Skip to main content
Log in

Genetic Predictors of the Clinical Response to Opioid Analgesics

Clinical Utility and Future Perspectives

  • Review Article
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

This review uses a candidate gene approach to identify possible pharmacogenetic modulators of opioid therapy, and discusses these modulators together with demonstrated genetic causes for the variability in clinical effects of opioids.

Genetically caused inactivity of cytochrome P450 (CYP) 2D6 renders codeine ineffective (lack of morphine formation), slightly decreases the efficacy of tramadol (lack of formation of the active O-desmethyl-tramadol) and slightly decreases the clearance of methadone. MDR1 mutations often demonstrate pharmacogenetic consequences, and since Opioids are among the P-glycoprotein substrates, opioid pharmacology may be affected by MDR1 mutations. The single nucleotide polymorphism A118G of the μ opioid receptor gene has been associated with decreased potency of morphine and morphine-6-glucuronide, and with decreased analgesic effects and higher alfentanil dose demands in carriers of the mutated Gl 18 allele. Genetic causes may also trigger or modify drug interactions, which in turn can alter the clinical response to opioid therapy. For example, by inhibiting CYP2D6, paroxetine increases the steady-state plasma concentrations of (R)-methadone in extensive but not in poor metabolisers of debrisoquine/sparteine.

So far, the clinical consequences of the pharmacogenetics of opioids are limited to codeine, which should not be administered to poor metabolisers of debrisoquine/sparteine. Genetically precipitated drug interactions might render a standard opioid dose toxic and should, therefore, be taken into consideration. Mutations affecting opioid receptors and pain perception/processing are of interest for the study of opioid actions, but with modern practice of on-demand administration of Opioids their utility may be limited to explaining why some patients need higher opioid doses; however, the adverse effects profile may be modified by these mutations. Nonetheless, at a limited level, pharmacogenetics can be expected to facilitate individualised opioid therapy.

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
Table I
Fig. 2
Table II
Fig. 3
Table III
Table IV
Table V

Similar content being viewed by others

Notes

  1. IV2 C691G was named C1031G in the review by Szeto et al.;[194] it has been changed for consistency with the numbering of the other SNPs.

References

  1. Lander ES, Linton LM, Birren B, et al. Initial sequencing and analysis of the human genome. Nature 2001; 409(6822): 860–921

    Article  CAS  PubMed  Google Scholar 

  2. Venter JC, Adams MD, Myers EW, et al. The sequence of the human genome. Science 2001; 291(5507): 1304–51

    Article  CAS  PubMed  Google Scholar 

  3. Stein C. The control of pain in peripheral tissue by Opioids. N Engl J Med 1995; 332(25): 1685–90

    Article  CAS  PubMed  Google Scholar 

  4. Ohara H, Miyabe Y, Deyashiki Y, et al. Reduction of drug ketones by dihydrodiol dehydrogenases, carbonyl reductase and aldehyde reductase of human liver. Biochem Pharmacol 1995; 50(2): 221–7

    Article  CAS  PubMed  Google Scholar 

  5. Yamano S, Ichinose F, Todaka T, et al. Purification and characterization of two major forms of naloxone reductase from rabbit liver cytosol, new members of aldo-keto reductase superfamily. Biol Pharm Bull 1999; 22(10): 1038–46

    Article  CAS  PubMed  Google Scholar 

  6. Yamano S, Nakamoto N, Toki S. Purification and characterization of rat liver naloxone reductase that is identical to 3α-hydroxysteroid dehydrogenase. Xenobiotica 1999; 29(9): 917–30

    Article  CAS  PubMed  Google Scholar 

  7. Eichelbaum M. Polymorphic drug oxidation in humans. Fed Proc 1984; 43(8): 2298–302

    CAS  PubMed  Google Scholar 

  8. Liu Z, Mortimer O, Smith CA, et al. Evidence for a role of cytochrome P450 2D6 and 3A4 in ethylmorphine metabolism. Br J Clin Pharmacol 1995; 39(1): 77–80

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Flockhart DA. Drug interactions: cytochrome P450 system [online]. Available from URL: http://medicine.iupui.edu/flockhart [Accesssed 2004 Aug 17]

  10. Gough AC, Smith CA, Howell SM, et al. Localization of the CYP2D gene locus to human chromosome 22q13.1 by Polymerase chain reaction, in situ hybridization, and linkage analysis. Genomics 1993; 15(2): 430–2

    Article  CAS  PubMed  Google Scholar 

  11. Kimura S, Umeno M, Skoda RC, et al. The human debrisoquine 4-hydroxylase (CYP2D) locus: sequence and identification of the polymorphic CYP2D6 gene, a related gene, and a pseudogene. Am J Hum Genet 1989; 45(6): 889–904

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Murphy MP, Beaman ME, Clark LS, et al. Prospective CYP2D6 genotyping as an exclusion criterion for enrollment of a phase III clinical trial. Pharmacogenetics 2000; 10(7): 583–90

    Article  CAS  PubMed  Google Scholar 

  13. Griese EU, Zanger UM, Brudermanns U, et al. Assessment of the predictive power of genotypes for the in-vivo catalytic function of CYP2D6 in a German population. Pharmacogenetics 1998; 8(1): 15–26

    Article  CAS  PubMed  Google Scholar 

  14. Katoh T, Higashi K. Ethnic differences of the primary gene defect at the cytochrome P-450 2D6. J UOEH 1992; 14(3): 205–9

    Article  CAS  PubMed  Google Scholar 

  15. Gaedigk A, Bradford LD, Marcucci KA, et al. Unique CYP2D6 activity distribution and genotype-phenotype discordance in black Americans. Clin Pharmacol Ther 2002; 72(1): 76–89

