Skip to main content
Erschienen in: CNS Drugs 10/2019

01.10.2019 | Current Opinion

Opioid-Induced Tolerance and Hyperalgesia

verfasst von: Sebastiano Mercadante, Edoardo Arcuri, Angela Santoni

Erschienen in: CNS Drugs | Ausgabe 10/2019

Einloggen, um Zugang zu erhalten

Abstract

Opioids are very potent and efficacious drugs, traditionally used for both acute and chronic pain conditions. However, the use of opioids is frequently associated with the occurrence of adverse effects or clinical problems. Other than adverse effects and dependence, the development of tolerance is a significant problem, as it requires increased opioid drug doses to achieve the same effect. Mechanisms of opioid tolerance include drug-induced adaptations or allostatic changes at the cellular, circuitry, and system levels. Dose escalation in long-term opioid therapy might cause opioid-induced hyperalgesia (OIH), which is a state of hypersensitivity to painful stimuli associated with opioid therapy, resulting in exacerbation of pain sensation rather than relief of pain. Various strategies may provide extra-opioid analgesia. There are drugs that may produce independent analgesic effects. A tailored treatment provided by skilled personnel, in accordance with the individual condition, is mandatory. Any treatment aimed at reducing opioid consumption may be indicated in these circumstances. Interventional techniques able to decrease the pain input may allow a decrease in the opioid dose, thus reverting the mechanisms producing tolerance of OIH. Intrathecal therapy with local anesthetics and a sympathetic block are the most common techniques utilized in these circumstances.
Literatur
1.
Zurück zum Zitat Breivik H, Eisenberg E, O’Brien T. The individual and societal burden of chronic pain in Europe: the case for strategic prioritisation and action to improve knowledge and availability of appropriate care. BMC Public Health. 2013;13:1229.CrossRefPubMedPubMedCentral Breivik H, Eisenberg E, O’Brien T. The individual and societal burden of chronic pain in Europe: the case for strategic prioritisation and action to improve knowledge and availability of appropriate care. BMC Public Health. 2013;13:1229.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Johannes CB, Le TK, Zhou X, Johnston JA, Dworkin RH. The prevalence of chronic pain in United States adults: results of an Internet-based survey. J Pain. 2010;11:1230–9.CrossRefPubMed Johannes CB, Le TK, Zhou X, Johnston JA, Dworkin RH. The prevalence of chronic pain in United States adults: results of an Internet-based survey. J Pain. 2010;11:1230–9.CrossRefPubMed
3.
Zurück zum Zitat Mercadante S, Portenoy RK. Opioid poorly-responsive cancer pain: part 2: basic mechanisms that could shift dose response for analgesia. J Pain Symptom Manag. 2001;21:255–64.CrossRef Mercadante S, Portenoy RK. Opioid poorly-responsive cancer pain: part 2: basic mechanisms that could shift dose response for analgesia. J Pain Symptom Manag. 2001;21:255–64.CrossRef
4.
Zurück zum Zitat Cahill CM, Walwyn W, Taylor AMW, Pradhan AAA, Evans CJ. Allostatic mechanisms of opioid tolerance beyond desensitization and downregulation. Trends Pharmacol Sci. 2016;37:963–76.CrossRefPubMedPubMedCentral Cahill CM, Walwyn W, Taylor AMW, Pradhan AAA, Evans CJ. Allostatic mechanisms of opioid tolerance beyond desensitization and downregulation. Trends Pharmacol Sci. 2016;37:963–76.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Gulur P, Williams L, Chaudhary S, Koury K, Jaff M. Opioid tolerance—a predictor of increased length of stay and higher readmission rates. Pain Physician. 2014;17:E503–7.PubMed Gulur P, Williams L, Chaudhary S, Koury K, Jaff M. Opioid tolerance—a predictor of increased length of stay and higher readmission rates. Pain Physician. 2014;17:E503–7.PubMed
6.
Zurück zum Zitat Volkow ND, McLellan AT. Opioid abuse in chronic pain—misconceptions and mitigation strategies. N Engl J Med. 2016;374:1253–63.CrossRefPubMed Volkow ND, McLellan AT. Opioid abuse in chronic pain—misconceptions and mitigation strategies. N Engl J Med. 2016;374:1253–63.CrossRefPubMed
7.
Zurück zum Zitat Siegel S, Hinson RE, Krank MD, McCully J. Heroin ‘overdose’ death: contribution of drug-associated environmental cues. Science. 1982;216:436–7.CrossRefPubMed Siegel S, Hinson RE, Krank MD, McCully J. Heroin ‘overdose’ death: contribution of drug-associated environmental cues. Science. 1982;216:436–7.CrossRefPubMed
8.
Zurück zum Zitat Voon P, Hayashi K, Milloy MJ, Nguyen P, Wood E, Montaner J, et al. Pain among high-risk patients on methadone maintenance treatment. J Pain. 2015;16:887–94.CrossRefPubMedPubMedCentral Voon P, Hayashi K, Milloy MJ, Nguyen P, Wood E, Montaner J, et al. Pain among high-risk patients on methadone maintenance treatment. J Pain. 2015;16:887–94.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Gupta S. Hyperalgesia induced by opioid drugs. J Ration Pharmacother Res. 2018;4:22–30. Gupta S. Hyperalgesia induced by opioid drugs. J Ration Pharmacother Res. 2018;4:22–30.
10.
Zurück zum Zitat Silverman SM. Opioid induced hyperalgesia: clinical implications for the pain practitioner. Physician. 2009;12:679–84. Silverman SM. Opioid induced hyperalgesia: clinical implications for the pain practitioner. Physician. 2009;12:679–84.
11.
