Skip to main content

Advertisement

Log in

Role of active metabolites in the use of opioids

  • Review Article
  • Published:
European Journal of Clinical Pharmacology Aims and scope Submit manuscript

Abstract

The opioid class of drugs, a large group, is mainly used for the treatment of acute and chronic persistent pain. All are eliminated from the body via metabolism involving principally CYP3A4 and the highly polymorphic CYP2D6, which markedly affects the drug’s function, and by conjugation reactions mainly by UGT2B7. In many cases, the resultant metabolites have the same pharmacological activity as the parent opioid; however in many cases, plasma metabolite concentrations are too low to make a meaningful contribution to the overall clinical effects of the parent drug. These metabolites are invariably more water soluble and require renal clearance as an important overall elimination pathway. Such metabolites have the potential to accumulate in the elderly and in those with declining renal function with resultant accumulation to a much greater extent than the parent opioid. The best known example is the accumulation of morphine-6-glucuronide from morphine. Some opioids have active metabolites but at different target sites. These are norpethidine, a neurotoxic agent, and nordextropropoxyphene, a cardiotoxic agent. Clinicians need to be aware that many opioids have active metabolites that will become therapeutically important, for example in cases of altered pathology, drug interactions and genetic polymorphisms of drug-metabolizing enzymes. Thus, dose individualisation and the avoidance of adverse effects of opioids due to the accumulation of active metabolites or lack of formation of active metabolites are important considerations when opioids are used.

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.

Institutional subscriptions

Fig. 1

Similar content being viewed by others

References

  1. Cowan A, Lewis JW, Macfarlane IR (1977) Agonist and antagonist properties of buprenorphine, a new antinociceptive agent. Brit J Pharmacol 60:537–545

    CAS  Google Scholar 

  2. Cowan A, Doxey JC, Harry EJ (1977) The animal pharmacology of buprenorphine, an oripavine analgesic agent. Br J Pharmacol 60:547–554

    PubMed  CAS  Google Scholar 

  3. Dahan A, Yassen A, Bijl H et al (2005) Comparison of the respiratory effects of intravenous buprenorphine and fentanyl in humans and rats. Br J Anaesth 94:825–834

    PubMed  CAS  Google Scholar 

  4. Dahan A, Yassen A, Romberg R et al (2006) Buprenorphine induces ceiling in respiratory depression but not in analgesia. Br J Anaesth 96:627–632

    PubMed  CAS  Google Scholar 

  5. Codd EE, Shank RP, Schupsky JJ et al (1995) Serotonin and norepinephrine uptake inhibiting activity of centrally acting analgesics: structural determinants and role in antinociception. J Pharmacol Exp Ther 274:1263–1270

    PubMed  CAS  Google Scholar 

  6. Katchman AN, McGroary KA, Kilborn MJ et al (2002) Influence of opioid agonists on cardiac human ether-a-go-go-related gene K(+) currents. J Pharmacol Exp Ther 303:688–694

    PubMed  CAS  Google Scholar 

  7. Huang P, Kehner GB, Cowan A et al (2001) Comparison of pharmacological activities of buprenorphine and norbuprenorphine: norbuprenorphine is a potent opioid agonist. J Pharmacol Exp Ther 297:688–695

    PubMed  CAS  Google Scholar 

  8. Leander JD (1987) Buprenorphine has potent kappa opioid receptor antagonist activity. Neuropharmacol 26:1445–1447

    CAS  Google Scholar 

  9. Jensen ML, Foster D, Upton R et al (2007) Comparison of cerebral pharmacokinetics of buprenorphine and norbuprenorphine in an in vivo sheep model. Xenobiotica 37:441–457

    PubMed  CAS  Google Scholar 

  10. Budd K, Collett BJ (2003) Old dog - new (ma)trix. Br J Anaesth 90:722–724

    PubMed  CAS  Google Scholar 

  11. Vachharajani NN, Shyu WC, Greene DS et al (1997) The pharmacokinetics of butorphanol and its metabolites at steady state following nasal administration in humans. Biopharm Drug Dispos 18:191–202

    PubMed  CAS  Google Scholar 

  12. Pachter IJ, Evens RP (1985) Butorphanol. Drug Alcohol Depend 14:325–338

    PubMed  CAS  Google Scholar 

  13. Gaver RC, Vasiljev M, Wong H et al (1980) Disposition of parenteral butorphanol in man. Drug Metab Dispos 8:230–235

    PubMed  CAS  Google Scholar 

  14. Ameer B, Salter FJ (1979) Drug therapy reviews: evaluation of butorphanol tartrate. Am J Hosp Pharm 36:1683–1691

    PubMed  CAS  Google Scholar 

  15. Chen ZR, Somogyi AA, Reynolds G et al (1991) Disposition and metabolism of codeine after single and chronic doses in one poor and seven extensive metabolisers. Br J Clin Pharmacol 31:381–390

    PubMed  CAS  Google Scholar 

  16. Yue QY, Hasselstrom J, Svensson JO et al (1991) Pharmacokinetics of codeine and its metabolites in Caucasian healthy volunteers: comparisons between extensive and poor hydroxylators of debrisoquine. Br J Clin Pharmacol 31:635–642

    PubMed  CAS  Google Scholar 

  17. Vree TB, Verwey-van Wissen CP (1992) Pharmacokinetics and metabolism of codeine in humans. Biopharm Drug Dispos 13:445–460