    Article  CAS  PubMed  Google Scholar 

  16. Ji L, Pan S, Marti-Jaun J, et al. Single-step assays to analyze CYP2D6 gene polymorphisms in Asians: allele frequencies and a novel *14B allele in mainland Chinese. Clin Chem 2002; 48(7): 983–8

    CAS  PubMed  Google Scholar 

  17. Johansson I, Oscarson M, Yue QY, et al. Genetic analysis of the Chinese cytochrome P4502D locus: characterization of variant CYP2D6 genes present in subjects with diminished capacity for debrisoquine hydroxylation. Mol Pharmacol 1994; 46(3): 452–9

    CAS  PubMed  Google Scholar 

  18. Eichelbaum M, Spannbrucker N, Steincke B, et al. Defective N-oxidation of sparteine in man: a new pharmacogenetic defect. Eur J Clin Pharmacol 1979; 16(3): 183–7

    Article  CAS  PubMed  Google Scholar 

  19. Mahgoub A, Idle JR, Dring LG, et al. Polymorphic hydroxylation of debrisoquine in man. Lancet 1977; II(8038): 584-6

    Google Scholar 

  20. Bertilsson L, Lou YQ, Du YL, et al. Pronounced differences between native Chinese and Swedish populations in the polymorphic hydroxylations of debrisoquin and S-mephenytoin. Clin Pharmacol Ther 1992; 51(4): 388–97

    Article  CAS  PubMed  Google Scholar 

  21. Lou YC, Ying L, Bertilsson L, et al. Low frequency of slow debrisoquine hydroxylation in a native Chinese population. Lancet 1987; II(8563): 852-3

    Google Scholar 

  22. Nakamura K, Goto F, Ray WA, et al. Interethnic differences in genetic polymorphism of debrisoquin and mephenytoin hydroxylation between Japanese and Caucasian populations. Clin Pharmacol Ther 1985; 38(4): 402–8

    Article  CAS  PubMed  Google Scholar 

  23. Ishizaki T, Eichelbaum M, Horai Y, et al. Evidence for polymorphic oxidation of sparteine in Japanese subjects. Br J Clin Pharmacol 1987; 23(4): 482–5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Johansson I, Lundqvist E, Bertilsson L, et al. Inherited amplification of an active gene in the cytochrome P450 CYP2D locus as a cause of ultrarapid metabolism of debrisoquine. Proc Natl Acad Sci U S A 1993; 90(24): 11825–9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dahl ML, Johansson I, Bertilsson L, et al. Ultrarapid hydroxylation of debrisoquine in a Swedish population: analysis of the molecular genetic basis. J Pharmacol Exp Ther 1995; 274(1): 516–20

    CAS  PubMed  Google Scholar 

  26. Ingelman-Sundberg M. Duplication, multiduplication, and amplification of genes encoding drug-metabolizing enzymes: evolutionary, toxicological, and clinical pharmacological aspects. Drug Metab Rev 1999; 31(2): 449–59

    Article  CAS  PubMed  Google Scholar 

  27. Lovlie R, Daly AK, Matre GE, et al. Polymorphisms in CYP2D6 duplication-negative individuals with the ultrarapid metabolizer phenotype: a role for the CYP2D6*35 allele in ultrarapid metabolism? Pharmacogenetics 2001; 11(1): 45–55

    Article  CAS  PubMed  Google Scholar 

  28. Bathum L, Johansson I, Ingelman-Sundberg M, et al. Ultrarapid metabolism of sparteine: frequency of alleles with duplicated CYP2D6 genes in a Danish population as determined by restriction fragment length polymorphism and long polymerase chain reaction. Pharmacogenetics 1998; 8(2): 119–23

    Article  CAS  PubMed  Google Scholar 

  29. Lovlie R, Daly AK, Molven A, et al. Ultrarapid metabolizers of debrisoquine: characterization and PCR-based detection of alleles with duplication of the CYP2D6 gene. FEBS Lett 1996 Aug 19; 392(1): 30–4

    Article  CAS  PubMed  Google Scholar 

  30. Human Cytochrome P450 Allele Nomenclature Committee. Nomenclature files for human cytochrome P450 alleles [online]. Available from URL: http://www.imm.ki.se/CYPalleles/ [Accesssed 2004 Aug 17]

  31. Bertilsson L, Dahl ML, Dalen P, et al. Molecular genetics of CYP2D6: clinical relevance with focus on psychotropic drugs. Br J Clin Pharmacol 2002; 53(2): 111–22

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bertilsson L. Geographical/interracial differences in polymorphic drug oxidation: current state of knowledge of cytochromes P450 (CYP) 2D6 and 2C19. Clin Pharmacokinet 1995; 29(3): 192–209

    Article  CAS  PubMed  Google Scholar 

  33. Mignat C, Wille U, Ziegler A. Affinity profiles of morphine, codeine, dihydrocodeine and their glucuronides at Opioid receptor subtypes. Life Sci 1995; 56(10): 793–9

    Article  CAS  PubMed  Google Scholar 

  34. Sindrup SH, Brosen K. The pharmacogenetics of codeine hypoalgesia. Pharmacogenetics 1995; 5(6): 335–46

    Article  CAS  PubMed  Google Scholar 

  35. Yue QY, Svensson JO, Alm C, et al. Codeine O-demethylation co-segregates with polymorphic debrisoquine hydroxylation. Br J Clin Pharmacol 1989; 28(6): 639–45

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chen ZR, Somogyi AA, Bochner F. Polymorphic O-demethylation of codeine. Lancet 1988; II(8616): 914–5

