You are on page 1of 7

Support Care Cancer (2005) 13: 153–159

DOI 10.1007/s00520-004-0690-6 REVIEW ARTICLE

Geoffrey K. Gourlay
Advances in opioid pharmacology

Received: 18 March 2004


Abstract Natural and synthetic opi- Recent research in molecular biology
Accepted: 9 August 2004 oid compounds, either alone or in and pharmacogenetics in relation to
Published online: 21 December 2004 combination with other drugs, are opioids and their receptors has helped
# Springer-Verlag 2004 widely used analgesics for patients clarify previous pharmacologic ob-
with both acute and chronic pain. servations and has laid the ground-
Decades of extensive pharmacologic work for new analgesic therapies with
G. K. Gourlay (*)
Pain Management Unit, Department of investigations have characterized improved therapeutic outcomes.
Anaesthesia and Pain Management, three high-affinity cell-surface neuro-
Flinders Medical Centre, The Flinders nal receptors, the activation of which Keywords Opioids . Pharmacology .
University of South Australia, is responsible for both the desirable Receptors . Metabolism
5042 Bedford Park,
South Australia, Australia
properties (antinociception) and un-
e-mail: Geoff.Gourlay@Flinders.edu.au desirable properties (respiratory de-
Tel.: +61-8-82017703 pression, nausea and vomiting,
Fax: +61-8-83741758 dependence, etc.) of opioid drugs.

Introduction endorphins) and, secondly, of their corresponding recep-


tors, which are expressed within specialized neurons of the
Opium and its derivative alkaloids have been known to ascending and descending pain transmission and inhibition
relieve pain and alter mood since the advent of recorded systems in the spinal cord, brainstem, medulla, periaque-
history. For centuries, these agents have been integrated ductal grey substance, thalamus, limbic system, and
into medical practice with varying efficacy. Recent devel- cerebral cortex [1, 2].
opments in opioid pharmacology and molecular biology The existence of three distinct opioid receptors was
promise to make this class of compounds even more originally deduced from the different pharmacologic ef-
clinically useful. New insights into receptor biology, fects of various opioid agonists and antagonists that se-
pharmacogenetics, and pharmacologic antagonism may lectively induce or inhibit different physiologic responses,
refine the use of established opioids and point toward the both experimentally and clinically [3–5]. Three separate
development of more selective compounds with improved receptors—μ (mu), δ (delta), and κ (kappa)—were iden-
clinical characteristics. Such improvements should be tified on the basis of pharmacologic response, in vitro
especially beneficial in treating patients with chronic pain, radioligand binding affinities, and in vivo localization of
such as cancer pain. labelled drug in tissue homogenates or sections [4–7]. This
allowed opioids to be grouped according to similarities
in the activation of their receptor types. Although all an-
New concepts in opioid receptors algesic agonists are bound by more than one receptor type
with varying degrees of affinity, the result is distinct dose-
The most important neurotransmitter system involved in response characteristics for each agonist while at the same
nociception is composed, firstly, of the endogenous opioid time accounting for the full range of recognized phar-
peptides (endomorphins, enkephalins, dynorphins, and macologic properties of each agent [1]. The μ-agonists
154