Zurück zum Zitat Compton P, Canamar CP, Hillhouse M, Ling W. Hyperalgesia in heroin dependent patients and the effects of opioid substitution therapy. J Pain. 2012;13:401–9.CrossRefPubMedPubMedCentral Compton P, Canamar CP, Hillhouse M, Ling W. Hyperalgesia in heroin dependent patients and the effects of opioid substitution therapy. J Pain. 2012;13:401–9.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Rieb LM, Norman WV, Martin RE, Berkowitz J, Wood E, McNeil R, et al. Withdrawal-associated injury site pain (WISP): a descriptive case series of an opioid cessation phenomenon. Pain. 2016;157:2865–74.CrossRefPubMedPubMedCentral Rieb LM, Norman WV, Martin RE, Berkowitz J, Wood E, McNeil R, et al. Withdrawal-associated injury site pain (WISP): a descriptive case series of an opioid cessation phenomenon. Pain. 2016;157:2865–74.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Arout C, Edens E, Ismene L, Petrakis IL, Sofuoglu M. Targeting opioid-induced hyperalgesia in clinical treatment: neurobiological considerations. CNS Drugs. 2015;29:465–86.CrossRefPubMed Arout C, Edens E, Ismene L, Petrakis IL, Sofuoglu M. Targeting opioid-induced hyperalgesia in clinical treatment: neurobiological considerations. CNS Drugs. 2015;29:465–86.CrossRefPubMed
14.
Zurück zum Zitat Wilson-Poe AR, Lau BK, Vaughan CW. Repeated morphine treatment alters cannabinoid modulation of GABAergic synaptic transmission within the rat periaqueductal grey. Br J Pharmacol. 2015;172:681–90.CrossRefPubMed Wilson-Poe AR, Lau BK, Vaughan CW. Repeated morphine treatment alters cannabinoid modulation of GABAergic synaptic transmission within the rat periaqueductal grey. Br J Pharmacol. 2015;172:681–90.CrossRefPubMed
15.
Zurück zum Zitat Wilson-Poe AR, Jeong HI, Vaughan CW. Chronic morphine reduces the readily releasable pool of GABA, a presynaptic mechanism of opioid tolerance. J Physiol. 2017;595:6541–55.CrossRefPubMedPubMedCentral Wilson-Poe AR, Jeong HI, Vaughan CW. Chronic morphine reduces the readily releasable pool of GABA, a presynaptic mechanism of opioid tolerance. J Physiol. 2017;595:6541–55.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Lau BK, Vaughan CW. Descending modulation of pain: the GABA disinhibition hypothesis of analgesia. Curr Opin Neurobiol. 2014;29:159–64.CrossRefPubMed Lau BK, Vaughan CW. Descending modulation of pain: the GABA disinhibition hypothesis of analgesia. Curr Opin Neurobiol. 2014;29:159–64.CrossRefPubMed
17.
Zurück zum Zitat Chieng B, Christie MJ. Inhibition by opioids acting on mu-receptors of GABAergic and glutamatergic postsynaptic potentials in single rat periaqueductal gray neurones in vitro. Br J Pharmacol. 1994;113:303–9.CrossRefPubMedPubMedCentral Chieng B, Christie MJ. Inhibition by opioids acting on mu-receptors of GABAergic and glutamatergic postsynaptic potentials in single rat periaqueductal gray neurones in vitro. Br J Pharmacol. 1994;113:303–9.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Vaughan CW, Christie MJ. Presynaptic inhibitory action of opioids on synaptic transmission in the rat periaqueductal grey in vitro. J Physiol. 1997;498:463–72.CrossRefPubMedPubMedCentral Vaughan CW, Christie MJ. Presynaptic inhibitory action of opioids on synaptic transmission in the rat periaqueductal grey in vitro. J Physiol. 1997;498:463–72.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Connor M, Bagley EE, Chieng BC, Christie MJ. β-Arrestin-2 knockout prevents development of cellular mu-opioid receptor tolerance but does not affect opioid-withdrawal-related adaptations in single PAG neurons. Brit J Pharmacol. 2015;172:492–500.CrossRef Connor M, Bagley EE, Chieng BC, Christie MJ. β-Arrestin-2 knockout prevents development of cellular mu-opioid receptor tolerance but does not affect opioid-withdrawal-related adaptations in single PAG neurons. Brit J Pharmacol. 2015;172:492–500.CrossRef
20.
Zurück zum Zitat Vanderah TW, Suenaga NM, Ossipov MH, Malan TP, Lai J, Porreca F. Tonic descending facilitation from the rostral ventromedial medulla mediates opioid-induced abnormal pain and antinociceptive tolerance. J Neurosci. 2001;21:279–86.CrossRefPubMedPubMedCentral Vanderah TW, Suenaga NM, Ossipov MH, Malan TP, Lai J, Porreca F. Tonic descending facilitation from the rostral ventromedial medulla mediates opioid-induced abnormal pain and antinociceptive tolerance. J Neurosci. 2001;21:279–86.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Meng ID, Harasawa I. Chronic morphine exposure increases the proportion of on-cells in the rostral ventromedial medulla in rats. Life Sci. 2007;80:1915–20.CrossRefPubMedPubMedCentral Meng ID, Harasawa I. Chronic morphine exposure increases the proportion of on-cells in the rostral ventromedial medulla in rats. Life Sci. 2007;80:1915–20.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Ippolito DL, Temkin PA, Rogalski SL, Chavkin C. N-terminal tyrosine residues within the potassium channel Kir3 modulate GTPase activity of Galphai. J Biol Chem. 2002;277:32692–6.CrossRefPubMed Ippolito DL, Temkin PA, Rogalski SL, Chavkin C. N-terminal tyrosine residues within the potassium channel Kir3 modulate GTPase activity of Galphai. J Biol Chem. 2002;277:32692–6.CrossRefPubMed
23.
Zurück zum Zitat Torrecilla M, Marker CL, Cintora SC, Stoffel M, Williams JT, Wickman K. G-protein-gated potassium channels containing Kir3.2 and Kir3.3 subunits mediate the acute inhibitory effects of opioids on locus ceruleus neurons. J Neurosci. 2002;22:4328–34.CrossRefPubMedPubMedCentral Torrecilla M, Marker CL, Cintora SC, Stoffel M, Williams JT, Wickman K. G-protein-gated potassium channels containing Kir3.2 and Kir3.3 subunits mediate the acute inhibitory effects of opioids on locus ceruleus neurons. J Neurosci. 2002;22:4328–34.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Zhao YL, Chen SR, Chen H, Pan HL. Chronic opioid potentiates presynaptic but impairs postsynaptic N-methyl-d-aspartic acid receptor activity in spinal cords: implications for opioid hyperalgesia and tolerance. J Biol Chem. 2012;287:25073–85.CrossRefPubMedPubMedCentral Zhao YL, Chen SR, Chen H, Pan HL. Chronic opioid potentiates presynaptic but impairs postsynaptic N-methyl-d-aspartic acid receptor activity in spinal cords: implications for opioid hyperalgesia and tolerance. J Biol Chem. 2012;287:25073–85.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Price DD, Mayer DJ, Mao J, Caruso FS. NMDA-receptor antagonists and opioid receptor interactions as related to analgesia and tolerance. J Pain Symptom Manag. 2000;19(1 Suppl.):S7–11.CrossRef Price DD, Mayer DJ, Mao J, Caruso FS. NMDA-receptor antagonists and opioid receptor interactions as related to analgesia and tolerance. J Pain Symptom Manag. 2000;19(1 Suppl.):S7–11.CrossRef