    PubMed  CAS  Google Scholar 

  18. Chen ZR, Somogyi AA, Bochner F (1988) Polymorphic O-demethylation of codeine. Lancet 2:914–915

    PubMed  CAS  Google Scholar 

  19. Sindrup S, Brøsen K (1995) The pharmacogenetics of codeine hypoalgesia. Pharmacogenetics 5:335–346

    PubMed  CAS  Google Scholar 

  20. Poulsen L, Riishede L, Brosen K et al (1998) Codeine in post-operative pain: study of the influence of sparteine phenotype and serum concentrations of morphine and morphine-6-glucuronide. Eur J Clin Pharmacol 54:451–454

    PubMed  CAS  Google Scholar 

  21. Sindrup SH, Poulsen L, Brosen K et al (1993) Are poor metabolisers of sparteine/debrisoquine less pain tolerant than extensive metabolisers? Pain 53:335–339

    PubMed  CAS  Google Scholar 

  22. Somogyi AA, Barratt DT, Coller JK (2007) Pharmacogenetics of opioids. Clin Pharmacol Ther 81:429–444

    PubMed  CAS  Google Scholar 

  23. Caraco Y, Sheller J, Wood AJ (1996) Pharmacogenetic determination of the effects of codeine and prediction of drug interactions. J Pharmacol Exp Ther 278:1165–1174

    PubMed  CAS  Google Scholar 

  24. Eckhardt K, Li S, Ammon S et al (1998) Same incidence of adverse drug events after codeine administration irrespective of the genetically determined differences in morphine formation. Pain 76:27–33

    PubMed  CAS  Google Scholar 

  25. Quiding H, Lundqvist G, Boreus LO et al (1993) Analgesic effect and plasma concentrations of codeine and morphine after two dose levels of codeine following oral surgery. Eur J Clin Pharmacol 44:319–323

    PubMed  CAS  Google Scholar 

  26. Srinivasan V, Wielbo D, Simpkins J et al (1996) Analgesic and immunomodulatory effects of codeine and codeine 6-glucuronide. Pharm Res 13:296–300

    PubMed  CAS  Google Scholar 

  27. Lötsch J, Skarke C, Schmidt H et al (2006) Evidence for morphine-independent central nervous opioid effects after administration of codeine: contribution of other codeine metabolites. Clin Pharmacol Ther 79:35–48

    PubMed  Google Scholar 

  28. Kirchheiner J, Schmidt H, Tzvetkov M et al (2007) Pharmacokinetics of codeine and its metabolite morphine in ultra-rapid metabolizers due to CYP2D6 duplication. Pharmacogenomics J 7:257–265

    PubMed  CAS  Google Scholar 

  29. Dalén P, Frengell C, Dahl M-L et al (1997) Quick onset of severe abdominal pain after codeine in an ultrarapid metabolizer of debrisoquine. Ther Drug Monit 19:543–544

    PubMed  Google Scholar 

  30. Gasche Y, Daali Y, Fathi M et al (2004) Codeine intoxication associated with ultrarapid CYP2D6 metabolism. N Engl J Med 351:2827–2831

    PubMed  CAS  Google Scholar 

  31. Koren G, Cairns J, Chitayat D et al (2006) Pharmacogenetics of morphine poisoning in a breastfed neonate of a codeine-prescribed mother. Lancet 368:704

    PubMed  Google Scholar 

  32. Leysen JE, Gommeren W, Niemegeers CJ (1983) [3H]Sufentanil, a superior ligand for mu-opiate receptors: binding properties and regional distribution in rat brain and spinal cord. Eur J Pharmacol 87:209–225

    PubMed  CAS  Google Scholar 

  33. Neil A (1984) Affinities of some common opioid analgesics towards four binding sites in mouse brain. Naunyn-Schmiedeberg’s Arch Pharmacol 328:24–29

    CAS  Google Scholar 

  34. Somogyi AA, Menelaou A, Fullston SV (2004) CYP3A4 mediates dextropropoxyphene N-demethylation to nordextropropoxyphene: human in vitro and in vivo studies and lack of CYP2D6 involvement. Xenobiotica 34:875–887

    PubMed  CAS  Google Scholar 

  35. Flanagan RJ, Johnston A, White AS et al (1989) Pharmacokinetics of dextropropoxyphene and nordextropropoxyphene in young and elderly volunteers after single and multiple dextropropoxyphene dosage. Br J Clin Pharmacol 28:463–469

    PubMed  CAS  Google Scholar 

  36. Ebert B, Andersen S, Hjeds H et al (1998) Dextropropoxyphene acts as a noncompetitive N-methyl-D-aspartate antagonist. J Pain Symptom Manage 15:269–274

    PubMed  CAS  Google Scholar 

  37. Ulens C, Daenens P, Tytgat J (1999) Norpropoxyphene-induced cardiotoxicity is associated with changes in ion-selectivity and gating of HERG currents. Cardiovasc Res 44:568–578

    PubMed  CAS  Google Scholar 

  38. Schmidt H, Vormfelde SV, Klinder K et al (2002) Affinities of dihydrocodeine and its metabolites to opioid receptors. Pharmacol Toxicol 91:57–63

    PubMed  CAS  Google Scholar 

  39. Fromm MF, Hofmann U, Griese E-U et al (1995) Dihydrocodeine: a new opioid substrate for the polymorphic CYP2D6 in humans. Clin Pharmacol Ther 58:374–382

    PubMed  CAS  Google Scholar 

  40. Mignat C, Wille U, Ziegler A (1995) Affinity profiles of morphine, codeine, dihydrocodeine and their glucuronides at opioid receptor subtypes. Life Sci 56:793–799

    PubMed  CAS  Google Scholar 

  41. Thompson CM, Wojno H, Greiner E et al (2004) Activation of G-proteins by morphine and codeine congeners: insights to the relevance of O- and N-demethylated metabolites at mu- and delta-opioid receptors. J Pharmacol Exp Ther 308:547–554

