Skip to main content
Erschienen in: Der Anaesthesist 5/2004

01.05.2004 | Schmerztherapie

Opioid-induzierte Hyperalgesie

Pathophysiologie und Klinik

verfasst von: Priv.-Doz. Dr. med. W. Koppert

Erschienen in: Die Anaesthesiologie | Ausgabe 5/2004

Einloggen, um Zugang zu erhalten

Zusammenfassung

Opioide sind Mittel der ersten Wahl in der Therapie mittelschwerer bis starker akuter und chronischer Schmerzzustände. Allerdings können Opioide auch zu einer Schmerzverstärkung führen, die auf einer Aktivierung pronozizeptiver Systeme beruht. Es konnte gezeigt werden, dass neben einer akute Rezeptordesensibilisierung und einer Hochregulation der Adenylylzyklaseaktivität insbesondere die Aktivierung des N-Methyl-D-Aspartat- (NMDA-)Rezeptor-Systems und die deszendierende Fazilitierung den antinozizeptiven Eigenschaften des Opioids entgegengerichtet sind. So können schon nach kurzzeitiger Anwendung Sensibilisierungsprozesse induziert werden, die einen Teil der analgetischen Wirkung des Opioids maskieren und noch viele Tage nach dem Absetzen nachweisbar sein können. Klinische Relevanz erhalten diese Befunde aus Studien, in denen nach der intraoperativen Anwendung hoher Dosen von μ-Agonisten vermehrte Schmerzen und ein erhöhter postoperativer Schmerzmittelverbrauch beobachtet wurde. Weiterhin werden nach länger dauernder Anwendung von μ-Agonisten oftmals neben einem ansteigenden Bedarf an Schmerzmitteln paradoxe Schmerzzustände beobachtet. Durch eine Kombination der Opioide mit Substanzen anderer Klassen, wie NMDA-Rezeptor-Antagonisten, α2-Agonisten oder nichtsteroidalen antiinflammatorischen Analgetika (NSAIDs), durch Opioidrotationen oder Kombinationen von Opioiden mit unterschiedlicher Rezeptorselektivität können diese Sensibilisierungsprozesse unterdrückt und die Schmerztherapie optimiert werden.
Literatur
1.
Zurück zum Zitat Abbadie C, Pasternak GW (2001) Differential in vivo internalization of MOR-1 and MOR-1C by morphine. Neuroreport 12:3069–3072CrossRefPubMed Abbadie C, Pasternak GW (2001) Differential in vivo internalization of MOR-1 and MOR-1C by morphine. Neuroreport 12:3069–3072CrossRefPubMed
2.
Zurück zum Zitat Abram SE, Yaksh T (1993) Morphine, but not inhalation anesthesia, blocks post-injury facilitation. Anesthesiology 78:713–721PubMed Abram SE, Yaksh T (1993) Morphine, but not inhalation anesthesia, blocks post-injury facilitation. Anesthesiology 78:713–721PubMed
3.
Zurück zum Zitat Adriaenssens G, Vermeyen KM, Hoffmann VL, Mertens E, Adriaensen HF (1999) Postoperative analgesia with i.v. patient-controlled morphine: effect of adding ketamine. Br J Anaesth 83:393–396PubMed Adriaenssens G, Vermeyen KM, Hoffmann VL, Mertens E, Adriaensen HF (1999) Postoperative analgesia with i.v. patient-controlled morphine: effect of adding ketamine. Br J Anaesth 83:393–396PubMed
4.
Zurück zum Zitat Ali NM (1986) Hyperalgesic response in a patient receiving high concentrations of spinal morphine. Anesthesiology 65:449–450 Ali NM (1986) Hyperalgesic response in a patient receiving high concentrations of spinal morphine. Anesthesiology 65:449–450
5.
Zurück zum Zitat Angers S, Salahpour A, Bouvier M (2002) Dimerization: an emerging concept for G protein-coupled receptor ontogeny and function. Annu Rev Pharmacol Toxicol 42:409–435CrossRefPubMed Angers S, Salahpour A, Bouvier M (2002) Dimerization: an emerging concept for G protein-coupled receptor ontogeny and function. Annu Rev Pharmacol Toxicol 42:409–435CrossRefPubMed
6.
Zurück zum Zitat Angst MS, Koppert W, Pahl I, Clark JD, Schmelz M (2003) Short-term infusion of the mu-opioid agonist remifentanil in humans causes hyperalgesia during withdrawal. Pain 106:49–57CrossRefPubMed Angst MS, Koppert W, Pahl I, Clark JD, Schmelz M (2003) Short-term infusion of the mu-opioid agonist remifentanil in humans causes hyperalgesia during withdrawal. Pain 106:49–57CrossRefPubMed
7.
Zurück zum Zitat Arain SR, Ruehlow RM, Uhrich TD, Ebert TJ (2004) The efficacy of dexmedetomidine versus morphine for postoperative analgesia after major inpatient surgery. Anesth Analg 98:153–158PubMed Arain SR, Ruehlow RM, Uhrich TD, Ebert TJ (2004) The efficacy of dexmedetomidine versus morphine for postoperative analgesia after major inpatient surgery. Anesth Analg 98:153–158PubMed
8.
Zurück zum Zitat Arner S, Rawal N, Gustafsson LL (1988) Clinical experience of long-term treatment with epidural and intrathecal opioids—a nationwide survey. Acta Anaesthesiol Scand 32:253–259PubMed Arner S, Rawal N, Gustafsson LL (1988) Clinical experience of long-term treatment with epidural and intrathecal opioids—a nationwide survey. Acta Anaesthesiol Scand 32:253–259PubMed
9.
Zurück zum Zitat Bartlett SE, Cramond T, Smith MT (1994) The excitatory effects of morphine-3-glucuronide are attenuated by LY274614, a competitive NMDA receptor antagonist, and by midazolam, an agonist at the benzodiazepine site on the GABAA receptor complex. Life Sci 54:687–694PubMed Bartlett SE, Cramond T, Smith MT (1994) The excitatory effects of morphine-3-glucuronide are attenuated by LY274614, a competitive NMDA receptor antagonist, and by midazolam, an agonist at the benzodiazepine site on the GABAA receptor complex. Life Sci 54:687–694PubMed
10.
Zurück zum Zitat Bernard JM, Hommeril JL, Passuti N, Pinaud M (1991) Postoperative analgesia by intravenous clonidine. Anesthesiology 75:577–582PubMed Bernard JM, Hommeril JL, Passuti N, Pinaud M (1991) Postoperative analgesia by intravenous clonidine. Anesthesiology 75:577–582PubMed
11.
Zurück zum Zitat Bie B, Pan ZZ (2003) Presynaptic mechanism for anti-analgesic and anti-hyperalgesic actions of k-opioid receptors. J Neurosci 23:7262–7268PubMed Bie B, Pan ZZ (2003) Presynaptic mechanism for anti-analgesic and anti-hyperalgesic actions of k-opioid receptors. J Neurosci 23:7262–7268PubMed
12.
Zurück zum Zitat Bie B, Fields HL, Williams JT, Pan ZZ (2003) Roles of alpha1- and alpha2-adrenoceptors in the nucleus raphe magnus in opioid analgesia and opioid abstinence-induced hyperalgesia. J Neurosci 23:7950–7957PubMed Bie B, Fields HL, Williams JT, Pan ZZ (2003) Roles of alpha1- and alpha2-adrenoceptors in the nucleus raphe magnus in opioid analgesia and opioid abstinence-induced hyperalgesia. J Neurosci 23:7950–7957PubMed
13.
Zurück zum Zitat Borgland SL (2001) Acute opioid receptor desensitization and tolerance: is there a link? Clin Exp Pharmacol Physiol 28:147–154CrossRefPubMed Borgland SL (2001) Acute opioid receptor desensitization and tolerance: is there a link? Clin Exp Pharmacol Physiol 28:147–154CrossRefPubMed
14.
Zurück zum Zitat Bot G, Blake AD, Li S, Reisine T (1998) Fentanyl and its analogs desensitize the cloned mu opioid receptor. J Pharmacol Exp Ther 285:1207–1218PubMed Bot G, Blake AD, Li S, Reisine T (1998) Fentanyl and its analogs desensitize the cloned mu opioid receptor. J Pharmacol Exp Ther 285:1207–1218PubMed
15.
Zurück zum Zitat Bruera E, Peirera J, Watanabe C, Belzile M, Kuehn N, Hanson J (1996) Opioid rotation in patients with cancer pain. A retrospective comparison of dose ratios between methadone, hydromorphone, and morphine. Cancer 78:852–857CrossRefPubMed Bruera E, Peirera J, Watanabe C, Belzile M, Kuehn N, Hanson J (1996) Opioid rotation in patients with cancer pain. A retrospective comparison of dose ratios between methadone, hydromorphone, and morphine. Cancer 78:852–857CrossRefPubMed