26.
Zurück zum Zitat Gintzler AR, Chakrabarti S. Chronic morphine-induced plasticity among signalling molecules. Novartis Found Symptom. 2004;261:191–3. Gintzler AR, Chakrabarti S. Chronic morphine-induced plasticity among signalling molecules. Novartis Found Symptom. 2004;261:191–3.
27.
Zurück zum Zitat Hull LC, Llorente J, Gabra BH, et al. The effect of protein kinase C and G protein-coupled receptor kinase inhibition on tolerance induced by mu-opioid agonists of different efficacy. J Pharmacol Exp Ther. 2010;332:1127–35.CrossRefPubMedPubMedCentral Hull LC, Llorente J, Gabra BH, et al. The effect of protein kinase C and G protein-coupled receptor kinase inhibition on tolerance induced by mu-opioid agonists of different efficacy. J Pharmacol Exp Ther. 2010;332:1127–35.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Melief EJ, Miyatake M, Bruchas MR, Chavkin C. Ligand-directed c-Jun N-terminal kinase activation disrupts opioid receptor signaling. Proc Natl Acad Sci USA. 2010;107:11608–13.CrossRefPubMedPubMedCentral Melief EJ, Miyatake M, Bruchas MR, Chavkin C. Ligand-directed c-Jun N-terminal kinase activation disrupts opioid receptor signaling. Proc Natl Acad Sci USA. 2010;107:11608–13.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Bobeck EN, Ingram SL, Hermes SM, Aicher SA, Morgan MM. Ligand-biased activation of extracellular signal-regulated kinase 1/2 leads to differences in opioid induced antinociception and tolerance. Behav Brain Res. 2016;298:17–24.CrossRefPubMed Bobeck EN, Ingram SL, Hermes SM, Aicher SA, Morgan MM. Ligand-biased activation of extracellular signal-regulated kinase 1/2 leads to differences in opioid induced antinociception and tolerance. Behav Brain Res. 2016;298:17–24.CrossRefPubMed
30.
Zurück zum Zitat Morgan MM, Bobeck EN, Ingram SL. Glutamate modulation of antinociception, but not tolerance, produced by morphine microinjection into the periaqueductal gray of the rat. Brain Res. 2009;1295:59–66.CrossRefPubMedPubMedCentral Morgan MM, Bobeck EN, Ingram SL. Glutamate modulation of antinociception, but not tolerance, produced by morphine microinjection into the periaqueductal gray of the rat. Brain Res. 2009;1295:59–66.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Xie JY, Herman DS, Stiller C-O, Gardell LR, Ossipov MH, Lai J, et al. Cholecystokinin in the rostral ventromedial medulla mediates opioid- induced hyperalgesia and antinociceptive tolerance. J Neurosci. 2005;25:409–16.CrossRefPubMedPubMedCentral Xie JY, Herman DS, Stiller C-O, Gardell LR, Ossipov MH, Lai J, et al. Cholecystokinin in the rostral ventromedial medulla mediates opioid- induced hyperalgesia and antinociceptive tolerance. J Neurosci. 2005;25:409–16.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Thomas J, Mustafa S, Johnson J, Nicotra L, Hutchinson M. The relationship between opioids and immune signaling in the spinal cord. Handb Exp Pharmacol. 2015;227:207–38.CrossRefPubMed Thomas J, Mustafa S, Johnson J, Nicotra L, Hutchinson M. The relationship between opioids and immune signaling in the spinal cord. Handb Exp Pharmacol. 2015;227:207–38.CrossRefPubMed
33.
Zurück zum Zitat Xu J-TT, Yaster M, Tao Y-XX, et al. Opioid receptor-triggered spinal mTORC1 activation contributes to morphine tolerance and hyperalgesia. J Clin Investig 2014;124:592–603. Xu J-TT, Yaster M, Tao Y-XX, et al. Opioid receptor-triggered spinal mTORC1 activation contributes to morphine tolerance and hyperalgesia. J Clin Investig 2014;124:592–603.
34.
Zurück zum Zitat Harada S, Nakamoto K, Tokuyama S. The involvement of midbrain astrocyte in the development of morphine tolerance. Life Sci. 2013;93:573–8.CrossRefPubMed Harada S, Nakamoto K, Tokuyama S. The involvement of midbrain astrocyte in the development of morphine tolerance. Life Sci. 2013;93:573–8.CrossRefPubMed
35.
Zurück zum Zitat Eidson LN, Murphy AZ. Blockade of toll-like receptor 4 attenuates morphine tolerance and facilitates the pain relieving properties of morphine. J Neurosci. 2013;33:15952–63.CrossRefPubMedPubMedCentral Eidson LN, Murphy AZ. Blockade of toll-like receptor 4 attenuates morphine tolerance and facilitates the pain relieving properties of morphine. J Neurosci. 2013;33:15952–63.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Beutler B, Du X, Poltorak A. Identification of Toll-like receptor 4 (Tlr4) as the sole conduit for LPS signal transduction: genetic and evolutionary studies. J Endotoxin Res. 2001;7:277–80.CrossRefPubMed Beutler B, Du X, Poltorak A. Identification of Toll-like receptor 4 (Tlr4) as the sole conduit for LPS signal transduction: genetic and evolutionary studies. J Endotoxin Res. 2001;7:277–80.CrossRefPubMed
37.
Zurück zum Zitat Thomas J, Mustafa S, Johnson J, Nicotra L, Hutchinson M. The relationship between opioids and immune signaling in the spinal cord. Handb Exp Pharmacol. 2015;227:207–38.CrossRefPubMed Thomas J, Mustafa S, Johnson J, Nicotra L, Hutchinson M. The relationship between opioids and immune signaling in the spinal cord. Handb Exp Pharmacol. 2015;227:207–38.CrossRefPubMed
38.
Zurück zum Zitat Tanga FY, Raghavendra V, DeLeo JA. Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain. Neurochem Int. 2004;45:397–407.CrossRefPubMed Tanga FY, Raghavendra V, DeLeo JA. Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain. Neurochem Int. 2004;45:397–407.CrossRefPubMed
39.