    PubMed  CAS  Google Scholar 

  42. Kirkwood LC, Nation RL, Somogyi AA (1997) Characterization of the human cytochrome P450 enzymes involved in the metabolism of dihydrocodeine. Br J Clin Pharmacol 44:549–555

    PubMed  CAS  Google Scholar 

  43. Webb JA, Rostami-Hodjegan A, Abdul-Manap R et al (2001) Contribution of dihydrocodeine and dihydromorphine to analgesia following dihydrocodeine administration in man: a PK-PD modelling analysis. Br J Clin Pharmacol 52:35–43

    PubMed  CAS  Google Scholar 

  44. Platten HP, Scheweizer E, Dilger K et al (1998) Pharmacokinetics and the pharmacodynamic action of midazolam in young and elderly patients undergoing tooth extraction. Clin Pharmacol Ther 63:552–560

    PubMed  CAS  Google Scholar 

  45. Wilder-Smith CH, Hufschmid E, Thormann W (1998) The visceral and somatic antinociceptive effects of dihydrocodeine and its metabolite, dihydromorphine. A cross-over study with extensive and quinidine-induced poor metabolizers. Br J Clin Pharmacol 45:575–581

    PubMed  CAS  Google Scholar 

  46. Magnan J, Paterson SJ, Tavani A et al (1982) The binding spectrum of nacrotic analgesic drugs with different agonist and antagonist properties. Naunyn-Schmiedeberg’s Arch Pharmacol 319:197–205

    CAS  Google Scholar 

  47. Kalvass JC, Olson ER, Cassidy MP et al (2007) Pharmacokinetics and pharmacodynamics of seven opioids in P-glycoprotein-competent mice: assessment of unbound brain EC50,u and correlation of in vitro, preclinical, and clinical data. J Pharmacol Exp Ther 323:346–355

    PubMed  CAS  Google Scholar 

  48. Battershill AJ, Keating GM (2006) Remifentanil: a review of its analgesic and sedative use in the intensive care unit. Drugs 66:365–385

    PubMed  CAS  Google Scholar 

  49. Hoke JF, Cunningham F, James MK et al (1997) Comparative pharmacokinetics and pharmacodynamics of remifentanil, its principle metabolite (GR90291) and alfentanil in dogs. J Pharmacol Exp Ther 281:226–232

    PubMed  CAS  Google Scholar 

  50. Cox EH, Langemeijer MW, Gubbens-Stibbe JM et al (1999) The comparative pharmacodynamics of remifentanil and its metabolite, GR90291, in a rat electroencephalographic model. Anesthesiology 90:535–544

    PubMed  CAS  Google Scholar 

  51. James MK, Feldman PL, Schuster SV et al (1991) Opioid receptor activity of GI 87084B, a novel ultra-short acting analgesic, in isolated tissues. J Pharmacol Exp Ther 259:712–718

    PubMed  CAS  Google Scholar 

  52. Guitton J, Buronfosse T, Desage M et al (1997) Possible involvement of multiple cytochrome P450S in fentanyl and sufentanil metabolism as opposed to alfentanil. Biochem Pharmacol 53:1613–1619

    PubMed  CAS  Google Scholar 

  53. Williams FM (1985) Clinical significance of esterases in man. Clin Pharmacokinet 10:392–403

    PubMed  CAS  Google Scholar 

  54. Lockridge O, Mottershaw-Jackson N, Eckerson HW et al (1980) Hydrolysis of diacetylmorphine (heroin) by human serum cholinesterase. J Pharmacol Exp Ther 215:1–8

    PubMed  CAS  Google Scholar 

  55. Yeh SY, McQuinn RL, Gorodetzky CW (1977) Identification of diacetylmorphine metabolites in humans. J Pharm Sci 66:201–204

    PubMed  CAS  Google Scholar 

  56. Rentsch KM, Kullak-Ublick GA, Reichel C et al (2001) Arterial and venous pharmacokinetics of intravenous heroin in subjects who are addicted to narcotics. Clin Pharmacol Ther 70:237–246

    PubMed  CAS  Google Scholar 

  57. Girardin F, Rentsch KM, Schwab MA et al (2003) Pharmacokinetics of high doses of intramuscular and oral heroin in narcotic addicts. Clin Pharmacol Ther 74:341–352

    PubMed  CAS  Google Scholar 

  58. Selley DE, Cao CC, Sexton T et al (2001) mu Opioid receptor-mediated G-protein activation by heroin metabolites: evidence for greater efficacy of 6-monoacetylmorphine compared with morphine. Biochem Pharmacol 62:447–455

    PubMed  CAS  Google Scholar 

  59. Rady JJ, Roerig SC, Fujimoto JM (1991) Heroin acts on different opioid receptors than morphine in Swiss Webster and ICR mice to produce antinociception. J Pharmacol Exp Ther 256:448–457

    PubMed  CAS  Google Scholar 

  60. Rady JJ, Aksu F, Fujimoto JM (1994) The heroin metabolite, 6-monoacetylmorphine, activates delta opioid receptors to produce antinociception in Swiss-Webster mice. J Pharmacol Exp Ther 268:1222–1231

    PubMed  CAS  Google Scholar 

  61. Rady JJ, Takemori AE, Portoghese PS et al (1994) Supraspinal delta receptor subtype activity of heroin and 6-monoacetylmorphine in Swiss Webster mice. Life Sci 55:603–609

    PubMed  CAS  Google Scholar 

  62. Cone EJ, Darwin WD, Gorodetzky CW et al (1978) Comparative metabolism of hydrocodone in man, rat, guinea pig, rabbit, and dog. Drug Metab Dispos 6:488–493