are responsible for analgesic properties at both spinal and Table 1 Opiate binding affinities. Note: the smaller the number, the
supraspinal sites, as well as for dependence, euphoria, greater the binding affinity for a particular receptor. Adapted from
reference 18 (ND, no data)
respiratory depression, sedation, miosis, and tolerance. The
δ-agonists, on the other hand, are associated with analgesia Opiate Affinity (Ki, 10−9 M) in the guinea pig
and euphoria, whereas the κ-agonists produce analgesia, μ δ κ
miosis, sedation, and, in contrast to the other receptors,
dysphoria [8, 9]. Morphine 1.8 90 317
Subsequently, similar observations led investigators to Normorphine 4.0 310 149
postulate the existence of multiple subtypes of each of the Levorphanol 0.6 5.6 9.6
three opioid receptors. Two subtypes of the μ-receptor (μ1 Codeine 2,700 >10,000 ND
and μ2) were initially proposed to explain how opioid Methadone 4.2 15.1 1,628
receptors demonstrated two different levels of in vitro Fentanyl 7.0 151 470
binding affinity, and how treatment with the μ-receptor Pethidine 385 4,345 5,140
antagonist naloxazone (an irreversible ligand) abolished Pentazocine 7.0 106 22.2
only the very high-affinity binding—i.e. the binding Buprenorphine 0.6 1.3 2.0
associated with typical opiate analgesia, or μ1. Lower Naloxone 1.8 27 17.2
affinity binding persisted for the μ-agonists as well for the
δ-agonists through a second, lower affinity μ-receptor, μ2
[10]. Pharmacologically, each subtype was purported to receptor types are highly conserved, whereas the extra-
be responsible for a portion of the responses typical of cellular loops are significantly dissimilar, and the primary
μ-agonist binding. Activation of the μ1-receptor is linked structure of each N-terminal extracellular segment is
to supraspinal analgesia, hypothermia, and prolactin re- essentially unique [21, 22]. Figure 2 depicts the same
lease, whereas activation of the μ2-receptor is responsible complex from above [11]. The highly conserved trans-
for spinal anaesthesia, respiratory depression, delayed membrane helices are arranged into the opioid binding
gastrointestinal tract transit, sedation, and bradycardia pocket. The poorly conserved extracellular loops provide a
[11]. Similar evidence has been used to support the ex- chemicophysical explanation for ligand discrimination due
istence of δ- and κ-receptor subtypes [12, 13]. to differences in noncovalent binding sites. The relatively
However, the presence of different receptor subtypes conserved intracellular loops and distinctive C-terminal
has been challenged by the failure to locate individually tails help account for the overall general similarities, as
distinct genes. Only three opioid receptor genes have been well as for the type-specific differences that occur in
isolated and cloned—a μ-receptor gene (MOR-1), a δ- intracellular signalling via G-coupled adenylyl cyclase
receptor gene (DOR-1), and a κ-receptor gene (KOR-1)
[14–16]. In addition, a variety of localization techniques
have demonstrated that each receptor type is expressed in
a distinct, but variably overlapping, distribution [17]. The
in vitro binding studies that suggested their existence had
been performed with crude tissue homogenates containing
more than a single receptor type. In light of this diver-
gence between the pharmacologic and molecular tech-
niques, three alternative hypotheses have been proposed
to account for opioid analgesia: (1) nearly all pharmaco-
logic and endogenous opioids bind to the high-affinity,
naloxone-sensitive μ1-receptor with similar affinity as
the source of analgesia [8]; (2) agonist binding affinities of
the three receptor types result in the analgesic response
[18]; and (3) receptor splice variants or receptor homo-
dimers and heterodimers might be responsible for anal-
gesic response [19, 20]. Table 1 lists the binding affinities
of different agonists and antagonists to the three opioid
receptors. Although the binding affinity is greatest to the
μ-receptor for each compound, variable (and often signif-
icant) binding occurs with the δ- and κ-receptors as well
(Table 1) [18].
Fig. 1 Cloned μ-, δ-, and κ-receptors from mammalian cells. Dark
Figure 1 demonstrates the degree of homology at each circles signify the same residue in all three opioid receptors, light
amino acid site from mammalian cells [21]. The trans- circles signify the same residue in two of three receptors, and
membrane domains and intracellular loops of all three open circles signify a unique residue. Adapted from reference 21
155

