: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

NSAIDs in Perisurgical Pain Management

COX-1 is produced in normal, quiescent condi­tions and is a constituent of healthy cells. It is im­portant in circumstances where prostaglandins have a protective function, such as gastric mucus production and renal blood flow maintenance. COX-2, the inducible form of the enzyme, is the major isozyme associated with inflammation. COX-2 is induced in endothelial cells, macro­phages and synovial fibroblasts by inflammatory agents.

The ratio of inhibition of COX-1 to COX-2 by NSAIDs will determine the likelihood of adverse effects. NSAIDs such as aspirin and indomethacin with a high ratio (indicating a greater degree of inhibition of the protective COX-1 enzyme) will exhibit more adverse effects than an NSAID such as ibuprofen that has alow ratio ofCOX-1 to COX-2 inhibition. The challenge must be to develop drugs selective for the inducible form of the en­zyme. It is of interest that dexamethasone, at con­centrations that are anti-inflammatory, inhibits COX_2.[13]

1.2 Inhibition of Prostaglandin Biosynthesis

NSAIDs inhibit the synthesis of prostaglandins at 1 or more points in the endoperoxide biosynthe­sis pathway. This property is a general charac­teristic of the aspirin-like drugs[l] and is believed to be the basis of their analgesic action since the products of the arachidonic acid cascade promote the pain associated with inflammation.

Up until the early 1970s, many attempts had been made to link the anti-inflammatory actions of substances like aspirin with the ability to inhibit the activity of endogenous substances such as ki­nins, slow reacting substance of anaphylaxis (SRS­A), adenosine triphosphate (ATP), arachidonic acid and prostaglandins. The hypothesis that anti­inflammatory substances such as aspirin might act by inhibition of the enzymes which synthesise prostaglandins was tested by Vane in 1971.[3] In cell-free homogenates of guinea-pig lung, dose­dependent inhibition of prostaglandin formation by (in order of potency) indomethacin, aspirin and sodium salicylate was demonstrated. Three other

© Adis International Limited. All rights reserved.

55

drugs, morphine, hydrocortisone and mepyramine, had little effect. Vane proposed that the pain reliev­ing action of aspirin-like drugs could be explained by inhibition of prostaglandin synthesisJ3,14-16]

At about the same time Ferreira and colleagues demonstrated that indomethacin and aspirin (but, interestingly, not sodium salicylate) blocked pros­taglandin release from isolated perfused dog spleen, an organ known to release large amounts of prostaglandins (mainly E2 and F2a) in response to sympathetic nervous system stimulation,l3] Hydrocortisone was again without effect. Using the same dog spleen model, Flower[17] demon­strated that, in addition to aspirin and indometha­cin, many other drugs of the aspirin type (including phenylbutazone and mefenamic acid) and paracet­amol (acetaminophen) blocked prostaglandin bio­synthesisJI5,17] Indomethacin is a more potent in­hibitor of prostaglandin synthetase than aspirin in the microsomal extract of dog spleen, while mefe­namic acid and phenylbutazone are intermediate in potency between aspirin and indomethacinJl5]

In a detailed examination of the mechanism of the anticyclo-oxygenase activity of NSAIDs, Lands proposed that cyclo-oxygenase inhibitors can be classified into at least 3 distinct catego­ries:[18]

• drugs which cause rapid, reversible competitive enzyme inhibition

• drugs which cause irreversible, time-dependent enzyme inactivation

• drugs which cause rapid, reversible noncompet­itive (free radical trapping) inhibition (fig. 2). Competitive inhibition occurs when a substrate

analogue, such as a closely related polyunsaturated fatty acid, has a binding constant for the enzyme that is similar to the substrate arachidonic acid, but which does not form a product. Drugs such as mef­enamic acid and the propionic acid derivative ibuprofen provide examples of this type of inhibi­tionJl9] Flufenamic acid and sulindac are consid­ered also to compete with arachidonic acid for binding to cyclo-oxygenaseJI9] A substituted car­boxylic acid residue probably mimics the terminal carboxyl of arachidonic acid whilst hydrophobic

Drugs 49 (1) 1995

Page 6: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

56

Substrate or substrate analogue

+ I Enzyme I

+ I Inhibitor I ~================

2

Enzymelinhlbitor complex

3

Cashman & McAnulty

Enzyme/substrate

Stable, inactive enzymelinhlbitor complex

Fig. 2. Suggested mechanism of action of nonsteroidal anti-inflammatory drugs (NSAIDs).[18] Symbols: 1 = drugs which cause rapid, reversible competitive inhibition; 2 = drugs which cause irreversible, time-dependent inactivation; 3 = drugs which cause rapid, reversible noncompetitive (free radical trapping) inhibition.

groups bind to the enzyme; this appears to be nec­essary for competitive inhibition. The presence of an aryl halogen enhances this activityJ19] Thus, for ibuprofen, binding to cyclo-oxygenase appears to be dependent on hydrophobic features rather than the ionic character of the moleculeJ18]

Aspirin, the best known example of the irre­versible, time-dependent class of inhibitors, is a site-specific acetylating agent. It forms a covalent, acetylated derivative of the enzyme by selectively acetylating the hydroxyl group of a single serine (ser 530) residue located 70 amino acids from the C terminus of the enzymeJ20] This causes irre­versible inactivation of cyclo-oxygenase. Once the enzyme has been inactivated by this irreversible acetylation, removal of the drug will not result in synthesis of further prostaglandin until new en­zyme is formed by protein synthesis in the tissues. Cyclo-oxygenase but not peroxidase activity is in­hibited when purified enzyme is acetylated. Thus, at low concentrations aspirin rapidly and selec­tively acetylates prostaglandin endoperoxidase synthesis. At high concentrations aspirin acetylates nonspecifically a variety of proteins and nucleic acidsJ21]

Other drugs are competitive, irreversible, time­dependent inhibitors by virtue of competing with the substrate for enzyme binding rather than induc­ing acetylation. Kulmacz and Lands[22] have estab­lished that indomethacin, meclofenamic acid and

© Adis International Limited, All rights reserved,

flurbiprofen interact in a time-dependent manner with prostaglandin H (PGH) synthetase to inhibit cyclo-oxygenase activity. The effect of these 3 agents is to diminish rather than destroy the cata­lytic activity of cyclo-oxygenase. Since indometh­acin can be recovered intact after inhibiting the en­zyme, the process probably does not involve covalent modification but rather a conformational change in the synthetase brought about by stereo­specific binding. Binding is initially rapid and re­versible, but the synthetase/inhibitor complex gradually decays to produce a stable form with only a fraction of the original activity. The majority of propionic acid derivatives, including naproxen, act as structural analogues of cyclic en do peroxides rather than arachidonic acidJ231

Lands[18] proposed that in inflammatory disor­ders the continual presence of lipid peroxide in­duces a free radical chain reaction mechanism which sustains cyclo-oxygenase biosynthesis of more peroxides. Hydroperoxides generated during arachidonic acid metabolism exert a positive feed­back mechanism and stimulate cyclo-oxygenase activityJ24] This peroxide tone can be blocked by the addition of free radical scavengers or antioxi­dant agents, which act as reversible noncompeti­tive inhibitors. Even when large amounts of cyclo­oxygenase are present together with substrate, little prostaglandin biosynthesis will occur in the pres­ence of a free-radical scavenger. Paracetamol may

Drugs 49 (1) 1995

Page 7: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

NSAIDs in Peri surgical Pain Management

block prostaglandin synthesis by this mecha­nism)25]

There are important differences between revers­ible inhibitors and irreversible, time-dependent in­hibitors. In general, a reversible inhibitor influ­ences cyclo-oxygenase only to the extent that the binding site is occupied by the agent. Dilution of the agent or competitive reversal of its binding by increased substrate allows formation of prosta­glandins to proceed. A time-dependent inhibitor appears to depend on the combined presence of a halogen with a free carboxylic acid groupJl9] At low concentrations they may not occupy more than 10% of enzyme sites at a given moment, but will lead to complete loss of activation with time.

1.3 Central Antinociceptive Mechanism of Action of NSAIDs

It is generally believed that the effects of NSAIDs are mediated through a peripheral mech­anism related to inhibition of prostaglandin syn­thesis. However, as early as 1899 Dreser suggested that aspirin might act on the central nervous sys­tem)26] There is increasing evidence that NSAIDs have a central mechanism of action which aug­ments the peripheral mechanism. Some of these drugs have an analgesic action that is dispropor­tionate to their anti-inflammatory action)27] Inhi­bition of prostaglandin release in the central nerv­ous system may be responsible for the antipyretic effect of NSAIDSJI6,28] It is also possible that a central action may involve inhibition of neural ac­tivity induced by excitatory amino acids or brady­kinins)29]

The behavioural responses of rats to subcutane­ous injection of formalin, acetic acid, phenol and carrageenin,[28] as well as evoked activity in re­sponse to electrical stimulation of nociceptive afferent nerves,[30,31] have been used to test the hypothesis that NSAIDs may have a central mech­anism of action. Ferreira et al)32] compared the peripheral and central effects of aspirin, indometh­acin, paracetamol and phenacetin on the hyperal­gesia induce by carrageenin injected into rat paws. Administration of these drugs into the cerebral

© Adis International Limited. All rights reserved.