    Article  Google Scholar 

  37. Yue QY, Hasselström J, Svensson JO, et al. Pharmacokinetics of codeine and its metabolites in Caucasian healthy volunteers: comparisons between extensive and poor hydroxylators of debrisoquine. Br J Clin Pharmacol 1991; 31(6): 635–42

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chen ZR, Irvine RJ, Bochner F, et al. Morphine formation from codeine in rat brain: a possible mechanism of codeine analgesia. Life Sci 1990; 46: 1067–74

    Article  CAS  PubMed  Google Scholar 

  39. Sindrup SH, Arendt-Nielsen L, Brosen K, et al. The effect of quinidine on the analgesic effect of codeine. Eur J Clin Pharmacol 1992; 42(6): 587–91

    Article  CAS  PubMed  Google Scholar 

  40. Sindrup SH, Hofmann U, Asmussen J, et al. Impact of quinidine on plasma and cerebrospinal fluid concentrations of codeine and morphine after codeine intake. Eur J Clin Pharmacol 1996; 49(6): 503–9

    Article  CAS  PubMed  Google Scholar 

  41. Sindrup SH, Brosen K, Bjerring P, et al. Codeine increases pain thresholds to copper vapor laser stimuli in extensive but not poor metabolizers of sparteine. Clin Pharmacol Ther 1990; 48(6): 686–93

    Article  CAS  PubMed  Google Scholar 

  42. Caraco Y, Sheller J, Wood AJ. Impact of ethnic origin and quinidine coadministration on codeine’s disposition and pharmacodynamic effects. J Pharmacol Exp Ther 1999; 290(1): 413–22

    CAS  PubMed  Google Scholar 

  43. Caraco Y, Sheller J, Wood AJ. Pharmacogenetic determination of the effects of codeine and prediction of drug interactions. J Pharmacol Exp Ther 1996; 278(3): 1165–74

    CAS  PubMed  Google Scholar 

  44. Desmeules J, Gascon MP, Dayer P, et al. Impact of environmental and genetic factors on codeine analgesia. Eur J Clin Pharmacol 1991; 41: 23–6

    Article  CAS  PubMed  Google Scholar 

  45. Dalen P, Frengell C, Dahl ML, et al. Quick onset of severe abdominal pain after codeine in an ultrarapid metabolizer of debrisoquine. Ther Drug Monit 1997; 19(5): 543–4

    Article  CAS  PubMed  Google Scholar 

  46. Hasselstrom J, Yue QY, Sawe J. The effect of codeine on gastrointestinal transit in extensive and poor metabolisers of debrisoquine. Eur J Clin Pharmacol 1997; 53(2): 145–8

    Article  CAS  PubMed  Google Scholar 

  47. Poulsen L, Brosen K, Arendt-Nielsen L, et al. Codeine and morphine in extensive and poor metabolizers of sparteine: pharmacokinetics, analgesic effect and side effects. Eur J Clin Pharmacol 1996; 51(3–4): 289–95

    Article  CAS  PubMed  Google Scholar 

  48. Mikus G, Trausch B, Rodewald C, et al. Effect of codeine on gastrointestinal motility in relation to CYP2D6 phenotype. Clin Pharmacol Ther 1997; 61(4): 459–66

    Article  CAS  PubMed  Google Scholar 

  49. Poulsen L, Riishede L, Brosen K, et al. Codeine in postoperative pain: study of the influence of sparteine phenotype and serum concentrations of morphine and morphine-6-glucuronide. Eur J Clin Pharmacol 1998; 54(6): 451–4

    Article  CAS  PubMed  Google Scholar 

  50. Persson K, Sjostrom S, Sigurdardottir I, et al. Patient-controlled analgesia (PCA) with codeine for postoperative pain relief in ten extensive metabolisers and one poor metaboliser of dextromethorphan. Br J Clin Pharmacol 1995; 39(2): 182–6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Romach MK, Otton SV, Somer G, et al. Cytochrome P450 2D6 and treatment of codeine dependence. J Clin Psychopharmacol 2000; 20(1): 43–5

    Article  CAS  PubMed  Google Scholar 

  52. Kathiramalainathan K, Kaplan HL, Romach MK, et al. Inhibition of cytochrome P450 2D6 modifies codeine abuse liability. J Clin Psychopharmacol 2000; 20(4): 435–44

    Article  CAS  PubMed  Google Scholar 

  53. Eckhardt K, Li S, Ammon S, et al. Same incidence of adverse drug events after codeine administration irrespective of the genetically determined differences in morphine formation. Pain 1998; 76(1–2): 27–33

    Article  CAS  PubMed  Google Scholar 

  54. Moore A, McQuay H, Gavaghan D. Deriving dichotomous outcome measures from continuous data in randomised controlled trials of analgesics. Pain 1996; 66(2–3): 229–37

    Article  CAS  PubMed  Google Scholar 

  55. McQuay HJ, Moore RA, Eccleston C, et al. Systematic review of outpatient services for chronic pain control. Health Technol Assess 1997; 1(6): 1–135

    Google Scholar 

  56. Rogers JF, Findlay JW, Hull JH, et al. Codeine disposition in smokers and nonsmokers. Clin Pharmacol Ther 1982; 32(2): 218–27

    Article  CAS  PubMed  Google Scholar 

  57. Quiding H, Lundqvist G, Boreus LO, et al. Analgesic effect and plasma concentrations of codeine and morphine after two dose levels of codeine following oral surgery. Eur J Clin Pharmacol 1993; 44: 319–23

    Article  CAS  PubMed  Google Scholar 

  58. Vree TB, van Dongen RT, Koopman-Kimenai PM. Codeine analgesia is due to codeine-6-glucuronide, not morphine. Int J Clin Pract 2000; 54(6): 395–8