three receptor types. The activities of highly selective μ-,


δ-, and κ-agonists should be eliminated in MOR-, DOR-,
and KOR-deficient mice, respectively, and any residual
responsiveness should be interpreted as evidence of cross-
activation of the nondisrupted receptors. For example, in
MOR-deficient mutants, the analgesic effect of morphine
is abolished. Respiratory depression, naloxone-precipi-
tated withdrawal, and reward are also absent, and mor-
phine administration becomes adversive, perhaps due to
weak binding of morphine at the κ-receptor. Conversely,
morphine analgesia is intact in DOR- and KOR-deficient
mice. Interestingly, μ-receptors are required, to a variable
extent, for full responses to δ-agonists, suggesting an
interdependence or cross-reactivity between μ- and δ-re-
ceptors. The results of morphine administration in μ-
Fig. 2 How fentanyl might fit into the binding pocket of the μ- receptor knockout mice demonstrate that morphine effects
opioid receptor between the seven transmembrane domains.
Adapted from reference 11 do not require δ- or κ-receptors [21].
The knowledge obtained from molecular genetic in-
vestigation may point towards the development of new
inhibition, ion channel modulation, and mitogen-activated opioid agonists with improved pharmacologic properties
protein (MAP) kinase activation. Activation of all three for clinical use. Unfortunately, because μ-receptor activa-
receptors leads downstream to decreased neurotransmitter tion by itself is responsible for both the analgesic, as well
release and nociceptive impulse propagation [23, 24]. as the adverse, effects of morphine, it may not be ther-
Although there is no evidence that specific genes apeutically possible to separate the desired from the un-
encode receptor subtypes, observed pharmacologic diver- desired effects [31]. On the other hand, the existence of
sity and differences in localization may be the result of pharmacologically (as opposed to genetically) distinct re-
receptor species that arise from alternatively spliced mes- ceptor subtypes may offer the possibility of creating more
senger ribonucleic acid variant transcripts, post-transla- selective and less toxic opioids for therapeutic use.
tionally modified proteins [25], or receptor homodimers or
heterodimers [19, 20]. Dimerization might result in varied
individual response and incomplete cross-tolerance and Metabolism of opioid drugs
may be the basis for the utility of opioid rotation for
chronic use, such as for cancer pain. The relevance of The opioid alkaloids are extensively metabolized mainly
alternatively spliced opioid receptor variants [24, 17] and in the liver (morphine is mainly metabolized in the gut
dimerization is still controversial. wall during absorption following oral administration) and
The identification and cloning of the three opioid predominantly excreted via the kidneys [18]. There are
receptors made it possible to study each receptor in two main types of reactions: oxidations (catalyzed by
isolation. Mutant mice lacking the individual correspond- cytochrome P450) [32, 33] and conjugations (catalyzed by
ing genes (i.e. knockout mice) were generated using transferases such as UDP-glucuronyl transferase). Opiates
homologous recombination [26–28]. Any disruption in an (drugs with a morphine-like structure) with a hydroxyl
opiate receptor produced mice with absent binding of group at the 3-position are metabolized by glucuronida-
selective agonists for that receptor type. Mice deficient in tion, whereas those with substitution of the C3 hydroxyl
one, two, or all three receptors have been generated [29] (e.g. a methyl group such as in either codeine or ox-
and their physiologic and behavioural responses to various ycodone) undergo an oxidative demethylation prior to
agonists have been studied. The absence of one receptor glucuronidation. Ester-type opioids (e.g. heroin) are rap-
type does not markedly alter the expression of the other idly hydrolyzed by tissue esterases. Table 2 gives the
two, although the potential exists for changes in receptor/ major routes of biotransformation and major metabolites
G-protein coupling efficiency. It is even possible to study for commonly prescribed opioids [18, 33–35].
the effects of disrupting various portions within the same
gene. It should be noted, however, that different results
can still be obtained with targeted knockouts of the same Cytochrome P450
gene in different mice, due to subtle differences in the
extent of the genetic disruption that is produced either in A number of commonly used opioid analgesics (e.g.
the coding region itself or in its controlling elements [30]. codeine, fentanyl, sufentanil, methadone) are oxidized
Particular opioids have been identified pharmacologi- in the liver by microsomal cytochrome P450 (CYP450)
cally as prototypically selective agonists for each of the enzymes as their major route of biotransformation. Cur-
156

Table 2 Metabolism of opioids used for cancer pain. Sources: references 18, 33–35
Opioid Primary metabolic pathway Metabolite

Morphine Hepatic glucuronide conjugation by UDP glucuronyl transferase Morphine-3-glucuronide