57

ventricles was associated with an enhanced anal­gesic effect. Combined administration of these drugs into paws and intraventricularly resulted in a synergistic rather than merely additive effect. Ad­ministration of the prostaglandin antagonist into the cerebral ventricles also inhibited hyperalgesia evoked by carrageenin.

Hunskaar[33] suggested that the behavioural re­sponse to intra-peritoneal injection offormalin and acetic acid shows 2 phases; the first due to direct stimulation of nociceptors and the second involv­ing pain secondary to inflammation. The short la­tency of the immediate response, he suggests, rep­resents a central mechanism of action. Acetyl salicylic acid reduced the direct response to acute noxious stimulation. Clear antinociceptive effects of aspirin were noted within 30 seconds of noxious stimulation, at a time when no established inflam­mation would be present and aspirin and morphine showed almost identical time-effect relationships.

Carlsson et al)30] recorded activity from as­cending axons of the spinal cord and single neu­rones in the dorsomedial part of the ventral nucleus (VDM) of rat thalamus elicited by supramaximal electrical stimulation of nociceptive afferent nerves in the sural nerve. They found that intra­venous aspirin, paracetamol, the pyrazolone de­rivatives metamizol and aminophenazone and morphine, dose-dependently depressed evoked ac­tivity in VDM neurones. Naloxone abolished the depressant effects of morphine but failed to reduce those of the nonopioids. Unlike morphine, the non­opioid analgesics, even at relatively high doses, did not completely block evoked activity in VDM neu­rones. It was suggested that so-called peripherally acting analgesic agents produce a central supraspi­nal analgesic effect which is weaker than mor­phine.

Jurna and Brune[31] investigated indomethacin, ibuprofen and diclofenac using the same rat model as Carlson et al. [30] and found that the 3 drugs produced a dose-dependent depression of evoked activity suggesting that a central action contributes to their analgesic effect. They observed that indo-

Drugs 49 (1) 1995

Page 8: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

58

methacin was more potent than diclofenac, which was more potent than ibuprofen.

McCormack and Brune[34] have reviewed a number of reports that support the existence of a central mechanism of action of NSAIDs in hu­mans. They quote studies which indicate that NSAIDs inhibit brain prostaglandin synthesis. However, the clinical relevance remains unclear since the pain-related role of prostaglandins in the central nervous system is not well established. In human volunteers the late pain response of the elec­troencephalogram (EEG) evoked by electrical stimulation of tooth pulp is decreased by diclofenac)35] Electrophysiological evidence for a supraspinal analgesic effect of keto prof en has been strongly suggested in a double-blind crossover study in paraplegic patients)27] Intrathecal admin­istration of lysine acetylsalicylate relieved intrac­table pain from a variety of aetiologies)36]

Various mechanisms may be suggested to ac­count for the central action of NSAIDs. Interfer­ence with the formation of prostaglandins or with transmitters or modulators in the nociceptive sys­tem may occur. Alternatively, the central action may be mediated in part by endogenous opioid pep­tides or they may block the release of serotonin by central secretion of bradykinin)3!]

2. Dose-Response Effects and Enantiomeric NSAIDs

2.1 Variability in Drug Response

Over the last 3 decades there has been a dramatic increase in the number of new NSAIDs available. Unfortunately, this increase has not been matched by a significant improvement in therapeutic in­dex)37] Furthermore, there is considerable inter­patient variability in pain relief obtained from NSAIDs.[37-39] As a result, ithas proved extremely difficult to construct any meaningful rank order of efficacy from the published data)37,40] Scott et al. [40] studied the variability in response of patients to aspirin, benory late, diclofenac, fenoprofen, flurbiprofen, ibuprofen, indomethacin, ketoprofen, naproxen and sulindac. They found that variation

© Adis International Limited. All rights reserved.

Cashman & McAnulty

between patients in response to the drugs was large whereas there was no discernible difference be­tween the drugs overall.

Early evidence in support of individual variabil­ity in response to NSAIDs was from a study[41] of the response of patients with rheumatoid arthritis to 4 different NSAIDs (fenoprofen, ibuprofen, ketoprofen and naproxen). There was striking vari­ation in the individual patient responses despite only minor differences in the mean outcome meas­ures. Adverse effects were less common with ibuprofen and naproxen, but naproxen was recom­mended as the first choice by these authors because it was slightly more effective.

In another comparative study of aspirin, feno­profen, ibuprofen, naproxen and tolmetin, Gall et al)42] also noted marked individual differences in response. There may be 2 possible, not necessarily mutually exclusive, causes for the interindividual variability in response to NSAIDs. These include pharmacodynamic actions and pharmacokinetic parameters. [37]

Capell et al)43] were unable to demonstrate a significant difference in serum concentration of unchanged flurbiprofen between responders and nonresponders. Several other studies have investi­gated the pharmacokinetics ofNSAIDs in respond­ing and nonresponding groups of patients. Baber et al)44] and Orme et al.l45] found no difference be­tween responders and nonresponders in the phar­macokinetics of indomethacin and flurbiprofen. In a dose-response study with ibuprofen, Grennan et al)46] reported considerable variation in plasma concentration and no significant correlation with clinical response. In contrast, Preston et al)47] studied patients' response to flurbiprofen on 2 sep­arate occasions and found that, whilst the response to flurbiprofen correlated with the degree of pain at commencement of the 2 study periods, there was no evidence to support the concept of responders and nonresponders. The basis of the lack of dose­response and concentration-response relationships in nonresponders may be pharmacokinetic.

Day and Brooks[37] have suggested that meas­urement of the active component of the drug will

Drugs 49 (1) 1995

Page 9: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

NSAIDs in Perisurgical Pain Management

be most likely to provide a correlation. This is likely to be most easily demonstrated with the pro­pionic acid derivatives which exist as active and inactive stereoisomers.

2.2 Stereoisomerism and NSAIDs

Stereoisomers possess a centre of asymmetry and exist as either right-handed [R or (-)] or left­handed [S or (+)] forms which are nonsuperimpos­able mirror images (enantiomers). Many synthetic chiral drugs, including a number of NSAIDs, are administered as racemic mixtures of Rand S iso­mers. There are often important differences in the activity of the isomers and the absorption, distri­bution, metabolism and excretion of such chiral mixtures may be stereospecific. In the case of pro­pionic acid derivatives chiral inversion from one isomer to the other may occur in vivo. Anti-inflam­matory analogues of propionic acid exhibit optical isomerism; only the S-enantiomers inhibit prosta­glandin synthesis.[48]

The propionic acid derivatives are not the only NSAIDs to exhibit isomerism. Enantiomers of ketorolac have also been identified.l49] However, naproxen is the only member of its class that is presented as the active S-enantiomer, whereas racemic mixtures (inactive R- and active S­enantiomers) of other propionic acid derivatives such as benoxaprofen, flurbiprofen, ibuprofen and ketoprofen are used in clinical practice.l50,51]

A unique unidirectional stereospecific inver­sion of the inactive R-enantiomer to the active S­enantiomer occurs to a variable degree in vivo.l48,52] The extent of this inversion varies de­pending on the drug and the individual per­son.[50,53] The R-enantiomer of fenoprofen is con­verted almost totally to the -enantiomer, whereas very little R-ketoprofen or R-flurbiprofen is con­verted to the S form. [5 1,54] In a study of the kinetics of chiral inversion in dogs Ahn et al.l55] have shown that 70 to 75% of an administered dose of ibuprofen undergoes R to S inversion. Furthermore feedback inhibition occurs, in which R to S inver­sion may be inhibited by the S-enantiomer.

© Adis International Limited. All rights reserved.

59

In rats, the bioavailability of the active S­enantiomer of pirprofen is greater than the inactive R-enantiomer.l56] No chiral inversion of pirprofen occurs and, because the S-enantiomer is less pro­tein bound, this suggests that first pass metabolism is also stereoselective. In humans, according to Lee et al.l48] an average of 63% of an administered dose of R-ibuprofen is inverted to the S-enantiomer.