    CAS  PubMed  Google Scholar 

  59. Srinivasan V, Wielbo D, Simpkins J, et al. Analgesic and immunomodulatory effects of codeine and codeine 6-glucuronide. Pharm Res 1996; 13(2): 296–300

    Article  CAS  PubMed  Google Scholar 

  60. Skarke C, Darimont J, Schmidt H, et al. Analgesic effects of morphine and morphine-6-glucuronide in a transcutaneous electrical pain model in healthy volunteers. Clin Pharmacol Ther 2003; 73(1): 107–21

    Article  CAS  PubMed  Google Scholar 

  61. Gillen C, Haurand M, Kobelt DJ, et al. Affinity, potency and efficacy of tramadol and its metabolites at the cloned human μ-opioid receptor. Naunyn Schmiedebergs Arch Pharmacol 2000; 362(2): 116–21

    Article  CAS  PubMed  Google Scholar 

  62. Lai J, Ma SW, Porreca F, et al. Tramadol, M1 metabolite and enantiomer affinities for cloned human Opioid receptors expressed in transfected HN9.10 neuroblastoma cells. Eur J Pharmacol 1996; 316(2–3): 369–72

    Article  CAS  PubMed  Google Scholar 

  63. Paar WD, Poche S, Gerloff J, et al. Polymorphic CYP2D6 mediates O-demethylation of the Opioid analgesic tramadol. Eur J Clin Pharmacol 1997; 53(3–4): 235–9

    Article  CAS  PubMed  Google Scholar 

  64. Abdel-Rahman SM, Leeder JS, Wilson JT, et al. Concordance between tramadol and dextromethorphan parent/metabolite ratios: the influence of CYP2D6 and non-CYP2D6 pathways on biotransformation. J Clin Pharmacol 2002; 42(1): 24–9

    Article  CAS  PubMed  Google Scholar 

  65. Ogunleye DS. Investigation of racial variations in the metabolism of tramadol. Eur J Drug Metab Pharmacokinet 2001; 26(1–2): 95–8

    Article  CAS  PubMed  Google Scholar 

  66. Willer JC. Studies on pain: effects of morphine on a spinal nociceptive flexion reflex and related pain sensation in man. Brain Res 1985; 331: 105–14

    Article  CAS  PubMed  Google Scholar 

  67. Collart L, Luthy C, Favario-Constantin C, et al. Duality of the analgesic effect of tramadol in humans [in French]. Schweiz Med Wochenschr 1993; 123(47): 2241–3

    CAS  PubMed  Google Scholar 

  68. Poulsen L, Arendt-Nielsen L, Brosen K, et al. The hypoalgesic effect of tramadol in relation to CYP2D6. Clin Pharmacol Ther 1996; 60(6): 636–44

    Article  CAS  PubMed  Google Scholar 

  69. Driessen B, Reimann W. Interaction of the central analgesic, tramadol, with the uptake and release of 5-hydroxytryptamine in the rat brain in vitro. Br J Pharmacol 1992; 105(1): 147–51

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Driessen B, Reimann W, Giertz H. Effects of the central analgesic tramadol on the uptake and release of noradrenaline and dopamine in vitro. Br J Pharmacol 1993; 108(3): 806–11

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Raffa RB, Friderichs E, Reimann W, et al. 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

    CAS  PubMed  Google Scholar 

  72. Schmidt H, Vormfelde SV, Klinder K, et al. Affinities of dihydrocodeine and its metabolites to Opioid receptors. Pharmacol Toxicol 2002; 91(2): 57–63

    Article  CAS  PubMed  Google Scholar 

  73. Fromm MF, Hofmann U, Griese EU, et al. Dihydrocodeine: a new Opioid substrate for the polymorphic CYP2D6 in humans. Clin Pharmacol Ther 1995; 58(4): 374–82

    Article  CAS  PubMed  Google Scholar 

  74. Wilder-Smith CH, Hufschmid E, Thormann W. 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 1998; 45(6): 575–81

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Heiskanen T, Olkkola KT, Kalso E. Effects of blocking CYP2D6 on the pharmacokinetics and pharmacodynamics of oxycodone. Clin Pharmacol Ther 1998; 64(6): 603–11

    Article  CAS  PubMed  Google Scholar 

  76. Otton SV, Schadel M, Cheung SW, et al. CYP2D6 phenotype determines the metabolic conversion of hydrocodone to hydromorphone. Clin Pharmacol Ther 1993; 54(5): 463–72

    Article  CAS  PubMed  Google Scholar 

  77. Chen ZR, Irvine RJ, Somogyi AA, et al. μ-Receptor binding of some commonly used Opioids and their metabolites. Life Sci 1991; 48: 2165–71

    Article  CAS  PubMed  Google Scholar 

  78. Kaplan HL, Busto UE, Baylon GJ, et al. Inhibition of cytochrome P450 2D6 metabolism of hydrocodone to hydromorphone does not importantly affect abuse liability. J Pharmacol Exp Ther 1997; 281(1): 103–8

    CAS  PubMed  Google Scholar 

  79. Begre S, von Bardeleben U, Ladewig D, et al. Paroxetine increases steady-state concentrations of (R)-methadone in CYP2D6 extensive but not poor metabolizers. J Clin Psychopharmacol 2002; 22(2): 211–5

    Article  CAS  PubMed  Google Scholar 

  80. Eap CB, Broly F, Mino A, et al. Cytochrome P450 2D6 genotype and methadone steady-state concentrations. J Clin Psychopharmacol 2001; 21(2): 229–34

    Article  CAS  PubMed  Google Scholar 

  81. Iribarne C, Dreano Y, Bardou LG, et al. Interaction of methadone with substrates of human hepatic cytochrome P450 3A4. Toxicology 1997; 117(1): 13–23