Morphine 6-glucuronide
Codeine Hepatic oxidative O-demethylation by CYP2D6 Morphine
Oxycodone Hepatic oxidative O-demethylation by CYP2D6 Oxymorphone
N-demethylation by CYP3A4 and reduction Noroxycodone, α and β oxycodol
Levorphanol Hepatic glucuronide conjugation
Hydromorphone Hepatic glucuronide conjugation
Fentanyl Hepatic oxidative N-dealkylation by CYP3A4 Inactive metabolites
Methadone Hepatic oxidative N-demethylation by CYP3A4 Inactive metabolites
Heroin Sequential two-step deacetylation by various tissue esterases Morphine
Propoxyphene N-demethylation by CYP3A4 Norpropoxyphene
Meperidine N-demethylation by CYP3A4 Normeperidine

rently, 49 separate P450 genes have been identified and Codeine


sequenced. They comprise 17 families of oxidative en-
zymes with >40% sequence homology at the protein level; CYP2D6 catalyzes the demethylation of codeine to the
subfamilies are further defined by >70% homology. P450 more active metabolite morphine. Extensive metabolizers
enzymes are identified numerically by family, alphabet- convert 6% to 9% of administered codeine to morphine,
ically by subfamily, and individually by number, e.g. compared with only a trace in poor metabolizers. Con-
CYP3A4 [32]. Many analgesic and psychoactive drugs are sequently, an analgesic response to codeine occurs in ex-
metabolized by one of three P450 enzymes. The major tensive metabolizers only, even though both groups are
cytochrome in humans is 3A4, which is involved in 50% of responsive to the effects of morphine itself. Furthermore,
all microsomal drug metabolism, and is responsible for the the adverse effects of codeine unrelated to its conversion
metabolism of methadone, fentanyl, and certain selective to morphine are observed in both genotypes; poor me-
serotonin reuptake inhibitors [32, 36]. Of importance for tabolizers, therefore, experience the side effects, but not
clinical therapeutics, all cytochrome P450 enzymes other the analgesic effects, of codeine [42].
than 2D6 are inducible [37].

Meperidine
Morphine
The clinical consequences of opioid metabolism may also
Morphine is glucuronidated by the hepatic enzyme UDP- impact medical practice. The synthetic opioid meperidine
glucuronyl transferase and produces two major metabolites, had been a widely used analgesic routinely prescribed for
morphine-6-glucuronide (M6G) and morphine-3-glucuro- moderate-to-severe pain in medical and surgical patients.
nide (M3G), both of which are excreted in the urine [38]. However, its metabolic product, normeperidine, produces
M6G is a highly potent analgesic opioid that activates the CNS hyperexcitability presenting as adverse effects rang-
μ-receptor and is inactive in MOR-deficient mice [38, 31]. ing from nervousness to tremors, twitches, multifocal
M3G, the predominant metabolite of morphine, has no myoclonus, and grand mal seizures. Although excreted
opioid properties and has been proposed to be responsible by the kidneys, the accumulation of normeperidine may
for neuroexcitatory effects, including allodynia, myoclo- lead to neurologic toxicity even with normal renal func-
nus, and seizures, that may accompany large doses of tion [1, 43]. The association with CNS toxicity was not
morphine [39, 40]. The results of a study [41] cast doubt noted earlier because meperidine had been traditionally
on this hypothesis by demonstrating that renal failure prescribed at subtherapeutic doses. Recognition of the
invariably leads to elevated concentrations of both metab- adverse effect of this metabolite has led to the current
olites, often resulting in respiratory depression, obtunda- recommendation that the use of meperidine be limited only
tion, and central nervous system (CNS) hyperexcitability, to patients allergic to or otherwise unable to tolerate all
and showing that M3G lacks pharmacologic activity in other opioids [44, 45]. Meperidine also has a potential
humans [1, 41]. lethal interaction with monoamine oxidase inhibitors [46].
157