The terminal half-lives of the 2 enantiomers of ibuprofen are similar in humans, but plasma con­centrations of the S-enantiomer exceed those of the R-enantiomer because of chiral inversion. The pharmacokinetics of each isomer are linear. How­ever, there is large interindividual variability in the ratio of R to S, which probably accounts for the lack of correlation between racemic ibuprofen concen­tration and therapeutic efficacy.l57]

The inversion reaction probably occurs for all arylpropionic acid analogues. Even naproxen, which is commercially available as the S-enantiomer under­goes chiral inversion.[52] Experiments with rat liver homogenates have shown that R-ibuprofen and R­fenoprofen but not S-ibuprofen or R- and S­flurbiprofen undergo chiral inversion, findings which are consistent with human experienceJ54,58] The pre­requisite for stereospecific inversion appears to be the highly stereoselective esterification of the R-enantio­mer via its coenzyme-A, thioester. Once the R-ester is formed, racemisation and hydrolysis oc­curs.l48,50,54,58] In the case of rapid inversion, inac­tive R-enantiomer acts as a prodrug for the active S-antipode. In the case of slow inversion, R is an unnecessary impurity.[52]

With the exception of naproxen and fenclo­fenac, no correlation has been established between dose, plasma concentration and effect for any NSAID of the 2-arylpropionic acid class.l45,46] Grennan et al.[46] found very poor correlation be­tween plasma concentration and clinical response to ibuprofen. A plateau effect was observed in rheumatoid arthritis patients, with ibuprofen 1600 mg/day seeming to be the optimal dose. Lee et al.[48] have stated that the pharmacokinetics of ibuprofen cannot be defined satisfactorily in terms of total ibuprofen concentrations. Hutt and Cald-

Drugs 49 (1) 1995

Page 10: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

60

Table I. Adverse effects of nonsteroidal anti·inflammatory drugs (NSAIDs)

Common Gastrointestinal disturbances

Gastric irritation

Increased bleeding time

Renal impairment

Uncommon

Asthma

Allergy

well [52] agree and state that it is essential to assay active isomer, since assay of the racemate is oflim­ited value. It is possible that measurement of the concentration of the active enantiomer rather than racemic drug in plasma will enable a correlation between plasma concentration and efficacy to be established for NSAIDs of the 2-ary lpropionic acid classJ53]

There may be benefits to using the S-enantiomer as opposed to racemic mixtures since toxicity of NSAIDs is related to the active formJ52] Chiral inversion might be expected to enhance toxicity. Hutt and Caldwell[52] suggest that use of the S-en­antiomer permits the use of a reduced dose and hence reduces variability of response. Stock et al.l59] found no difference between 2 groups of pa­tients treated with either racemic ibuprofen 600mg 3 times daily or S-ibuprofen 400mg 3 times daily with respect to efficacy or adverse effects. They concluded that S-ibuprofen was advantageous be­cause it reduced the metabolic load to the body. However, not everyone agrees. Geisslinger et al.l60] found that the interindividual variation in the pharmacokinetics of S-ibuprofen following race­mate administration was similar to that following administration of the single isomer, suggesting that chiral inversion was not a major factor contributing to variability in drug disposition. Geisslinger et al.l60] felt that the administration of a single isomer offered no advantage.

3. Adverse Responses to NSAIDs

NSAIDs can damage gastrointestinal mucosa, impair renal function and may be associated with

© Adis International Limited. All rights reserved.

Cashman & McAnulty

an increased risk of postoperative haemorrhage. They can also provoke acute asthma in susceptible patients (table I). However, gastrointestinal ad­verse effects, such as dyspepsia with a tendency to gastric erosions and ulceration, are the main ad­verse effects of NSAIDs. Lesions of the gastroin­testinal tract are not predictable, and all NSAIDs are incriminated to some extent. The association between peri operative NSAID treatment, upper gastrointestinal, renal, haematological and allergic adverse effects have been extensively reviewed previously in Drugs.l61 ,62]

The UK Committee on Safety of Medicines and the Medicines Control Agency reviewed the evi­dence on the relative safety of7 of the most widely used oral non-aspirin NSAIDs. Results from 10 ep­idemiological studies were combined with results from adverse drug reaction reporting in the UK over the last 5 years. The combined information indicates that azapropazone is associated with the highest risk and ibuprofen with the lowest risk of serious upper gastrointestinal toxicity. Diclofenac, indomethacin, ketoprofen, naproxen and piroxi-

Table II. Comparison of acute toxicity (expressed as LD50) and gastrointestinal toxicity (expressed as ED50) of various nonsteroi­dal anti-inflammatory drugs (NSAIDs)164]

Compound Mean LD50 Mean ED50 (mg/kg) (mg/kg)

Propionic acids Ibuprofen 969 15

Indoprofen 7.5

Flurbiprofen 1.0

Ketoprofen 160 3.5

Naproxen 543 3.2

Acetic acids

Diclofenac 240 6.5

Indomethacin 19

Fenamates

Mefenamic acid 790

Pyrazolones

Phenylbutazone 700

Oxicams

Piroxicam 255 2.9

Abbreviations: ED50 = dose that produces gastriC ulceration in 50% of animals; LD50 = dose that is lethal to 50% of animals.

Drugs 49 Cl) 1995

Page 11: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

NSAIDs in Peri surgical Pain Management 61

Table III. Ranking of nonsteroidal anti-inflammatory drugs (NSAIDs) according to total deaths and complications in which they have been implicated651

NSAID Total serious reactions (deaths)

Azapropazone 87.9 (9.9)

Piroxicam 68.1 (6.2)

Fenoprofen 43.7 (6.6)

Naproxen 41.1 (5.6)

Diclofenac 39.4 (3.1)

Flurbiprofen 35.8 (3.3)

Indomethacin na (3.3)

Ketoprofen 38.6 (1.6)

Ibuprofen 13.2 (0.7)

Abbreviation: na denotes data not available.

cam are associated with intermediate risk, al­though the latter may be associated with a higher risk than other NSAIDs in this group. Non-gastro­intestinal adverse reactions to NSAIDs (i.e. renal, hepatic, allergic and haematological) were less common. Spontaneous reporting in the UK over the last 14 years indicates that azapropazone fol­lowed by diclofenac are most likely to cause these adverse reactions, while ibuprofen, indomethacin and ketoprofen are least likely.[63]

3.1 Adverse Gastrointestinal Effects

The acute toxicity of NSAIDs in rats is re­viewed in table II. Gastrointestinal adverse effects are the commonest adverse reaction to NSAIDs and constitute the greatest risk of death (table III). The dosage and duration of NSAID treatment, as well as a history of peptic ulceration, are the most important determinants of the likelihood of gastric mucosal damage)65] Furthermore, the degree of enzyme inhibition of many NSAIDs correlates with their capacity to erode gastric mucosa)66] The effects on the gastric mucosa result from both a systemic and a direct local irritant action of NSAIDs.[67,68] The use of rectal and parenteral routes of administration, and preparations de­signed for intestinal rather than gastric release may reduce the extent of any local effect)67] Although gastric adverse effects ofNSAIDs correlate closely with the levels of circulating metabolites, rectal

© Adis International Limited. All rights reserved.

Gastrointestinal reactions Others

67.0 20.9

58.7 9.4

32.3 11.4

32.8 8.4

20.9 18.5

27.4 8.4

na na

33.2 5.3

6.6 6.6

administration of NSAIDs can reduce gastric intol­erance by 20 to 30%.

Concern about the risk of serious NSAID­induced gastrointestinal toxicity has led to the em­pirical use of various prophylactic therapies. Syn­thetic prostaglandins may be useful in the treatment and prevention of NSAID-induced gas­tropathy.[69] Misoprostol, a synthetic PGEI ana­logue, inhibits gastric acid secretion by binding to an E-type prostaglandin receptor on the surface of gastric parietal cells)70] There is endoscopic evi­dence that misoprostol can prevent NSAID-in­duced gastric and duodenal ulcers associated with long term treatment, whereas histamine H2 recep­tor antagonists only protect against duodenal ul­cers.[69,71,72]

In a controlled trial comparing omeprazole with ranitidine in the treatment of patients with gastric ulcer, Walan et al)73] included a subgroup who continued to receive NSAIDs during anti-ulcer therapy. Although the numbers in each subgroup were small, results with omeprazole were better than those with ranitidine. In addition, continued use of NSAIDs did not impair ulcer healing with omeprazole)73] At present, there are no data to jus­tify the use of sucralfate in NSAID-related gastric ulceration)68]

Although gastrointestinal haemorrhage is gen­erally rare after short term administration, the risk of bleeding in surgical patients following brief short term exposure to NSAIDs is unclear. Dyspep-

Drugs 49 (1) 1995

Page 12: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

62

sia and ulceration may occur following administra­tion of rectal and parenteral, as well as oral, formu­lations. Short term NSAID treatment in volunteers has been shown to result in endoscopically detect­able gastroduodenal mucosallesions.[74] However,

serious complications such as bleeding and per­

foration were not observed. Lanza et al.l74] found that short term administration of ibuprofen and naproxen in low doses did not induce lesions, but indomethacin and aspirin did.