    Article  CAS  PubMed  Google Scholar 

  82. Heelon MW, Meade LB. Methadone withdrawal when starting an antiretroviral regimen including nevirapine. Pharmacotherapy 1999; 19(4): 471–2

    Article  CAS  PubMed  Google Scholar 

  83. Geletko SM, Erickson AD. Decreased methadone effect after ritonavir initiation. Pharmacotherapy 2000; 20(1): 93–4

    Article  CAS  PubMed  Google Scholar 

  84. Holmes VF. Rifampin-induced methadone withdrawal in AIDS. J Clin Psychopharmacol 1990; 10(6): 443–4

    CAS  PubMed  Google Scholar 

  85. Bending MR, Skacel PO. Rifampicin and methadone withdrawal [letter]. Lancet 1977; I(8023): 1211

    Article  Google Scholar 

  86. Iribarne C, Berthou F, Baird S, et al. Involvement of cytochrome P450 3A4 enzyme in the N-demethylation of methadone in human liver microsomes. Chem Res Toxicol 1996; 9(2): 365–73

    Article  CAS  PubMed  Google Scholar 

  87. Eap CB, Buclin T, Baumann P. Interindividual variability of the clinical pharmacokinetics of methadone: implications for the treatment of Opioid dependence. Clin Pharmacokinet 2002; 41(14): 1153–93

    Article  CAS  PubMed  Google Scholar 

  88. Kobayashi K, Yamamoto T, Chiba K, et al. Human buprenorphine N-dealkylation is catalyzed by cytochrome P450 3A4. Drug Metab Dispos 1998; 26(8): 818–21

    CAS  PubMed  Google Scholar 

  89. Feierman DE, Lasker JM. Metabolism of fentanyl, a synthetic Opioid analgesic, by human liver microsomes: role of CYP3A4. Drug Metab Dispos 1996; 24(9): 932–9

    CAS  PubMed  Google Scholar 

  90. Kharasch ED, Thummel KE. Human alfentanil metabolism by cytochrome P450 3A3/4: an explanation for the interindividual variability in alfentanil clearance? Anesth Analg 1993; 76(5): 1033–9

    Article  CAS  PubMed  Google Scholar 

  91. Phimmasone S, Kharasch ED. A pilot evaluation of alfentanilinduced miosis as a noninvasive probe for hepatic cytochrome P450 3A4 (CYP3A4) activity in humans. Clin Pharmacol Ther 2001; 70(6): 505–17

    Article  CAS  PubMed  Google Scholar 

  92. Palkama VJ, Neuvonen PJ, Olkkola KT. The CYP 3A4 inhibitor itraconazole has no effect on the pharmacokinetics of i.v. fentanyl. Br J Anaesth 1998; 81(4): 598–600

    Article  CAS  PubMed  Google Scholar 

  93. Olkkola KT, Palkama VJ, Neuvonen PJ. Ritonavir’s role in reducing fentanyl clearance and prolonging its half-life. Anesthesiology 1999; 91(3): 681–5

    Article  CAS  PubMed  Google Scholar 

  94. Gellner K, Eiselt R, Hustert E, et al. Genomic organization of the human CYP3A locus: identification of a new, inducible CYP3A gene. Pharmacogenetics 2001; 11(2): 111–21

    Article  CAS  PubMed  Google Scholar 

  95. Sata F, Sapone A, Elizondo G, et al. CYP3A4 allelic variants with amino acid substitutions in exons 7 and 12: evidence for an allelic variant with altered catalytic activity. Clin Pharmacol Ther 2000; 67(1): 48–56

    Article  CAS  PubMed  Google Scholar 

  96. Aoyama T, Yamano S, Waxman DJ, et al. Cytochrome P-450 hPCN3, a novel cytochrome P-450 IIIA gene product that is differentially expressed in adult human liver: cDNA and deduced amino acid sequence and distinct specificities of cDNA-expressed hPCN1 and hPCN3 for the metabolism of steroid hormones and cyclosporine. J Biol Chem 1989; 264(18): 10388–95

    CAS  PubMed  Google Scholar 

  97. Kuehl P, Zhang J, Lin Y, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet 2001; 27(4): 383–91

    Article  CAS  PubMed  Google Scholar 

  98. Paul D, Standifer KM, Inturrisi CE, et al. Pharmacological characterization of morphine-6 β-glucuronide, a very potent morphine metabolite. J Pharmacol Exp Ther 1989; 251: 477–83

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  100. de Wildt SN, Kearns GL, Leeder JS, et al. Glucuronidation in humans: pharmacogenetic and developmental aspects. Clin Pharmacokinet 1999; 36(6): 439–52

    Article  PubMed  Google Scholar 

  101. Green MD, King CD, Mojarrabi B, et al. Glucuronidation of amines and other xenobiotics catalyzed by expressed human UDP-glucuronosyltransferase 1A3. Drug Metab Dispos 1998; 26(6): 507–12

    CAS  PubMed  Google Scholar 

  102. Kirkwood LC, Nation RL, Somogyi AA. Glucuronidation of dihydrocodeine by human liver microsomes and the effect of inhibitors. Clin Exp Pharmacol Physiol 1998; 25(3–4): 266–70

    Article  CAS  PubMed  Google Scholar 

  103. Ritter JK, Sheen YY, Owens IS. Cloning and expression of human liver UDP-glucuronosyltransferase in COS-1 cells: 3,4-catechol estrogens and estriol as primary substrates. J Biol Chem 1990; 265(14): 7900–6