    PubMed  CAS  Google Scholar 

  63. Hutchinson MR, Menelaou A, Foster DJR et al (2003) CYP2D6 and CYP3A4 involvement in the primary oxidative metabolism of hydrocodone by human liver microsomes. Br J Clin Pharmacol 57:287–297

    Google Scholar 

  64. Otton SV, Schadel M, Cheung SW et al (1993) CYP2D6 phenotype determines the metabolic conversion of hydrocodone to hydromorphone. Clin Pharmacol Ther 54:463–472

    PubMed  CAS  Google Scholar 

  65. Susce MT, Murray-Carmichael E, de Leon J (2006) Response to hydrocodone, codeine and oxycodone in a CYP2D6 poor metabolizer. Prog Neuro-Psychopharmacol Biol Psych 30:1356–1358

    CAS  Google Scholar 

  66. Foster A, Mobley E, Wang Z (2007) Complicated pain management in a CYP450 2D6 poor metabolizer. Pain Pract 7:352–356

    PubMed  Google Scholar 

  67. Zheng M, McErlane KM, Ong MC (2002) Hydromorphone metabolites: isolation and identification from pooled urine samples of a cancer patient. Xenobiotica 32:427–439

    PubMed  CAS  Google Scholar 

  68. Baldacci A, Thormann W (2006) Capillary electrophoresis contributions to the hydromorphone metabolism in man. Electrophoresis 27:2444–2457

    PubMed  CAS  Google Scholar 

  69. Wright AW, Mather LE, Smith MT (2001) Hydromorphone-3-glucuronide: a more potent neuro-excitant than its structural analogue, morphine-3-glucuronide. Life Sci 69:409–420

    PubMed  CAS  Google Scholar 

  70. Smith MT (2000) Neuroexcitatory effects of morphine and hydromorphone: evidence implicating the 3-glucuronide metabolites. Clin Exp Pharmacol Physiol 27:524–528

    PubMed  CAS  Google Scholar 

  71. Sundstrom I, Hedeland M, Bondesson U et al (2002) Identification of glucuronide conjugates of ketobemidone and its phase I metabolites in human urine utilizing accurate mass and tandem time-of-flight mass spectrometry. J Mass Spectrom 37:414–420

    PubMed  CAS  Google Scholar 

  72. Yasar U, Annas A, Svensson JO et al (2005) Ketobemidone is a substrate for cytochrome P4502C9 and 3A4, but not for P-glycoprotein. Xenobiotica 35:785–796

    PubMed  CAS  Google Scholar 

  73. Al-Shurbaji A, Sawe J (2002) The pharmacokinetics of ketobemidone are not affected by CYP2D6 or CYP2C19 phenotype. Eur J Clin Pharmacol 57:877–881

    PubMed  CAS  Google Scholar 

  74. Kharasch ED, Whittington D, Hoffer C et al (2005) Paradoxical role of cytochrome P450 3A in the bioactivation and clinical effects of levo-alpha-acetylmethadol: importance of clinical investigations to validate in vitro drug metabolism studies. Clin Pharmacokinet 44:731–751

    PubMed  CAS  Google Scholar 

  75. Newcombe DA, Bochner F, White JM et al (2004) Evaluation of levo-alpha-acetylmethadol (LAAM) as an alternative treatment for methadone maintenance patients who regularly experience withdrawal: a pharmacokinetic and pharmacodynamic analysis. Drug Alcohol Depend 76:63–72

    PubMed  CAS  Google Scholar 

  76. Stringer M, Makin MK, Miles J et al (2000) d-Morphine, but not l-morphine has low micromolar affinity for the non-competitive N-methyl-D-aspartate site in rat forebrain. Possible clinical implications for the management of neuropathic pain. Neurosci Lett 295:21–24

    PubMed  CAS  Google Scholar 

  77. Dixon R, Crews T, Inturrisi C et al (1983) Levorphanol: pharmacokinetics and steady-state plasma concentrations in patients with pain. Res Commun Chem Pathol Pharmacol 41:3–17

    PubMed  CAS  Google Scholar 

  78. Coffman BL, Rios GR, King CD et al (1997) Human UGT2B7 catalyzes morphine glucuronidation. Drug Metab Dispos 25:1–4

    PubMed  CAS  Google Scholar 

  79. DeHaven-Hudkins DL, Burgos LC, Cassel JA et al (1999) Loperamide (ADL 2–1294), an opioid antihyperalgesic agent with peripheral selectivity. J Pharmacol Exp Ther 289:494–502

    PubMed  CAS  Google Scholar 

  80. Kim KA, Chung J, Jung DH et al (2004) Identification of cytochrome P450 isoforms involved in the metabolism of loperamide in human liver microsomes. Eur J Clin Pharmacol 60:575–581

    PubMed  CAS  Google Scholar 

  81. Dollery C (1991) Therapeutic drugs. Churchill Livingstone, Edinburgh

    Google Scholar 

  82. Lötsch J, Skarke C, Wieting J et al (2006) Modulation of the central nervous effects of levomethadone by genetic polymorphisms potentially affecting its metabolism, distribution, and drug action. Clin Pharmacol Ther 79:72–89

    PubMed  Google Scholar 

  83. de Vos JW, Geerlings PJ, van den Brink W et al (1995) Pharmacokinetics of methadone and its primary metabolite in 20 opiate addicts. Eur J Clin Pharmacol 48:361–366

    PubMed  Google Scholar 

  84. Foster DJR (2001) An examination of the metabolism and pharmacokinetics of methadone with respect to stereoselectivity. PhD Thesis, University of Adelaide, Adelaide