There have been numerous studies of the risk of gastroduodenal complications during long term treatment with NSAIDs. Meta-analysis of the pub­lished data indicates that the odds ratio for overall risk of serious gastroduodenal complications asso­ciated with long term NSAID treatment is in­creased 3-fold compared with untreated pa­tients.l61 ] However, there does not appear to be any increase in the incidence of serious complications during the first week of treatment.

Kehlet and Dahl[61] have analysed the results of 15 studies (927 patients in all) in which periopera­tive NSAIDs were used for 2 to 7 days for analge­sia. Patients who underwent moderate or major sur­gery and did not receive NSAIDs (603 patients) acted as controls. Of the 1530 patients included in the analysis there was only 1 reported case of haematemesis. This occurred in a patient who had received perioperative rectal indomethacin, but the haematemesis was transient and required no treat­ment.[7S] None of the control patients had a serious gastrointestinal event. Kehlet and Dahl suggest that NSAID treatment of less than 1 week may lead to superficial gastrointestinal mucosal damage in volunteers, but that there does not appear to be any evidence of an increased risk of severe gastrointes­tinal complications during perioperative NSAID treatment of similar duration.

Ingham and Portenoy[76] have reviewed the re­sults of long term administration and found that there is a higher risk associated with the use of piroxicam than aspirin, ibuprofen, indomethacin or naproxen. The risk associated with piroxicam is up to 18 times that of ibuprofen, which is associated with less risk than aspirin.

© Adis International Limited. All rights reserved.

Cashman & McAnulty

Two large studies have recently reported on the odds ratio for the risk of gastrointestinal haemor­rhage associated with 7 commonly prescribed NSAIDs.l77,78] Although NSAIDs are associated with an excess risk of bleeding, ibuprofen tends to carry the lowest risk.

3.2 Adverse Renal Effects

Prostaglandins have the capacity to influence all elements of renal function, but under normal con­ditions prostaglandin synthesis plays only a minor role in the maintenance of renal function. In animal studies, NSAID-induced reduction of renal prosta­glandin levels was of little physiological conse­quenceV9] Renal effects of NSAIDs are dose and duration dependent and tend to recover on cessa­tion of treatment. The majority of clinical studies have not shown any significant effects of NSAIDs on haemodynamics in normal human kidneys.lsO] However, prostaglandin control of renal function becomes significant when there are disturbances in fluid or electrolyte balance with a compensatory increase in vasodilatory prostaglandins to support renal blood flow.

Administration of NSAIDs reduces glomerular filtration rate, can cause renal ischaemia and may lead to acute renal failure.l sO,Sl] NSAIDs, such as indomethacin, have been shown to decrease glo­merular filtration rate and effective renal plasma flow when there is disturbed renal function.[l4] In patients with mild chronic renal failure, even a brief course of ibuprofen may precipitate acute re­nal failure.l S2] Acute renal failure following ad­ministration of ketorolac in patients with known risk factors has been reported.lS3]

The stress of anaesthesia and surgery in patients without compromised renal blood flow may in­crease the risk of renal ischaemia following admin­istration of NSAIDs. Transient postoperative renal impairment after intravenous infusion of diclo­fenac has been reported following thoracotomy.lS4] Smith et al.lS5] reported, in 2 patients who had pre­viously taken NSAIDs without adverse effect, de­velopment of renal failure when they received ketorolac in association with surgery. This con-

Drugs 49 (1) 1995

Page 13: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

NSAIDs in Perisurgical Pain Management

trasts with the findings of Aitken et aU86] who sug­gest that ketorolac does not cause the same degree of renal impairment as other NSAIDs. One case of reversible nephrotic syndrome with interstitial ne­phritis has been reported in a 74-year-old woman who had received topical piroxicamJ81] It is of note that topical application of piroxicam over 14 days in volunteers resulted in blood concentrations 5% of that achieved by the oral routeJ87] Elderly patients may achieve higher concentrations be­cause of reduced plasma clearance, thin skin and frequent application of the drug over large areasJ81]

3.3 Aspirin and Bronchospasm

Aspirin and other NSAIDs may precipitate acute bronchospasm in asthmatic patientsJ16,88] The prevalence of sensitivity to NSAIDs in adults with asthma is of the order of 5 to 10% and is com­mon in those with atopic allergy. The mechanism of the reaction is unclear. Once thought to be aller­gic in nature,[16] aspirin-related bronchospasm is now considered to have a pharmacological ba­sisJ88] Repeated administration of aspirin induces tolerance and may even be associated with devel­opment of a bronchodilator response.l89]

4. Choosing an NSAID for Use in the Peri operative Period

4.1 Rational Prescribing of NSAIDs

Rational prescribing of NSAIDs in the peri­operative period is particularly important. The ba­sic tenets for prescribing the drugs for long term use outlined by Orme[90] and by Ingham and Portenoy[76] can be adapted for short term postop­erative use.

4. 1. 1 Drug Selection • Choose from a limited selection of NSAIDs

spanning the different chemical groups. • Prefer established drugs with long clinical

experience and good safety profile (e.g. ibu­profen).

© Adis International Limited. All rights reserved.

63

• Record concurrent drug therapy and be aware of possible pharmacokinetic and pharmacody­namic interactions.

• Avoid NSAIDs in patients with known contra­indications to their use.

• Use only one NSAID at a time, and ensure ad­equate dosage.

4.1.2 Route of Administration

• Be aware of available preparations. Many of the newer NSAIDs have the potential advantage of being available in a range of formulations in­cluding oral, rectal, parenteral and topical.

• Use the least invasive route possible.

4.1.3 Dosage

• Adapt dosages to suit patients' requirements, particularly with respect to duration of action.

• Increase dose until adequate analgesia occurs or maximum recommended dose is reached.

• Review therapy frequently and change to an al­ternative NSAID, possibly from another class, if there is poor response to treatment.

4.1.4 Toxicity • Observe for potential toxicity (gastrointestinal,

renal, haematological, etc.). • Increase frequency of monitoring for at-risk pa­

tients (the elderly and those with concurrent dis­ease).

• Consider prophylaxis against adverse gastroin­testinal events in these patients.

4.2 Pharmacokinetic-Pharmacodynamic Relationships

In general, the dosage of a drug should be tai­lored to the individual patient's response, based on clinical assessment. However, the concentration of almost any drug may fluctuate markedly during each dose interval as a result of continuing absorp­tion, distribution and elimination of the drug. An understanding of the pharmacokinetics of drugs is therefore necessary in order to achieve steady-state plasma concentrations.

For single doses of aspirin, analgesic effect has been shown to increase with increasing doses up to

Drugs 49 (l) 1995

Page 14: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

64

1200mg.l91 ,92]It may be more appropriate to con­sider this dose-response relationship as a relation­ship between drug concentration and response,[93] However, in the case of aspirin and other NSAIDs there is a delay in the time to maximum pharmaco­logical effect relative to the time to peak plasma concentration of the drug. It has been shown using radio-labelled ketorolac that the time to peak plasma concentration after intravenous administra­tion is achieved in less than 5 minutes and more slowly after intramuscular (45 minutes) and oral (30 minutes) administration.l94] However, the time to peak plasma concentration correlates poorly with onset of analgesia which does not reach a peak until 30 minutes after intravenous administra­tion.[94] The time to peak plasma concentration af­ter oral administration in humans has been det~r­mined for a number of NSAIDs (table IV).

Levy[93] argues that there is a concentration­effect relationship for aspirin which is complex and time dependent. He suggests a number of reasons for the observed disparity between clinical effect and plasma concentration:

• the site of action of the drug may have the phar­macokinetic characteristics of a compartment distinct from the central compartment;

• the drug may act indirectly; or • the effects may be mediated by an active meta­

bolite. It is possible that aspirin is an example of this

last mechanism since there is still discussion as to whether aspirin is the active compound or merely a pro-drug for salicylate.

In a review of the temporal pattern of pain relief produced by analgesic drugs, Levy[96] concluded that the rate of decline in pharmacological effect was unrelated to the specific analgesic agent. He proposed that the larger the dose (and hence pre­sumably the higher the peak concentration of the drug in the biophase), the greater the analgesic ef­fect would be. Moreover, the greater the maximum analgesic effect, the longer the duration of effect and the greater the total area under the plasma con­centration versus time curve. Levy argued that the logical extension of this reasoning is that sustained

© Adis International Lirnited, All rights reserved,

Cashman & McAnulty

Table IV. Pharmacokinetics of various nonsteroidal anti-inflamma­tory drugs (NSAIDs)[95J

Compound Time to peak plasma Half-life concentration (h) (h)

Flurbiprofen 1.5-3,0 3-6

Ibuprofen 0,5-1,5 2-5

Indomethacin 1.0-2,0 4-8

Ketoprofen 0,3 2-4

Naproxen 1,0-2,0 10-20

Phenylbutazone 2,0 40-80

Piroxicam 2,0 40-80

release preparations are unlikely to provide pro­longed analgesia. Rapidly absorbed preparations should produce not only more immediate but also more pronounced analgesia. However, Cass and Frederikl97] found identical durations of analgesia produced by both regular and sustained release as­pirin.

