Skip to main content
Top
Published in: CNS Drugs 4/2014

01-04-2014 | Review Article

The Mu-Opioid Receptor Agonist/Noradrenaline Reuptake Inhibition (MOR–NRI) Concept in Analgesia: The Case of Tapentadol

Authors: Thomas M. Tzschentke, Thomas Christoph, Babette Y. Kögel

Published in: CNS Drugs | Issue 4/2014

Login to get access

Abstract

Tapentadol is a novel, centrally-acting analgesic drug, with an analgesic efficacy comparable to that of strong opioids such as oxycodone and morphine. Its high efficacy has been demonstrated in a range of animal models of acute and chronic, nociceptive, inflammatory, and neuropathic pain as well as in clinical studies with moderate to severe pain arising from a number of different etiologies. At the same time, a favorable gastrointestinal tolerability has been demonstrated in rodents and humans, and advantages over morphine regarding tolerance development and physical dependence were shown in animal studies. Furthermore, a low level of abuse and diversion is beginning to emerge from first post-marketing data. Tapentadol acts as a μ-opioid receptor (MOR) agonist and noradrenaline reuptake inhibitor (NRI). Both mechanisms of action have been shown to contribute to the analgesic activity of tapentadol and to produce analgesia in a synergistic manner, such that relatively moderate activity at the two target sites (MOR and noradrenaline reuptake transporter) is sufficient to produce strong analgesic effects. It has been suggested that tapentadol is the first representative of a proposed new class of analgesics, MOR–NRI. This review presents the evidence that has led to this suggestion, and outlines how the pharmacology of tapentadol can explain its broad analgesic activity profile and high analgesic potency as well as its favorable tolerability.
Literature
3.
go back to reference Bottegoni G, Favia AD, Recanatini M, Cavalli A. The role of fragment-based and computational methods in polypharmacology. Drug Discov Today. 2012;17(1–2):23–34.PubMedCrossRef Bottegoni G, Favia AD, Recanatini M, Cavalli A. The role of fragment-based and computational methods in polypharmacology. Drug Discov Today. 2012;17(1–2):23–34.PubMedCrossRef
4.
go back to reference Barkin RL. Acetaminophen, aspirin, or ibuprofen in combination analgesic products. Am J Ther. 2001;8(6):433–42.PubMedCrossRef Barkin RL. Acetaminophen, aspirin, or ibuprofen in combination analgesic products. Am J Ther. 2001;8(6):433–42.PubMedCrossRef
5.
go back to reference Raffa RB, Clark-Vetri R, Tallarida RJ, Wertheimer AI. Combination strategies for pain management. Expert Opin Pharmacother. 2003;4(10):1697–708.PubMedCrossRef Raffa RB, Clark-Vetri R, Tallarida RJ, Wertheimer AI. Combination strategies for pain management. Expert Opin Pharmacother. 2003;4(10):1697–708.PubMedCrossRef
6.
go back to reference Gilron I, Max MB. Combination pharmacotherapy for neuropathic pain: current evidence and future directions. Expert Rev Neurother. 2005;5(6):823–30.PubMedCrossRef Gilron I, Max MB. Combination pharmacotherapy for neuropathic pain: current evidence and future directions. Expert Rev Neurother. 2005;5(6):823–30.PubMedCrossRef
7.
go back to reference Vorobeychik Y, Gordin V, Mao J, Chen L. Combination therapy for neuropathic pain: a review of current evidence. CNS Drugs. 2011;25(12):1023–34.PubMedCrossRef Vorobeychik Y, Gordin V, Mao J, Chen L. Combination therapy for neuropathic pain: a review of current evidence. CNS Drugs. 2011;25(12):1023–34.PubMedCrossRef
8.
9.
go back to reference Morlion B. Pharmacotherapy of low back pain: targeting nociceptive and neuropathic pain components. Curr Med Res Opin. 2011;27(1):11–33.PubMedCrossRef Morlion B. Pharmacotherapy of low back pain: targeting nociceptive and neuropathic pain components. Curr Med Res Opin. 2011;27(1):11–33.PubMedCrossRef
10.
go back to reference Romano CL, Romano D, Lacerenza M. Antineuropathic and antinociceptive drugs combination in patients with chronic low back pain: a systematic review. Pain Res Treat. 2012;2012:154781.PubMedCentralPubMed Romano CL, Romano D, Lacerenza M. Antineuropathic and antinociceptive drugs combination in patients with chronic low back pain: a systematic review. Pain Res Treat. 2012;2012:154781.PubMedCentralPubMed
11.
go back to reference Lotsch J, Skarke C, Tegeder I, Geisslinger G. Drug interactions with patient-controlled analgesia. Clin Pharmacokinet. 2002;41(1):31–57.PubMedCrossRef Lotsch J, Skarke C, Tegeder I, Geisslinger G. Drug interactions with patient-controlled analgesia. Clin Pharmacokinet. 2002;41(1):31–57.PubMedCrossRef
12.
go back to reference Ossipov MH, Malseed RT, Goldstein FJ. Augmentation of central and peripheral morphine analgesia by desipramine. Arch Int Pharmacodyn Ther. 1982;259(2):222–9.PubMed Ossipov MH, Malseed RT, Goldstein FJ. Augmentation of central and peripheral morphine analgesia by desipramine. Arch Int Pharmacodyn Ther. 1982;259(2):222–9.PubMed
13.
