Skip to main content
Log in

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

  • Review Article
  • Published:
CNS Drugs Aims and scope Submit manuscript

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.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Costigan M, Scholz J, Woolf CJ. Neuropathic pain: a maladaptive response of the nervous system to damage. Annu Rev Neurosci. 2009;32:1–32.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  2. Woolf CJ. Central sensitization: implications for the diagnosis and treatment of pain. Pain. 2011;152(3 Suppl):S2–15.

    Article  PubMed Central  PubMed  Google Scholar 

  3. 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.

    Article  PubMed  CAS  Google Scholar 

  4. Barkin RL. Acetaminophen, aspirin, or ibuprofen in combination analgesic products. Am J Ther. 2001;8(6):433–42.

    Article  PubMed  CAS  Google Scholar 

  5. Raffa RB, Clark-Vetri R, Tallarida RJ, Wertheimer AI. Combination strategies for pain management. Expert Opin Pharmacother. 2003;4(10):1697–708.

    Article  PubMed  CAS  Google Scholar 

  6. Gilron I, Max MB. Combination pharmacotherapy for neuropathic pain: current evidence and future directions. Expert Rev Neurother. 2005;5(6):823–30.

    Article  PubMed  CAS  Google Scholar 

  7. 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.

    Article  PubMed  CAS  Google Scholar 

  8. Lui F, Ng KF. Adjuvant analgesics in acute pain. Expert Opin Pharmacother. 2011;12(3):363–85.

    Article  PubMed  CAS  Google Scholar 

  9. Morlion B. Pharmacotherapy of low back pain: targeting nociceptive and neuropathic pain components. Curr Med Res Opin. 2011;27(1):11–33.

    Article  PubMed  CAS  Google Scholar 

  10. 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.

    PubMed Central  PubMed  Google Scholar 

  11. Lotsch J, Skarke C, Tegeder I, Geisslinger G. Drug interactions with patient-controlled analgesia. Clin Pharmacokinet. 2002;41(1):31–57.

    Article  PubMed  CAS  Google Scholar 

  12. 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  CAS  Google Scholar 

  13. 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  CAS  Google Scholar 

  14. 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.

    Article  PubMed  CAS  Google Scholar 

  15. 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.

    Article  PubMed  CAS  Google Scholar 

  16. Lambert DG. The nociceptin/orphanin FQ receptor: a target with broad therapeutic potential. Nat Rev Drug Discov. 2008;7(8):694–710.

    Article  PubMed  CAS  Google Scholar 

  17. Bannister K, Bee LA, Dickenson AH. Preclinical and early clinical investigations related to monoaminergic pain modulation. Neurotherapeutics. 2009;6(4):703–12.

    Article  PubMed  CAS  Google Scholar 

  18. Benarroch EE. Descending monoaminergic pain modulation: bidirectional control and clinical relevance. Neurology. 2008;71(3):217–21.

    Article  PubMed  Google Scholar 

  19. Millan MJ. Descending control of pain. Prog Neurobiol. 2002;66(6):355–474.

    Article  PubMed  CAS  Google Scholar 

  20. 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.

    Article  PubMed  CAS  Google Scholar 

  21. Mico JA, Ardid D, Berrocoso E, Eschalier A. Antidepressants and pain. Trends Pharmacol Sci. 2006;27(7):348–54.

    Article  PubMed  CAS  Google Scholar 

  22. 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.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  23. 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.

    Article  PubMed  CAS  Google Scholar 

  24. 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).

    Google Scholar 

  25. Klotz U. Tramadol: the impact of its pharmacokinetic and pharmacodynamic properties on the clinical management of pain. Arzneimittelforschung. 2003;53(10):681–7.

    PubMed  CAS  Google Scholar 

  26. 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.

    Article  PubMed  CAS  Google Scholar 

  27. 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  CAS  Google Scholar 

  28. 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  CAS  Google Scholar 

  29. 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.

    CAS  Google Scholar 

  30. 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.

    Article  PubMed  CAS  Google Scholar 

  31. 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. 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.

    Article  PubMed  CAS  Google Scholar 

  33. 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.

    Article  PubMed  CAS  Google Scholar 

  34. 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  CAS  Google Scholar 

  35. 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.