Agonist-antagonist combinations the first 24 h after surgery [53]. However, in a second trial
of general surgical patients, treatment with low-dose
Opioid antagonists are molecules that block or reverse the naloxone (average dose 0.5 μg/kg per hour for the first
pharmacologic effects of opioid agonists by competing for 2 h; 0.06 μg/kg per hour from hours 2 to 24 of ob-
opioid receptor sites. Unlike agonist binding, which leads servation) plus morphine via PCA-administered intermit-
to decreased neurotransmitter release and impulse propa- tent boluses resulted in higher morphine requirements,
gation, antagonist binding does not result in the usual in- greater pain intensity, and less pain relief and satisfaction
tracellular responses brought about by inhibitory G-protein than treatment with morphine plus placebo [54].
(Gi/Go) activation. The antagonist naloxone has a binding Unfortunately, differences in naloxone and morphine
affinity for the μ-receptor that is similar to that of dosages and modes of administration do not allow a direct
morphine (Table 1) [18]. Experimentally, however, low comparison of the contradictory findings from these two
concentrations of naloxone have been shown, paradoxi- trials. Low naloxone doses above a certain critical level
cally, to induce analgesia in mice by blocking the pre- still appear to allow opioid antagonism to predominate,
synaptic autoinhibition of enkephalin release thus causing whereas ultra-low doses may be necessary for the desired
an exaggerated release of endogenous opioids [47, 48]. proanalgesic effect. Also, synergism was apparent only at
Recent in vitro and in vivo data from animals have lower, but not at higher, morphine doses. Furthermore, an
demonstrated that extremely low doses of the opioid intermittent bolus may precipitate a period of hyperalgesia
antagonist naltrexone can enhance the antinociceptive due to higher initial naloxone levels, followed by a period
effects of morphine and other opioid agonists, allowing of excitatory opioid receptor sensitization by opioid ex-
such agents to be administered at far lower doses than posure [55]. At present, evidence for the use of low-dose
those normally required for analgesia. The basis for these antagonism is contradictory, and the routine introduction of
observations is a bimodal effect of opioid agonists. At this approach into clinical practice remains controversial.
extremely low doses, morphine can produce hyperalgesia
to painful stimuli in mice, whereas at normal doses, typical
analgesic effects are elicited [49]. An ultra-low dose of an Conclusions
opioid antagonist appears to block only the hyperalgesic
effect, unmasking the analgesic effect of the low-dose The opioid alkaloids are potent clinical analgesics the use
agonist. At higher doses, antagonists block both excitatory of which can be accompanied by significant adverse
and inhibitory activities. It has been suggested that no- effects, including respiratory depression, nausea, vomit-
ciceptive neurons contain small numbers of excitatory GS- ing, pruritus, and neuroexcitation. Use of these agents
protein-coupled opioid receptors responsible for adverse is also associated with mood elevation, tolerance, and
opioid effects, in addition to the more abundant inhibitory dependency. However, the rate of development of tol-
Gi/Go-protein-coupled receptors responsible for initiating erance between patients is highly variable and opioid
the ordinary antinociceptive response [49]. In neuronal dependency is not considered a major clinical problem in
cell culture, the opioid antagonist naloxone selectively appropriately selected patients. Opioid action is mediated
blocks excitatory GS-coupled responses elicited by very through high-affinity G-coupled receptors that produce
low concentrations of morphine and unmasks the analge- inhibition of the nociception pathways. Three receptor
sic inhibitory Gi/Go-coupled effects of these low opioid types—μ, δ, and κ—have been defined pharmacologically
concentrations [49, 50]. and their respective genes have been cloned. Several sub-
It has been proposed that coadministration of an ultra- types of each receptor have been proposed on the basis
low dose of naloxone or naltrexone may allow the clinical of differential pharmacologic effects; although no sepa-
use of morphine doses that are low enough to reduce rate genes have been identified, evidence for alternatively
tolerance, dependence, and other adverse effects [49]. spliced variants has been presented.
Several clinical trials of low-dose opioid antagonists Many opioid analgesics, including codeine, fentanyl,
coadministered with morphine have suggested that this and methadone, are metabolized by hepatic cytochrome
approach might reduce morphine requirements and side P450 enzymes, mainly 3A4 and 2D6. The presence of
effects associated with morphine administration, including polymorphisms in CYP2D6 defines poor versus extensive
respiratory depression, urinary retention, nausea, vomit- metabolizers. Poor metabolizers do not obtain analgesic
ing, and pruritus in patients requiring pain management relief from codeine, which must be transformed to
[51, 52]. In one randomized, placebo-controlled study, morphine, but they do experience adverse effects. Mor-
patients undergoing total abdominal hysterectomy who phine is metabolized through glucuronidation into the
were treated with morphine via patient-controlled analge- active analgesic M6G and the inactive metabolite M3G,
sia (PCA) plus naloxone (0.25 μg/kg per hour) adminis- and accumulation of these metabolites may actually induce
tered as a continuous intravenous infusion experienced an neuroexcitability. Renal failure may lead to accumulation
approximately 30% decrease in cumulative morphine use of both metabolites, thus resulting in combined toxicities.
compared with morphine plus placebo-treated patients in
158