go back to reference Ossipov MH, Harris S, Lloyd P, Messineo E. An isobolographic analysis of the antinociceptive effect of systemically and intrathecally administered combinations of clonidine and opiates. J Pharmacol Exp Ther. 1990;255(3):1107–16.PubMed Ossipov MH, Harris S, Lloyd P, Messineo E. An isobolographic analysis of the antinociceptive effect of systemically and intrathecally administered combinations of clonidine and opiates. J Pharmacol Exp Ther. 1990;255(3):1107–16.PubMed
14.
go back to reference Ossipov MH, Lopez Y, Bian D, Nichols ML, Porreca F. Synergistic antinociceptive interactions of morphine and clonidine in rats with nerve-ligation injury. Anesthesiology. 1997;86(1):196–204.PubMedCrossRef Ossipov MH, Lopez Y, Bian D, Nichols ML, Porreca F. Synergistic antinociceptive interactions of morphine and clonidine in rats with nerve-ligation injury. Anesthesiology. 1997;86(1):196–204.PubMedCrossRef
15.
go back to reference Eisenach JC, D’Angelo R, Taylor C, Hood DD. An isobolographic study of epidural clonidine and fentanyl after cesarean section. Anesth Analg. 1994;79(2):285–90.PubMedCrossRef Eisenach JC, D’Angelo R, Taylor C, Hood DD. An isobolographic study of epidural clonidine and fentanyl after cesarean section. Anesth Analg. 1994;79(2):285–90.PubMedCrossRef
16.
go back to reference Lambert DG. The nociceptin/orphanin FQ receptor: a target with broad therapeutic potential. Nat Rev Drug Discov. 2008;7(8):694–710.PubMedCrossRef Lambert DG. The nociceptin/orphanin FQ receptor: a target with broad therapeutic potential. Nat Rev Drug Discov. 2008;7(8):694–710.PubMedCrossRef
17.
go back to reference Bannister K, Bee LA, Dickenson AH. Preclinical and early clinical investigations related to monoaminergic pain modulation. Neurotherapeutics. 2009;6(4):703–12.PubMedCrossRef Bannister K, Bee LA, Dickenson AH. Preclinical and early clinical investigations related to monoaminergic pain modulation. Neurotherapeutics. 2009;6(4):703–12.PubMedCrossRef
18.
go back to reference Benarroch EE. Descending monoaminergic pain modulation: bidirectional control and clinical relevance. Neurology. 2008;71(3):217–21.PubMedCrossRef Benarroch EE. Descending monoaminergic pain modulation: bidirectional control and clinical relevance. Neurology. 2008;71(3):217–21.PubMedCrossRef
20.
go back to reference Suzuki R, Rygh LJ, Dickenson AH. Bad news from the brain: descending 5-HT pathways that control spinal pain processing. Trends Pharmacol Sci. 2004;25(12):613–7.PubMedCrossRef Suzuki R, Rygh LJ, Dickenson AH. Bad news from the brain: descending 5-HT pathways that control spinal pain processing. Trends Pharmacol Sci. 2004;25(12):613–7.PubMedCrossRef
21.
go back to reference Mico JA, Ardid D, Berrocoso E, Eschalier A. Antidepressants and pain. Trends Pharmacol Sci. 2006;27(7):348–54.PubMedCrossRef Mico JA, Ardid D, Berrocoso E, Eschalier A. Antidepressants and pain. Trends Pharmacol Sci. 2006;27(7):348–54.PubMedCrossRef
22.
go back to reference Hall FS, Schwarzbaum JM, Perona MT, Templin JS, Caron MG, Lesch KP, et al. A greater role for the norepinephrine transporter than the serotonin transporter in murine nociception. Neuroscience. 2011;175:315–27.PubMedCentralPubMedCrossRef Hall FS, Schwarzbaum JM, Perona MT, Templin JS, Caron MG, Lesch KP, et al. A greater role for the norepinephrine transporter than the serotonin transporter in murine nociception. Neuroscience. 2011;175:315–27.PubMedCentralPubMedCrossRef
23.
go back to reference Pettersen VL, Zapata-Sudo G, Raimundo JM, Trachez MM, Sudo RT. The synergistic interaction between morphine and maprotiline after intrathecal injection in rats. Anesth Analg. 2009;109(4):1312–7.PubMedCrossRef Pettersen VL, Zapata-Sudo G, Raimundo JM, Trachez MM, Sudo RT. The synergistic interaction between morphine and maprotiline after intrathecal injection in rats. Anesth Analg. 2009;109(4):1312–7.PubMedCrossRef
24.
go back to reference Suzuki T, Ueta K, Tamagaki S, Mashimo T. Antiallodynic and antihyperalgesic effect of milnacipran in mice with spinal nerve ligation. Anesth Analg. 2008;106(4):1309–15 (table). Suzuki T, Ueta K, Tamagaki S, Mashimo T. Antiallodynic and antihyperalgesic effect of milnacipran in mice with spinal nerve ligation. Anesth Analg. 2008;106(4):1309–15 (table).