16.
Zurück zum Zitat Carpenter KJ, Chapman V, Dickenson AH (2000) Neuronal inhibitory effects of methadone are predominantly opioid receptor mediated in the rat spinal cord in vivo. Eur J Pain 4:19–26CrossRefPubMed Carpenter KJ, Chapman V, Dickenson AH (2000) Neuronal inhibitory effects of methadone are predominantly opioid receptor mediated in the rat spinal cord in vivo. Eur J Pain 4:19–26CrossRefPubMed
17.
Zurück zum Zitat Celerier E, Laulin J, Larcher A, Le Moal M, Simonnet G (1999) Evidence for opiate-activated NMDA processes masking opiate analgesia in rats. Brain Res 847:18–25CrossRefPubMed Celerier E, Laulin J, Larcher A, Le Moal M, Simonnet G (1999) Evidence for opiate-activated NMDA processes masking opiate analgesia in rats. Brain Res 847:18–25CrossRefPubMed
18.
Zurück zum Zitat Celerier E, Rivat C, Jun Y, Laulin JP, Larcher A, Reynier P, Simonnet G (2000) Long-lasting hyperalgesia induced by fentanyl in rats: preventive effect of ketamine. Anesthesiology 92:465–472PubMed Celerier E, Rivat C, Jun Y, Laulin JP, Larcher A, Reynier P, Simonnet G (2000) Long-lasting hyperalgesia induced by fentanyl in rats: preventive effect of ketamine. Anesthesiology 92:465–472PubMed
19.
Zurück zum Zitat Celerier E, Laulin JP, Corcuff JB, Le Moal M, Simonnet G (2001) Progressive enhancement of delayed hyperalgesia induced by repeated heroin administration: a sensitization process. J Neurosci 21:4074–4080PubMed Celerier E, Laulin JP, Corcuff JB, Le Moal M, Simonnet G (2001) Progressive enhancement of delayed hyperalgesia induced by repeated heroin administration: a sensitization process. J Neurosci 21:4074–4080PubMed
20.
Zurück zum Zitat Chang HM, Berde CB, Holz GG 4th, Steward GF, Kream RM (1989) Sufentanil, morphine, met-enkephalin, and kappa-agonist (U-50,488H) inhibit substance P release from primary sensory neurons: a model for presynaptic spinal opioid actions. Anesthesiology 70:672–677PubMed Chang HM, Berde CB, Holz GG 4th, Steward GF, Kream RM (1989) Sufentanil, morphine, met-enkephalin, and kappa-agonist (U-50,488H) inhibit substance P release from primary sensory neurons: a model for presynaptic spinal opioid actions. Anesthesiology 70:672–677PubMed
21.
Zurück zum Zitat Cheng HYM, Pitcher GM, Laviolette SR et al. (2002) DREAM is a critical transcriptional repressor for pain modulation. Cell 108:31–43PubMed Cheng HYM, Pitcher GM, Laviolette SR et al. (2002) DREAM is a critical transcriptional repressor for pain modulation. Cell 108:31–43PubMed
22.
Zurück zum Zitat Chia YY, Liu K, Chow LH, Lee TY (1999) The preoperative administration of intravenous dextromethorphan reduces postoperative morphine consumption. Anesth Analg 89:748–752PubMed Chia YY, Liu K, Chow LH, Lee TY (1999) The preoperative administration of intravenous dextromethorphan reduces postoperative morphine consumption. Anesth Analg 89:748–752PubMed
23.
Zurück zum Zitat Chia YY, Liu K, Wang JJ, Kuo MC, Ho ST (1999) Intraoperative high dose fentanyl induces postoperative fentanyl tolerance. Can J Anaesth 46:872–877PubMed Chia YY, Liu K, Wang JJ, Kuo MC, Ho ST (1999) Intraoperative high dose fentanyl induces postoperative fentanyl tolerance. Can J Anaesth 46:872–877PubMed
24.
Zurück zum Zitat Clapham DE, Neer EJ (1997) G protein beta gamma subunits. Annu Rev Pharmacol Toxicol 37:167–203CrossRefPubMed Clapham DE, Neer EJ (1997) G protein beta gamma subunits. Annu Rev Pharmacol Toxicol 37:167–203CrossRefPubMed
25.
Zurück zum Zitat Clark JD (2002) Comment on: Doverty et al., Hyperalgesic responses in methadone maintenance patients. Pain 99:608–609CrossRefPubMed Clark JD (2002) Comment on: Doverty et al., Hyperalgesic responses in methadone maintenance patients. Pain 99:608–609CrossRefPubMed
26.
Zurück zum Zitat Colpaert FC (1996) System theory of pain and of opiate analgesia: No tolerance to opiates. Pharmacol Rev 48:355–402PubMed Colpaert FC (1996) System theory of pain and of opiate analgesia: No tolerance to opiates. Pharmacol Rev 48:355–402PubMed
27.
Zurück zum Zitat Compton P, Charuvastra VC, Ling W (2001) Pain intolerance in opioid-maintained former opiate addicts: effect of long-acting maintenance agent. Drug Alcohol Depend 63:139–146 Compton P, Charuvastra VC, Ling W (2001) Pain intolerance in opioid-maintained former opiate addicts: effect of long-acting maintenance agent. Drug Alcohol Depend 63:139–146
28.
Zurück zum Zitat Connor M, Christie MJ (1999) Opiod receptor signalling mechanisms. Clin Exp Pharmacol Physiol 26:493–499CrossRefPubMed Connor M, Christie MJ (1999) Opiod receptor signalling mechanisms. Clin Exp Pharmacol Physiol 26:493–499CrossRefPubMed
29.
Zurück zum Zitat Cortinez LI, Brandes V, Munoz HR, Guerrero ME, Mur M (2001) No clinical evidence of acute opioid tolerance after remifentanil-based anaesthesia. Br J Anaesth 87:866–869 Cortinez LI, Brandes V, Munoz HR, Guerrero ME, Mur M (2001) No clinical evidence of acute opioid tolerance after remifentanil-based anaesthesia. Br J Anaesth 87:866–869
30.
Zurück zum Zitat Crain SM, Shen KF (1998) Modulation of opioid analgesia, tolerance and dependence by Gs-coupled, GM1 ganglioside-regulated opioid receptor functions. Trends Pharmacol Sci 19:358–365CrossRefPubMed Crain SM, Shen KF (1998) Modulation of opioid analgesia, tolerance and dependence by Gs-coupled, GM1 ganglioside-regulated opioid receptor functions. Trends Pharmacol Sci 19:358–365CrossRefPubMed
31.
Zurück zum Zitat Dahl JB, Rosenberg J, Dirkes WE, Morgensen T, Kehlet H (1990) Prevention of postoperative pain by balanced analgesia. Br J Anaesth 64:518–520PubMed Dahl JB, Rosenberg J, Dirkes WE, Morgensen T, Kehlet H (1990) Prevention of postoperative pain by balanced analgesia. Br J Anaesth 64:518–520PubMed
32.
Zurück zum Zitat Davis AM, Inturrisi CE (1999) d-Methadone blocks morphine tolerance and N-methyl-D-aspartate-induced hyperalgesia. J Pharmacol Exp Ther 289:1048–1053PubMed Davis AM, Inturrisi CE (1999) d-Methadone blocks morphine tolerance and N-methyl-D-aspartate-induced hyperalgesia. J Pharmacol Exp Ther 289:1048–1053PubMed
33.
Zurück zum Zitat Devillers JP, Labrouche SA, Castes E, Simonnet G (1995) Release of neuropeptide FF, an anti-opioid peptide, in rat spinal cord slices is voltage- and Ca(2+)-sensitive: possible involvement of P-type Ca2+ channels. J Neurochem 64:1567–1575PubMed Devillers JP, Labrouche SA, Castes E, Simonnet G (1995) Release of neuropeptide FF, an anti-opioid peptide, in rat spinal cord slices is voltage- and Ca(2+)-sensitive: possible involvement of P-type Ca2+ channels. J Neurochem 64:1567–1575PubMed
34.
Zurück zum Zitat Dickenson AH (1995) Spinal cord pharmacology of pain. Br J Anaesth 75:193–200PubMed Dickenson AH (1995) Spinal cord pharmacology of pain. Br J Anaesth 75:193–200PubMed
35.
Zurück zum Zitat Dickenson AH, Chapman V, Green GM (1997) The pharmacology of excitatory and inhibitory amino acid-mediated events in the transmission and modulation of pain in the spinal cord. Gen Pharmacol 28:633–638 Dickenson AH, Chapman V, Green GM (1997) The pharmacology of excitatory and inhibitory amino acid-mediated events in the transmission and modulation of pain in the spinal cord. Gen Pharmacol 28:633–638
36.