Although the pharmacokinetics of repeat doses may have been elucidated for many analgesics, lit­tle is known about the pharmacodynamics. Follow­ing oral administration of aspirin, salicylate con­centrations are up to 50 times higher than aspirin levels and, unlike aspirin, are detectable for several hours.l9] Peak plasma concentrations of salicylate increase after each dose until a steady-state is reached. By contrast, aspirin concentrations de­cline rapidly to zero during each dose interval be­cause of the very short biological half-life of the drug. Thus, salicylic acid but not aspirin accumu­lates in the plasma with repeat drug administration. If the analgesic effect of aspirin were due to sali­cylic acid, a gradually increasing analgesic effect during repeated aspirin administration would re­sult, whereas there would be no such increase in analgesia if the effect were produced primarily by aspirin itself.

Most observations of repeated administration of aspirin are consistent with the view that the anal­gesic effect is produced mainly by the drug itself rather than its hydrolysis product, salicylic acid. However, salicylic acid seems to be primarily re­sponsible for the anti-inflammatory and toxic ef­fects of aspirin. Salicylate exhibits capacity-limited

Drugs 49 (l) 1995

Page 15: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

NSAIDs in Perisurgical Pain Management

elimination; hence plasma salicylate concentra­tions decline relatively more slowly with an in­creasing daily dose.

5. Dosage Regimens

5.1 Oral and Rectal

Drugs given at fixed dose intervals, rather than in response to patient demand, may be associated with better analgesia. Regular oral ketorolac 5mg and lOmg 4 times daily has been shown to be as effective as regular oral diflunisal 500mg twice daily for the treatment of postmeniscectomy pain. [98] Lovett et al. [99] demonstrated that despite similar pain scores, postoperative papaveretum re­quirements were reduced 25 to 30% by concurrent regular mandatory doses with intramuscular diclofenac 75mg pre- and postoperatively on the day of surgery, followed thereafter by oral diclofenac 50mg 3 times daily for the next 3 days. This reduction in opioid demand was comparable to that obtained by Owen et al. [1 00] using oral ibuprofen pre- and postoperatively in patients un­dergoing gynaecological surgery and similar to that demonstrated in a study in abdominal surgery using 2 postoperative doses of dic1ofenacJIOI] However, this degree of morphine-sparing is less impressive than the 50% reduction reported by Derbyshire and Richardsonll02] with only a single (50mg oral) pre- and (lOOmg rectal) postoperative dose of dic1ofenac.

Rectal administration of indomethacin in a rel­atively high dose (lOOmg 8-hourly) commenced either preoperatively[I03] or postoperatively[75] re­duced opioid requirements and improved pain re­lief after major abdominal surgery. Rectal indo­methacin (200mg postoperatively followed by 100mg twice daily) reduced pain scores and opioid requirements compared with placebo following thoracic surgery)104] Preoperative administration of intravenous indomethacin followed by suppos­itories 3 times daily postoperatively reduced op­ioid requirement by 29% compared with placebo after gynaecological surgery) 105] In I of these studies there was a significantly higher incidence

© Adis International Limited. All rights reserved.

65

of haemorrhagic complications with the NSAID than with placebo, but gastrointestinal adverse ef­fects were similar between groups.[75]

5.2 Parenteral

5.2. 1 Intramuscular Parenteral administration of NSAIDs results in

the rapid attainment of a therapeutic plasma con­centrations. Postoperative intramuscular diclo­fenac has been shown to be effective in relieving pain following orthopaedic, gynaecological and major abdominal surgery)106-108] Lindgren and Djupso[106] demonstrated that intramuscular di­clofenac 75mg was superior to pethidine (meperid­ine) 50mg and placebo in relieving postoperative orthopaedic pain. As a result of their findings Lindgren and Djupso suggested that NSAIDs, ad­ministered routinely, should be preferred to op­ioids in the early postoperative period.

Bossi et al.[107] showed that in the management of post cholecystectomy pain, the analgesic effect of a single injection of intramuscular diclofenac 75mg was almost identical to that of intramuscular pentazocine 50mg with regard to potency, onset time and duration of effect. Carlos[108] found that, after abdominal surgery, intramuscular dic10fenac 75mg provided equivalent but longer lasting anal­gesia to intramuscular pethidine 1 mg/kg and intra­muscular nalbuphine 0.14 mg/kg.

Burns et al. [1 09] showed that a continuous intra­muscular infusion of ketorolac is superior to inter­mittent intramuscular injections. Continuous intra­muscular infusions of ketorolac were superior to intermittent intramuscular injections and resulted in significant morphine sparing effect[ 109, 110]

5.2.2 Intravenous Intravenous infusions are preferable to bolus

doses because the duration of analgesia following a single intravenous bolus is short. The administra­tion of an analgesic as an infusion results in more stable blood concentrations and may be expected to provide superior analgesia. Harris and Riegel­man[1ll] have suggested that a constant rate infu­sion of aspirin is necessary to achieve a steady­state where the tissues come into distribution

Drugs 49 (1) 1995

Page 16: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

66

equilibrium with the plasma. Combining a loading dose with a zero-order (fixed rate) infusion is one way of accelerating the attainment of steady-state drug concentrations.

Loo and Riegelman[l12] and Rowland and Riegelman[113] used healthy volunteers to investi­gate absorption, distribution, metabolism and ex­cretion kinetics of intravenous aspirin. The methylglucamine salt was administered both as a rapid intravenous injection and as an intravenous infusion. In both studies aspirin was given intrave­nously at known logarithmic infusion rates (in which the rate of infusion was reduced at fixed in­tervals with the duration of the intervals determin­ing the half-life) and linear infusion rates, so as to mimic first- and zero-order absorption conditions. These studies showed that the kinetics of aspirin could be described using a 2-compartment model, and that it should be possible to predict plasma lev­els using a mathematical model. Other investiga­tors[114-116] have investigated the effectiveness of continuous intravenous infusions of aspirin. It was found that intravenous aspirin provided pain relief equivalent to that of pethidine and morphine in a variety of surgical conditions.

Postoperative intravenous indomethacin 25mg followed by a continuous intravenous infusion was more effective in reducing pain scores and increas­ing pain free periods than oxycodone administered according to a similar regimen. [117] Dic10fenac has been successfully administered by slow intrave­nous injection[llS,119] and as an intravenous infu­sion,D20] without apparent adverse effect. Camp­bell et al.[lIS] administered diclofenac 75mg, diluted to 18ml, by slow intravenous injection after dental surgery to good effect.

Hovorka et al,l119] found that intravenous diclofenac 100mg successfully reduced the need for postoperative analgesia in diagnostic laparos­copy but not in interventional laparoscopy. Laitinen et al.[l20] found that on completion of sur­gery an infusion of diclofenac (75mg in 100mi iso­tonic saline over 30 minutes) provided equivalent analgesia to an intravenous infusion of oxycodone and was superior to an intravenous infusion of in-

© Adis International Limited. All rights reserved.

Cashman & McAnulty

domethacin after orthopaedic surgery. A 2-day in­travenous infusion of diclofenac provided effective analgesia after thoracotomy and was associated with a significant reduction in morphine consump­tion compared with a control group,lS4]

Although many NSAIDs are presented in a va­riety of formulations, no study has specifically compared the analgesic efficacy of alternative routes of administration of the same drug. On the­oretical grounds, continuous intravenous infusions of NSAIDs should provide analgesia that is supe­rior to that of intermittent bolus injections. Several studies have shown that intravenous infusions re­sult in effective pain relief,lS4,1l7-120] In addition, the results of Burns et al,l109] and Gillies et al.[lIO] indicate that constant plasma drug levels are achieved using intramuscular infusions, and are as­sociated with effective analgesia.

6. Preoperative Administration

There seems to be little benefit from administra­tion of an NSAID before major surgery. Murphy and Medley[121] compared pain relief provided by indomethacin given pre- or postoperatively to pa­tients undergoing thoracotomy and found no differ­ence between the 2 groups. Even when NSAIDs were given preoperatively as part of a multimodal pre-emptive analgesic regimen to thoracotomy pa­tients, no significant improvement was found in overall postoperative pain scores,l122] Further­more, although the preoperative administration of NSAIDs may appear logical, there is a risk of in­creased bleeding particularly in patients in whom extensive surgical resection is anticipated.[75]

7. Conclusions

There are a number of general conclusions which can be drawn with regard to prescription of NSAIDs for postoperative pain relief. These relate to timing of dose, the regimen and the route of ad­ministration.