25.
go back to reference Klotz U. Tramadol: the impact of its pharmacokinetic and pharmacodynamic properties on the clinical management of pain. Arzneimittelforschung. 2003;53(10):681–7.PubMed Klotz U. Tramadol: the impact of its pharmacokinetic and pharmacodynamic properties on the clinical management of pain. Arzneimittelforschung. 2003;53(10):681–7.PubMed
26.
go back to reference Raffa RB, Buschmann H, Christoph T, Eichenbaum G, Englberger W, Flores CM, et al. Mechanistic and functional differentiation of tapentadol and tramadol. Expert Opin Pharmacother. 2012;13(10):1437–49.PubMedCrossRef Raffa RB, Buschmann H, Christoph T, Eichenbaum G, Englberger W, Flores CM, et al. Mechanistic and functional differentiation of tapentadol and tramadol. Expert Opin Pharmacother. 2012;13(10):1437–49.PubMedCrossRef
27.
go back to reference Frink MC, Hennies HH, Englberger W, Haurand M, Wilffert B. Influence of tramadol on neurotransmitter systems of the rat brain. Arzneimittelforschung. 1996;46(11):1029–36.PubMed Frink MC, Hennies HH, Englberger W, Haurand M, Wilffert B. Influence of tramadol on neurotransmitter systems of the rat brain. Arzneimittelforschung. 1996;46(11):1029–36.PubMed
28.
go back to reference Raffa RB, Friderichs E, Reimann W, Shank RP, Codd EE, Vaught JL. Opioid and nonopioid components independently contribute to the mechanism of action of tramadol, an ‘atypical’ opioid analgesic. J Pharmacol Exp Ther. 1992;260(1):275–85.PubMed Raffa RB, Friderichs E, Reimann W, Shank RP, Codd EE, Vaught JL. Opioid and nonopioid components independently contribute to the mechanism of action of tramadol, an ‘atypical’ opioid analgesic. J Pharmacol Exp Ther. 1992;260(1):275–85.PubMed
29.
go back to reference Raffa RB, Nayak RK, Liao S, Minn FL. The mechanism(s) of action and pharmacokinetics of tramadol hydrochloride. Rev Contemp Pharmacother. 1995;6:485–97. Raffa RB, Nayak RK, Liao S, Minn FL. The mechanism(s) of action and pharmacokinetics of tramadol hydrochloride. Rev Contemp Pharmacother. 1995;6:485–97.
30.
go back to reference Oliva P, Aurilio C, Massimo F, Grella A, Maione S, Grella E, et al. The antinociceptive effect of tramadol in the formalin test is mediated by the serotonergic component. Eur J Pharmacol. 2002;445(3):179–85.PubMedCrossRef Oliva P, Aurilio C, Massimo F, Grella A, Maione S, Grella E, et al. The antinociceptive effect of tramadol in the formalin test is mediated by the serotonergic component. Eur J Pharmacol. 2002;445(3):179–85.PubMedCrossRef
31.
go back to reference Arcioni R, della RM, Romano S, Romano R, Pietropaoli P, Gasparetto A. Ondansetron inhibits the analgesic effects of tramadol: a possible 5-HT(3) spinal receptor involvement in acute pain in humans. Anesth Analg. 2002;94(6):1553–7 (table). Arcioni R, della RM, Romano S, Romano R, Pietropaoli P, Gasparetto A. Ondansetron inhibits the analgesic effects of tramadol: a possible 5-HT(3) spinal receptor involvement in acute pain in humans. Anesth Analg. 2002;94(6):1553–7 (table).
32.
go back to reference Yanarates O, Dogrul A, Yildirim V, Sahin A, Sizlan A, Seyrek M, et al. Spinal 5-HT7 receptors play an important role in the antinociceptive and antihyperalgesic effects of tramadol and its metabolite, O-desmethyltramadol, via activation of descending serotonergic pathways. Anesthesiology. 2010;112(3):696–710.PubMedCrossRef Yanarates O, Dogrul A, Yildirim V, Sahin A, Sizlan A, Seyrek M, et al. Spinal 5-HT7 receptors play an important role in the antinociceptive and antihyperalgesic effects of tramadol and its metabolite, O-desmethyltramadol, via activation of descending serotonergic pathways. Anesthesiology. 2010;112(3):696–710.PubMedCrossRef
33.
go back to reference Suzuki R, Rahman W, Hunt SP, Dickenson AH. Descending facilitatory control of mechanically evoked responses is enhanced in deep dorsal horn neurons following peripheral nerve injury. Brain Res. 2004;1019(1–2):68–76.PubMedCrossRef Suzuki R, Rahman W, Hunt SP, Dickenson AH. Descending facilitatory control of mechanically evoked responses is enhanced in deep dorsal horn neurons following peripheral nerve injury. Brain Res. 2004;1019(1–2):68–76.PubMedCrossRef
34.
go back to reference Raffa RB, Friderichs E, Reimann W, Shank RP, Codd EE, Vaught JL, et al. Complementary and synergistic antinociceptive interaction between the enantiomers of tramadol. J Pharmacol Exp Ther. 1993;267(1):331–40.PubMed Raffa RB, Friderichs E, Reimann W, Shank RP, Codd EE, Vaught JL, et al. Complementary and synergistic antinociceptive interaction between the enantiomers of tramadol. J Pharmacol Exp Ther. 1993;267(1):331–40.PubMed
35.
go back to reference Beier H, Garrido MJ, Christoph T, Kasel D, Trocóniz IF. Semi-mechanistic pharmacokinetic/pharmacodynamic modelling of the antinociceptive response in the presence of competitive antagonism: the interaction between tramadol and its active metabolite on mu-opioid agonism and monoamine reuptake inhibition, in the rat. Pharm Res. 2008;25(8):1789–97.PubMedCrossRef Beier H, Garrido MJ, Christoph T, Kasel D, Trocóniz IF. Semi-mechanistic pharmacokinetic/pharmacodynamic modelling of the antinociceptive response in the presence of competitive antagonism: the interaction between tramadol and its active metabolite on mu-opioid agonism and monoamine reuptake inhibition, in the rat. Pharm Res. 2008;25(8):1789–97.PubMedCrossRef
36.