Zurück zum Zitat Doverty M, Somogyi AA, White JM, Bochner F, Beare CH, Menelaou A, Ling W (2001) Methadone maintenance patients are cross-tolerant to the antinociceptive effects of morphine. Pain 93:155–163CrossRefPubMed Doverty M, Somogyi AA, White JM, Bochner F, Beare CH, Menelaou A, Ling W (2001) Methadone maintenance patients are cross-tolerant to the antinociceptive effects of morphine. Pain 93:155–163CrossRefPubMed
37.
Zurück zum Zitat Doverty M, White JM, Somogyi AA, Bochner F, Ali R, Ling W (2001) Hyperalgesic responses in methadone maintenance patients. Pain 90:91–96CrossRefPubMed Doverty M, White JM, Somogyi AA, Bochner F, Ali R, Ling W (2001) Hyperalgesic responses in methadone maintenance patients. Pain 90:91–96CrossRefPubMed
38.
Zurück zum Zitat Duttaroy A, Yoburn BC (1995) The effects of intrinsic efficacy on opioid tolerance. Anesthesiology 82:1226–1236PubMed Duttaroy A, Yoburn BC (1995) The effects of intrinsic efficacy on opioid tolerance. Anesthesiology 82:1226–1236PubMed
39.
Zurück zum Zitat Fairbanks CA, Wilcox GL (2000) Spinal plasticity of acute opioid tolerance. J Biomed Sci 7:200–212CrossRefPubMed Fairbanks CA, Wilcox GL (2000) Spinal plasticity of acute opioid tolerance. J Biomed Sci 7:200–212CrossRefPubMed
40.
Zurück zum Zitat Fields HL, Heinricher MM, Mason P (1991) Neurotransmitters in nociceptive modulatory circuits. Annu Rev Neurosci 14:219–245PubMed Fields HL, Heinricher MM, Mason P (1991) Neurotransmitters in nociceptive modulatory circuits. Annu Rev Neurosci 14:219–245PubMed
41.
Zurück zum Zitat Freye E, Latasch L (2003) Toleranzentwicklung unter Opioidgabe—Molekulare Mechanismen und klinische Bedeutung. Anasthesiol Intensivmed Notfallmed Schmerzther 38:14–26 Freye E, Latasch L (2003) Toleranzentwicklung unter Opioidgabe—Molekulare Mechanismen und klinische Bedeutung. Anasthesiol Intensivmed Notfallmed Schmerzther 38:14–26
42.
Zurück zum Zitat Gardell LR, Wang R, Burgess SE et al. (2002) Sustained morphine exposure induces a spinal dynorphin-dependent enhancement of excitatory transmitter release from primary afferent fibers. J Neurosci 22:6747–6755PubMed Gardell LR, Wang R, Burgess SE et al. (2002) Sustained morphine exposure induces a spinal dynorphin-dependent enhancement of excitatory transmitter release from primary afferent fibers. J Neurosci 22:6747–6755PubMed
43.
Zurück zum Zitat Goldstein A, Tachibana S, Lowney LI, Hunkapiller M, Hood L (1979) Dynorphin-(1–13), an extraordinarily potent opioid peptide. Proc Natl Acad Sci U S A 76:6666–6670PubMed Goldstein A, Tachibana S, Lowney LI, Hunkapiller M, Hood L (1979) Dynorphin-(1–13), an extraordinarily potent opioid peptide. Proc Natl Acad Sci U S A 76:6666–6670PubMed
44.
Zurück zum Zitat Gorman AL, Elliott KJ, Inturrisi CE (1997) The d- and l-isomers of methadone bind to the non-competitive site on the N-methyl-D-aspartate (NMDA) receptor in rat forebrain and spinal cord. Neurosci Lett 223:1–4CrossRefPubMed Gorman AL, Elliott KJ, Inturrisi CE (1997) The d- and l-isomers of methadone bind to the non-competitive site on the N-methyl-D-aspartate (NMDA) receptor in rat forebrain and spinal cord. Neurosci Lett 223:1–4CrossRefPubMed
45.
Zurück zum Zitat Gouarderes C, Tafani JA, Meunier JC, Jhamandas K, Zajac JM (1999) Nociceptin receptors in the rat spinal cord during morphine tolerance. Brain Res 838:85–94CrossRefPubMed Gouarderes C, Tafani JA, Meunier JC, Jhamandas K, Zajac JM (1999) Nociceptin receptors in the rat spinal cord during morphine tolerance. Brain Res 838:85–94CrossRefPubMed
46.
Zurück zum Zitat Gowing LR, Farrell M, Ali RL, White JM (2002) Alpha2-adrenergic agonists in opioid withdrawal. Addiction 97:49–58CrossRefPubMed Gowing LR, Farrell M, Ali RL, White JM (2002) Alpha2-adrenergic agonists in opioid withdrawal. Addiction 97:49–58CrossRefPubMed
47.
Zurück zum Zitat Guignard B, Bossard AE, Coste C et al. (2000) Acute opioid tolerance: intraoperative remifentanil increases postoperative pain and morphine requirement. Anesthesiology 93:409–417PubMed Guignard B, Bossard AE, Coste C et al. (2000) Acute opioid tolerance: intraoperative remifentanil increases postoperative pain and morphine requirement. Anesthesiology 93:409–417PubMed
48.
Zurück zum Zitat Guignard B, Coste C, Costes H et al. (2002) Supplementing desflurane-remifentanil anesthesia with small-dose ketamine reduces perioperative opioid analgesic requirements. Anesth Analg 95:103–108PubMed Guignard B, Coste C, Costes H et al. (2002) Supplementing desflurane-remifentanil anesthesia with small-dose ketamine reduces perioperative opioid analgesic requirements. Anesth Analg 95:103–108PubMed
49.
Zurück zum Zitat Gustorff B, Felleiter P, Nahlik G, Brannath W, Hoerauf KH, Spacek A, Kress HG (2001) The effect of remifentanil on the heat pain threshold in volunteers. Anesth Analg 92:369–374PubMed Gustorff B, Felleiter P, Nahlik G, Brannath W, Hoerauf KH, Spacek A, Kress HG (2001) The effect of remifentanil on the heat pain threshold in volunteers. Anesth Analg 92:369–374PubMed
50.
Zurück zum Zitat Gustorff B, Nahlik G, Hoerauf KH, Kress HG (2002) The absence of acute tolerance during remifentanil infusion in volunteers. Anesth Analg 94:1223–1228 Gustorff B, Nahlik G, Hoerauf KH, Kress HG (2002) The absence of acute tolerance during remifentanil infusion in volunteers. Anesth Analg 94:1223–1228
51.
Zurück zum Zitat He L, Fong J, Zastrow M von (2002) Regulation of opioid receptor trafficking and morphine tolerance by receptor oligomerization. Cell 108:271–282PubMed He L, Fong J, Zastrow M von (2002) Regulation of opioid receptor trafficking and morphine tolerance by receptor oligomerization. Cell 108:271–282PubMed
52.
Zurück zum Zitat Heinricher MM, Morgan MM, Tortorici V, Fields HL (1994) Disinhibition of off-cells and antinociception produced by an opioid action within the rostral ventromedial medulla. Neuroscience 63:279–288 Heinricher MM, Morgan MM, Tortorici V, Fields HL (1994) Disinhibition of off-cells and antinociception produced by an opioid action within the rostral ventromedial medulla. Neuroscience 63:279–288
53.
Zurück zum Zitat Heinricher MM, McGaraughty S, Grandy DK (1997) Circuitry underlying antiopioid actions of orphanin FQ in the rostral ventromedial medulla. J Neurophysiol 78:3351–3358PubMed Heinricher MM, McGaraughty S, Grandy DK (1997) Circuitry underlying antiopioid actions of orphanin FQ in the rostral ventromedial medulla. J Neurophysiol 78:3351–3358PubMed
54.
Zurück zum Zitat Heinricher MM, McGaraughty S, Tortorici V (2001) Circuitry underlying antiopioid actions of cholecystokinin within the rostral ventromedial medulla. J Neurophysiol 85:280–286PubMed Heinricher MM, McGaraughty S, Tortorici V (2001) Circuitry underlying antiopioid actions of cholecystokinin within the rostral ventromedial medulla. J Neurophysiol 85:280–286PubMed
55.
Zurück zum Zitat Hemstapat K, Monteith GR, Smith D, Smith MT (2003) Morphine-3-glucuronide’s neuro-excitatory effects are mediated via indirect activation of N-methyl-D-aspartic acid receptors: mechanistic studies in embryonic cultured hippocampal neurones. Anesth Analg 97:494–505PubMed Hemstapat K, Monteith GR, Smith D, Smith MT (2003) Morphine-3-glucuronide’s neuro-excitatory effects are mediated via indirect activation of N-methyl-D-aspartic acid receptors: mechanistic studies in embryonic cultured hippocampal neurones. Anesth Analg 97:494–505PubMed