Growing pharmacologic and molecular understanding conflicting clinical results on the efficacy of this combined
of the opioid system may bring the development of novel therapy, and additional trials in humans are warranted.
opioids that may reduce the adverse effects that occasion-
ally result from opioid treatment. One strategy, which has Acknowledgements The preparation of this article was supported
experimental support, suggests that morphine doses can be by Endo Pharmaceuticals, Chadds Ford, Pennsylvania, with editorial
lowered by the coadministration of an ultra-low dose of an assistance provided by Accel Medical Education, New York, New
York.
opioid antagonist such as naloxone; however, there are

References
1. Inturrisi CE (2002) Clinical pharma- 12. Jiang Q, Takemori AE, Sultana M, 22. Gaveriaux-Ruff C, Kieffer BL (2002)
cology of opioids for pain. Clin J Pain Portoghese PS, Bowen WD, Mosberg Opioid receptor genes inactivated in
18:S3–13 HI, Porreca F (1991) Differential mice: the highlights. Neuropeptides
2. Przewlocki R, Przewlocka B (2001) antagonism of opioid delta antinoci- 36:62–71
Opioids in chronic pain. Eur J Phar- ception by [D-Ala2, Leu5, Cys6]. 23. Trapaidze N, Gomes I, Cvejic S,
macol 429:79–91 enkephalin and naltrindole 5′-isothio- Bansinath M, Devi LA (2000) Opioid
3. Gilbert PE, Martin WR (1976) Sigma cyanate: evidence for delta receptor receptor endocytosis and activation of
effects of nalorphine in the chronic subtypes. J Pharmacol Exp Ther MAP kinase pathway. Brain Res Mol
spinal dog. Drug Alcohol Depend 257:1069–1075 Brain Res 76:220–228
1:373–376 13. Attali B, Gouarderes C, Mazarguil H, 24. Chevlen E (2003) Opioids: a review.
4. Martin WR, Eades CG, Thompson JA, Audigier Y, Cros J (1982) Evidence for Curr Pain Headache Rep 7:15–23
Huppler RE, Gilbert PE (1976) The multiple “kappa” binding sites by use 25. Kaufman DL, Keith DE Jr, Anton B,
effects of morphine- and nalorphine- of opioid peptides in the guinea-pig Tian J, Magendzo K, Newman D, Tran
like drugs in the nondependent and lumbo-sacral spinal cord. Neuropep- TH, Lee DS, Wen C, Xia YR (1995)
morphine-dependent chronic spinal tides 3:53–64 Characterization of the murine mu
dog. J Pharmacol Exp Ther 197:517– 14. Chen Y, Mestek A, Liu J, Hurley JA, opioid receptor gene. J Biol Chem
532 Yu L (1993) Molecular cloning and 270:15877–15883
5. Lord JA, Waterfield AA, Hughes J, functional expression of a mu-opioid 26. Sora I, Takahashi N, Funada M, Ujike
Kosterlitz HW (1977) Endogenous receptor from rat brain. Mol Pharmacol H, Revay RS, Donovan DM, Miner LL,
opioid peptides: multiple agonists and 44:8–12 Uhl GR (1997) Opiate receptor knock-
receptors. Nature 267:495–499 15. Evans CJ, Keith DE Jr, Morrison H, out mice define mu receptor roles in
6. Pasternak GW (1980) Multiple opiate Magendzo K, Edwards RH (1992) endogenous nociceptive responses and