go back to reference Grond S, Meuser T, Zech D, Hennig U, Lehmann KA. Analgesic efficacy and safety of tramadol enantiomers in comparison with the racemate: a randomised, double-blind study with gynaecological patients using intravenous patient-controlled analgesia. Pain. 1995;62(3):313–20.PubMedCrossRef Grond S, Meuser T, Zech D, Hennig U, Lehmann KA. Analgesic efficacy and safety of tramadol enantiomers in comparison with the racemate: a randomised, double-blind study with gynaecological patients using intravenous patient-controlled analgesia. Pain. 1995;62(3):313–20.PubMedCrossRef
37.
go back to reference Kalso E, Edwards JE, Moore RA, McQuay HJ. Opioids in chronic non-cancer pain: systematic review of efficacy and safety. Pain. 2004;112(3):372–80.PubMedCrossRef Kalso E, Edwards JE, Moore RA, McQuay HJ. Opioids in chronic non-cancer pain: systematic review of efficacy and safety. Pain. 2004;112(3):372–80.PubMedCrossRef
38.
go back to reference Carter GT, Sullivan MD. Antidepressants in pain management. Curr Opin Investig Drugs. 2002;3(3):454–8.PubMed Carter GT, Sullivan MD. Antidepressants in pain management. Curr Opin Investig Drugs. 2002;3(3):454–8.PubMed
40.
go back to reference Bee LA, Bannister K, Rahman W, Dickenson AH. Mu-opioid and noradrenergic alpha(2)-adrenoceptor contributions to the effects of tapentadol on spinal electrophysiological measures of nociception in nerve-injured rats. Pain. 2011;152(1):131–9.PubMedCrossRef Bee LA, Bannister K, Rahman W, Dickenson AH. Mu-opioid and noradrenergic alpha(2)-adrenoceptor contributions to the effects of tapentadol on spinal electrophysiological measures of nociception in nerve-injured rats. Pain. 2011;152(1):131–9.PubMedCrossRef
41.
go back to reference Schröder W, Tzschentke TM, Terlinden R, De VJ, Jahnel U, Christoph T, et al. Synergistic interaction between the two mechanisms of action of tapentadol in analgesia. J Pharmacol Exp Ther. 2011;337(1):312–20.PubMedCentralPubMedCrossRef Schröder W, Tzschentke TM, Terlinden R, De VJ, Jahnel U, Christoph T, et al. Synergistic interaction between the two mechanisms of action of tapentadol in analgesia. J Pharmacol Exp Ther. 2011;337(1):312–20.PubMedCentralPubMedCrossRef
42.
go back to reference Tzschentke TM, Christoph T, Kögel B, Schiene K, Hennies H-H, Englberger W, et al. (−)-(1R,2R)-3-(3-dimethylamino-1-ethyl-2-methyl-propyl)-phenol hydrochloride (tapentadol HCl): a novel μ-opioid receptor agonist/norepinephrine reuptake inhibitor with broad-spectrum analgesic properties. J Pharmacol Exp Ther. 2007;323(1):265–76.PubMedCrossRef Tzschentke TM, Christoph T, Kögel B, Schiene K, Hennies H-H, Englberger W, et al. (−)-(1R,2R)-3-(3-dimethylamino-1-ethyl-2-methyl-propyl)-phenol hydrochloride (tapentadol HCl): a novel μ-opioid receptor agonist/norepinephrine reuptake inhibitor with broad-spectrum analgesic properties. J Pharmacol Exp Ther. 2007;323(1):265–76.PubMedCrossRef
43.
go back to reference Tzschentke TM, Folgering JH, Flik G, De VJ. Tapentadol increases levels of noradrenaline in the rat spinal cord as measured by in vivo microdialysis. Neurosci Lett. 2012;507(2):151–5.PubMedCrossRef Tzschentke TM, Folgering JH, Flik G, De VJ. Tapentadol increases levels of noradrenaline in the rat spinal cord as measured by in vivo microdialysis. Neurosci Lett. 2012;507(2):151–5.PubMedCrossRef
44.
go back to reference Schroder W, Vry JD, Tzschentke TM, Jahnel U, Christoph T. Differential contribution of opioid and noradrenergic mechanisms of tapentadol in rat models of nociceptive and neuropathic pain. Eur J Pain. 2010;14(8):814–21.PubMedCrossRef Schroder W, Vry JD, Tzschentke TM, Jahnel U, Christoph T. Differential contribution of opioid and noradrenergic mechanisms of tapentadol in rat models of nociceptive and neuropathic pain. Eur J Pain. 2010;14(8):814–21.PubMedCrossRef
45.
go back to reference Schiene K, De VJ, Tzschentke TM. Antinociceptive and antihyperalgesic effects of tapentadol in animal models of inflammatory pain. J Pharmacol Exp Ther. 2011;339(2):537–44.PubMedCrossRef Schiene K, De VJ, Tzschentke TM. Antinociceptive and antihyperalgesic effects of tapentadol in animal models of inflammatory pain. J Pharmacol Exp Ther. 2011;339(2):537–44.PubMedCrossRef
46.
go back to reference Kogel B, De VJ, Tzschentke TM, Christoph T. The antinociceptive and antihyperalgesic effect of tapentadol is partially retained in OPRM1 (mu-opioid receptor) knockout mice. Neurosci Lett. 2011;491(2):104–7.PubMedCrossRef Kogel B, De VJ, Tzschentke TM, Christoph T. The antinociceptive and antihyperalgesic effect of tapentadol is partially retained in OPRM1 (mu-opioid receptor) knockout mice. Neurosci Lett. 2011;491(2):104–7.PubMedCrossRef
47.
go back to reference Meneghini V, Cuccurazzu B, Bortolotto V, Ramazzotti V, Ubezio F, Tzschentke TM, Canonico PL, Grilli M. The noradrenergic component in tapentadol action counteracts Mor-mediated adverse effects on adult neurogenesis. Mol Pharmacol. 2014 (in press). Meneghini V, Cuccurazzu B, Bortolotto V, Ramazzotti V, Ubezio F, Tzschentke TM, Canonico PL, Grilli M. The noradrenergic component in tapentadol action counteracts Mor-mediated adverse effects on adult neurogenesis. Mol Pharmacol. 2014 (in press).
