5
The impact of opioid-induced hyperalgesia for postoperative pain

https://doi.org/10.1016/j.bpa.2006.12.004Get rights and content

Clinical evidence suggests that – besides their well known analgesic activity – opioids can increase rather than decrease sensitivity to noxious stimuli. Based on the observation that opioids can activate pain inhibitory and pain facilitatory systems, this pain hypersensitivity has been attributed to a relative predominance of pronociceptive mechanisms. Acute receptor desensitization via uncoupling of the receptor from G-proteins, upregulation of the cAMP pathway, activation of the N-methyl-d-aspartate (NMDA)-receptor system, as well as descending facilitation, have been proposed as potential mechanisms underlying opioid-induced hyperalgesia. Numerous reports exist demonstrating that opioid-induced hyperalgesia is observed both in animal and human experimental models. Brief exposures to μ-receptor agonists induce long-lasting hyperalgesic effects for days in rodents, and also in humans large-doses of intraoperative μ-receptor agonists were found to increase postoperative pain and morphine consumption. Furthermore, the prolonged use of opioids in patients is often associated with a requirement for increasing doses and the development of abnormal pain. Successful strategies that may decrease or prevent opioid-induced hyperalgesia include the concomitant administration of drugs like NMDA-antagonists, α2-agonists, or non-steroidal anti-inflammatory drugs (NSAIDs), opioid rotation or combinations of opioids with different receptor selectivity.

Introduction

Tissue damage during surgery elicits an activation of nociceptive systems. High threshold mechano-, thermo- and chemosensors, the nociceptors (lat.: nocereb = to damage), rapidly transmit information about the degree and site of damage to the central nervous system. Depending on the type and extent of damage, sensitization processes leading to increased pain sensitivity, i.e. hyperalgesia, can be observed in the peripheral and central nervous systems (Figure 1A). Peripheral sensitization can be observed particularly during inflammation and other pathological tissue changes. They can sensitize nociceptors locally by lowering their activation thresholds or de-novo sensitize primarily insensitive, so-called ‘sleeping’ nociceptors in.1, 2, 3, 4, 5, 6Central sensitization is characterized by the increased spontaneous activity, and expansion of receptive fields of dorsal horn neurons.7, 8, 9, 10 One crucial event of this process is the activation of spinal N-Methyl-d-Aspartate (NMDA-) receptors by glutamate.11, 12, 13 Central sensitization processes can, thus, not only initiate but also maintain pain conditions that long out-last the triggering event. Furthermore, sensitization processes also appear, independent of pain perception during anesthesia and are often the basis for the development of postoperative pain.

Several publications show that perioperative antinociceptive therapy with opioids can reduce postoperative pain.14, 15, 16, 17, 18, 19, 20 Furthermore, opioids are the drugs of choice in tumor pain therapy, for the treatment of strong trauma pain, and for concomitant medication in patients with long-term artificial respiration. Thus, it is remarkable that patients with similar pain conditions often require very different quantities of opioids. Factors that influence this variability include pain type (nociceptive-, inflammatory- or neuropathic pain), psychosocial condition, and genetic disposition (gender or ethnicity).21, 22 Habituation to opioids or use of concomitant medication can also cause variation in opioid requirement.

The concept of ‘habituation’ is based on a multitude of adaptive responses of the organism to exogenous opioids. In this discussion, we include these adaptive responses in the term tolerance development.23 The (analgesic) tolerance is characterized by decreasing analgesic effect during long-term application of opioids, necessitating dose increases (Figure 1B). Tolerance development is not based on intensified pain sensation, but can be observed even without overt pain experience.24 However, not only decreasing analgesic effects are observed clinically following administration of opioids, but pain may also increase above the preexisting level of hyperalgesia.25, 26, 27, *28, *29 This would imply that the decreasing analgesic effect is based not only on a reduced antinociceptive potency of opiods, but additionally on the activation of opposing, i.e. pronociceptive systems.*30, 31, *32, *33 The basic idea of such compensatory reactions to drug application has been described in the “Opponent Process Theory”.34 In this theory the interplay between a drug-induced central effect and the induced counteracting endogenous response is discussed. The drug induced effects (such as opioid-induced analgesia) have a short onset and are stable upon repetition, whereas the counteracting process (such as opioid-induced hyperalgesia) has a delayed onset, but increased upon repetition (Figure 2). According to this theory the observed effect of opioids would be determined by the interaction of the two opposing anti- and pro-nociceptive processes.33

The molecular mechanisms underlying these anti- and pronociceptive mechanisms will be discussed below. Additionally, the relevance of these mechanisms to human and animal experimental investigation will be described and approaches to therapeutic modulation of opioid-induced hyperalgesia will be introduced.