  85. Lötsch J, Stockmann A, Kobal G et al (1996) Pharmacokinetics of morphine and its glucuronides after intravenous infusion of morphine and morphine-6-glucuronide in healthy volunteers. Clin Pharmacol Ther 60:316–325

    PubMed  Google Scholar 

  86. Osborne R, Joel S, Trew D et al (1990) Morphine and metabolite behavior after different routes of morphine administration: demonstration of the importance of the active metabolite morphine-6-glucuronide. Clin Pharmacol Ther 47:12–19

    PubMed  CAS  Google Scholar 

  87. Miller JW, Anderson HH (1954) The effect of N-demethylation on certain pharmacologic actions of morphine, codeine, and meperidine in the mouse. J Pharmacol Exp Ther 112:191–196

    PubMed  CAS  Google Scholar 

  88. Loser SV, Meyer J, Freudenthaler S et al (1996) Morphine-6-O-beta-D-glucuronide but not morphine-3-O-beta-D-glucuronide binds to mu-, delta- and kappa- specific opioid binding sites in cerebral membranes. Naunyn-Schmiedeberg’s Arch Pharmacol 354:192–197

    CAS  Google Scholar 

  89. Ulens C, Baker L, Ratka A et al (2001) Morphine-6beta-glucuronide and morphine-3-glucuronide, opioid receptor agonists with different potencies. Biochem Pharmacol 62:1273–1282

    PubMed  CAS  Google Scholar 

  90. Gong QL, Hedner J, Bjorkman R et al (1992) Morphine-3-glucuronide may functionally antagonize morphine-6-glucuronide induced antinociception and ventilatory depression in the rat. Pain 48:249–255

    PubMed  CAS  Google Scholar 

  91. 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 54:687–694

    PubMed  CAS  Google Scholar 

  92. Halliday AJ, Bartlett SE, Colditz P et al (1999) Brain region-specific studies of the excitatory behavioral effects of morphine-3-glucuronide. Life Sci 65:225–236

    PubMed  CAS  Google Scholar 

  93. Faura CC, Olaso MJ, Garcia Cabanes C et al (1996) Lack of morphine-6-glucuronide antinociception after morphine treatment. Is morphine-3-glucuronide involved? Pain 65:25–30

    PubMed  CAS  Google Scholar 

  94. Gardmark M, Karlsson MO, Jonsson F et al (1998) Morphine-3-glucuronide has a minor effect on morphine antinociception. Pharmacodynamic modeling. J Pharm Sci 87:813–820

    PubMed  CAS  Google Scholar 

  95. Ouellet DM, Pollack GM (1997) Effect of prior morphine-3-glucuronide exposure on morphine disposition and antinociception. Biochem Pharmacol 53:1451–1457

    PubMed  CAS  Google Scholar 

  96. Penson RT, Joel SP, Bakhshi K et al (2000) Randomized placebo-controlled trial of the activity of the morphine glucuronides. Clin Pharmacol Ther 68:667–676

    PubMed  CAS  Google Scholar 

  97. Penson RT, Joel SP, Clark S et al (2001) Limited phase I study of morphine-3-glucuronide. J Pharm Sci 90:1810–1816

    PubMed  CAS  Google Scholar 

  98. Osborne R, Joel S, Grebenik K et al (1993) The pharmacokinetics of morphine and morphine glucuronides in kidney failure. Clin Pharmacol Ther 54:158–167

    PubMed  CAS  Google Scholar 

  99. Ashby M, Fleming B, Wood M et al (1997) Plasma morphine and glucuronide (M3G and M6G) concentrations in hospice inpatients. J Pain Symptom Manage 14:157–167

    PubMed  CAS  Google Scholar 

  100. Frances B, Gout R, Campistron G et al (1990) Morphine-6-glucuronide is more mu-selective and potent in analgesic tests than morphine. Prog Clin Biol Res 328:477–480

    PubMed  CAS  Google Scholar 

  101. Frances B, Gout R, Monsarrat B et al (1992) Further evidence that morphine-6 beta-glucuronide is a more potent opioid agonist than morphine. J Pharmacol Exp Ther 262:25–31

    PubMed  CAS  Google Scholar 

  102. Oguri K, Yamada-Mori I, Shigezane J et al (1987) Enhanced binding of morphine and nalorphine to opioid delta receptor by glucuronate and sulfate conjugations at the 6-position. Life Sci 41:1457–1464

    PubMed  CAS  Google Scholar 

  103. Christensen CB, Reiff L (1991) Morphine-6-glucuronide: receptor binding profile in bovine caudate nucleus. Pharmacol Toxicol 68:151–153

    Article  PubMed  CAS  Google Scholar 

  104. Christensen CB, Jorgensen LN (1987) Morphine-6-glucuronide has high affinity for the opioid receptor. Pharmacol Toxicol 60:75–76

    PubMed  CAS  Google Scholar 

  105. Pasternak GW, Bodnar RJ, Clark JA et al (1987) Morphine-6-glucuronide, a potent mu agonist. Life Sci 41:2845–2849

    PubMed  CAS  Google Scholar 

  106. Hucks D, Thompson PI, McLoughlin L et al (1992) Explanation at the opioid receptor level for differing toxicity of morphine and morphine 6-glucuronide. Br J Cancer 65:122–126

    PubMed  CAS  Google Scholar 

  107. Chen ZR, Irvine RJ, Somogyi AA et al (1991) Mu receptor binding of some commonly used opioids and their metabolites. Life Sci 48:2165–2171