    CAS  PubMed  Google Scholar 

  104. Bhasker CR, McKinnon W, Stone A, et al. Genetic polymorphism of UDP-glucuronosyltransferase 2B7 (UGT2B7) at amino acid 268: ethnic diversity of alleles and potential clinical significance. Pharmacogenetics 2000; 10(8): 679–85

    Article  CAS  PubMed  Google Scholar 

  105. Innocenti F, Iyer L, Ramirez J, et al. Epirubicin glucuronidation is catalyzed by human UDP-glucuronosyltransferase 2B7. Drug Metab Dispos 2001; 29(5): 686–92

    CAS  PubMed  Google Scholar 

  106. Holthe M, Rakvag TN, Klepstad P, et al. Sequence variations in the UDP-glucuronosyltransferase 2B7 (UGT2B7) gene: identification of 10 novel single nucleotide polymorphisms (SNPs) and analysis of their relevance to morphine glucuronidation in cancer patients. Pharmacogenomics J 2003; 3(1): 17–26

    Article  CAS  PubMed  Google Scholar 

  107. Coffman BL, King CD, Rios GR, et al. The glucuronidation of Opioids, other xenobiotics, and androgene by human UGT2B7Y (268) and UGT2B7H (268). Drug Metab Dispos 1998; 26(1): 73–7

    CAS  PubMed  Google Scholar 

  108. Jin C, Miners JO, Lillywhite KJ, et al. Complementary deoxyribonucleic acid cloning and expression of a human liver uridine diphosphate-glucuronosyltransferase glucuronidating carboxylic acid-containing drugs. J Pharmacol Exp Ther 1993; 264(1): 475–9

    CAS  PubMed  Google Scholar 

  109. Holthe M, Klepstad P, Zahlsen K, et al. Morphine glucuronide-to-morphine plasma ratios are unaffected by the UGT2B7 H268Y and UGT1A1*28 polymorphisms in cancer patients on chronic morphine therapy. Eur J Clin Pharmacol 2002; 58(5): 353–6

    Article  CAS  PubMed  Google Scholar 

  110. Cepeda MS, Farrar JT, Roa JH, et al. Ethnicity influences morphine pharmacokinetics and pharmacodynamics. Clin Pharmacol Ther 2001; 70(4): 351–61

    Article  CAS  PubMed  Google Scholar 

  111. Yue QY, Svensson JO, Sjoqvist F, et al. A comparison of the pharmacokinetics of codeine and its metabolites in healthy Chinese and Caucasian extensive hydroxylators of debrisoquine. Br J Clin Pharmacol 1991; 31(6): 643–7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Nishimura TG, Jackson SH, Cohen SN. Prolongation of morphine anaesthesia in a patient with Gilbert’s disease: report of a case. Can Anaesth Soc J 1973; 20(5): 709–12

    Article  CAS  PubMed  Google Scholar 

  113. Bosma PJ, Chowdhury JR, Bakker C, et al. The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1 in Gilbert’s syndrome. N Engl J Med 1995; 333(18): 1171–5

    Article  CAS  PubMed  Google Scholar 

  114. Burchell B, Hume R. Molecular genetic basis of Gilbert’s syndrome. J Gastroenterol Hepatol 1999; 14(10): 960–6

    Article  CAS  PubMed  Google Scholar 

  115. Skarke C, Schmidt H, Geisslinger G, et al. Pharmacokinetics of morphine are not altered in persons with Gilbert’s syndrome. Br J Clin Pharmacol 2003; 56(2): 228–31

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Juliano RL, Ling V. A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochim Biophys Acta 1976; 455(1): 152–62

    Article  CAS  PubMed  Google Scholar 

  117. King M, Su W, Chang A, et al. Transport of Opioids from the brain to the periphery by P-glycoprotein: peripheral actions of central drugs. Nat Neurosci 2001; 4(3): 268–74

    Article  CAS  PubMed  Google Scholar 

  118. Wandel C, Kim R, Wood M, et al. Interaction of morphine, fentanyl, sufentanil, alfentanil, and loperamide with the efflux drug transporter P-glycoprotein. Anesthesiology 2002; 96(4): 913–20

    Article  CAS  PubMed  Google Scholar 

  119. Thompson SJ, Koszdin K, Bernards CM. Opiate-induced analgesia is increased and prolonged in mice lacking P-glycoprotein. Anesthesiology 2000; 92(5): 1392–9

    Article  CAS  PubMed  Google Scholar 

  120. Huwyler J, Drewe J, Klusemann C, et al. Evidence for P-glycoprotein-modulated penetration of morphine-6- glucuronide into brain capillary endothelium. Br J Pharmacol 1996; 118(8): 1879–85

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Lötsch J, Tegeder I, Angst MS, et al. Antinociceptive effects of morphine-6-glucuronide in homozygous MDR1a P-glycoprotein knockout and in wildtype mice in the hotplate test. Life Sci 2000; 66(24): 2393–403

    Article  PubMed  Google Scholar 

  122. Lötsch J, Schmidt R, Vetter G, et al. Increased CNS uptake and enhanced antinociception of morphine-6-glucuronide in rats after inhibition of P-glycoprotein. J Neurochem 2002; 83(2): 241–8

    Article  PubMed  Google Scholar 

  123. Drewe J, Ball HA, Beglinger C, et al. Effect of P-glycoprotein modulation on the clinical pharmacokinetics and adverse effects of morphine. Br J Clin Pharmacol 2000; 50(3): 237–46

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Sadeque AJ, Wandel C, He H, et al. Increased drug delivery to the brain by P-glycoprotein inhibition. Clin Pharmacol Ther 2000; 68(3): 231–7