Levy's work with aspirin[96] suggests a complex relationship between plasma levels and analgesic effect. However, later studies of steady-state infu­sions with ketorolac indicate that superior pain re-

Drugs 49 (1) 1995

Page 17: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

NSAIDs in Peri surgical Pain Management

lief is associated with maintenance of constant rather than fluctuating plasma NSAID concentra­tions.l 109,1l0] Thus, analgesia is likely to be most effective when infusions (either intravenous or in­tramuscular) or mandatory oral dosage regimens are used.

In comparison with the oral route, the paren­teral, rectal and topical routes of administration of NSAIDs are associated with a reduced incidence of adverse effects including gastric effects. Gastric adverse effects correlate closely with the concen­trations of circulating metabolites. Nevertheless, rectal administration ofNSAIDs can reduce gastric intolerance by 20 to 30%. Furthermore, sustained release suppository formulations of NSAIDs can be effective alternatives to parenteral administra­tion. The choice of which formulation of analgesic to use for postoperative pain relief is dictated by a number of factors. In general, the oral route cannot be used in the immediate postoperative period al­though some drugs, notably the oxicams, are ab­sorbed across gastric mucosa independent of the factors that alter gastric emptying.

Future improvements in the therapeutic index of NSAIDs may derive from the development of drugs which more selectively inhibit the inducible form of CYclo-OXygenase (COX-2). Drugs avail­able now, such as ibuprofen, which inhibit COX-2 relatively more than COX-I, can be expected to have a better safety profile than drugs which inhibit COX-I preferentially.

References I. Vane JR, Botting RM. The mode of action of anti-inflammatory

drugs. Postgrad Med J 1990; 66 Suppl. 4: S2-S 17 2. Ferreira SH, Moncada S, Vane JR. Indomethacin and aspirin

abolish prostaglandin release from spleen. Nature 1971; 231: 237-9

3. Vane JR. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nature (New Bioi) 1971; 231: 232-5

4. Ferreira SH. Peripheral analgesia: mechanism of the analgesic action of aspirin like drugs and opiate-antagonists. Br J Clin Pharmacol 1980; 10: 237-245S

5. Von Euler US. On the specific vasodilating and plain muscle stimulating substance from accessory genital glands in man and certain animals (prostaglandin and vesiglandin). J Phys­io11937; 88: 213-34

6. Hemler M, Lands WEM, Smith WL. Purification of the cyclo­oxygenase that forms prostaglandins. Demonstration of two

© Adis International Limited. All rights reserved.

67

forms of iron in the holoenzyme. J Bioi Chern 1976; 251: 2629-36

7. Ferreira SH. Prostaglandins, aspirin-like drugs and analgesia. Nature 1972; 240: 200-3

8. Higgs GA, Higgs EA, Salmon JA. Prostacyciin in inflamma­tion. In: Vane JAR, Bergstrom S, editors. Prostacyclin. New York: Raven Press, 1979: 187-92

9. Higgs GA, Salmon JA, Henderson B, et al. Pharmacokinetics of aspirin and salicylate in relation to inhibition of arachidon­ate cyclooxygenase and antiinflammatory activity. Proc Natl Acad Sci USA 1987; 84: 1417-20

10. Higgs GA. Arachidonic acid metabolism, pain and hyperalge­sia: the mode of action of non-steroid mild analgesics. Br J Pharmacol 1980; 10: 233-235S

II. Higgs GA, Moncada S, Vane JR. Prostacyclin as a potent dilator of arterioles in the hamster cheek pouch. J Physiol (Lond) 1978; 275: 30-1

12. Higgs EA, Moncada S, Vane JR. Inflammatory effects of pro­stacyclin (PGI2) and 6-oxo-PGFla in the rat paw. Prostaglan­dins 1978; 16: 153-62

13. Dray A, Bevan S. Inflammation and hyperalgesia: the team effort. Trends Pharm Sci 1993; 14: 287-90

14. Vane JR, Flower RJ, Botting RM. The mechanism of action of aspirin. In: Vane JR, Botting RM, editors. Aspirin and other salicylates. London: Chapman & Hall Medical, 1992

IS. Flower R, Gryglewski R, Herbaczynska-Cedro K, et al. Effects of anti-inflammatory drugs on prostaglandin biosynthesis. Nature (New Bioi) 1972; 238: 104-6

16. Vane JR. The mode of action of aspirin and similar compounds. J Allergy Clin Immunol1976; 58: 691-712

17. Flower RJ. Drugs which inhibit prostaglandin biosynthesis. Pharmacol Rev 1974; 26: 33-67

18. Lands WEM. Actions of anti-inflammatory drugs. Trends Phar­macol Sci 1981; 2: 78-80

19. Rome LH, Lands WEM. Structural requirements for time-de­pendent inhibition of prostaglandin biosynthesis by anti-in­flammatory drugs. Proc Nat Acad Sci USA 1975; 72: 4863-4865

20. Roth GJ, Majerus pw. The mechanism of the effect of aspirin on human platelets - I: acetylation of a particulate fraction protein. J Clin Invest 1975; 56: 624-32

21. Roth GJ, Stanford N, Majerus pw. Acetylation of prostaglandin synthase by aspirin. Proc Nat! Acad Sci USA 1975; 72: 3073-6

22. Kulmacz RJ, Lands WEM. Stoichometry and kinetics of the interaction of prostaglandin H synthase with anti-inflamma­tory agents. J Bioi Chern 1985; 260: 12572-8

23. Appleton RA, Brown K. Conformational requirements at the prostaglandin cyclo-oxygenase receptor site: a template for designing non-steroidal anti-inflammatory drugs. Prostaglan­dins 1979; 18: 29-34

24. Hemler ME, Lands WEM. Evidence for peroxide-initiated free radical mechanism of prostaglandin biosynthesis. J Bioi Chern 1980; 255: 6253-61

25. Lands WEM, Cook HW, Rome LH. Prostaglandin biosynthesis: consequences of oxygenase mechanisms on in vitro assays of drug effectiveness. Adv Prostaglandin Thromboxane Leukot Res 1976; 1: 7-17

26. Dreser H. Pharmacologisches uber Aspirin (Acetylsalicyl-sa­ure). Pfluger's Arch Gesamte Physiol Menschen Tiere 1899; 76: 306-18

27. Wiler JC, De Brouckner T, Bussel B, et al. Central analgesic effect of ketoprofen in humans - electrophysiological evi­dence for a supraspinal mechanism in a double-blind and cross-over study. Pain 1989; 38: 1-7

Drugs49(lJ 1995

Page 18: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

68

28. Ferreira SH, Nakamura M, de Abreu Castro MS. The hyperal­gesic effects of prostacyclin and prostaglandin E2. Prosta­glandins 1978; 16: 31-7

29. Malmbergh AB,Yaksh TL. Hyperalgesia mediated by spinal glutamate or substance receptor blocked by spinal cyclo-ox­ygenase inhibition. Science 1992; 257: 1276-9

30. Carlsson K-H, Mangel W, Jurna I. Depression by morphine and the non-opioid analgesic agents, metamizol (dipyrone), lysine acetylsalicylate, and paracetamol, of activity in rat thalamus neurones by electrical stimulation of nociceptive afferents. Pain 1988; 32: 313-26

31. Jurna I, Brune K. Central effect of the non-steroid anti-inflam­matory agents, indometacin, ibuprofen, and diclofenac, deter­mined in C fibre-evoked activity in single neurones of the rat thalamus. Pain 1990; 41: 71-80

32. Ferreira SH, Lorenzetti BB, Correa FEM. Central and periph­eral antialgesic action of aspirin-like drugs. Eur J Pharmacol 1978; 53: 39-48

33. Hunskaar S. Similar effects of acetylsalicylic acid and morphine on immediate responses to acute noxious stimulation. Phar­macol Toxicol1987; 60: 167-70

34. McCormack K, Brune K. Dissociation between the antinocicep­tive and anti-inflammatory effects of the non-steroidal anti­inflammatory drugs. Drugs 1991; 41: 533-47

35. Attal N, Kayser V, Eschalier A, et al. Behavioural and electro­physiological evidence for an analgesic effect of a non-steroi­dal anti-inflammatory agent, diclofenac. Pain 1988; 35: 341-8

36. Devoghel J-e. Small intrathecal doses oflysine-acetylsalicylate relieve intractable pain in man. J Int Med Res 1983; II: 90-1

37. Day RO, Brooks PM. Variations in response to non-steroidal anti­inflammatory drugs. Br J Clin Pharmacol 1987; 23: 655-8

38. Bellamy N. Variance in NSAID studies: contribution of patient variance. Agents Actions Supp11985; 17: 21-8

39. Day RO. Variability in response to NSAID. Agents Actions Suppl 1985; 17: 15-9

40. Scott DL, Roden S, Marshall T, et al. Variations in response to non-steroidal anti-inflammatory drugs. Br J Clin Pharmacol 1982; 14: 691-4