    PubMed  CAS  Google Scholar 

  108. Paul D, Standifer KM, Inturrisi CE et al (1989) Pharmacological characterization of morphine-6 beta-glucuronide, a very potent morphine metabolite. J Pharmacol Exp Ther 251:477–483

    PubMed  CAS  Google Scholar 

  109. Shimomura K, Kamata O, Ueki S et al (1971) Analgesic effect of morphine glucuronides. Tohoku J Exp Med 105:45–52

    Article  PubMed  CAS  Google Scholar 

  110. Abbott FV, Palmour RM (1988) Morphine-6-glucuronide: analgesic effects and receptor binding profile in rats. Life Sci 43:1685–1695

    PubMed  CAS  Google Scholar 

  111. Hasselstrom J, Alexander N, Bringel C et al (1991) Single-dose and steady-state kinetics of morphine and its metabolites in cancer patients–a comparison of two oral formulations. Eur J Clin Pharmacol 40:585–591

    PubMed  CAS  Google Scholar 

  112. Skarke C, Darimont J, Schmidt H et al (2003) Analgesic effects of morphine and morphine-6-glucuronide in a transcutaneous electrical pain model in healthy volunteers. Clin Pharmacol Ther 73:107–121

    PubMed  CAS  Google Scholar 

  113. Romberg R, Olofsen E, Sarton E et al (2004) Pharmacokinetic-pharmacodynamic modeling of morphine-6-glucuronide-induced analgesia in healthy volunteers. Anesthesiology 100:120–133

    PubMed  CAS  Google Scholar 

  114. Thompson PI, Joel SP, John L et al (1995) Respiratory depression following morphine and morphine-6-glucuronide in normal subjects. Br J Clin Pharmacol 40:145–152

    PubMed  CAS  Google Scholar 

  115. Buetler TM, Wilder-Smith OH, Wilder-Smith CH et al (2000) Analgesic action of i.v. morphine-6-glucuronide in healthy volunteers. Br J Anaesth 84:97–99

    PubMed  CAS  Google Scholar 

  116. Lötsch J, Kobal G, Stockmann A et al (1997) Lack of analgesic activity of morphine-6-glucuronide after short-term intravenous administration in healthy volunteers. Anesthesiology 87:1348–1358

    PubMed  Google Scholar 

  117. Motamed C, Mazoit X, Ghanouchi K et al (2000) Preemptive intravenous morphine-6-glucuronide is ineffective for postoperative pain relief. Anesthesiology 92:355–360

    PubMed  CAS  Google Scholar 

  118. Tegeder I, Meier S, Burian M et al (2003) Peripheral opioid analgesia in experimental human pain models. Brain 126:1092–1102

    PubMed  Google Scholar 

  119. Osborne R, Thompson P, Joel S et al (1992) The analgesic activity of morphine-6-glucuronide. Br J Clin Pharmacol 34:130–138

    PubMed  CAS  Google Scholar 

  120. Hanna MH, Peat SJ, Woodham M et al (1990) Analgesic efficacy and CSF pharmacokinetics of intrathecal morphine-6-glucuronide: comparison with morphine. Br J Anaesth 64:547–550

    PubMed  CAS  Google Scholar 

  121. Grace D, Fee JP (1996) A comparison of intrathecal morphine-6-glucuronide and intrathecal morphine sulfate as analgesics for total hip replacement. Anesth Analg 83:1055–1059

    PubMed  CAS  Google Scholar 

  122. Romberg R, van Dorp E, Hollander J et al (2007) A randomized, double-blind, placebo-controlled pilot study of IV morphine-6-glucuronide for postoperative pain relief after knee replacement surgery. Clin J Pain 23:197–203

    PubMed  Google Scholar 

  123. Cann C, Curran J, Milner T et al (2002) Unwanted effects of morphine-6-glucoronide and morphine. Anaesthesia 57:1200–1203

    PubMed  CAS  Google Scholar 

  124. Peat SJ, Hanna MH, Woodham M et al (1991) Morphine-6-glucuronide: effects on ventilation in normal volunteers. Pain 45:101–104

    PubMed  CAS  Google Scholar 

  125. Romberg R, Olofsen E, Sarton E et al (2003) Pharmacodynamic effect of morphine-6-glucuronide versus morphine on hypoxic and hypercapnic breathing in healthy volunteers. Anesthesiology 99:788–798

    PubMed  CAS  Google Scholar 

  126. Fromm MF, Eckhardt K, Li S et al (1997) Loss of analgesic effect of morphine due to coadministration of rifampin. Pain 72:261–267

    PubMed  CAS  Google Scholar 

  127. Koopman-Kimenai PM, Vree TB, Booij LH et al (1993) Pharmacokinetics of intravenously administered nicomorphine and its metabolites in man. Eur J Anaesthesiol 10:125–132

    PubMed  CAS  Google Scholar 

  128. Lobbezoo MW, Van Rooy HH, Van Wijngaarden I et al (1982) Opiate receptor binding of nicomorphine and its hydrolysis products in rat brain. Eur J Pharmacol 82:207–211

    PubMed  CAS  Google Scholar 

  129. Rasmussen I (2000) Identification of cytochrome P450 isoforms involved in the metabolism of oxycodone. Master of Science Thesis, University of Adelaide, Adelaide

  130. Lalovic B, Phillips B, Risler L et al (2004) Quantitative contribution of CYP2D6 and CYP3A to oxycodone metabolism in human liver and intestinal microsomes. Drug Metab Dispos 32:447–454

    PubMed  CAS  Google Scholar 

  131. Ishida T, Oguri K, Yoshimura H (1979) Isolation and identification of urinary metabolites of oxycodone in rabbits. Drug Metab Dispos 7:162–165