56.
Zurück zum Zitat Hood DD, Curry R, Eisenach JC (2003) Intravenous remifentanil produces withdrawal hyperalgesia in volunteers with capsaicin-induced hyperalgesia. Anesth Analg 97:810–815PubMed Hood DD, Curry R, Eisenach JC (2003) Intravenous remifentanil produces withdrawal hyperalgesia in volunteers with capsaicin-induced hyperalgesia. Anesth Analg 97:810–815PubMed
57.
Zurück zum Zitat Jordan BA, Devi LA (1999) G-protein coupled receptor heterodimerization modulates receptor function. Nature 399:697–700PubMed Jordan BA, Devi LA (1999) G-protein coupled receptor heterodimerization modulates receptor function. Nature 399:697–700PubMed
58.
Zurück zum Zitat Joshi W, Connelly NR, Reuben SS, Wolckenhaar M, Thakkar N (2003) An evaluation of the safety and efficacy of administering rofecoxib for postoperative pain management. Anesth Analg 97:35–38 Joshi W, Connelly NR, Reuben SS, Wolckenhaar M, Thakkar N (2003) An evaluation of the safety and efficacy of administering rofecoxib for postoperative pain management. Anesth Analg 97:35–38
59.
Zurück zum Zitat Kaplan H, Fields HL (1991) Hyperalgesia during acute opioid abstinence: evidence for a nociceptive facilitating function of the rostral ventromedial medulla. J Neurosci 11:1433–1439PubMed Kaplan H, Fields HL (1991) Hyperalgesia during acute opioid abstinence: evidence for a nociceptive facilitating function of the rostral ventromedial medulla. J Neurosci 11:1433–1439PubMed
60.
Zurück zum Zitat Katz NP (2000) Morphidex (MS:DM) double-blind, multiple-dose studies in chronic pain patients. J Pain Symptom Manage 19 [Suppl 1]:37–41 Katz NP (2000) Morphidex (MS:DM) double-blind, multiple-dose studies in chronic pain patients. J Pain Symptom Manage 19 [Suppl 1]:37–41
61.
Zurück zum Zitat Kehlet H (1997) Multimodal approach to control postoperative pathophysiology and rehabilitation. Br J Anaesth 708:606–617 Kehlet H (1997) Multimodal approach to control postoperative pathophysiology and rehabilitation. Br J Anaesth 708:606–617
62.
Zurück zum Zitat Kehlet H, Dahl JB (1993) The value of multimodal or balanced analgesia in postoperative pain treatment. Anesth Analg 77:1048–1056PubMed Kehlet H, Dahl JB (1993) The value of multimodal or balanced analgesia in postoperative pain treatment. Anesth Analg 77:1048–1056PubMed
63.
Zurück zum Zitat Keith DE, Anton B, Murray SR et al. (1998) mu-Opioid receptor internalization: opiate drugs have differential effects on a conserved endocytic mechanism in vitro and in the mammalian brain. Mol Pharmacol 53:377–384PubMed Keith DE, Anton B, Murray SR et al. (1998) mu-Opioid receptor internalization: opiate drugs have differential effects on a conserved endocytic mechanism in vitro and in the mammalian brain. Mol Pharmacol 53:377–384PubMed
64.
Zurück zum Zitat Kest B, Sarton E, Dahan A (2000) Gender differences in opioid-mediated analgesia: animal and human studies. Anesthesiology 93:539–547PubMed Kest B, Sarton E, Dahan A (2000) Gender differences in opioid-mediated analgesia: animal and human studies. Anesthesiology 93:539–547PubMed
65.
Zurück zum Zitat Kieffer BL, Evans CJ (2002) Opioid tolerance—In search of the holy grail. Cell 108:587–590PubMed Kieffer BL, Evans CJ (2002) Opioid tolerance—In search of the holy grail. Cell 108:587–590PubMed
66.
Zurück zum Zitat Kissin I, Brown PT, Bradley EL Jr (1992) Does midazolam inhibit the development of acute tolerance to the analgesic effect of alfentanil? Life Sci 52:55–60 Kissin I, Brown PT, Bradley EL Jr (1992) Does midazolam inhibit the development of acute tolerance to the analgesic effect of alfentanil? Life Sci 52:55–60
67.
Zurück zum Zitat Kissin I, Lee SS, Arthur GR, Bradley EL Jr (1996) Time course characteristics of acute tolerance development to continuously infused alfentanil in rats. Anesth Analg 83:600–605PubMed Kissin I, Lee SS, Arthur GR, Bradley EL Jr (1996) Time course characteristics of acute tolerance development to continuously infused alfentanil in rats. Anesth Analg 83:600–605PubMed
68.
Zurück zum Zitat Kissin I, Bright CA, Bradley EL Jr (2000) Acute tolerance to continuously infused alfentanil: the role of cholecystokinin and N-methyl-D-aspartate-nitric oxide systems. Anesth Analg 91:110–116PubMed Kissin I, Bright CA, Bradley EL Jr (2000) Acute tolerance to continuously infused alfentanil: the role of cholecystokinin and N-methyl-D-aspartate-nitric oxide systems. Anesth Analg 91:110–116PubMed
69.
Zurück zum Zitat Kissin I, Bright CA, Bradley EL Jr (2001) Can inflammatory pain prevent the development of acute tolerance to alfentanil? Anesth Analg 92:1296–1300PubMed Kissin I, Bright CA, Bradley EL Jr (2001) Can inflammatory pain prevent the development of acute tolerance to alfentanil? Anesth Analg 92:1296–1300PubMed
70.
Zurück zum Zitat Koch T, Schulz S, Pfeiffer M, Klutzny M, Schröder H, Kahl E, Höllt V (2001) C-terminal splice variants of the mouse mu-opioid receptor differ in morphine-induced internalization and receptor resensitization. J Biol Chem 276:31408–31414CrossRefPubMed Koch T, Schulz S, Pfeiffer M, Klutzny M, Schröder H, Kahl E, Höllt V (2001) C-terminal splice variants of the mouse mu-opioid receptor differ in morphine-induced internalization and receptor resensitization. J Biol Chem 276:31408–31414CrossRefPubMed
71.
Zurück zum Zitat Kock MF de, Pichon G, Scholtes JL (1992) Intraoperative clonidine enhances postoperative morphine patient-controlled analgesia. Can J Anaesth 39:537–544PubMed Kock MF de, Pichon G, Scholtes JL (1992) Intraoperative clonidine enhances postoperative morphine patient-controlled analgesia. Can J Anaesth 39:537–544PubMed
72.
Zurück zum Zitat Kolesnikov YA, Pan YX, Babey AM, Jain S, Wilson R, Pasternak GW (1997) Functionally differentiating two neuronal nitric oxide synthase isoforms through antisense mapping: evidence for opposing NO actions on morphine analgesia and tolerance. Proc Natl Acad Sci U S A 94:8220–8225 Kolesnikov YA, Pan YX, Babey AM, Jain S, Wilson R, Pasternak GW (1997) Functionally differentiating two neuronal nitric oxide synthase isoforms through antisense mapping: evidence for opposing NO actions on morphine analgesia and tolerance. Proc Natl Acad Sci U S A 94:8220–8225
73.
Zurück zum Zitat Koppert W, Dern SK, Sittl R, Albrecht S, Schuttler J, Schmelz M (2001) A new model of electrically evoked pain and hyperalgesia in human skin: the effects of intravenous alfentanil, S(+)-ketamine, and lidocaine. Anesthesiology 95:395–402CrossRefPubMed Koppert W, Dern SK, Sittl R, Albrecht S, Schuttler J, Schmelz M (2001) A new model of electrically evoked pain and hyperalgesia in human skin: the effects of intravenous alfentanil, S(+)-ketamine, and lidocaine. Anesthesiology 95:395–402CrossRefPubMed
74.
Zurück zum Zitat Koppert W, Angst MS, Alsheimer M, Sittl R, Albrecht S, Schüttler J, Schmelz M (2003) Naloxone provokes similar pain facilitation as observed after short-term infusion of remifentanil in humans. Pain 106:91–99CrossRefPubMed Koppert W, Angst MS, Alsheimer M, Sittl R, Albrecht S, Schüttler J, Schmelz M (2003) Naloxone provokes similar pain facilitation as observed after short-term infusion of remifentanil in humans. Pain 106:91–99CrossRefPubMed