Zurück zum Zitat Tanga FY, Nutile-McMenemy N, DeLeo JA. The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proc Natl Acad Sci USA. 2005;102:5856–61.CrossRefPubMedPubMedCentral Tanga FY, Nutile-McMenemy N, DeLeo JA. The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proc Natl Acad Sci USA. 2005;102:5856–61.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Lewis SS, Hutchinson MR, Rezvani N, Loram LC, Zhang Y, Maier SF, et al. Evidence that intrathecal morphine-3-glucuronide may cause pain enhancement via toll-like receptor 4/MD-2 and interleukin-1beta. Neuroscience. 2010;165:569–83.CrossRefPubMed Lewis SS, Hutchinson MR, Rezvani N, Loram LC, Zhang Y, Maier SF, et al. Evidence that intrathecal morphine-3-glucuronide may cause pain enhancement via toll-like receptor 4/MD-2 and interleukin-1beta. Neuroscience. 2010;165:569–83.CrossRefPubMed
41.
Zurück zum Zitat Raghavendra V, Tanga FY, DeLeo JA. Complete Freunds adjuvant induced peripheral inflammation evokes glial activation and proinflammatory cytokine expression in the CNS. Eur J Neurosci. 2004;20:467–73.CrossRefPubMed Raghavendra V, Tanga FY, DeLeo JA. Complete Freunds adjuvant induced peripheral inflammation evokes glial activation and proinflammatory cytokine expression in the CNS. Eur J Neurosci. 2004;20:467–73.CrossRefPubMed
42.
Zurück zum Zitat Wang X, Loram LC, Ramos K, et al. Morphine activates neuroinflammation in a manner parallel to endotoxin. Proc Natl Acad Sci USA. 2012;109:6325–30.CrossRefPubMedPubMedCentral Wang X, Loram LC, Ramos K, et al. Morphine activates neuroinflammation in a manner parallel to endotoxin. Proc Natl Acad Sci USA. 2012;109:6325–30.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Little JW, Cuzzocrea S, Bryant L, Esposito E, Doyle T, Rausaria S, et al. Spinal mitochondrial-derived peroxynitrite enhances neuroimmune activation during morphine hyperalgesia and antinociceptive tolerance. Pain. 2013;154:978–86.CrossRefPubMedPubMedCentral Little JW, Cuzzocrea S, Bryant L, Esposito E, Doyle T, Rausaria S, et al. Spinal mitochondrial-derived peroxynitrite enhances neuroimmune activation during morphine hyperalgesia and antinociceptive tolerance. Pain. 2013;154:978–86.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Salvemini D, Doyle T, Kress M, Nicol G. Therapeutic targeting of the ceramide-to-sphingosine 1-phosphate pathway in pain. Trends Pharmacol Sci. 2013;34:110–8.CrossRefPubMed Salvemini D, Doyle T, Kress M, Nicol G. Therapeutic targeting of the ceramide-to-sphingosine 1-phosphate pathway in pain. Trends Pharmacol Sci. 2013;34:110–8.CrossRefPubMed
45.
Zurück zum Zitat You MH, Kim BM, Chen CH, Begley MJ, Cantley LC, Lee TH. Death-associated protein kinase 1 phosphorylates NDRG2 and induces neuronal cell death. Cell Death Differ. 2017;24:238–50.CrossRefPubMed You MH, Kim BM, Chen CH, Begley MJ, Cantley LC, Lee TH. Death-associated protein kinase 1 phosphorylates NDRG2 and induces neuronal cell death. Cell Death Differ. 2017;24:238–50.CrossRefPubMed
46.
Zurück zum Zitat Muscoli C, Doyle T, Dagostino C, Bryant L, Chen Z, Watkins LR, et al. Counterregulation of opioid analgesia by glial-derived bioactive sphingolipids. J Neurosci. 2010;30:15400–8.CrossRefPubMedPubMedCentral Muscoli C, Doyle T, Dagostino C, Bryant L, Chen Z, Watkins LR, et al. Counterregulation of opioid analgesia by glial-derived bioactive sphingolipids. J Neurosci. 2010;30:15400–8.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Grace PM, Gaudet AD, Staikopoulos V, Maier SF, Hutchinson MR, Salvemini D, et al. Nitroxidative signaling mechanisms in pathological pain. Trends Neurosci. 2016;39:862–79.CrossRefPubMedPubMedCentral Grace PM, Gaudet AD, Staikopoulos V, Maier SF, Hutchinson MR, Salvemini D, et al. Nitroxidative signaling mechanisms in pathological pain. Trends Neurosci. 2016;39:862–79.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Watkins LR, Hutchinson MR, Rice KC, Maier SF. The “toll” of opioid-induced glial activation: improving the clinical efficacy of opioids by targeting glia. Trends Pharmacol. Sci. 2009;30:581–91.CrossRefPubMedPubMedCentral Watkins LR, Hutchinson MR, Rice KC, Maier SF. The “toll” of opioid-induced glial activation: improving the clinical efficacy of opioids by targeting glia. Trends Pharmacol. Sci. 2009;30:581–91.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Nakamoto K, Kawasaki S, Kobori T, et al. Involvement of matrix metalloproteinase-9 in the development of morphine tolerance. Eur J Pharmacol. 2012;683:86–92.CrossRefPubMed Nakamoto K, Kawasaki S, Kobori T, et al. Involvement of matrix metalloproteinase-9 in the development of morphine tolerance. Eur J Pharmacol. 2012;683:86–92.CrossRefPubMed
50.
Zurück zum Zitat Hutchinson MR, Zhang Y, Shridhar M, et al. Evidence that opioids may have toll-like receptor 4 and MD-2 effects. Brain Behav. Immun. 2010;24:83–95.CrossRefPubMed Hutchinson MR, Zhang Y, Shridhar M, et al. Evidence that opioids may have toll-like receptor 4 and MD-2 effects. Brain Behav. Immun. 2010;24:83–95.CrossRefPubMed
51.
Zurück zum Zitat Watkins LR, Hutchinson MR, Johnston IN, Maier SF. Glia: novel counter-regulators of opioid analgesia. Trends Neurosci. 2005;28:661–9.CrossRefPubMed Watkins LR, Hutchinson MR, Johnston IN, Maier SF. Glia: novel counter-regulators of opioid analgesia. Trends Neurosci. 2005;28:661–9.CrossRefPubMed
52.
Zurück zum Zitat Eidson LN, Inoue K, Young LJ, Tansey MG, Murphy AZ. Toll-like receptor 4 mediates morphine-induced neuroinflammation and tolerance via soluble tumor necrosis factor signaling. Neuropsychopharmacology. 2017;42:661–70.CrossRefPubMed Eidson LN, Inoue K, Young LJ, Tansey MG, Murphy AZ. Toll-like receptor 4 mediates morphine-induced neuroinflammation and tolerance via soluble tumor necrosis factor signaling. Neuropsychopharmacology. 2017;42:661–70.CrossRefPubMed
53.