    PubMed  CAS  Google Scholar 

  132. Lalovic B, Kharasch E, Hoffer C et al (2006) Pharmacokinetics and pharmacodynamics of oral oxycodone in healthy human subjects: role of circulating active metabolites. Clin Pharmacol Ther 79:461–479

    PubMed  CAS  Google Scholar 

  133. Beaver WT, Wallenstein SL, Rogers A et al (1978) Analgesic studies of codeine and oxycodone in patients with cancer. II. Comparisons of intramuscular oxycodone with intramuscular morphine and codeine. J Pharmacol Exp Ther 207:101–108

    PubMed  CAS  Google Scholar 

  134. Leow KP, Smith MT (1994) The antinociceptive potencies of oxycodone, noroxycodone and morphine after intracerebroventricular administration to rats. Life Sci 54:1229–1236

    PubMed  CAS  Google Scholar 

  135. Nielsen CK, Ross FB, Lotfipour S et al (2007) Oxycodone and morphine have distinctly different pharmacological profiles: radioligand binding and behavioural studies in two rat models of neuropathic pain. Pain 132:289–300

    PubMed  CAS  Google Scholar 

  136. Ross FB, Smith MT (1997) The intrinsic antinociceptive effects of oxycodone appear to be kappa-opioid receptor mediated. Pain 73:151–157

    PubMed  CAS  Google Scholar 

  137. Poyhia R, Olkkola KT, Seppala T et al (1991) The pharmacokinetics of oxycodone after intravenous injection in adults. Br J Clin Pharmacol 32:516–518

    PubMed  CAS  Google Scholar 

  138. Poyhia R, Seppala T, Olkkola KT et al (1992) The pharmacokinetics and metabolism of oxycodone after intramuscular and oral administration to healthy subjects. Br J Clin Pharmacol 33:617–621

    PubMed  CAS  Google Scholar 

  139. Twistler ST, Enggaard TP, Noehr-Jensen L et al (2008) The hypoalgesic effect of oxycodone in human experimental pain models in relation to the CYP2D6 oxidation polymorphism. In: Scandinavian Association for the Study of Pain, 31st annual meeting

  140. Beaver WT, Wallenstein SL, Houde RW et al (1977) Comparisons of the analgesic effects of oral and intramuscular oxymorphone and of intramuscular oxymorphone and morphine in patients with cancer. J Clin Pharmacol 17:186–198

    PubMed  CAS  Google Scholar 

  141. Prommer E (2006) Oxymorphone: a review. Support Care Cancer 14:109–115

    PubMed  Google Scholar 

  142. Goldstein G (1985) Pentazocine. Drug Alcohol Depend 14:313–324

    PubMed  CAS  Google Scholar 

  143. Berkowitz B (1973) Pharmacokinetics and neurochemical effects of pentazocine and its optical isomers. Adv Biochem Psychopharmacol 8:495–501

    PubMed  CAS  Google Scholar 

  144. MacDonald AD, Woolfe G, Bergel F et al (1946) Analgesic action of pethidine derivatives and related compounds. Brit J Pharmacol 1:4–14

    PubMed  CAS  Google Scholar 

  145. Ramirez J, Innocenti F, Schuetz EG et al (2004) CYP2B6, CYP3A4, and CYP2C19 are responsible for the in vitro N-demethylation of meperidine in human liver microsomes. Drug Metab Dispos 32:930–936

    PubMed  CAS  Google Scholar 

  146. Latta KS, Ginsberg B, Barkin RL (2002) Meperidine: a critical review. Am J Ther 9:53–68

    PubMed  Google Scholar 

  147. Thierry C, Boeynaems J-M, Paolo M (2005) Actions of tilidine and nortilidine on cloned opioid receptors. Eur J Pharmacol 506:205–208

    PubMed  CAS  Google Scholar 

  148. Vollmer KO, Thomann P, Hengy H (1989) Pharmacokinetics of tilidine and metabolites in man. Arzneimittelforschung 39:1283–1288

    PubMed  CAS  Google Scholar 

  149. Hadja JP, Jahncen E, Ole S et al (2002) Sequential first-pass metabolism of nortilidine: the active metabolite of the synthetic opioid drug tilidine. J Clin Pharmacol 42:1257–1261

    Google Scholar 

  150. Seiler KU, Jahncen E, Trenk D et al (2001) Pharmacokinetics of tilidine in terminal renal failure. J Clin Pharmacol 41:79–84

    PubMed  CAS  Google Scholar 

  151. Brennscheidt U, Brunnmuller U, Proppe D et al (2007) Pharmacokinetics of tilidine and naloxone in patients with severe hepatic impairment. Arzneimittelforschung 57:106–111

    PubMed  CAS  Google Scholar 

  152. Gillen C, Haurand M, Kobelt DJ et al (2000) Affinity, potency and efficacy of tramadol and its metabolites at the cloned human mu-opioid receptor. Naunyn-Schmiedeberg’s Arch Pharmacol 362:116–121

    CAS  Google Scholar 

  153. Poulsen L, Arendt-Nielsen L, Brøsen K et al (1996) The hypoalgesic effect of tramadol in relation to CYP2D6. Clin Pharmacol Ther 60:636–644

    PubMed  CAS  Google Scholar 

  154. Borlak J, Hermann R, Erb K et al (2003) A rapid and simple CYP2D6 genotyping assay - case study with analgetic tramadol. Metabolism 52:1439–1443

    PubMed  CAS  Google Scholar 

  155. Fliegert F, Kurth B, Gohler K (2005) The effects of tramadol on static and dynamic pupillometry in healthy subjects-the relationship between pharmacodynamics, pharmacokinetics and CYP2D6 metaboliser status. Eur J Pharmacol 61:257–266