41. Huskisson EC, Woolf DL, Balme HW, Scott J, Franklyn S. Four new anti-inflammatory drugs: responses and variations. BMJ 1976; 48: 1048-9

42. Gall EP, Caperton JF, McComb JE, et al. Clinical comparison of ibuprofen, fenoprofen calcium, naproxen and tolmetin so­dium in rheumatoid arthritis. J Rheumatol 1982; 9: 402-7

43. Capell HA, Konetschnik B, Glass Re. Anti-inflammatory anal­gesic drug responders and non-responders: a clinico-pharma­cological study offlurbiprofen. Br J Clin PharmacoI1977; 15: 311-6

44. Baber N, Halliday LDC, Van Den Heuvel WJA, et al. Indometh­acin in rheumatoid arthritis: clinical effects, pharmacokinet­ics, and platelet studies in responders and non-responders. Ann Rheum 1979; 13: 128-37

45. Orme ML'E, Baber N, Keenan L, et al. Pharmacokinetics and biochemical effects in responders and non-responders to non­steroidal anti-inflammatory drugs. Scand J Rheumatol 1981; (Suppl. 39): 19-27

46. Grennan DM, Aarons L, Siddiqui M, et al. Dose-response study with ibuprofen in rheumatoid arthritis: clinical and pharma­cokinetic findings. Br J Clin Pharmacol 1983; 18: 311-6

47. Preston SJ, Arnold MH, Beller EM, etal. Variability in response to non-steroidal anti-inflammatory analgesics: evidence from controlled clinical therapeutic trial of flurbiprofen in rheuma­toid arthritis. Br J Clin Pharmacol 1988; 26: 759-64

© Adis International Limited. All rights reserved.

Cashman & McAnulty

48. Lee EJD, Williams K, Day R, et al. Stereoselective disposition of ibuprofen enantiomers in man. Br J Clin Pharmacol 1985; 19: 669-74

49. Hayball PJ, Tamblyn JG, Holden Y, et al. Stereoselective anal­ysis ofketorolac in human plasma by high-performance liquid chromotography. Chirality 1993; 5: 31-5

50. Williams K, Day R, Knihinicki R, et al. The stereoselective uptake of ibuprofen enantiomers into adipose tissue. Biochem Pharmacol 1986; 35: 3403-5

51. Oates JA, Wood AJJ. Nonsteroidal antiinflammatory drugs -differences and similarities. N Eng J Med 1991; 24: 1716-25

52. Hutt AJ, Caldwell J. The metabolic chiral inversion of 2-aryl propionic acids - a novel route with pharmacological con­sequences. J Pharm Pharmacol 1983; 35: 693-704

53. Day RO, Williams KM, Graham GG, et al. Stereoselective dis­position of ibuprofen enantiomers in synovial fluid. Clin Pharmacol Ther 1988; 43: 480-7

54. Knihinicki RD, Williams KM, Day RO. Chiral inversion of 2-arylpropionic acid non-steroidal anti-inflammatory drugs - I: in vitro studies of ibuprofen and flurbiprofen. Biochem Phar­macol 1989; 38: 4389-95

55. Ahn AHY, Amidon GL, Smith DE. Stereoselective systemic disposition of ibuprofen enantiomers in the dog. Pharm Res 1991; 8: 1186-90

56. Brocks DR, Liang WT, Jamali F. Influence of the route of ad­ministration on the pharmacokinetics of pirprofen enantio­mers in the rat. Chirality 1993; 5: 61-4

57. Olliary J, Tod M, Nicolas P, et al. Pharmacokinetics of ibuprofen enantiomers after single and repeated doses in man. Biopharm Drug Dispos 1992; 13: 337-44

58. Knihinicki RD, Day RO, Williams KM. Chiral inversion of 2-arylpropionic acid non-steroidal anti-inflammatory drugs - II: racemization and hydrolysis of (R)- and (S)-ibuprofen-CoA thioesters. Biochem Pharmacol 1991; 42: 1905-11

59. Stock KP, Geisslinger G, Loew D, et al. S-ibuprofen versus ibuprofen-racemate: a randomized double-blind study in pa­tients with rheumatoid arthritis. Rheumatol Int 1991; 11: 199-202

60. Geisslinger G, Stock KP, Loew D, et a1. Variability in the stereoselective disposition of ibuprofen in patients with rheu­matoid arthritis. Br J Clin Pharmacol 1993; 35: 603-7

61. Kehlet H, Dahl JB. Are perioperative nonsteroidal anti-inflam­matory drugs ulcerogenic in the short term? Drugs 1992; 44 Supp1. 5: 38-41

62. Kenny GNe. Potential renal, haematological and allergic ad­verse effects associated with nonsteroidal anti-inflammatory drugs. Drugs 1992; 44 Suppl. 5: 31-7

63. Committee on Safety of Medicines and the Medicines Control Agency. Relative Safety of oral non-aspirin NSAIDs. Current Problems Pharmacovig 1994; 20 Aug: 9-11

64. Jahn U. Survey of toxicological investigations of azapropozone. In: Rainsford KD, editor. Azapropazone. Dordrecht: Kluwer Academic Publishers, 1989: 21-9

65. Which NSAID? [editorial]. Drug Ther Bull 1978; 25: 81-4 66. Carson JL, Strom BL, Soper KA, et al. The association of non­

steroidal antiinflammatory drugs with upper gastrointestinal tract bleeding. Arch Intern Med 1987; 147: 85-8

67. Aspirin and the stomach [editorial]. BMJ 1981; 282: 91-2 68. Soll AH, Weinstein WM, Kursta J, et a1. Nonsteroidal anti-in­

flammatory drugs and peptic ulcer disease. Ann Intern Med 1991; 114: 307-19

69. Fenn GC, Robinson Ge. Misoprostol - a logical therapeutic approach to gastroduodenal mucosal injury induced by non-

Drugs 49 (1) 1995

Page 19: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

NSAIDs in Perisurgical Pain Management

steroidal anti-inflammatory drugs? J Clin Pharm Ther 1991; 16: 385-409

70. Tsai BS, Kessler LK, Schoenhard G, et al. Demonstration of specific E-type prostaglandin receptors using enriched prep­arations of canine parietal cells and [3H] misoprostol free acid. AmJ Med 1987; 83 Suppl. IA: 9-14

71. Roth S, Agrawal N, Mahowald M, et al. Misoprostal heals gas­troduodenal injury in patients with rheumatoid arthritis re­ceiving aspirin. Arch Intern Med 1989; 149: 775-9

72. Agrawal NM, Saggioro A. Treatment and prevention ofNSAID induced gastroduodenal mucosal damage. J Rheumatol 1991; 18 (Suppl.): 15-8

73. Walan A, Bader J-P, Classen M, et al. Effect of omeprazole and ranitidine on ulcer healing and relapse rates in patients with benign gastric ulcer. N Engl J Med 1989; 320: 69-75

74. Lanza FL, Royer Jr GL, Nelson RS, et al. The effects of ibuprofen, indomethacin, aspirin, naproxen and placebo on the gastric mucosa of normal volunteers. Digest Dis Sci 1979; 24: 823-8

75. Reasbeck PG, Rice ML, Reasbeck Jc. Double-blind controlled trial of indomethacin as an adjunct to narcotic analgesia after major abdominal surgery. Lancet 1982; i: 115-8

76. Ingham JM, Portenoy RK. Drugs in the treatment of pain: NSAIDS and opioids. CUff Opin Anaesthesiol 1993; 6: 838-44

77. Garcia Rodriguez LA, Jick H. Risk of upper gastrointestinal bleeding and perforation associated with individual non-ste­roidal anti-inflammatory drugs. Lancet 1994; 343: 769-72

78. Langman MJ, Weil J, Wainwright P, et al. Risks of bleeding peptic ulcer associated with individual non-steroidal anti-in­flammatory drugs. Lancet 1994; 343: 1075-8

79. Gray DA, Langrieger N, Reschmaier J, et al. The effect of ace­tylsalicylic acid on renal function in the Pekin duck. Br J Pharmacol 1984; 82: 329-38

80. Harris K. The role of prostaglandins in the control of renal function [editorial]. Br J Anaesth 1992; 69: 233-5

81. 0' Callaghan CA, Andrews PA, Ogg CS. Renal disease and use of topical non-steroidal anti-inflammatory drugs. BMJ 1994; 308: 110-1

82. Whelton A, Stout RL, Spilman PS, et al. Renal effects of ibuprofen, piroxicam and sulindac in patients with asymp­tomatic renal failure. Ann Intern Med 1990; 112: 568-76