receptors: [3H].ethylketocyclazocine Cloning of a delta opioid receptor by morphine-induced analgesia. Proc Natl
receptor binding and ketocyclazocine functional expression. Science Acad Sci U S A 94:1544–1549
analgesia. Proc Natl Acad Sci U S A 258:1952–1955 27. Zhu Y, King MA, Schuller AG, Nitsche
77:3691–3694 16. Minami M, Toya T, Katao Y, Maekawa JF, Reidl M, Elde RP, Unterwald E,
7. Pasternak GW (1993) Pharmacological K, Nakamura S, Onogi T, Kaneko S, Pasternak GW, Pintar JE (1999) Re-
mechanisms of opioid analgesics. Clin Satoh M (1993) Cloning and expres- tention of supraspinal delta-like anal-
Neuropharmacol 16:1–18 sion of a cDNA for the rat kappa-opioid gesia and loss of morphine tolerance in
8. Knapp RJ, Porreca F, Burks TF, receptor. FEBS Lett 329:291–295 delta opioid receptor knockout mice.
Yamamura HI (1989) Mediation of 17. Wei LN, Loh HH (2002) Regulation of Neuron 24:243–252
analgesia by multiple opioid receptors. opioid receptor expression. Curr Opin 28. Simonin F, Valverde O, Smadja C,
In: Hill CS, Fields WS (eds) Advances Pharmacol 2:69–75 Slowe S, Kitchen I, Dierich A, Le Meur
in pain research and therapy. Raven 18. Gourlay GK (2002) Clinical pharma- M, Roques BP, Maldonado R, Kieffer
Press, New York, pp 247–289 cology of the treatment of chronic non- BL (1998) Disruption of the kappa-
9. Freye E (1987) New concepts in opioid cancer pain. In: Giamberardino MA opioid receptor gene in mice enhances
activity. Opioid agonists, antagonists, (ed) Pain 2002—an updated review sensitivity to chemical visceral pain,
and mixed narcotic analgesics: theoret- (refresher course syllabus). IASP Press, impairs pharmacological actions of the
ical background and consideration for Seattle, pp 381–394 selective kappa-agonist U-50, 488H
practical use. Springer, New York, pp 19. Filizola M, Olmea O, Weinstein H and attenuates morphine withdrawal.
64–66 (2002) Prediction of heterodimerization EMBO J 17:886–897
10. Wolozin BL, Pasternak GW (1981) interfaces of G-protein coupled recep- 29. Simonin F, Slowe S, Becker JA,
Classification of multiple morphine and tors with a new subtractive correlated Matthes HW, Filliol D, Chluba J,
enkephalin binding sites in the central mutation method. Protein Eng 15:881– Kitchen I, Kieffer BL (2001) Analysis
nervous system. Proc Natl Acad Sci U 885 of [3H] bremazocine binding in single
S A 78:6181–6185 20. Jordan BA, Devi LA (1999) G-protein- and combinatorial opioid receptor
11. Traynor JR (1996) The u-opioid re- coupled receptor heterodimerization knockout mice. Eur J Pharmacol
ceptor. Pain Rev 3:221–248 modulates receptor function. Nature 414:189–195
399:697–700
21. Gaveriaux-Ruff C, Kieffer BL (1999)
Opioid receptors: gene structure and
function. In: Stein C (ed) Opioids in
pain control: basic and clinical aspects.
Cambridge University Press,
Cambridge, pp 1–20
159