75.
Zurück zum Zitat Koppert W, Sittl R, Scheuber K, Alsheimer M, Schmelz M, Schüttler J (2003) Differential modulation of remifentanil-induced analgesia and postinfusion hyperalgesia by S-ketamine and clonidine in humans. Anesthesiology 99:152–159CrossRefPubMed Koppert W, Sittl R, Scheuber K, Alsheimer M, Schmelz M, Schüttler J (2003) Differential modulation of remifentanil-induced analgesia and postinfusion hyperalgesia by S-ketamine and clonidine in humans. Anesthesiology 99:152–159CrossRefPubMed
76.
Zurück zum Zitat Larcher A, Laulin JP, Celerier E, Moal M le, Simonnet G (1998) Acute tolerance associated with a single opiate administration: involvement of N-methyl-D-aspartate-dependent pain facilitatory systems. Neuroscience 84:583–589PubMed Larcher A, Laulin JP, Celerier E, Moal M le, Simonnet G (1998) Acute tolerance associated with a single opiate administration: involvement of N-methyl-D-aspartate-dependent pain facilitatory systems. Neuroscience 84:583–589PubMed
77.
Zurück zum Zitat Laulin JP, Larcher A, Celerier E, Moal M le, Simonnet G (1998) Long-lasting increased pain sensitivity in rat following exposure to heroin for the first time. Eur J Neurosci 10:782–785PubMed Laulin JP, Larcher A, Celerier E, Moal M le, Simonnet G (1998) Long-lasting increased pain sensitivity in rat following exposure to heroin for the first time. Eur J Neurosci 10:782–785PubMed
78.
Zurück zum Zitat Laulin JP, Maurette P, Corcuff JB, Rivat C, Chauvin M, Simonnet G (2002) The role of ketamine in preventing fentanyl-induced hyperalgesia and subsequent acute morphine tolerance. Anesth Analg 94:1263–1269 Laulin JP, Maurette P, Corcuff JB, Rivat C, Chauvin M, Simonnet G (2002) The role of ketamine in preventing fentanyl-induced hyperalgesia and subsequent acute morphine tolerance. Anesth Analg 94:1263–1269
79.
Zurück zum Zitat Lauretti GR, Perez MV, Reis MP, Pereira NL (2002) Double-blind evaluation of transdermal nitroglycerine as adjuvant to oral morphine for cancer pain management. J Clin Anesth 14:83–86CrossRefPubMed Lauretti GR, Perez MV, Reis MP, Pereira NL (2002) Double-blind evaluation of transdermal nitroglycerine as adjuvant to oral morphine for cancer pain management. J Clin Anesth 14:83–86CrossRefPubMed
80.
Zurück zum Zitat Lee SC, Wang JJ, Ho ST, Tao PL (1997) Nalbuphine coadministered with morphine prevents tolerance and dependence. Anesth Analg 84:810–815PubMed Lee SC, Wang JJ, Ho ST, Tao PL (1997) Nalbuphine coadministered with morphine prevents tolerance and dependence. Anesth Analg 84:810–815PubMed
81.
Zurück zum Zitat Li X, Angst MS, Clark JD (2001) A murine model of opioid-induced hyperalgesia. Brain Res Mol Brain Res 86:56–62CrossRefPubMed Li X, Angst MS, Clark JD (2001) A murine model of opioid-induced hyperalgesia. Brain Res Mol Brain Res 86:56–62CrossRefPubMed
82.
Zurück zum Zitat Likar R, Griessinger N, Sadjak A, Sittl R (2003) Transdermales Buprenorphin für die Behandlung chronischer Tumor- und Nicht-Tumorschmerzen. Wien Med Wochenschr 153:317–322CrossRefPubMed Likar R, Griessinger N, Sadjak A, Sittl R (2003) Transdermales Buprenorphin für die Behandlung chronischer Tumor- und Nicht-Tumorschmerzen. Wien Med Wochenschr 153:317–322CrossRefPubMed
83.
Zurück zum Zitat Luger TJ, Hayashi T, Weiss CG, Hill HF (1995) The spinal potentiating effect and the supraspinal inhibitory effect of midazolam on opioid-induced analgesia. Eur J Pharmacol 275:153–162CrossRefPubMed Luger TJ, Hayashi T, Weiss CG, Hill HF (1995) The spinal potentiating effect and the supraspinal inhibitory effect of midazolam on opioid-induced analgesia. Eur J Pharmacol 275:153–162CrossRefPubMed
84.
Zurück zum Zitat Luginbühl M, Gerber A, Schnider TW, Petersen-Felix S, Arendt-Nielsen L (2003) Modulation of remifentanil-induced analgesia, hyperalgesia and tolerance by small-dose ketamine in humans. Anesth Analg 96:726–732PubMed Luginbühl M, Gerber A, Schnider TW, Petersen-Felix S, Arendt-Nielsen L (2003) Modulation of remifentanil-induced analgesia, hyperalgesia and tolerance by small-dose ketamine in humans. Anesth Analg 96:726–732PubMed
85.
Zurück zum Zitat Majeed NH, Przewlocka B, Machelska H, Przewlocki R (1994) Inhibition of nitric oxide synthetase attenuates the development of morphine tolerance and dependence in mice. Neuropharmacology 32:189–192CrossRef Majeed NH, Przewlocka B, Machelska H, Przewlocki R (1994) Inhibition of nitric oxide synthetase attenuates the development of morphine tolerance and dependence in mice. Neuropharmacology 32:189–192CrossRef
86.
Zurück zum Zitat Malmberg AB, Yaksh TL (1992) Hyperalgesia mediated by spinal glutamate or substance P receptor blocked by spinal cyclooxygenase inhibition. Science 257:1276–1279PubMed Malmberg AB, Yaksh TL (1992) Hyperalgesia mediated by spinal glutamate or substance P receptor blocked by spinal cyclooxygenase inhibition. Science 257:1276–1279PubMed
87.
Zurück zum Zitat Manning B, Mao J, Frenk H, Price DD, Mayer DJ (1996) Continuous co-administration of dextromethorphan or MK-801 with morphine: attenuation of morphine dependence and naloxone-reversible attenuation of morphine tolerance. Pain 67:79–88CrossRefPubMed Manning B, Mao J, Frenk H, Price DD, Mayer DJ (1996) Continuous co-administration of dextromethorphan or MK-801 with morphine: attenuation of morphine dependence and naloxone-reversible attenuation of morphine tolerance. Pain 67:79–88CrossRefPubMed
88.
Zurück zum Zitat Mao J, Price DD, Caruso F, Mayer DJ (1996) Oral administration of dextromethorphan prevents the development of morphine tolerance and dependence in rats. Pain 67:361–368CrossRefPubMed Mao J, Price DD, Caruso F, Mayer DJ (1996) Oral administration of dextromethorphan prevents the development of morphine tolerance and dependence in rats. Pain 67:361–368CrossRefPubMed
89.
Zurück zum Zitat Mao J, Sung B, Ji RR, Lim G (2002) Neuronal apoptosis associated with morphine tolerance: evidence for an opioid-induced neurotoxic mechanism. J Neurosci 22:7650–7661PubMed Mao J, Sung B, Ji RR, Lim G (2002) Neuronal apoptosis associated with morphine tolerance: evidence for an opioid-induced neurotoxic mechanism. J Neurosci 22:7650–7661PubMed
90.
Zurück zum Zitat Martin WJ, Liu H, Wang H, Malmberg AB, Basbaum AI (1999) Inflammation-induced up-regulation of protein kinase Cgamma immunoreactivity in rat spinal cord correlates with enhanced nociceptive processing. Neuroscience 88:1267–1274CrossRefPubMed Martin WJ, Liu H, Wang H, Malmberg AB, Basbaum AI (1999) Inflammation-induced up-regulation of protein kinase Cgamma immunoreactivity in rat spinal cord correlates with enhanced nociceptive processing. Neuroscience 88:1267–1274CrossRefPubMed
91.
Zurück zum Zitat Mayer DJ, Mao J, Holt J, Price DD (1999) Cellular mechanisms of neuropathic pain, morphine tolerance, and their interactions. Proc Natl Acad Sci U S A 96:7731–7736CrossRefPubMed Mayer DJ, Mao J, Holt J, Price DD (1999) Cellular mechanisms of neuropathic pain, morphine tolerance, and their interactions. Proc Natl Acad Sci U S A 96:7731–7736CrossRefPubMed
92.
Zurück zum Zitat Mercadante S (1999) Opioid rotation for cancer pain: rationale and clinical aspects. Cancer 86:1856–1866CrossRefPubMed Mercadante S (1999) Opioid rotation for cancer pain: rationale and clinical aspects. Cancer 86:1856–1866CrossRefPubMed