48.
go back to reference Tzschentke TM, De Vry J, Terlinden R, Hennies HH, Lange C, Strassburger W, et al. Tapentadol hydrochloride: analgesic, mu-opioid receptor agonist, noradrenaline reuptake inhibitor. Drugs Future. 2006;31(12):1053–61.CrossRef Tzschentke TM, De Vry J, Terlinden R, Hennies HH, Lange C, Strassburger W, et al. Tapentadol hydrochloride: analgesic, mu-opioid receptor agonist, noradrenaline reuptake inhibitor. Drugs Future. 2006;31(12):1053–61.CrossRef
49.
go back to reference Tzschentke TM, Jahnel U, Kogel B, Christoph T, Englberger W, De VJ, et al. Tapentadol hydrochloride: a next-generation, centrally acting analgesic with two mechanisms of action in a single molecule. Drugs Today (Barc). 2009;45(7):483–96. Tzschentke TM, Jahnel U, Kogel B, Christoph T, Englberger W, De VJ, et al. Tapentadol hydrochloride: a next-generation, centrally acting analgesic with two mechanisms of action in a single molecule. Drugs Today (Barc). 2009;45(7):483–96.
50.
go back to reference Christoph T, De VJ, Tzschentke TM. Tapentadol, but not morphine, selectively inhibits disease-related thermal hyperalgesia in a mouse model of diabetic neuropathic pain. Neurosci Lett. 2010;470(2):91–4.PubMedCrossRef Christoph T, De VJ, Tzschentke TM. Tapentadol, but not morphine, selectively inhibits disease-related thermal hyperalgesia in a mouse model of diabetic neuropathic pain. Neurosci Lett. 2010;470(2):91–4.PubMedCrossRef
51.
go back to reference Christoph T, De Vry J, Schiene K, Tallarida RJ, Tzschentke TM. Synergistic antihypersensitive effects of pregabalin and tapentadol in a rat model of neuropathic pain. Eur J Pharmacol. 2011;666(1–3):72–9. Christoph T, De Vry J, Schiene K, Tallarida RJ, Tzschentke TM. Synergistic antihypersensitive effects of pregabalin and tapentadol in a rat model of neuropathic pain. Eur J Pharmacol. 2011;666(1–3):72–9.
52.
go back to reference Arner S, Meyerson BA. Lack of analgesic effect of opioids on neuropathic and idiopathic forms of pain. Pain. 1988;33(1):11–23.PubMedCrossRef Arner S, Meyerson BA. Lack of analgesic effect of opioids on neuropathic and idiopathic forms of pain. Pain. 1988;33(1):11–23.PubMedCrossRef
53.
go back to reference Xu XJ, Puke MJ, Wiesenfeld-Hallin Z. The depressive effect of intrathecal clonidine on the spinal flexor reflex is enhanced after sciatic nerve section in rats. Pain. 1992;51(2):145–51.PubMedCrossRef Xu XJ, Puke MJ, Wiesenfeld-Hallin Z. The depressive effect of intrathecal clonidine on the spinal flexor reflex is enhanced after sciatic nerve section in rats. Pain. 1992;51(2):145–51.PubMedCrossRef
54.
go back to reference Ossipov MH, Lopez Y, Nichols ML, Bian D, Porreca F. The loss of antinociceptive efficacy of spinal morphine in rats with nerve ligation injury is prevented by reducing spinal afferent drive. Neurosci Lett. 1995;199(2):87–90.PubMedCrossRef Ossipov MH, Lopez Y, Nichols ML, Bian D, Porreca F. The loss of antinociceptive efficacy of spinal morphine in rats with nerve ligation injury is prevented by reducing spinal afferent drive. Neurosci Lett. 1995;199(2):87–90.PubMedCrossRef
55.
go back to reference Portenoy RK, Foley KM, Inturrisi CE. The nature of opioid responsiveness and its implications for neuropathic pain: new hypotheses derived from studies of opioid infusions. Pain. 1990;43(3):273–86.PubMedCrossRef Portenoy RK, Foley KM, Inturrisi CE. The nature of opioid responsiveness and its implications for neuropathic pain: new hypotheses derived from studies of opioid infusions. Pain. 1990;43(3):273–86.PubMedCrossRef
56.
go back to reference Bantel C, Eisenach JC, Duflo F, Tobin JR, Childers SR. Spinal nerve ligation increases alpha2-adrenergic receptor G-protein coupling in the spinal cord. Brain Res. 2005;1038(1):76–82.PubMedCrossRef Bantel C, Eisenach JC, Duflo F, Tobin JR, Childers SR. Spinal nerve ligation increases alpha2-adrenergic receptor G-protein coupling in the spinal cord. Brain Res. 2005;1038(1):76–82.PubMedCrossRef
57.