    CAS  Google Scholar 

  156. Slanar O, Nobilis M, Kvetina J et al (2006) CYP2D6 polymorphism, tramadol pharmacokinetics and pupillary response. Eur J Clin Pharmacol 62:75–76

    PubMed  Google Scholar 

  157. Slanar O, Nobilis M, Kvetina J et al (2007) Miotic action of tramadol is determined by CYP2D6 genotype. Physiol Res 56:129–136

    PubMed  CAS  Google Scholar 

  158. Levo A, Koski A, Ojanpera I et al (2003) Post-mortem SNP analysis of CYP2D6 gene reveals correlation between genotype and opioid drug (tramadol) metabolite ratios in blood. Forensic Sci Int 135:9–15

    PubMed  CAS  Google Scholar 

  159. Pedersen RS, Damkier P, Brøsen K (2005) Tramadol as a new probe for cytochrome P450 2D6 phenotyping: a population study. Clin Pharmacol Ther 77:458–467

    PubMed  CAS  Google Scholar 

  160. Enggaard TP, Poulsen L, Arendt-Nielsen L et al (2006) The analgesic effect of tramadol after intravenous injection in healthy volunteers in relation to CYP2D6. Anesth Analg 102:146–150

    PubMed  CAS  Google Scholar 

  161. Stamer UM, Musshoff F, Kobilay M et al (2007) Concentrations of tramadol and O-desmethyltramadol enantiomers in different CYP2D6 genotypes. Clin Pharmacol Ther 82:41–47

    PubMed  CAS  Google Scholar 

  162. Coffman BL, King CD, Rios GR et al (1998) The glucuronidation of opioids, other xenobiotics, and androgens by human UGT2B7Y(268) and UGT2B7H(268). Drug Metab Dispos 26:73–77

    PubMed  CAS  Google Scholar 

  163. Moffat AC, Osselton MD, Widdop B (eds) (2004) Clarke’s analysis of drugs and poisons, 3rd ed., vol. 2. Pharmaceutical Press, London

  164. Oguri K, Yamada-Mori I, Shigezane J et al (1984) Potentiation of physical dependence by conjugation at the 6-position of nalorphine. Eur J Pharmacol 102:229–235

    PubMed  CAS  Google Scholar 

  165. Konno F, Kobayashi C, Morimoto R et al (1986) Pharmacological effects of nalorphine and nalorphine-7,8-oxide (nalorphine-epoxide): interaction of the intrinsic activity, affinity and pharmacological responses. Arch Int Pharmacodyn Ther 282:219–232

    PubMed  CAS  Google Scholar 

  166. Wang D, Raehal KM, Bilsky EJ et al (2001) Inverse agonists and neutral antagonists at mu opioid receptor (MOR): possible role of basal receptor signaling in narcotic dependence. J Neurochem 77:1590–1600

    PubMed  CAS  Google Scholar 

  167. Sadee W, Wang D, Bilsky EJ (2005) Basal opioid receptor activity, neutral antagonists, and therapeutic opportunities. Life Sci 76:1427–1437

    PubMed  CAS  Google Scholar 

  168. Weinstein SH, Pfeffer M, Schor JM (1973) Metabolism and pharmacokinetics of naloxone. Adv Biochem Psychopharmacol 8:525–535

    PubMed  CAS  Google Scholar 

  169. Wahlstrom A, Winblad B, Bixo M et al (1988) Human brain metabolism of morphine and naloxone. Pain 35:121–127

    PubMed  CAS  Google Scholar 

  170. Reber P, Brenneisen R, Flogerzi B et al (2007) Effect of naloxone-3-glucuronide and N-methylnaloxone on the motility of the isolated rat colon after morphine. Dig Dis Sci 52:502–507

    PubMed  CAS  Google Scholar 

  171. Yamano S, Ichinose F, Todaka T et al (1999) Purification and characterization of two major forms of naloxone reductase from rabbit liver cytosol, new members of aldo-keto reductase superfamily. Biol Pharm Bull 22:1038–1046

    PubMed  CAS  Google Scholar 

  172. Olsen LD, Klein P, Nelson WL et al (1990) Conjugate addition ligands of opioid antagonists. Methacrylate esters and ethers of 6 alpha- and 6 beta-naltrexol. J Med Chem 33:737–741

    PubMed  CAS  Google Scholar 

  173. Porter SJ, Somogyi AA, White JM (2002) In vivo and in vitro potency studies of 6beta-naltrexol, the major human metabolite of naltrexone. Addict Biol 7:219–225

    PubMed  CAS  Google Scholar 

  174. Raehal KM, Lowery JJ, Bhamidipati CM et al (2005) In vivo characterization of 6beta-naltrexol, an opioid ligand with less inverse agonist activity compared with naltrexone and naloxone in opioid-dependent mice. J Pharmacol Exp Ther 313:1150–1162

    PubMed  CAS  Google Scholar 

  175. Ferrari A, Bertolotti M, Dell’Utri A et al (1998) Serum time course of naltrexone and 6 beta-naltrexol levels during long-term treatment in drug addicts. Drug Alcohol Depend 52:211–220

    PubMed  CAS  Google Scholar 

Download references

Acknowledgements

Supported by the National Health and Medical Research Council of Australia. J.C. is a FTT Fricker Research Fellow at the University of Adelaide.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Andrew A. Somogyi.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Coller, J.K., Christrup, L.L. & Somogyi, A.A. Role of active metabolites in the use of opioids. Eur J Clin Pharmacol 65, 121–139 (2009). https://doi.org/10.1007/s00228-008-0570-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00228-008-0570-y

Keywords

Navigation