83. Pearce CJ, Gonzales FM, Wallin JD. Renal failure and hyper­kalaemia associated with ketorolac tromethamine. Arch In­tern Med 1993; 153: 1000-2

84. Perttunen K, Kalso E, Heinonen J, et al. LV. diclofenac in post­thoracotomy pain. Br J Anaesth 1992; 68: 474-80

85. Smith K, Halliwell RMT, Lawrence S, et al. Acute renal failure associated with intramuscular ketorolac. Anaesth Intens Care 1993; 21: 700-3

86. Aitken HA, Burns JW, McArdle CS, et al. Effects of ketorolac trometamol on renal function. Br J Anaesth 1992; 68: 481-5

87. Doogan DP. Topical non-steroidal anti-inflammatory drugs. Lancet 1989; ii: 1270-1

88. Power L Aspirin-induced asthma. Br J Anaesth 1993; 71: 619-21

89. Asad SI, Kemeny OM, Youlten L, et al. Effect of aspirin in 'aspirin sensitive' patients. BMJ 1984; 288: 745-8

90. Orme M. Profile of non-steroidal anti-inflammatory drugs. Pre­scribers J 1990; 30: 95-100

91. Parkhouse J, Rees-Lewis M, Skolinik M, et al. The clinical dose response to aspirin. Br J Anaesth 1968; 40: 433-41

92. Seymour RA, Rawlins MD. Efficacy and pharmacokinetics of aspirin in post-operative dental pain. Br J Clin Pharmacol 1982; 13: 807-10

© Adis International Limited. All rights reserved.

69

93. Levy G. Clinical pharmacokinetics of salicylates: a reassess­ment. Br J Clin Pharmacol 1980; 10: 285-290S

94. Greenwald RA. Ketorolac: an innovative non-steroidal analge­sic. Drugs Today 1992; 28: 41-6

95. McCormack K. Mathematical model for assessing risk of gas­trointestinal reactions to NSAIDs. In: Rainsford KD, editor. Azopropizone. Dordrecht: Kluwer Academic Publishers, 1989: 81-93

96. Levy G. Kinetics of pharmacologic effects. Clin Pharmacol Ther 1967; 7: 362-72

97. Cass LJ, Frederik WS. A clinical evaluation of a sustained-re­lease aspirin. Curr Ther Res 1965; 7: 683-92

98. Honig WJ, Van Ochten J. A multiple-dose comparison of ketorolac tromethamine with diflunisal and placebo in postmeniscectomy pain. J Clin Pharmacol 1986; 26: 700-5

99. Lovett PE, Stanton SL, Hennessey 0, et al. Pain relief after major gynaecological surgery. Br J Nurs 1994; 3: 159-62

100. Owen H, Glavin RJ, Shaw NA. Ibuprofen in the management of postoperative pain. Br J Anaesth 1986; 58: 1371-5

101. Hodsman NBA, Burns J, Blyth A, et al. The morphine sparing effects of diclofenac sodium following abdominal surgery. Anaesthesia 1987; 42: 1005-8

102. Derbyshire DR, Richardson J. Voltarol (diclofenac) as a peri opera­tive analgesic supplement. Br J Anaesth 1987; 59: 1327-8

103. Thind P, Sigsgaard T. The analgesic effect of indomethacin in the early postoperative following abdominal surgery. Acta Chirug Scand 1988; 154: 9-12

104. Pavy T, Medley C, Murphy OF. Effect of indomethacin on pain relief after thoracotomy. Br J Anaesth 1990; 65: 624-7

105. Engel C, Lund B, Kristensen AA, et al. Indomethacin as an analgesic after hysterectomy. Acta Anaesthesiol Scand 1989; 33: 498-501

106. Lindgren U, Djupso H. Diclofenac for pain after hip surgery. Acta Orthop Scand 1985; 55: 28-31

107. Bossi L, Galante G, Conoscente F, et al. Tratamento del dol ore postoperatorio. Confronto in doppio cieco fra diclofenac e pentazocin. Minerva Anest 1984; 50: 373-8

108. Carlos DE. A comparative study of the efficacy of diclofenac sodium, meperidine HCI and nalbuphine HCI in postopera­tive analgesia. In: Birdwood GFB, Gantmacher JV, editors. Further experience with voltaren. Bern: Hans Huber, 1984: 73-8

109. Burns JW, Aitken HA, Bullingham RES, et al. Double-blind comparison of the morphine sparing effect of continuous and intermittent i.m. administration of ketorolac. Br J Anaesth 1991; 67: 235-8

110. Gillies GW, Kenny GN, Bullingham RE, et al. The morphine sparing effect of ketorolac tromethamine: a study of a new non-steroidal anti-inflammatory agent after abdominal sur­gery. Anaesthesia 1987; 42: 727-31

Ill. Harris PA, Riegelman. Influence of the route of administration on the area under the plasma concentration-time curve. J Pharm Sci 1969; 58: 71-5

112. Loo JCK, Riegelman S. New method for calculating the intrin­sic absorption rate of drugs. J Pharm Sci 1968; 57: 918-28

113. Rowland M, Riegelman S. Pharmacokinetics of acetylsalicylic acid and salicylic acid after intravenous administration in man. J Pharmaceut Sci 1968; 57: 1313-9

114. Miralles FS, Carceles MD, Nicol JA, et al. Administration of lysine acetylsalicylate and pethidine in acute postoperative pain. Rev Esp Anestiol Reanim 1992; 39: 149-54

115. Cashman IN, Jones RM, Foster JMG, et al. Comparison of infusions of morphine and lysine acetyl salicylate for the re­lief of pain after surgery. Br J Anaesth 1984; 57: 255-8

Drugs 49 (1) 1995

Page 20: Nonsteroidal Anti-Inflammatory Drugs in Perisurgical Pain Management

70

116. Jones RM, Cashman IN, Foster JMG, et al. Comparisons of infusions of morphine and lysine acetyl salicylate for the relief of pain following thoracic surgery. Br J Anaesth 1984; 57: 259-63

117. Mattila MK, Ahlstrom-Bengs E, Pekkola P. Intravenous indo­methacin in prevention of post-operative pain. BMJ 1993; 287: 1026

118. Campbell WI, Kendrick R, Patterson C. Intravenous diclofenac sodium. Anaesthesia 1990; 45: 763-6

119. Hovorka J, Kallela H, Kortilla K. Effect of intravenous diclofenac on pain and recovery profile after day-case lapa­roscopy. Eur J Anaesthiol 1993; 10: 105-8

120. Laitinan J, Nuutinen L, Kiiskial E-L, et al. Comparison of in­travenous diclofenac, indomethecin and oxycodone as post-

Errata

Cashman & McAnulty

operative analgesics in patients undergoing knee surgery. Eur J Anaesthiol 1992; 9: 29-34

121. Murphy DF, Medley C. Preoperative indomethacin for pain re­lief after thoracotomy: comparison with postoperative indo­methacin. Br J Anaesth 1993; 70: 298-300

122. Kavanagh BP, Katz J, Sandler AN, et al. Multimodal analgesia before thoracic surgery does not reduce postoperative pain. Br J Anaesth 1994; 73: 184-9

Correspondence and reprints: Dr Jeremy Cashman, Consult­ant Anaesthetist, Department of Anaesthesia, St George's Hospital, Blackshaw Road, London SW17 OQT, England.

Vol. 47, No.5, p. 827: In the summary of Dosage and Administration, the second sentence should read, 'In adults with normal renal function, a loading dose of 400mg (approximately 6 mg/kg) every 12 hours for the first 3 doses, followed by 400mg once daily, is recommended.' [Brogden RN, Peters DH. Teicoplanin: a reappraisal of its antimicrobial activity, pharmacokinetic properties and ther­apeutic efficacy. Drugs 1994 May; 47 (5): 823-54J

Vol. 48, No.3, p. 389: The first sentence in the Clinical Tolerability summary should read, 'PLS administration has been well tolerated in multicentre clinical trials (including approximately 3200 patients), in which it did .. .'. p. 397: The first sentence in section 4 Clinical Tolerability should read, 'PLS has been well tolerated in large multicentre clinical trials to date (including a total of approximately 3200 infants), in which it did ... '. p. 397: In section 3.2 Prophylactic Use, the sentence beginning on line 7 of the second column should read, 'The findings of a recent meta-analysis of 3 clinical studies, including the trial discussed abov/49J and evaluating a total of 671 neonates, indicate, however, that PLS prophylaxis does significantly improve clinical outcome versus rescue treat­mentY°J,

[Wiseman LR, Bryson HM. Porcine-derived lung surfactant: a review of the therapeutic efficacy and clinical tolerability of a natural surfactant preparation (Curosur!) in neonatal respiratory distress syndrome. Drugs 1994 Sept; 48 (3): 386-403J

© Adis International Limited. All rights reserved. Drugs 49 (1) 1995