go back to reference Tzschentke TM, Christoph T, Schroder W, Englberger W, De VJ, Jahnel U, et al. Tapentadol: with two mechanisms of action in one molecule effective against nociceptive and neuropathic pain. Preclinical overview. Schmerz. 2011;25(1):19–25.PubMedCrossRef Tzschentke TM, Christoph T, Schroder W, Englberger W, De VJ, Jahnel U, et al. Tapentadol: with two mechanisms of action in one molecule effective against nociceptive and neuropathic pain. Preclinical overview. Schmerz. 2011;25(1):19–25.PubMedCrossRef
58.
go back to reference Daniels SE, Upmalis D, Okamoto A, Lange C, Haeussler J. A randomized, double-blind, phase III study comparing multiple doses of tapentadol IR, oxycodone IR, and placebo for postoperative (bunionectomy) pain. Curr Med Res Opin. 2009;25(3):765–76.PubMedCrossRef Daniels SE, Upmalis D, Okamoto A, Lange C, Haeussler J. A randomized, double-blind, phase III study comparing multiple doses of tapentadol IR, oxycodone IR, and placebo for postoperative (bunionectomy) pain. Curr Med Res Opin. 2009;25(3):765–76.PubMedCrossRef
59.
go back to reference Afilalo M, Etropolski MS, Kuperwasser B, Kelly K, Okamoto A, Van H, I et al. Efficacy and safety of tapentadol extended release compared with oxycodone controlled release for the management of moderate to severe chronic pain related to osteoarthritis of the knee: a randomized, double-blind, placebo- and active-controlled phase III study. Clin Drug Investig. 2010;30(8):489–505. Afilalo M, Etropolski MS, Kuperwasser B, Kelly K, Okamoto A, Van H, I et al. Efficacy and safety of tapentadol extended release compared with oxycodone controlled release for the management of moderate to severe chronic pain related to osteoarthritis of the knee: a randomized, double-blind, placebo- and active-controlled phase III study. Clin Drug Investig. 2010;30(8):489–505.
60.
go back to reference Lange B, Kuperwasser B, Okamoto A, Steup A, Haufel T, Ashworth J, et al. Efficacy and safety of tapentadol prolonged release for chronic osteoarthritis pain and low back pain. Adv Ther. 2010;27(6):381–99.PubMedCrossRef Lange B, Kuperwasser B, Okamoto A, Steup A, Haufel T, Ashworth J, et al. Efficacy and safety of tapentadol prolonged release for chronic osteoarthritis pain and low back pain. Adv Ther. 2010;27(6):381–99.PubMedCrossRef
61.
go back to reference Wild JE, Grond S, Kuperwasser B, Gilbert J, McCann B, Lange B, et al. Long-term safety and tolerability of tapentadol extended release for the management of chronic low back pain or osteoarthritis pain. Pain Pract. 2010;10(5):416–27.PubMedCrossRef Wild JE, Grond S, Kuperwasser B, Gilbert J, McCann B, Lange B, et al. Long-term safety and tolerability of tapentadol extended release for the management of chronic low back pain or osteoarthritis pain. Pain Pract. 2010;10(5):416–27.PubMedCrossRef
62.
go back to reference Schwartz S, Etropolski M, Shapiro DY, Okamoto A, Lange R, Haeussler J, et al. Safety and efficacy of tapentadol ER in patients with painful diabetic peripheral neuropathy: results of a randomized-withdrawal, placebo-controlled trial. Curr Med Res Opin. 2011;27(1):151–62.PubMedCrossRef Schwartz S, Etropolski M, Shapiro DY, Okamoto A, Lange R, Haeussler J, et al. Safety and efficacy of tapentadol ER in patients with painful diabetic peripheral neuropathy: results of a randomized-withdrawal, placebo-controlled trial. Curr Med Res Opin. 2011;27(1):151–62.PubMedCrossRef
63.
go back to reference Ashworth J, Kuperwasser B, Etropolski M, Lange B, Lange R, Haufel T. Assessment of opioid withdrawal in patients treated with tapentadol prolonged release during an open-label extension study. In: Poster presented at: the Osteoarthritis Research Society International (OARSI) 2010 World Congress On Osteoarthritis; September 23–26, 2010. Brussels, Belgium; 2010. Ashworth J, Kuperwasser B, Etropolski M, Lange B, Lange R, Haufel T. Assessment of opioid withdrawal in patients treated with tapentadol prolonged release during an open-label extension study. In: Poster presented at: the Osteoarthritis Research Society International (OARSI) 2010 World Congress On Osteoarthritis; September 23–26, 2010. Brussels, Belgium; 2010.
64.
go back to reference Ashworth J, Lange B, Lange R, Okamoto A, Etropolski M, Haufel T. Pooled analysis of opioid withdrawal outcomes in phase 2/3 trials of tapentadol prolonged release. In: Poster presented at: the Annual Scientific Meeting of the British Pain Society (BPS); April 13–16, 2010. Manchester, England; 2010. Ashworth J, Lange B, Lange R, Okamoto A, Etropolski M, Haufel T. Pooled analysis of opioid withdrawal outcomes in phase 2/3 trials of tapentadol prolonged release. In: Poster presented at: the Annual Scientific Meeting of the British Pain Society (BPS); April 13–16, 2010. Manchester, England; 2010.
65.
go back to reference Cowan A, Raffa RB, Tallarida CS, Tallarida RJ, Christoph T, Schröder W, Tzschentke TM. Lack of synergistic interaction between the two mechanisms of action of tapentadol in gastrointestinal transit. Eur J Pain. 2014 (in press). Cowan A, Raffa RB, Tallarida CS, Tallarida RJ, Christoph T, Schröder W, Tzschentke TM. Lack of synergistic interaction between the two mechanisms of action of tapentadol in gastrointestinal transit. Eur J Pain. 2014 (in press).