93.
Zurück zum Zitat Moiniche S, Kehlet H, Dahl JB (2002) A qualitative and quantitative systematic review of preemptive analgesia for postoperative pain relief. Anesthesiology 96:725–741PubMed Moiniche S, Kehlet H, Dahl JB (2002) A qualitative and quantitative systematic review of preemptive analgesia for postoperative pain relief. Anesthesiology 96:725–741PubMed
94.
Zurück zum Zitat Morley JS, Watt JW, Wells JC, Miles JB, Finnegan MJ, Leng G (1993) Methadone in pain uncontrolled by morphine. Lancet 342:1243CrossRefPubMed Morley JS, Watt JW, Wells JC, Miles JB, Finnegan MJ, Leng G (1993) Methadone in pain uncontrolled by morphine. Lancet 342:1243CrossRefPubMed
95.
Zurück zum Zitat Motte RH la, Shain CN, Simone DA, Tsai EF (1991) Neurogenic hyperalgesia: psychophysical studies of underlying mechanisms. J Neurophysiol 66:190–211PubMed Motte RH la, Shain CN, Simone DA, Tsai EF (1991) Neurogenic hyperalgesia: psychophysical studies of underlying mechanisms. J Neurophysiol 66:190–211PubMed
96.
Zurück zum Zitat Ossipov MH, Lai J, Vanderah TW, Porreca F (2003) Induction of pain facilitation by sustained opioid exposure: relationship to opioid antinociceptive tolerance. Life Sci 73:783–800CrossRefPubMed Ossipov MH, Lai J, Vanderah TW, Porreca F (2003) Induction of pain facilitation by sustained opioid exposure: relationship to opioid antinociceptive tolerance. Life Sci 73:783–800CrossRefPubMed
97.
Zurück zum Zitat Przewlocki R, Machelska H, Przewlocka B (1993) Inhibition of nitric oxide synthase enhances morphine antinociception in the rat spinal cord. Life Sci 53:1–5CrossRefPubMed Przewlocki R, Machelska H, Przewlocka B (1993) Inhibition of nitric oxide synthase enhances morphine antinociception in the rat spinal cord. Life Sci 53:1–5CrossRefPubMed
98.
Zurück zum Zitat Rattan AK, Tejwani GA (1997) Effect of chronic treatment with morphine, midazolam and both together on dynorphin(1–13) levels in the rat. Brain Res 754:239–244CrossRefPubMed Rattan AK, Tejwani GA (1997) Effect of chronic treatment with morphine, midazolam and both together on dynorphin(1–13) levels in the rat. Brain Res 754:239–244CrossRefPubMed
99.
Zurück zum Zitat Reeh PW, Bayer J, Kocher L, Handwerker HO (1987) Sensitization of nociceptive cutaneous nerve fibers from the rat’s tail by noxious mechanical stimulation. Exp Brain Res 65:505–512PubMed Reeh PW, Bayer J, Kocher L, Handwerker HO (1987) Sensitization of nociceptive cutaneous nerve fibers from the rat’s tail by noxious mechanical stimulation. Exp Brain Res 65:505–512PubMed
100.
Zurück zum Zitat Reuben SS, Bhopatkar M, Maciolek H, Joshi W, Sklar J (2002) Preemptive analgesic effect of refecoxib after ambulatory arthroscopic knee surgery. Anesth Analg 94:55–59PubMed Reuben SS, Bhopatkar M, Maciolek H, Joshi W, Sklar J (2002) Preemptive analgesic effect of refecoxib after ambulatory arthroscopic knee surgery. Anesth Analg 94:55–59PubMed
101.
Zurück zum Zitat Richebe P, Rivat C, Creton C, Maurette P, Simonnet G (2003) Nitrous oxide revisited: preventive effects on fentanyl induced hyperalgesia and morphine acute tolerance. Anesthesiology 99:A940 Richebe P, Rivat C, Creton C, Maurette P, Simonnet G (2003) Nitrous oxide revisited: preventive effects on fentanyl induced hyperalgesia and morphine acute tolerance. Anesthesiology 99:A940
102.
Zurück zum Zitat Richebe P, Rivat C, Laulin JP, Maurette P, Simonnet G (2003) Acute morphine tolerance in rats operated under fentanyl. Preventive effect of ketamine. Anesthesiology 99:A941 Richebe P, Rivat C, Laulin JP, Maurette P, Simonnet G (2003) Acute morphine tolerance in rats operated under fentanyl. Preventive effect of ketamine. Anesthesiology 99:A941
103.
Zurück zum Zitat Rivat C, Laulin JP, Corcuff JB, Celerier E, Pain L, Simonnet G (2002) Fentanyl enhancement of carrageenan-induced long-lasting hyperalgesia in rats: prevention by the N-methyl-d-aspartate receptor antagonist ketamine. Anesthesiology 96:381–391CrossRefPubMed Rivat C, Laulin JP, Corcuff JB, Celerier E, Pain L, Simonnet G (2002) Fentanyl enhancement of carrageenan-induced long-lasting hyperalgesia in rats: prevention by the N-methyl-d-aspartate receptor antagonist ketamine. Anesthesiology 96:381–391CrossRefPubMed
104.
Zurück zum Zitat Rizzi A, Bigoni R, Marzola G, Guerrini R, Salvadori S, Regoli D, Calo G (2000) The nociceptin/orphanin FQ receptor antagonist, [Nphe1]NC(1–13)NH2, potentiates morphine analgesia. Neuroreport 11:2369–2372PubMed Rizzi A, Bigoni R, Marzola G, Guerrini R, Salvadori S, Regoli D, Calo G (2000) The nociceptin/orphanin FQ receptor antagonist, [Nphe1]NC(1–13)NH2, potentiates morphine analgesia. Neuroreport 11:2369–2372PubMed
105.
Zurück zum Zitat Schmelz M, Schmidt R, Ringkamp M, Forster C, Handwerker HO, Torebjörk HE (1996) Limitation of sensitization to injured parts of receptive fields in human skin C-nociceptors. Exp Brain Res 109:141–147PubMed Schmelz M, Schmidt R, Ringkamp M, Forster C, Handwerker HO, Torebjörk HE (1996) Limitation of sensitization to injured parts of receptive fields in human skin C-nociceptors. Exp Brain Res 109:141–147PubMed
106.
Zurück zum Zitat Schmid RL, Sandler AN, Katz J (1999) Use and efficacy of low-dose ketamine in the management of acute postoperative pain: a review of current techniques and outcomes. Pain 82:111–125CrossRefPubMed Schmid RL, Sandler AN, Katz J (1999) Use and efficacy of low-dose ketamine in the management of acute postoperative pain: a review of current techniques and outcomes. Pain 82:111–125CrossRefPubMed
107.
Zurück zum Zitat Schmidt R, Schmelz M, Forster C, Ringkamp M, Torebjörk HE, Handwerker HO (1995) Novel classes of responsive and unresponsive C nociceptors in human skin. J Neurosci 15:333–341PubMed Schmidt R, Schmelz M, Forster C, Ringkamp M, Torebjörk HE, Handwerker HO (1995) Novel classes of responsive and unresponsive C nociceptors in human skin. J Neurosci 15:333–341PubMed
108.
Zurück zum Zitat Schraag S, Checketts MR, Kenny GN (1999) Lack of rapid development of opioid tolerance during alfentanil and remifentanil infusions for postoperative pain. Anesth Analg 89:753–757PubMed Schraag S, Checketts MR, Kenny GN (1999) Lack of rapid development of opioid tolerance during alfentanil and remifentanil infusions for postoperative pain. Anesth Analg 89:753–757PubMed
109.
Zurück zum Zitat Simonnet G, Rivat C (2003) Opioid-induced hyperalgesia: abnormal or normal pain. Neuroreport 14:1–7CrossRefPubMed Simonnet G, Rivat C (2003) Opioid-induced hyperalgesia: abnormal or normal pain. Neuroreport 14:1–7CrossRefPubMed
110.
Zurück zum Zitat Sinatra RS, Shen QJ, Halaszynski T, Luther MA, Shaheen Y (2004) Preoperative rofecoxib oral suspension as an analgesic adjunct after lower abdominal surgery: the effects on effort-dependent pain and pulmonary function. Anesth Analg 98:135–140PubMed Sinatra RS, Shen QJ, Halaszynski T, Luther MA, Shaheen Y (2004) Preoperative rofecoxib oral suspension as an analgesic adjunct after lower abdominal surgery: the effects on effort-dependent pain and pulmonary function. Anesth Analg 98:135–140PubMed
111.