    Article  CAS  PubMed  Google Scholar 

  125. Ueda K, Clark DP, Chen CJ, et al. The human multidrug resistance (MDR1) gene: cDNA cloning and transcription initiation. J Biol Chem 1987; 262(2): 505–8

    CAS  PubMed  Google Scholar 

  126. Hoffmeyer S, Burk O, von Richter O, et al. Functional polymorphisms of the human multidrug-resistance gene: multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc Natl Acad Sci U S A 2000; 97(7): 3473–8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Cascorbi I, Gerloff T, Johne A, et al. Frequency of single nucleotide polymorphisms in the P-glycoprotein drug transporter MDR1 gene in white subjects. Clin Pharmacol Ther 2001; 69(3): 169–74

    Article  CAS  PubMed  Google Scholar 

  128. Brinkmann U, Roots I, Eichelbaum M. Pharmacogenetics of the human drug-transporter gene MDR1: impact of polymorphisms on pharmacotherapy. Drug Discov Today 2001; 6(16): 835–9

    Article  CAS  PubMed  Google Scholar 

  129. Sakaeda T, Nakamura T, Okumura K. MDR1 genotype-related pharmacokinetics and pharmacodynamics. Biol Pharm Bull 2002; 25(11): 1391–400

    Article  CAS  PubMed  Google Scholar 

  130. Schwab M, Eichelbaum M, Fromm MF. Genetic polymorphisms of the human MDR1 drug transporter. Annu Rev Pharmacol Toxicol 2003; 43: 285–307

    Article  CAS  PubMed  Google Scholar 

  131. Ito S, Ieiri I, Tanabe M, et al. Polymorphism of the ABC transporter genes, MDR1, MRP1 and MRP2/cMOAT, in healthy Japanese subjects. Pharmacogenetics 2001; 11(2): 175–84

    Article  CAS  PubMed  Google Scholar 

  132. Tanabe M, Ieiri I, Nagata N, et al. Expression of P-glycoprotein in human placenta: relation to genetic polymorphism of the multidrug resistance (MDR)-1 gene. J Pharmacol Exp Ther 2001; 297(3): 1137–43

    CAS  PubMed  Google Scholar 

  133. Tang K, Ngoi SM, Gwee PC, et al. Distinct haplotype profiles and strong linkage disequilibrium at the MDR1 multidrug transporter gene locus in three ethnic Asian populations. Pharmacogenetics 2002; 12(6): 437–50

    Article  CAS  PubMed  Google Scholar 

  134. Siegmund W, Ludwig K, Giessmann T, et al. The effects of the human MDR1 genotype on the expression of duodenal P-glycoprotein and disposition of the probe drug talinolol. Clin Pharmacol Ther 2002; 72(5): 572–83

    Article  CAS  PubMed  Google Scholar 

  135. Roberts RL, Joyce PR, Mulder RT, et al. A common P-glycoprotein polymorphism is associated with nortriptylineinduced postural hypotension in patients treated for major depression. Pharmacogenomics J 2002; 2(3): 191–6

    Article  CAS  PubMed  Google Scholar 

  136. Schaeffeler E, Eichelbaum M, Brinkmann U, et al. Frequency of C3435T polymorphism of MDR1 gene in African people. Lancet 2001; 358(9279): 383–4

    Article  CAS  PubMed  Google Scholar 

  137. Hitzl M, Drescher S, et al. The C3435T mutation in the human MDR1 gene is associated with altered efflux of the P-glycoprotein substrate rhodamine 123 from CD56+ natural killer cells. Pharmacogenetics 2001; 11(4): 293–8

    Article  CAS  PubMed  Google Scholar 

  138. Kim RB, Leake BF, Choo EF, et al. Identification of functionally variant MDR1 alleles among European Americans and African Americans. Clin Pharmacol Ther 2001; 70(2): 189–99

    Article  CAS  PubMed  Google Scholar 

  139. Goto M, Masuda S, Saito H, et al. C3435T polymorphism in the MDR1 gene affects the enterocyte expression level of CYP3A4 rather than Pgp in recipients of living-donor liver transplantation. Pharmacogenetics 2002; 12(6): 451–7

    Article  CAS  PubMed  Google Scholar 

  140. Ameyaw MM, Regateiro F, Li T, et al. MDR1 pharmacogenetics: frequency of the C3435T mutation in exon 26 is significantly influenced by ethnicity. Pharmacogenetics 2001; 11(3): 217–21

    Article  CAS  PubMed  Google Scholar 

  141. Nauck M, Stein U, von Karger S, et al. Rapid detection of the C3435T polymorphism of multidrug resistance gene 1 using fluorogenic hybridization probes. Clin Chem 2000; 46(12): 1995–7

    CAS  PubMed  Google Scholar 

  142. Min DI, Ellingrod VL. C3435T mutation in exon 26 of the human MDR1 gene and cyclosporine pharmacokinetics in healthy subjects. Ther Drug Monit 2002; 24(3): 400–4

    Article  CAS  PubMed  Google Scholar 

  143. Darimont J, Skarke C, Grösch S, et al. Simple detection of single nucleotide polymorphisms C3935T of the MDR-1 gene and A1186 of the OPRM1 gene and allele frequencies among Frankfurt medical studies [abstract]. Naunyn Schmiedebergs Arch Pharmacol 2002; 365 Suppl. 1: R116

    Google Scholar 

  144. Wada M, Toh S, Taniguchi K, et al. Mutations in the canilicular multispecific organic anion transporter (cMOAT) gene, a novel ABC transporter, in patients with hyperbilirubinemia II/Dubin-Johnson syndrome. Hum Mol Genet 1998; 7(2): 203–7