66.
go back to reference Williams JT, Christie MJ, Manzoni O. Cellular and synaptic adaptations mediating opioid dependence. Physiol Rev. 2001;81(1):299–343.PubMed Williams JT, Christie MJ, Manzoni O. Cellular and synaptic adaptations mediating opioid dependence. Physiol Rev. 2001;81(1):299–343.PubMed
67.
go back to reference Aghajanian GK. Tolerance of locus coeruleus neurones to morphine and suppression of withdrawal response by clonidine. Nature. 1978;276(5684):186–8.PubMedCrossRef Aghajanian GK. Tolerance of locus coeruleus neurones to morphine and suppression of withdrawal response by clonidine. Nature. 1978;276(5684):186–8.PubMedCrossRef
68.
go back to reference Gold MS, Pottash AL, Sweeney DR, Kleber HD. Efficacy of clonidine in opiate withdrawal: a study of thirty patients. Drug Alcohol Depend. 1980;6(4):201–8.PubMedCrossRef Gold MS, Pottash AL, Sweeney DR, Kleber HD. Efficacy of clonidine in opiate withdrawal: a study of thirty patients. Drug Alcohol Depend. 1980;6(4):201–8.PubMedCrossRef
69.
go back to reference Cruz HG, Berton F, Sollini M, Blanchet C, Pravetoni M, Wickman K, et al. Absence and rescue of morphine withdrawal in GIRK/Kir3 knock-out mice. J Neurosci. 2008;28(15):4069–77.PubMedCentralPubMedCrossRef Cruz HG, Berton F, Sollini M, Blanchet C, Pravetoni M, Wickman K, et al. Absence and rescue of morphine withdrawal in GIRK/Kir3 knock-out mice. J Neurosci. 2008;28(15):4069–77.PubMedCentralPubMedCrossRef
70.
go back to reference Dart RC, Cicero TJ, Surratt HL, Rosenblum A, Bucher Bartelson B, Adams EH. Assessment of the abuse of tapentadol immediate release: the first 24 months. J Opioid Manag. 2012;8(6):395–402.PubMedCrossRef Dart RC, Cicero TJ, Surratt HL, Rosenblum A, Bucher Bartelson B, Adams EH. Assessment of the abuse of tapentadol immediate release: the first 24 months. J Opioid Manag. 2012;8(6):395–402.PubMedCrossRef
71.
go back to reference Dart R, Adams E, Bucher Bartelson B, Baker G, Pitner J, Vorsanger G. Trends in the non-medical use of tapentadol immediate release by college students. In: Poster presented at: the American Academy of Pain Medicine (AAPM) 28th Annual Meeting; February 23–26, 2012. Palm Springs, CA; 2012. Dart R, Adams E, Bucher Bartelson B, Baker G, Pitner J, Vorsanger G. Trends in the non-medical use of tapentadol immediate release by college students. In: Poster presented at: the American Academy of Pain Medicine (AAPM) 28th Annual Meeting; February 23–26, 2012. Palm Springs, CA; 2012.
72.
go back to reference Tzschentke TM, Magalas Z, De VJ. Effects of venlafaxine and desipramine on heroin-induced conditioned place preference in the rat. Addict Biol. 2006;11(1):64–71.PubMedCrossRef Tzschentke TM, Magalas Z, De VJ. Effects of venlafaxine and desipramine on heroin-induced conditioned place preference in the rat. Addict Biol. 2006;11(1):64–71.PubMedCrossRef
73.
go back to reference Magalas Z, De Vry J, Tzschentke TM. The serotonin/noradrenaline reuptake inhibitor venlafaxine attenuates acquisition, but not maintenance, of intravenous self-administration of heroin in rats. Eur J Pharmacol. 2005;528(1–3):103–9.PubMedCrossRef Magalas Z, De Vry J, Tzschentke TM. The serotonin/noradrenaline reuptake inhibitor venlafaxine attenuates acquisition, but not maintenance, of intravenous self-administration of heroin in rats. Eur J Pharmacol. 2005;528(1–3):103–9.PubMedCrossRef
74.
go back to reference Lu L, Su WJ, Yue W, Ge X, Su F, Pei G, et al. Attenuation of morphine dependence and withdrawal in rats by venlafaxine, a serotonin and noradrenaline reuptake inhibitor. Life Sci. 2001;69(1):37–46.PubMedCrossRef Lu L, Su WJ, Yue W, Ge X, Su F, Pei G, et al. Attenuation of morphine dependence and withdrawal in rats by venlafaxine, a serotonin and noradrenaline reuptake inhibitor. Life Sci. 2001;69(1):37–46.PubMedCrossRef
75.
go back to reference de WH. Impulsivity as a determinant and consequence of drug use: a review of underlying processes. Addict Biol 2009;14(1):22–31. de WH. Impulsivity as a determinant and consequence of drug use: a review of underlying processes. Addict Biol 2009;14(1):22–31.