Section snippets

Antinociceptive systems

Opioids activate peripheral, spinal, and supraspinal opioid receptors. To date, four different groups of opioid receptors have been identified (μ, δ, κ, ORL-1). In addition, eight isoforms (μ 1−3, δ 1−2, κ 1−3) and numerous subtypes have been pharmacologically characterized. The opioid receptors mediate their effects via an activation of guanine-nucleotide-binding protein (G-proteins), particularly – but not exclusively – pertussis toxin-sensitive Gi/o-protein.35 Opioid receptor-initiated

Pronociceptive systems

Long before Pert and Snyder (1974) first described opioid receptors, it was known that long-term opioid therapy could cause a renewed increase of initially suppressed pain, which was initially attributed to a loss of the antinociceptive capacity of opioids. In recent years a new hypothesis was brought up that proposed activation of pronociceptive systems*30, 31, *33 by opioids. Possible mechanisms of pronociceptive opioid effects are summarized in Figure 3.

Receptor-Desensitization. Opioid

Experimental investigation of opioid-induced hyperalgesia

Opioid-induced hyperalgesia has been investigated studying pain behaviour in animal models and using psychophysics in humans. Below, we focus on the effects of 4-Anilinopiperidine opioids used today in the perioperative setting (fentanyl, alfentanil and remifentanil) and the opioids used in pain and substitution therapy (morphine, methadone and heroin) will be characterized based on animal and human experimental research.

Fentanyl. In animal experiments, after repeated application of fentanyl, a

Therapeutic implications

In many cases, an increased demand for opioids can be attributed to increasing input of nociceptive afferents or, particularly in chronic and tumor pain therapy, a situational variation of pain experiences (Fear, Grief, Isolation)23, but not to an opioid-induced hyperalgesia. However, in these clinical conditions increased pain can be mediated by activation of the same pronociceptive systems (NMDA-Receptor system, descending facilitation) and thus, therapeutically may have the same implications.

Conclusion

Opioid-mediated analgesia causes a reduction and even a reversal of pain sensation, thus playing a significant role for the integrity of the human body. However, opioids can also cause hyperalgesic pain conditions in both animals and humans29 and opioid therapy can be complicated by tolerance development. Even after a short-term application, opioids have been shown to initiate sensitization processes that are still detectable after several days.108, 109 Both, hyperalgesic effects and tolerance

References (184)

  • M.H. Ossipov et al.

    Induction of pain facilitation by sustained opioid exposure: relationship to opioid antinociceptive tolerance

    Life Sciences

    (2003)
  • L.D. Aimone et al.

    Opioid modulation of capsaicin-evoked release of substance P from rat spinal cord in vivo

    Peptides

    (1989)
  • L. He et al.

    Regulation of opioid receptor trafficking and morphine tolerance by receptor oligomerization

    Cell

    (2002)
  • B.L. Kieffer et al.

    Opioid tolerance – in search of the holy grail

    Cell

    (2002)
  • D.E. Keith et al.

    Morphine activates opioid receptors without causing their rapid internalization

    The Journal of Biological Chemistry

    (1996)
  • J. Whistler et al.

    Functional dissociation of m Opioid receptor signalling and endocytosis: implications for the biology of opiate tolerance and addiction

    Neuron

    (1999)
  • M.M. Heinricher et al.

    Direct and indirect actions of morphine on medullary neurons that modulate nociception

    Neuroscience

    (1992)
  • M.M. Heinricher et al.

    Disinhibition of off-cells and antinociception produced by an opioid action within the rostral ventromedial medulla

    Neuroscience

    (1994)
  • A.H. Dickenson et al.

    The pharmacology of excitatory and inhibitory amino acid-mediated events in the transmission and modulation of pain in the spinal cord

    General Pharmacology

    (1997)
  • W.J. Martin et al.

    Inflammation-induced up-regulation of protein kinase Cgamma immunoreactivity in rat spinal cord correlates with enhanced nociceptive processing

    Neuroscience

    (1999)
  • D.J. Mayer et al.

    The development of morphine tolerance and dependence is associated with translocation of protein kinase C

    Pain

    (1995)
  • T. Avidor-Reiss et al.

    Chronic opioid treatment induces adenyl cyclase V superactivation

    The Journal of Biological Chemistry

    (1996)
  • S.M. Crain et al.

    Modulation of opioid analgesia, tolerance and dependence by Gs-coupled, GM1ganglioside-regulated opioid receptor functions

    Trends in Pharmacological Sciences

    (1998)
  • T.J. Luger et al.

    Mechanisms of the influence of midazolame on morphine antinociception at spinal and supraspinal levels in rats

    European Journal of Pharmacology

    (1994)
  • T.J. Luger et al.