Zurück zum Zitat Sjogren P, Dragsted L, Christensen CB (1993) Myoclonic spasms during treatment with high doses of intravenous morphine in renal failure. Acta Anaesthesiol Scand 37:780–782PubMed Sjogren P, Dragsted L, Christensen CB (1993) Myoclonic spasms during treatment with high doses of intravenous morphine in renal failure. Acta Anaesthesiol Scand 37:780–782PubMed
112.
Zurück zum Zitat Sjogren P, Jensen NH, Jensen TS (1994) Disappearence of morphine-induced hyperalgesia after discontinuing or substituting with other opioid agonists. Pain 59:313–316CrossRefPubMed Sjogren P, Jensen NH, Jensen TS (1994) Disappearence of morphine-induced hyperalgesia after discontinuing or substituting with other opioid agonists. Pain 59:313–316CrossRefPubMed
113.
Zurück zum Zitat Smith MT (2000) Neuroexcitatory effects of morphine and hydromorphone: evidence implicating the 3-glucuronide metabolites. Clin Exp Pharmacol Physiol 27:524–528CrossRefPubMed Smith MT (2000) Neuroexcitatory effects of morphine and hydromorphone: evidence implicating the 3-glucuronide metabolites. Clin Exp Pharmacol Physiol 27:524–528CrossRefPubMed
114.
Zurück zum Zitat Smith MT, Watt JA, Crammond T (1990) Morphine-3-glucuronide—a potent antagonist of morphine analgesia. Life Sci 47:579–585PubMed Smith MT, Watt JA, Crammond T (1990) Morphine-3-glucuronide—a potent antagonist of morphine analgesia. Life Sci 47:579–585PubMed
115.
Zurück zum Zitat Solomon RL, Corbit JD (1974) An opponent-process theory of motivation. I. Temporal dynamics of affect. Psychol Rev 81:119–145PubMed Solomon RL, Corbit JD (1974) An opponent-process theory of motivation. I. Temporal dynamics of affect. Psychol Rev 81:119–145PubMed
116.
Zurück zum Zitat Tejwani GA, Rattan AK, Sribanditmongkol P, Sheu MJ, Zuniga J, McDonald JS (1993) Inhibition of morphine-induced tolerance and dependence by a benzodiazepine receptor agonist midazolam in the rat. Anesth Analg 76:1052–1060PubMed Tejwani GA, Rattan AK, Sribanditmongkol P, Sheu MJ, Zuniga J, McDonald JS (1993) Inhibition of morphine-induced tolerance and dependence by a benzodiazepine receptor agonist midazolam in the rat. Anesth Analg 76:1052–1060PubMed
117.
Zurück zum Zitat Vanderah TW, Ossipov MH, Lai J, Malan TP, Porreca F (2001) Mechanisms of opioid-induced pain and antinociceptive tolerance: descending facilitation and spinal dynorphin. Pain 92:5–9CrossRefPubMed Vanderah TW, Ossipov MH, Lai J, Malan TP, Porreca F (2001) Mechanisms of opioid-induced pain and antinociceptive tolerance: descending facilitation and spinal dynorphin. Pain 92:5–9CrossRefPubMed
118.
Zurück zum Zitat Vanderah TW, Suenaga NMH, Ossipov MH, Malan TP, Lai J, Porreca F (2001) Tonic descending facilitation from the rostral ventromedial medulla mediates opioid-induced abnormal pain and antinociceptive tolerance. J Neurosci 21:279–286PubMed Vanderah TW, Suenaga NMH, Ossipov MH, Malan TP, Lai J, Porreca F (2001) Tonic descending facilitation from the rostral ventromedial medulla mediates opioid-induced abnormal pain and antinociceptive tolerance. J Neurosci 21:279–286PubMed
119.
Zurück zum Zitat Vaughan CW, Ingram SL, Connor MA (1997) How opioids inhibit GABA-mediated neurotransmission. Nature 360:611–614CrossRef Vaughan CW, Ingram SL, Connor MA (1997) How opioids inhibit GABA-mediated neurotransmission. Nature 360:611–614CrossRef
120.
Zurück zum Zitat Vinik HR, Kissin I (1998) Rapid development of tolerance to analgesia during remifentanil infusion in human. Anesth Analg 86:1307–1311PubMed Vinik HR, Kissin I (1998) Rapid development of tolerance to analgesia during remifentanil infusion in human. Anesth Analg 86:1307–1311PubMed
121.
Zurück zum Zitat Watkins LR, Kinscheck IB, Mayer DJ (1984) Potentiation of opiate analgesia and apparent reversal of morphine tolerance by proglumide. Science 224:395–396PubMed Watkins LR, Kinscheck IB, Mayer DJ (1984) Potentiation of opiate analgesia and apparent reversal of morphine tolerance by proglumide. Science 224:395–396PubMed
122.
Zurück zum Zitat Weinbroum AA, Gorodetzky A, Nirkin A et al. (2002) Dextromethorphan for the reduction of immediate and late postoperative pain and morphine consumption in orthopedic oncology patients: a randomized, placebo-controlled, double-blind study. Cancer 95:1164–1170CrossRefPubMed Weinbroum AA, Gorodetzky A, Nirkin A et al. (2002) Dextromethorphan for the reduction of immediate and late postoperative pain and morphine consumption in orthopedic oncology patients: a randomized, placebo-controlled, double-blind study. Cancer 95:1164–1170CrossRefPubMed
123.
Zurück zum Zitat Whistler J, Chuang HH, Chu P, Jan LY, Zastrow M von (1999) Functional dissociation of mu opioid receptor signaling and endocytosis: implications for the biology of opiate tolerance and addiction. Neuron 23:737–746CrossRefPubMed Whistler J, Chuang HH, Chu P, Jan LY, Zastrow M von (1999) Functional dissociation of mu opioid receptor signaling and endocytosis: implications for the biology of opiate tolerance and addiction. Neuron 23:737–746CrossRefPubMed
124.
Zurück zum Zitat Wilhelm W, Dorscheid E, Schlaich N, Niederprüm P, Deller D (1999) Remifentanil zur Analgosedierung von Intensivpatienten. Anaesthesist 48:625–629CrossRefPubMed Wilhelm W, Dorscheid E, Schlaich N, Niederprüm P, Deller D (1999) Remifentanil zur Analgosedierung von Intensivpatienten. Anaesthesist 48:625–629CrossRefPubMed
125.
Zurück zum Zitat Woolf CJ, Thompson SW (1991) The induction and maintenance of central sensitization is dependent on N-methyl-D-aspartic acid receptor activation; implications for the treatment of post-injury pain hypersensitivity states. Pain 44:293–299PubMed Woolf CJ, Thompson SW (1991) The induction and maintenance of central sensitization is dependent on N-methyl-D-aspartic acid receptor activation; implications for the treatment of post-injury pain hypersensitivity states. Pain 44:293–299PubMed
126.
Zurück zum Zitat Xu XJ, Puke MJC, Verge VMK, Wiesenfeld-Hallin Z, Hughes J, Hokfelt T (1993) Up-regulation of cholecystokinin in primary sensory neurons is associated with morphine insensitivity in experimental neuropathic pain in the rat. Neurosci Lett 152:129–132CrossRefPubMed Xu XJ, Puke MJC, Verge VMK, Wiesenfeld-Hallin Z, Hughes J, Hokfelt T (1993) Up-regulation of cholecystokinin in primary sensory neurons is associated with morphine insensitivity in experimental neuropathic pain in the rat. Neurosci Lett 152:129–132CrossRefPubMed
127.
Zurück zum Zitat Yaksh TL, Harty GJ (1988) Pharmacology of the allodynia in rats evoked by high dose intrathecal morphine. J Pharmacol Exp Ther 244:501–507PubMed Yaksh TL, Harty GJ (1988) Pharmacology of the allodynia in rats evoked by high dose intrathecal morphine. J Pharmacol Exp Ther 244:501–507PubMed
128.
Zurück zum Zitat Yamamoto T, Ohno M, Ueki S (1988) A selective k-agonist, U-50,488H, blocks the development of tolerance to morphine analgesia in rats. Eur J Pharmacol 156:173–176CrossRefPubMed Yamamoto T, Ohno M, Ueki S (1988) A selective k-agonist, U-50,488H, blocks the development of tolerance to morphine analgesia in rats. Eur J Pharmacol 156:173–176CrossRefPubMed
Metadaten
Titel
Opioid-induzierte Hyperalgesie
Pathophysiologie und Klinik
verfasst von
Priv.-Doz. Dr. med. W. Koppert
Publikationsdatum
01.05.2004
Verlag
Springer-Verlag
Erschienen in
Die Anaesthesiologie / Ausgabe 5/2004
Print ISSN: 2731-6858
Elektronische ISSN: 2731-6866
DOI
https://doi.org/10.1007/s00101-004-0669-1