76.
go back to reference Leeman RF, Potenza MN. Similarities and differences between pathological gambling and substance use disorders: a focus on impulsivity and compulsivity. Psychopharmacology (Berl). 2012;219(2):469–90.CrossRef Leeman RF, Potenza MN. Similarities and differences between pathological gambling and substance use disorders: a focus on impulsivity and compulsivity. Psychopharmacology (Berl). 2012;219(2):469–90.CrossRef
77.
go back to reference Baarendse PJ, Vanderschuren LJ. Dissociable effects of monoamine reuptake inhibitors on distinct forms of impulsive behavior in rats. Psychopharmacology (Berl). 2012;219(2):313–26.CrossRef Baarendse PJ, Vanderschuren LJ. Dissociable effects of monoamine reuptake inhibitors on distinct forms of impulsive behavior in rats. Psychopharmacology (Berl). 2012;219(2):313–26.CrossRef
78.
go back to reference Fernando AB, Economidou D, Theobald DE, Zou MF, Newman AH, Spoelder M, et al. Modulation of high impulsivity and attentional performance in rats by selective direct and indirect dopaminergic and noradrenergic receptor agonists. Psychopharmacology (Berl). 2012;219(2):341–52.CrossRef Fernando AB, Economidou D, Theobald DE, Zou MF, Newman AH, Spoelder M, et al. Modulation of high impulsivity and attentional performance in rats by selective direct and indirect dopaminergic and noradrenergic receptor agonists. Psychopharmacology (Berl). 2012;219(2):341–52.CrossRef
79.
go back to reference Robinson ES. Blockade of noradrenaline re-uptake sites improves accuracy and impulse control in rats performing a five-choice serial reaction time tasks. Psychopharmacology (Berl). 2012;219(2):303–12.CrossRef Robinson ES. Blockade of noradrenaline re-uptake sites improves accuracy and impulse control in rats performing a five-choice serial reaction time tasks. Psychopharmacology (Berl). 2012;219(2):303–12.CrossRef
80.
go back to reference Kress HG. Tapentadol and its two mechanisms of action: is there a new pharmacological class of centrally-acting analgesics on the horizon? Eur J Pain. 2010;14(8):781–3.PubMedCrossRef Kress HG. Tapentadol and its two mechanisms of action: is there a new pharmacological class of centrally-acting analgesics on the horizon? Eur J Pain. 2010;14(8):781–3.PubMedCrossRef
81.
go back to reference Fields HL, Martin JB. In: Gaul C, Endres M, Erbguth F, editors. Harrison′s neurology in clinical medicine, German ed. Berlin, Germany: ABW Wissenschaftsverlag; 2012. Fields HL, Martin JB. In: Gaul C, Endres M, Erbguth F, editors. Harrison′s neurology in clinical medicine, German ed. Berlin, Germany: ABW Wissenschaftsverlag; 2012.
82.
go back to reference Pergolizzi J, Alon E, Baron R, Bonezzi C, Dobrogowski J, Galvez R, et al. Tapentadol in the management of chronic low back pain: a novel approach to a complex condition? J Pain Res. 2011;4:203–10.PubMedCentralPubMed Pergolizzi J, Alon E, Baron R, Bonezzi C, Dobrogowski J, Galvez R, et al. Tapentadol in the management of chronic low back pain: a novel approach to a complex condition? J Pain Res. 2011;4:203–10.PubMedCentralPubMed
83.
go back to reference Pergolizzi J, Alegre C, Blake D, Alen JC, Caporali R, Casser HR, et al. Current considerations for the treatment of severe chronic pain: the potential for tapentadol. Pain Pract. 2012;12(4):290–306.PubMedCrossRef Pergolizzi J, Alegre C, Blake D, Alen JC, Caporali R, Casser HR, et al. Current considerations for the treatment of severe chronic pain: the potential for tapentadol. Pain Pract. 2012;12(4):290–306.PubMedCrossRef
84.
go back to reference Steigerwald I, Muller M, Davies A, Samper D, Sabatowski R, Baron R, et al. Effectiveness and safety of tapentadol prolonged release for severe, chronic low back pain with or without a neuropathic pain component: results of an open-label, phase 3b study. Curr Med Res Opin. 2012;28(6):911–36.PubMedCrossRef Steigerwald I, Muller M, Davies A, Samper D, Sabatowski R, Baron R, et al. Effectiveness and safety of tapentadol prolonged release for severe, chronic low back pain with or without a neuropathic pain component: results of an open-label, phase 3b study. Curr Med Res Opin. 2012;28(6):911–36.PubMedCrossRef
85.
go back to reference Freynhagen R, Baron R, Gockel U, Tolle TR. painDETECT: a new screening questionnaire to identify neuropathic components in patients with back pain. Curr Med Res Opin. 2006;22(10):1911–20.PubMedCrossRef Freynhagen R, Baron R, Gockel U, Tolle TR. painDETECT: a new screening questionnaire to identify neuropathic components in patients with back pain. Curr Med Res Opin. 2006;22(10):1911–20.PubMedCrossRef
86.
go back to reference Gillen C, Haurand M, Kobelt DJ, Wnendt S. Affinity, potency and efficacy of tramadol and its metabolites at the cloned human mu-opioid receptor. Naunyn Schmiedebergs Arch Pharmacol. 2000;362(2):116–21.PubMedCrossRef Gillen C, Haurand M, Kobelt DJ, Wnendt S. Affinity, potency and efficacy of tramadol and its metabolites at the cloned human mu-opioid receptor. Naunyn Schmiedebergs Arch Pharmacol. 2000;362(2):116–21.PubMedCrossRef
Metadata
Title
The Mu-Opioid Receptor Agonist/Noradrenaline Reuptake Inhibition (MOR–NRI) Concept in Analgesia: The Case of Tapentadol
Authors
Thomas M. Tzschentke
Thomas Christoph
Babette Y. Kögel
Publication date
01-04-2014
Publisher
Springer International Publishing
Published in
CNS Drugs / Issue 4/2014
Print ISSN: 1172-7047
Electronic ISSN: 1179-1934
DOI
https://doi.org/10.1007/s40263-014-0151-9

Other articles of this Issue 4/2014

CNS Drugs 4/2014 Go to the issue