Zurück zum Zitat Hutchinson MR, Lewis SS, Coats BD, Skyba DA, Crysdale NY, Berkelhammer DL, et al. Reduction of opioid withdrawal and potentiation of acute opioid analgesia by systemic AV411 (ibudilast). Brain Behav Immun. 2009;23:240–50.CrossRefPubMed Hutchinson MR, Lewis SS, Coats BD, Skyba DA, Crysdale NY, Berkelhammer DL, et al. Reduction of opioid withdrawal and potentiation of acute opioid analgesia by systemic AV411 (ibudilast). Brain Behav Immun. 2009;23:240–50.CrossRefPubMed
54.
Zurück zum Zitat Johnston IN, Milligan ED, Wieseler-Frank J, et al. A role for proinflammatory cytokines and fractalkine in analgesia, tolerance, and subsequent pain facilitation induced by chronic intrathecal morphine. J Neurosci. 2004;24:7353–65.CrossRefPubMedPubMedCentral Johnston IN, Milligan ED, Wieseler-Frank J, et al. A role for proinflammatory cytokines and fractalkine in analgesia, tolerance, and subsequent pain facilitation induced by chronic intrathecal morphine. J Neurosci. 2004;24:7353–65.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Johnson JL, Rolan PE, Johnson ME, Bobrovskaya L, Williams DB, Johnson K, et al. Codeine-induced hyperalgesia and allodynia: investigating the role of glial activation. Transl Psychiatry. 2014;4:e482.CrossRefPubMedPubMedCentral Johnson JL, Rolan PE, Johnson ME, Bobrovskaya L, Williams DB, Johnson K, et al. Codeine-induced hyperalgesia and allodynia: investigating the role of glial activation. Transl Psychiatry. 2014;4:e482.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Gruber-Schoffnegger D, Drdla-Schutting R, Honigsperger C, Wunderbaldinger G, Gassner M, Sandkuhler J. Induction of thermal hyperalgesia and synaptic long-term potentiation in the spinal cord lamina I by TNF-alpha and IL-1beta is mediated by glial cells. J Neurosci. 2013;33:6540–51.CrossRefPubMedPubMedCentral Gruber-Schoffnegger D, Drdla-Schutting R, Honigsperger C, Wunderbaldinger G, Gassner M, Sandkuhler J. Induction of thermal hyperalgesia and synaptic long-term potentiation in the spinal cord lamina I by TNF-alpha and IL-1beta is mediated by glial cells. J Neurosci. 2013;33:6540–51.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Melik Parsadaniantz S, Rivat C, Rostene W, Reaux-Le Goazigo A. Opioid and chemokine receptor crosstalk: a promising target for pain therapy? Nat Rev Neurosci. 2015;16:69–78.CrossRefPubMed Melik Parsadaniantz S, Rivat C, Rostene W, Reaux-Le Goazigo A. Opioid and chemokine receptor crosstalk: a promising target for pain therapy? Nat Rev Neurosci. 2015;16:69–78.CrossRefPubMed
58.
Zurück zum Zitat Zhang N, Rogers TJ, Caterina M, Oppenheim JJ. Proinflammatory chemokines, such as C-C chemokine ligand 3, desensitize mu-opioid receptors on dorsal root ganglia neurons. J Immunol. 2004;173:594–9.CrossRefPubMed Zhang N, Rogers TJ, Caterina M, Oppenheim JJ. Proinflammatory chemokines, such as C-C chemokine ligand 3, desensitize mu-opioid receptors on dorsal root ganglia neurons. J Immunol. 2004;173:594–9.CrossRefPubMed
59.
Zurück zum Zitat Rivat C, Sebaihi S, Van Steenwinckel J, et al. Src family kinases involved in CXCL12-induced loss of acute morphine analgesia. Brain Behav Immun. 2014;38:38–52.CrossRefPubMed Rivat C, Sebaihi S, Van Steenwinckel J, et al. Src family kinases involved in CXCL12-induced loss of acute morphine analgesia. Brain Behav Immun. 2014;38:38–52.CrossRefPubMed
60.
Zurück zum Zitat Lin CP, Kang KH, Lin TH, et al. Role of spinal CXCL1 (GROalpha) in opioid tolerance: a human-to-rodent translational study. Anesthesiology. 2015;122:666–76.CrossRefPubMed Lin CP, Kang KH, Lin TH, et al. Role of spinal CXCL1 (GROalpha) in opioid tolerance: a human-to-rodent translational study. Anesthesiology. 2015;122:666–76.CrossRefPubMed
61.
Zurück zum Zitat Luo X, Wang X, Xia Z, Chung SK, Cheung CW. CXCL12/ CXCR61 axis: an emerging neuromodulator in pathological pain. Rev Neurosci. 2016;27:83–92.CrossRefPubMed Luo X, Wang X, Xia Z, Chung SK, Cheung CW. CXCL12/ CXCR61 axis: an emerging neuromodulator in pathological pain. Rev Neurosci. 2016;27:83–92.CrossRefPubMed
62.
63.
Zurück zum Zitat Horvath RJ, Romero-Sandoval EA, De Leo JA. Inhibition of microglial P2X4 receptors attenuates morphine tolerance, Iba1, GFAP and mu opioid receptor protein expression while enhancing perivascular microglial ED2. Pain. 2010;150:401–13.CrossRefPubMedPubMedCentral Horvath RJ, Romero-Sandoval EA, De Leo JA. Inhibition of microglial P2X4 receptors attenuates morphine tolerance, Iba1, GFAP and mu opioid receptor protein expression while enhancing perivascular microglial ED2. Pain. 2010;150:401–13.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Zhou D, Chen ML, Zhang YQ, Zhao ZQ. Involvement of spinal microglial P2X7 receptor in generation of tolerance to morphine analgesia in rats. J Neurosci. 2010;30:8042–7.CrossRefPubMedPubMedCentral Zhou D, Chen ML, Zhang YQ, Zhao ZQ. Involvement of spinal microglial P2X7 receptor in generation of tolerance to morphine analgesia in rats. J Neurosci. 2010;30:8042–7.CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Chen ML, Cao H, Chu YX, et al. Role of P2X7 receptor-mediated IL-18/IL-18R signaling in morphine tolerance: Multiple glial-neuronal dialogues in the rat spinal cord. J Pain. 2012;13:945–58.CrossRefPubMed Chen ML, Cao H, Chu YX, et al. Role of P2X7 receptor-mediated IL-18/IL-18R signaling in morphine tolerance: Multiple glial-neuronal dialogues in the rat spinal cord. J Pain. 2012;13:945–58.CrossRefPubMed
66.