    Article  PubMed  CAS  Google Scholar 

  36. 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.

    Article  PubMed  CAS  Google Scholar 

  37. 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.

    Article  PubMed  CAS  Google Scholar 

  38. Carter GT, Sullivan MD. Antidepressants in pain management. Curr Opin Investig Drugs. 2002;3(3):454–8.

    PubMed  CAS  Google Scholar 

  39. Vanderah TW. Pathophysiology of pain. Med Clin North Am. 2007;91(1):1–12.

    Article  PubMed  Google Scholar 

  40. 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.

    Article  PubMed  CAS  Google Scholar 

  41. 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.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  42. 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.

    Article  PubMed  CAS  Google Scholar 

  43. 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.

    Article  PubMed  CAS  Google Scholar 

  44. 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.

    Article  PubMed  CAS  Google Scholar 

  45. 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.

    Article  PubMed  CAS  Google Scholar 

  46. 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.

    Article  PubMed  CAS  Google Scholar 

  47. 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. 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.

    Article  CAS  Google Scholar 

  49. 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.

    CAS  Google Scholar 

  50. 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.

    Article  PubMed  CAS  Google Scholar 

  51. 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.

    Google Scholar 

Ref Type: In Press

  1. Arner S, Meyerson BA. Lack of analgesic effect of opioids on neuropathic and idiopathic forms of pain. Pain. 1988;33(1):11–23.

    Article  PubMed  CAS  Google Scholar 

  2. 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.

    Article  PubMed  CAS  Google Scholar 

  3. 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.

    Article  PubMed  CAS  Google Scholar 

  4. 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.

    Article  PubMed  CAS  Google Scholar 

  5. 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.

    Article  PubMed  CAS  Google Scholar 

  6. 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.

    Article  PubMed  CAS  Google Scholar 

  7. 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.

    Article  PubMed  CAS  Google Scholar 

  8. 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.

  9. 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.

    Article  PubMed  CAS  Google Scholar 

  10. 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.

    Article  PubMed  Google Scholar 

  11. 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.

    Article  PubMed  CAS  Google Scholar 

  12. 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.

  13. 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.

  14. 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).

  15. Williams JT, Christie MJ, Manzoni O. Cellular and synaptic adaptations mediating opioid dependence. Physiol Rev. 2001;81(1):299–343.

    PubMed  CAS  Google Scholar 

  16. Aghajanian GK. Tolerance of locus coeruleus neurones to morphine and suppression of withdrawal response by clonidine. Nature. 1978;276(5684):186–8.

    Article  PubMed  CAS  Google Scholar 

  17. 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.

    Article  PubMed  CAS  Google Scholar 

  18. 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.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  19. 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.

    Article  PubMed  Google Scholar 

  20. 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.

  21. 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.

    Article  PubMed  CAS  Google Scholar 

  22. 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.

    Article  PubMed  CAS  Google Scholar 

  23. 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.

    Article  PubMed  CAS  Google Scholar 

  24. de WH. Impulsivity as a determinant and consequence of drug use: a review of underlying processes. Addict Biol 2009;14(1):22–31.

  25. 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.

    Article  CAS  Google Scholar 

  26. 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.

    Article  CAS  Google Scholar 

  27. 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.

    Article  CAS  Google Scholar 

  28. 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.

    Article  CAS  Google Scholar 

  29. 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.

    Article  PubMed  CAS  Google Scholar 

  30. 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.

  31. 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.

    PubMed Central  PubMed  CAS  Google Scholar 

  32. 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.

    Article  PubMed  Google Scholar 

  33. 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.

    Article  PubMed  CAS  Google Scholar 

  34. 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.

    Article  PubMed  Google Scholar 

  35. 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.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

Written and funded by Grünenthal GmbH.

Conflict of interest

The authors are employees of Grünenthal GmbH, the inventor and manufacturer of tapentadol.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Thomas M. Tzschentke.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Tzschentke, T.M., Christoph, T. & Kögel, B.Y. The Mu-Opioid Receptor Agonist/Noradrenaline Reuptake Inhibition (MOR–NRI) Concept in Analgesia: The Case of Tapentadol. CNS Drugs 28, 319–329 (2014). https://doi.org/10.1007/s40263-014-0151-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40263-014-0151-9

Keywords

Navigation