30. Pasternak GW (2001) The pharmacol- 40. Bartlett SE, Smith MT (1995) The 49. Crain SM, Shen KF (2001) Acute
ogy of mu analgesics: from patients to apparent affinity of morphine-3-glucu- thermal hyperalgesia elicited by low-
genes. Neuroscientist 7:220–231 ronide at mu1-opioid receptors results dose morphine in normal mice is
31. Kieffer BL, Gaveriaux-Ruff C (2002) from morphine contamination: demon- blocked by ultra-low-dose naltrexone,
Exploring the opioid system by gene stration using HPLC and radioligand unmasking potent opioid analgesia.
knockout. Prog Neurobiol 66:285–306 binding. Life Sci 57:609–615 Brain Res 888:75–82
32. Ma MK, Woo MH, McLeod HL (2002) 41. Osborne RJ, Joel SP, Slevin ML (1986) 50. Crain SM, Shen KF (1995) Ultra-low
Genetic basis of drug metabolism. Am Morphine intoxication in renal failure: concentrations of naloxone selectively
J Health Syst Pharm 59:2061–2069 the role of morphine-6-glucuronide. antagonize excitatory effects of mor-
33. Heiskanen T, Olkkola KT, Kalso E BMJ 292:1548–1549 phine on sensory neurons, thereby
(1998) Effects of blocking CYP2D6 on 42. Eckhardt K, Li S, Ammon S, Schanzle increasing its antinociceptive potency
the pharmacokinetics and pharmaco- G, Mikus G, Eichelbaum M (1998) and attenuating tolerance/dependence
dynamics of oxycodone. Clin Pharma- Same incidence of adverse drug events during chronic cotreatment. Proc Natl
col Ther 64:603–611 after codeine administration irrespec- Acad Sci U S A 92:10540–10544
34. Wright AW, Mather LE, Smith MT tive of the genetically determined dif- 51. Wright PM, O’Toole DP, Barron DW
(2001) Hydromorphone-3-glucuronide: ferences in morphine formation. Pain (1992) The influence of naloxone in-
a more potent neuro-excitant than its 76:27–33 fusion on the action of intrathecal
structural analogue, morphine-3-glucu- 43. Kaiko RF, Foley KM, Grabinski PY, diamorphine: low-dose naloxone and
ronide. Life Sci 69:409–420 Heidrich G, Rogers AG, Inturrisi CE, neuroendocrine responses. Acta Ana-
35. Roxane Laboratories (2000) Levorpha- Reidenberg MM (1983) Central ner- esthesiol Scand 36:230–233
nol (prescribing information). Roxane vous system excitatory effects of me- 52. Joshi GP, Duffy L, Chehade J,
Laboratories, Columbus, OH peridine in cancer patients. Ann Neurol Wesevich J, Gajraj N, Johnson ER
36. Foster DJ, Somogyi AA, Bochner F 13:180–185 (1999) Effects of prophylactic nalme-
(1999) Methadone N-demethylation in 44. Waitman J, McCaffery M (2001) Me- fene on the incidence of morphine-
human liver microsomes: lack of peridine—a liability. Am J Nurs related side effects in patients receiving
stereoselectivity and involvement of 101:57–58 intravenous patient-controlled analge-
CYP3A4. Br J Clin Pharmacol 47:403– 45. Agency for Healthcare Policy and sia. Anesthesiology 90:1007–1011
412 Research (1994) Management of cancer 53. Gan TJ, Ginsberg B, Glass PS, Fortney
37. Barkin RL, Barkin D (2001) Pharma- pain, clinical guideline number 9 J, Jhaveri R, Perno R (1997) Opioid-
cologic management of acute and (AHCPR publication number 94-0592) sparing effects of a low-dose infusion
chronic pain: focus on drug interactions 46. Boden R, Botting R, Coulson P, of naloxone in patient-administered
and patient-specific pharmacotherapeu- Spanswick G (1984) Effect of nonse- morphine sulfate. Anesthesiology
tic selection. South Med J 94:756–770 lective and selective inhibitors of 87:1075–1081
38. Penson RT, Joel SP, Bakhshi K, Clark monoamine oxidases A and B on 54. Cepeda MS, Africano JM, Manrique
SJ, Langford RM, Slevin ML (2000) pethidine toxicity in mice. Br J Phar- AM, Fragoso W, Carr DB (2002) The
Randomized placebo-controlled trial of macol 82:151–154 combination of low dose of naloxone
the activity of the morphine glucuro- 47. Ueda H, Fukushima N, Kitao T, Ge M, and morphine in PCA does not de-
nides. Clin Pharmacol Ther 68:667– Takagi H (1986) Low doses of nalox- crease opioid requirements in the post-
676 one produce analgesia in the mouse operative period. Pain 96:73–79
39. Smith MT (2000) Neuroexcitatory ef- brain by blocking presynaptic autoin- 55. Mehlisch DR (2003) The combination
fects of morphine and hydromorphone: hibition of enkephalin release. Neurosci of low dose of naloxone and morphine
evidence implicating the 3-glucuronide Lett 65:247–252 in patient-controlled (PCA) does not
metabolites. Clin Exp Pharmacol 48. Vaughan CW, Christie MJ (1997) Pre- decrease opioid requirements in the
Physiol 27:524–528 synaptic inhibitory action of opioids on postoperative period. Pain 101:209–
synaptic transmission in the rat periaq- 211
ueductal grey in vitro. J Physiol 498
(Pt 2):463–472

You might also like