Zurück zum Zitat Ferrini F, Trang T, Mattioli TA. Morphine hyperalgesia gated through microglia-mediated disruption of neuronal Cl– homeostasis. Nat Neurosci. 2013;16:183–92.CrossRefPubMedPubMedCentral Ferrini F, Trang T, Mattioli TA. Morphine hyperalgesia gated through microglia-mediated disruption of neuronal Cl homeostasis. Nat Neurosci. 2013;16:183–92.CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat Mattioli TA, Leduc-Pessah H, Skelhorne-Gross G, et al. Toll-like receptor 4 mutant and null mice retain morphine-induced tolerance, hyperalgesia, and physical dependence. PLoS One. 2014;9:e97361.CrossRefPubMedPubMedCentral Mattioli TA, Leduc-Pessah H, Skelhorne-Gross G, et al. Toll-like receptor 4 mutant and null mice retain morphine-induced tolerance, hyperalgesia, and physical dependence. PLoS One. 2014;9:e97361.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Ossipov MH, Lai J, Vanderah TW, Porreca F. Induction of pain facilitation by sustained opioid exposure: relationship to opioid antinociceptive tolerance. Life Sci. 2003;73:783–800.CrossRefPubMed Ossipov MH, Lai J, Vanderah TW, Porreca F. Induction of pain facilitation by sustained opioid exposure: relationship to opioid antinociceptive tolerance. Life Sci. 2003;73:783–800.CrossRefPubMed
70.
Zurück zum Zitat Toll L, Bruchas MR, Calo G, Cox BM, Zaveri NT. Nociceptin/orphanin FQ receptor structure, signaling, ligands, functions, and interactions with opioid systems. Pharmacol Rev. 2016;68:419–57.CrossRefPubMedPubMedCentral Toll L, Bruchas MR, Calo G, Cox BM, Zaveri NT. Nociceptin/orphanin FQ receptor structure, signaling, ligands, functions, and interactions with opioid systems. Pharmacol Rev. 2016;68:419–57.CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat Faris PL, Komisaruk BR, Watkins LR, Mayer DJ. Evidence for the neuropeptide cholecystokinin as an antagonist of opiate analgesia. Science. 1983;219:310–2.CrossRefPubMed Faris PL, Komisaruk BR, Watkins LR, Mayer DJ. Evidence for the neuropeptide cholecystokinin as an antagonist of opiate analgesia. Science. 1983;219:310–2.CrossRefPubMed
72.
Zurück zum Zitat Rothman RB. A review of the role of anti-opioid peptides in morphine tolerance and dependence. Synapse. 1992;12:129–38.CrossRefPubMed Rothman RB. A review of the role of anti-opioid peptides in morphine tolerance and dependence. Synapse. 1992;12:129–38.CrossRefPubMed
73.
Zurück zum Zitat McNally GP. Pain facilitatory circuits in the mammalian central nervous system: their behavioral significance and role in morphine analgesic tolerance. Neurosci Biobehav Rev. 1999;23:1059–78.CrossRefPubMed McNally GP. Pain facilitatory circuits in the mammalian central nervous system: their behavioral significance and role in morphine analgesic tolerance. Neurosci Biobehav Rev. 1999;23:1059–78.CrossRefPubMed
74.
Zurück zum Zitat Dourish CT, O’Neill MF, Schaffer LW, Siegl PK, Iversen SD. The cholecystokinin receptor antagonist devazepide enhances morphine-induced analgesia but not morphine-induced respiratory depression in the squirrel monkey. J Pharmacol Exp Ther. 1990;255:1158–65.PubMed Dourish CT, O’Neill MF, Schaffer LW, Siegl PK, Iversen SD. The cholecystokinin receptor antagonist devazepide enhances morphine-induced analgesia but not morphine-induced respiratory depression in the squirrel monkey. J Pharmacol Exp Ther. 1990;255:1158–65.PubMed
75.
Zurück zum Zitat Hoffmann O, Wiesenfeld-Hallin Z. The CCK-B receptor antagonist Cl 988 reverses tolerance to morphine in rats. NeuroReport. 1994;5:2565–8.CrossRefPubMed Hoffmann O, Wiesenfeld-Hallin Z. The CCK-B receptor antagonist Cl 988 reverses tolerance to morphine in rats. NeuroReport. 1994;5:2565–8.CrossRefPubMed
76.
Zurück zum Zitat Simonin F, Schmitt M, Laulin J-P, et al. RF9, a potent and selective neuropeptide FF receptor antagonist, prevents opioid-induced tolerance associated with hyperalgesia. Proc Natl Acad Sci USA. 2006;103:466–71.CrossRefPubMedPubMedCentral Simonin F, Schmitt M, Laulin J-P, et al. RF9, a potent and selective neuropeptide FF receptor antagonist, prevents opioid-induced tolerance associated with hyperalgesia. Proc Natl Acad Sci USA. 2006;103:466–71.CrossRefPubMedPubMedCentral
77.
Zurück zum Zitat Elhabazi K, Trigo JM, Mollereau C, et al. Involvement of neuropeptide FF receptors in neuroadaptive responses to acute and chronic opiate treatments. Br J Pharmacol. 2012;165:424–35.CrossRefPubMedPubMedCentral Elhabazi K, Trigo JM, Mollereau C, et al. Involvement of neuropeptide FF receptors in neuroadaptive responses to acute and chronic opiate treatments. Br J Pharmacol. 2012;165:424–35.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Depner UB, Reinscheid RK, Takeshima H, Brune K, Zeilhofer HU. Normal sensitivity to acute pain, but increased inflammatory hyperalgesia in mice lacking the nociception precursor polypeptide or the nociceptin receptor. Eur J Neurosci. 2003;17:2381–7.CrossRefPubMed Depner UB, Reinscheid RK, Takeshima H, Brune K, Zeilhofer HU. Normal sensitivity to acute pain, but increased inflammatory hyperalgesia in mice lacking the nociception precursor polypeptide or the nociceptin receptor. Eur J Neurosci. 2003;17:2381–7.CrossRefPubMed
79.
Zurück zum Zitat Rizzi A, Nazzaro C, Marzola GG, et al. Endogenous nociceptin/orphanin FQ signalling produces opposite spinal antinociceptive and supraspinal pronociceptive effects in the mouse formalin test: pharmacological and genetic evidences. Pain. 2006;124:100–8.CrossRefPubMed Rizzi A, Nazzaro C, Marzola GG, et al. Endogenous nociceptin/orphanin FQ signalling produces opposite spinal antinociceptive and supraspinal pronociceptive effects in the mouse formalin test: pharmacological and genetic evidences. Pain. 2006;124:100–8.CrossRefPubMed
81.
Zurück zum Zitat Tumati S, Largent-Milnes TM, Keresztes AI, et al. Tachykinin NK(1) receptor antagonist co-administration attenuates opioid withdrawal-mediated spinal microglia and astrocyte activation. Eur J Pharmacol. 2012;684:64–70.CrossRefPubMedPubMedCentral Tumati S, Largent-Milnes TM, Keresztes AI, et al. Tachykinin NK(1) receptor antagonist co-administration attenuates opioid withdrawal-mediated spinal microglia and astrocyte activation. Eur J Pharmacol. 2012;684:64–70.CrossRefPubMedPubMedCentral
82.
83.