Weitere Artikel der Ausgabe 5/2004

Der Anaesthesist 5/2004 Zur Ausgabe

Weiterbildung · Zertifizierte Fortbildung

Blutgasanalyse

Ähnliche Überlebensraten nach Reanimation während des Transports bzw. vor Ort

29.05.2024 Reanimation im Kindesalter Nachrichten

Laut einer Studie aus den USA und Kanada scheint es bei der Reanimation von Kindern außerhalb einer Klinik keinen Unterschied für das Überleben zu machen, ob die Wiederbelebungsmaßnahmen während des Transports in die Klinik stattfinden oder vor Ort ausgeführt werden. Jedoch gibt es dabei einige Einschränkungen und eine wichtige Ausnahme.

Häusliche Gewalt in der orthopädischen Notaufnahme oft nicht erkannt

28.05.2024 Häusliche Gewalt Nachrichten

In der Notaufnahme wird die Chance, Opfer von häuslicher Gewalt zu identifizieren, von Orthopäden und Orthopädinnen offenbar zu wenig genutzt. Darauf deuten die Ergebnisse einer Fragebogenstudie an der Sahlgrenska-Universität in Schweden hin.

Fehlerkultur in der Medizin – Offenheit zählt!

28.05.2024 Fehlerkultur Podcast

Darüber reden und aus Fehlern lernen, sollte das Motto in der Medizin lauten. Und zwar nicht nur im Sinne der Patientensicherheit. Eine negative Fehlerkultur kann auch die Behandelnden ernsthaft krank machen, warnt Prof. Dr. Reinhard Strametz. Ein Plädoyer und ein Leitfaden für den offenen Umgang mit kritischen Ereignissen in Medizin und Pflege.

Mehr Frauen im OP – weniger postoperative Komplikationen

21.05.2024 Allgemeine Chirurgie Nachrichten

Ein Frauenanteil von mindestens einem Drittel im ärztlichen Op.-Team war in einer großen retrospektiven Studie aus Kanada mit einer signifikanten Reduktion der postoperativen Morbidität assoziiert.

Update AINS

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