    The spinal potentiating effect and the supraspinal inhibitory effect of midazolam on opioid-induced analgesia

    European Journal of Pharmacology

    (1995)
  • K. Elliott et al.

    The NMDA receptor antagonists, LY274614 and MK-801, and the nitric oxide synthase inhibitor, NG-nitro-L-arginine, attenuate analgesic tolerance to th mu-opioid morphine but not to kappa opioids

    Pain

    (1994)
  • N.H. Majeed et al.

    Inhibition of nitric oxide synthetase attenuates the development of morphine tolerance and dependence in mice

    Neuropharmacology

    (1994)
  • J. Mao et al.

    The inhibition of nitric oxide-activated poly(ADP-ribose) synthetase attenuates transsynaptic alteration of spinal cord dorsal horn neurons and neuropathic pain in the rat

    Pain

    (1997)
  • S.R. Skilling et al.

    Selective potentiation on NMDA-induced activity and release of excitatory amino acids by dynorphin: possible roles in paralysis and neurotoxicity

    Brain Research

    (1992)
  • J.J. Benoliel et al.

    Differential inhibitory/stimulatory modulation of spinal CCK release by mu and delta opioid agonists, and selective blockade of mu-dependent inhibition by kappa receptor stimulation

    Neuroscience Letters

    (1991)
  • J.P. Devillers et al.

    Simultaneous activation of spinal antiopioid system (neuropeptide FF) and pain facilitatory circuitry by stimulation of opioid receptors in rats

    Brain Research

    (1995)
  • C. Gouarderes et al.

    Nociceptin receptors in the rat spinal cord during morphine tolerance

    Brain Research

    (1999)
  • A.K. Rattan et al.

    Effect of chronic treatment with morphine, midazolame and both together on dynorphin(1-3) levels in the rat

    Brain Research

    (1997)
  • X.J. Xu et al.

    Up-regulation of cholecystokinin in primary sensory neurons is associated with morphine insensitivity in experimental neuropathic pain in the rat

    Neuroscience Letters

    (1993)
  • L. Yuan et al.

    Accelerated release and production of orphanin FQ in the brain of chronic morphine tolerant rats

    Brain Research

    (1999)
  • T.W. Vanderah et al.

    Mechanisms of opioid-induced pain and antinociceptive tolerance: descending facilitation and spinal dynorphin

    Pain

    (2001)
  • L.R. Watkins et al.

    Potentiation of morphine analgesia by the cholecystokinin antagonists proglumide

    Brain Research

    (1985)
  • P.W. Beck et al.

    Bradykinin and serotonin effects on various types of cutaneous nerve fibers

    Pflügers Archiv: European Journal of Physiology

    (1974)
  • P. Bessou et al.

    Responses of cutaneous sensory units with unmyelinated fibers to noxious stimuli

    Journal of Neurophysiology

    (1969)
  • R.A. Meyer et al.

    Myelinated nociceptive afferents account for the hyperalgesia that follows a burn to the hand

    Science

    (1981)
  • P.W. Reeh et al.

    Sensitization of nociceptive cutaneous nerve fibers from the rat's tail by noxious mechanical stimulation

    Experimental Brain Research

    (1987)
  • M. Schmelz et al.

    Limitation of sensitization to injured parts of receptive fields in human skin C-nociceptors

    Experimental Brain Research

    (1996)
  • R. Schmidt et al.

    Novel classes of responsive and unresponsive C nociceptors in human skin

    The Journal of Neuroscience

    (1995)
  • R.H. LaMotte et al.

    Neurogenic hyperalgesia psychophysical studies of underlying mechanisms

    Journal of Neurophysiology

    (1991)
  • S.N. Raja et al.

    Evidence for different mechanisms of primary and secondary hyperalgesia following heat injury to the glabrous skin

    Brain

    (1984)
  • H.G. Schaible et al.

    The effects of NMDA antagonists on neuronal activity in cats spinal cord evoked by acute inflammation in the knee joint

    The European Journal of Neuroscience

    (1991)
  • S.E. Abram et al.

    Morphine, but not inhalation anesthesia, blocks post-injury facilitation

    Anesthesiology

    (1993)
  • H. Kehlet et al.

    The value of multimodal or balanced analgesia in postoperative pain treatment

    Anesthesia and Analgesia

    (1993)
  • C.J. Woolf et al.

    Preemptive analgesia – treating postoperative pain by preventing the establishment of central sensitization

    Anesthesia and Analgesia

    (1993)
  • B. Kest et al.

    Gender-differences in opioid-mediated analgesia: animal and human studies

    Anesthesiology

    (2000)
  • Cited by (131)

    View all citing articles on Scopus
    View full text