Zurück zum Zitat Doehring A, Geisslinger G, Lotsch J. Epigenetics in pain and analgesia: an imminent research field. Eur J Pain. 2011;15:11–6.CrossRefPubMed Doehring A, Geisslinger G, Lotsch J. Epigenetics in pain and analgesia: an imminent research field. Eur J Pain. 2011;15:11–6.CrossRefPubMed
84.
Zurück zum Zitat Doehring A, Oertel BG, Sittl R, Lotsch J. Chronic opioid use is associated with increased DNA methylation correlating with increased clinical pain. Pain. 2013;154:15–23.CrossRefPubMed Doehring A, Oertel BG, Sittl R, Lotsch J. Chronic opioid use is associated with increased DNA methylation correlating with increased clinical pain. Pain. 2013;154:15–23.CrossRefPubMed
85.
Zurück zum Zitat Viet CT, Dang D, Aouizerat BE, Miaskowski C, Ye Y, Viet DT, et al. OPRM1 methylation contributes to opioid tolerance in cancer patients. J Pain. 2017;18:1046–59.CrossRefPubMedPubMedCentral Viet CT, Dang D, Aouizerat BE, Miaskowski C, Ye Y, Viet DT, et al. OPRM1 methylation contributes to opioid tolerance in cancer patients. J Pain. 2017;18:1046–59.CrossRefPubMedPubMedCentral
86.
Zurück zum Zitat Chao YC, Xie F, Li X, Guo R, Yang N, Zhang C, et al. Demethylation regulation of BDNF gene expression in dorsal root ganglion neurons is implicated in opioid-induced pain hypersensitivity in rats. Neurochem Int. 2016;97:91–8.CrossRefPubMed Chao YC, Xie F, Li X, Guo R, Yang N, Zhang C, et al. Demethylation regulation of BDNF gene expression in dorsal root ganglion neurons is implicated in opioid-induced pain hypersensitivity in rats. Neurochem Int. 2016;97:91–8.CrossRefPubMed
88.
Zurück zum Zitat He Y, Yang C, Kirkmire CM, Wang ZJ. Regulation of opioid tolerance by let-7 family microRNA targeting the mu opioid receptor. J Neurosci. 2010;30:10251–8.CrossRefPubMedPubMedCentral He Y, Yang C, Kirkmire CM, Wang ZJ. Regulation of opioid tolerance by let-7 family microRNA targeting the mu opioid receptor. J Neurosci. 2010;30:10251–8.CrossRefPubMedPubMedCentral
89.
Zurück zum Zitat Wu Q, Zhang L, Law PY, Wei LN, Loh HH. Long-term morphine treatment decreases the association of mu-opioid receptor (MOR1) mRNA with polysomes through miRNA23b. Mol Pharmacol. 2009;75:744–50.CrossRefPubMedPubMedCentral Wu Q, Zhang L, Law PY, Wei LN, Loh HH. Long-term morphine treatment decreases the association of mu-opioid receptor (MOR1) mRNA with polysomes through miRNA23b. Mol Pharmacol. 2009;75:744–50.CrossRefPubMedPubMedCentral
90.
Zurück zum Zitat Mercadante S. Portenoy RK Opioid poorly-responsive cancer pain. Part 3. Clinical strategies to improve opioid responsiveness. J Pain Symptom Manag. 2001;21:338–54.CrossRef Mercadante S. Portenoy RK Opioid poorly-responsive cancer pain. Part 3. Clinical strategies to improve opioid responsiveness. J Pain Symptom Manag. 2001;21:338–54.CrossRef
92.
Zurück zum Zitat Lee JT, Sanderson CR, Xuan W, Agar M. Lidocaine for cancer pain in adults: a systematic review and meta-analysis. J Palliat Med. 2019;22:326–34.CrossRefPubMed Lee JT, Sanderson CR, Xuan W, Agar M. Lidocaine for cancer pain in adults: a systematic review and meta-analysis. J Palliat Med. 2019;22:326–34.CrossRefPubMed
93.
Zurück zum Zitat Mercadante S, Fulfaro F, Casuccio A. A randomized controlled study on the use of anti-inflammatory drugs in patients with cancer pain on morphine therapy: effects on dose escalation and a pharmacoeconomic analysis. Eur J Cancer. 2002;38:1358–66.CrossRefPubMed Mercadante S, Fulfaro F, Casuccio A. A randomized controlled study on the use of anti-inflammatory drugs in patients with cancer pain on morphine therapy: effects on dose escalation and a pharmacoeconomic analysis. Eur J Cancer. 2002;38:1358–66.CrossRefPubMed
94.
Zurück zum Zitat Chabot-Doré AJ, Schuster DJ, Stone LS, Wilcox GL. Analgesic synergy between opioid and α2-adrenoceptors. Br J Pharmacol. 2015;172:388–402.CrossRefPubMed Chabot-Doré AJ, Schuster DJ, Stone LS, Wilcox GL. Analgesic synergy between opioid and α2-adrenoceptors. Br J Pharmacol. 2015;172:388–402.CrossRefPubMed
95.
Zurück zum Zitat Belgrade M, Hall S. Dexmedetomidine infusion for the management of opioid-induced hyperalgesia. Pain Med. 2010;11:1819–26.CrossRefPubMed Belgrade M, Hall S. Dexmedetomidine infusion for the management of opioid-induced hyperalgesia. Pain Med. 2010;11:1819–26.CrossRefPubMed
97.
Zurück zum Zitat Mercadante S, Caruselli A, Casuccio A. The use of ketamine in a palliative-supportive care unit: a retrospective analysis. Ann Palliat Med. 2018;7:205–10.CrossRefPubMed Mercadante S, Caruselli A, Casuccio A. The use of ketamine in a palliative-supportive care unit: a retrospective analysis. Ann Palliat Med. 2018;7:205–10.CrossRefPubMed
98.
Zurück zum Zitat Pasternak GW. Molecular biology of opioid analgesia. J Pain Symptom Manag. 2005;19:S2.CrossRef Pasternak GW. Molecular biology of opioid analgesia. J Pain Symptom Manag. 2005;19:S2.CrossRef
100.
Zurück zum Zitat Hashimoto T, Saito Y, Yamada K, et al. Enhancement of morphine analgesic effect with induction of mu-opioid receptor endocytosis in rats. Anesthesiology. 2006;105:574–80.CrossRefPubMed Hashimoto T, Saito Y, Yamada K, et al. Enhancement of morphine analgesic effect with induction of mu-opioid receptor endocytosis in rats. Anesthesiology. 2006;105:574–80.CrossRefPubMed
101.
Zurück zum Zitat Kieffer BL, Evans CJ. Opioid tolerance—in search of the holy grail. Cell. 2002;108:587–90.CrossRefPubMed Kieffer BL, Evans CJ. Opioid tolerance—in search of the holy grail. Cell. 2002;108:587–90.CrossRefPubMed
102.
Zurück zum Zitat Mercadante S, Bruera E. Opioid switching in cancer pain: from the beginning to nowadays. Crit Rev Oncol Hematol. 2016;99:241–8.CrossRefPubMed Mercadante S, Bruera E. Opioid switching in cancer pain: from the beginning to nowadays. Crit Rev Oncol Hematol. 2016;99:241–8.CrossRefPubMed
103.
Zurück zum Zitat Mercadante S. Managing difficult pain conditions in the cancer patient. Curr Pain Headache Rep. 2014;18:395.CrossRefPubMed Mercadante S. Managing difficult pain conditions in the cancer patient. Curr Pain Headache Rep. 2014;18:395.CrossRefPubMed
104.
Zurück zum Zitat Mercadante S. Switching methadone: a 10-year experience of 345 patients in an acute palliative care unit. Pain Med. 2012;13:399–404.CrossRefPubMed Mercadante S. Switching methadone: a 10-year experience of 345 patients in an acute palliative care unit. Pain Med. 2012;13:399–404.CrossRefPubMed
105.
Zurück zum Zitat Reddy A, Yennurajalingam S, Pulivarthi K, Palla SL, Wang X, Kwon JH, et al. Frequency, outcome, and predictors of success within 6 weeks of an opioid rotation among outpatients with cancer receiving strong opioids. Oncologist. 2013;18:212–20.CrossRefPubMed Reddy A, Yennurajalingam S, Pulivarthi K, Palla SL, Wang X, Kwon JH, et al. Frequency, outcome, and predictors of success within 6 weeks of an opioid rotation among outpatients with cancer receiving strong opioids. Oncologist. 2013;18:212–20.CrossRefPubMed
106.
Zurück zum Zitat Mercadante S, Arcuri E. Hyperalgesia and opioid switching. Am J Hosp Palliat Care. 2005;22:291–4.CrossRefPubMed Mercadante S, Arcuri E. Hyperalgesia and opioid switching. Am J Hosp Palliat Care. 2005;22:291–4.CrossRefPubMed
107.
Zurück zum Zitat Prommer E. Rotating methadone to other opioids: a lesson in the mechanisms of opioid tolerance and opioid-induced pain. J Palliat Med. 2006;9:488–93.CrossRefPubMed Prommer E. Rotating methadone to other opioids: a lesson in the mechanisms of opioid tolerance and opioid-induced pain. J Palliat Med. 2006;9:488–93.CrossRefPubMed
108.
Zurück zum Zitat Mercadante S, Caraceni A. Conversion ratios for opioid switching in the treatment of cancer pain: a systematic review. Palliat Med. 2011;25:494–503.CrossRef Mercadante S, Caraceni A. Conversion ratios for opioid switching in the treatment of cancer pain: a systematic review. Palliat Med. 2011;25:494–503.CrossRef
109.
Zurück zum Zitat Dale O, Moksnes K, Kaasa S. European Palliative Care Research Collaborative pain guidelines: opioid switching to improve analgesia or reduce side effects. A systematic review. Palliat Med. 2011;25:494–503.CrossRefPubMed Dale O, Moksnes K, Kaasa S. European Palliative Care Research Collaborative pain guidelines: opioid switching to improve analgesia or reduce side effects. A systematic review. Palliat Med. 2011;25:494–503.CrossRefPubMed
110.
Zurück zum Zitat Bruera E, Pereira J, Watanabe S, Belzile M, Kuehn N, Hanson J. Opioid rotation in patients with cancer pain. A retrospective comparison of dose ratios between methadone, hydromorphone, and morphine. Cancer. 1996;78:852–7.CrossRefPubMed Bruera E, Pereira J, Watanabe S, Belzile M, Kuehn N, Hanson J. Opioid rotation in patients with cancer pain. A retrospective comparison of dose ratios between methadone, hydromorphone, and morphine. Cancer. 1996;78:852–7.CrossRefPubMed
111.
Zurück zum Zitat Mercadante S, Casuccio A, Calderone L. Rapid switching from morphine to methadone in cancer patients with poor response to morphine. J Clin Oncol. 1999;17:3307–12.CrossRefPubMed Mercadante S, Casuccio A, Calderone L. Rapid switching from morphine to methadone in cancer patients with poor response to morphine. J Clin Oncol. 1999;17:3307–12.CrossRefPubMed
112.
Zurück zum Zitat Mercadante S, Bianchi M, Villari P, Ferrera P, Casuccio A, Fulfaro F, et al. Opioid plasma concentration during switching from morphine to methadone: preliminary data. Support Care Cancer. 2003;11:326–31.PubMed Mercadante S, Bianchi M, Villari P, Ferrera P, Casuccio A, Fulfaro F, et al. Opioid plasma concentration during switching from morphine to methadone: preliminary data. Support Care Cancer. 2003;11:326–31.PubMed
113.
Zurück zum Zitat Kurita GP, Sjøgren P, Klepstad P, Mercadante S. Interventional techniques to management of cancer-related pain: clinical and critical aspects. Cancers (Basel). 2019;11(4):443.CrossRefPubMedCentral Kurita GP, Sjøgren P, Klepstad P, Mercadante S. Interventional techniques to management of cancer-related pain: clinical and critical aspects. Cancers (Basel). 2019;11(4):443.CrossRefPubMedCentral
114.
Zurück zum Zitat Mercadante S, Klepstad P, Kurita GP, Sjøgren P, Giarratano A. Sympathetic blocks for visceral cancer pain management: a systematic review and EAPC recommendations. Crit Rev Oncol Hematol. 2015;96:577–83.CrossRefPubMed Mercadante S, Klepstad P, Kurita GP, Sjøgren P, Giarratano A. Sympathetic blocks for visceral cancer pain management: a systematic review and EAPC recommendations. Crit Rev Oncol Hematol. 2015;96:577–83.CrossRefPubMed
Metadaten
Titel
Opioid-Induced Tolerance and Hyperalgesia
verfasst von
Sebastiano Mercadante
Edoardo Arcuri
Angela Santoni
Publikationsdatum
01.10.2019
Verlag
Springer International Publishing
Erschienen in
CNS Drugs / Ausgabe 10/2019
Print ISSN: 1172-7047
Elektronische ISSN: 1179-1934
DOI
https://doi.org/10.1007/s40263-019-00660-0

Weitere Artikel der Ausgabe 10/2019

CNS Drugs 10/2019 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Thrombektomie auch bei großen Infarkten von Vorteil

16.05.2024 Ischämischer Schlaganfall Nachrichten

Auch ein sehr ausgedehnter ischämischer Schlaganfall scheint an sich kein Grund zu sein, von einer mechanischen Thrombektomie abzusehen. Dafür spricht die LASTE-Studie, an der Patienten und Patientinnen mit einem ASPECTS von maximal 5 beteiligt waren.

Schwindelursache: Massagepistole lässt Otholiten tanzen

14.05.2024 Benigner Lagerungsschwindel Nachrichten

Wenn jüngere Menschen über ständig rezidivierenden Lagerungsschwindel klagen, könnte eine Massagepistole der Auslöser sein. In JAMA Otolaryngology warnt ein Team vor der Anwendung hochpotenter Geräte im Bereich des Nackens.

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Viertel reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.