    Article  CAS  PubMed  Google Scholar 

  145. Paulusma CC, Kool M, Bosma PJ, et al. A mutation in the human canalicular multispecific organic anion transporter gene causes the Dubin-Johnson syndrome. Hepatology 1997; 25(6): 1539–42

    Article  CAS  PubMed  Google Scholar 

  146. Mor-Cohen R, Zivelin A, Rosenberg N, et al. Identification and functional analysis of two novel mutations in the multidrug resistance protein 2 gene in Israeli patients with Dubin-Johnson syndrome. J Biol Chem 2001; 276(40): 36923–30

    Article  CAS  PubMed  Google Scholar 

  147. Tirona RG, Leake BF, Merino G, et al. Polymorphisms in OATP-C: identification of multiple allelic variants associated with altered transport activity among European- and African-Americans. J Biol Chem 2001; 276(38): 35669–75

    Article  CAS  PubMed  Google Scholar 

  148. Drescher S, Schaeffeler E, Hitzl M, et al. MDR1 gene polymorphisms and disposition of the P-glycoprotein substrate fexofenadine. Br J Clin Pharmacol 2002; 53(5): 526–34

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Fellay J, Marzolini C, Meaden ER, et al. Response to antiretroviral treatment in HIV-1-infected individuals with allelic variants of the multidrug resistance transporter 1: a pharmacogenetics study. Lancet 2002; 359(9300): 30–6

    Article  CAS  PubMed  Google Scholar 

  150. Sakaeda T, Nakamura T, Horinouchi M, et al. MDR1 genotyperelated pharmacokinetics of digoxin after single oral administration in healthy Japanese subjects. Pharm Res 2001; 18(10): 1400–4

    Article  CAS  PubMed  Google Scholar 

  151. Kimchi-Sarfaty C, Gribar JJ, Gottesman MM. Functional characterization of coding polymorphisms in the human MDR1 gene using a vaccinia virus expression system. Mol Pharmacol 2002; 62(1): 1–6

    Article  CAS  PubMed  Google Scholar 

  152. Moriya Y, Nakamura T, Horinouchi M, et al. Effects of polymorphisms of MDR1, MRP1, and MRP2 genes on their mRNA expression levels in duodenal enterocytes of healthy Japanese subjects. Biol Pharm Bull 2002; 25(10): 1356–9

    Article  CAS  PubMed  Google Scholar 

  153. Kurata Y, Ieiri I, Kimura M, et al. Role of human MDR1 gene polymorphism in bioavailability and interaction of digoxin, a substrate of P-glycoprotein. Clin Pharmacol Ther 2002; 72(2): 209–19

    Article  CAS  PubMed  Google Scholar 

  154. Johne A, Kopke K, Gerloff T, et al. Modulation of steady-state kinetics of digoxin by haplotypes of the P-glycoprotein MDR1 gene. Clin Pharmacol Ther 2002; 72(5): 584–94

    Article  CAS  PubMed  Google Scholar 

  155. Cole SP, Bhardwaj G, Gerlach JH, et al. Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line. Science 1992; 258(5088): 1650–4

    Article  CAS  PubMed  Google Scholar 

  156. Taniguchi K, Wada M, Kohno K, et al. A human canalicular multispecific organic anion transporter (cMOAT) gene is overexpressed in cisplatin-resistant human cancer cell lines with decreased drug accumulation. Cancer Res 1996; 56(18): 4124–9

    CAS  PubMed  Google Scholar 

  157. Konig J, Nies AT, Cui Y, et al. Conjugate export pumps of the multidrug resistance protein (MRP) family: localization, substrate specificity, and MRP2-mediated drug resistance. Biochim Biophys Acta 1999; 1461(2): 377–94

    Article  CAS  PubMed  Google Scholar 

  158. Flens MJ, Zaman GJ, van der Valk P, et al. Tissue distribution of the multidrug resistance protein. Am J Pathol 1996; 148(4): 1237–47

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Klein I, Sarkadi B, Varadi A. An inventory of the human ABC proteins. Biochim Biophys Acta 1999; 1461(2): 237–62

    Article  CAS  PubMed  Google Scholar 

  160. Borst P, Evers R, Kool M, et al. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst 2000; 92(16): 1295–302

    Article  CAS  PubMed  Google Scholar 

  161. Müller M. Human ATP-binding cassette transporters [online]. Available from URL: http://nutrigene.4t.com/humanabc.htm [Accesssed 2004 Aug 17]

  162. Sweet DH, Bush KT, Nigam SK. The organic anion transporter family: from physiology to ontogeny and the clinic. Am J Physiol Renal Physiol 2001; 281(2): F197–205

    CAS  PubMed  Google Scholar 

  163. Tamai I, Nozawa T, Koshida M, et al. Functional characterization of human organic anion transporting Polypeptide B (OATP-B) in comparison with liver-specific OATP-C. Pharm Res 2001; 18(9): 1262–9

    Article  CAS  PubMed  Google Scholar 

  164. Race JE, Grassl SM, Williams WJ, et al. Molecular cloning and characterization of two novel human renal organic anion transporters (hOAT1 and hOAT3). Biochem Biophys Res Commun 1999; 255(2): 508–14

    Article  CAS  PubMed  Google Scholar 

  165. Kullak-Ublick GA, Hagenbuch B, Stieger B, et al. Molecular and functional characterization of an organic anion transporting Polypeptide cloned from human liver. Gastroenterology 1995; 109(4): 1274–82

    Article  CAS  PubMed  Google Scholar 

  166. Kusuhara H, Sugiyama Y. Role of transporters in the tissueselective distribution and elimination of drugs: transporters in the liver, small intestine, brain and kidney. J Control Release 2002; 78(1–3): 43–54

    Article  CAS  PubMed  Google Scholar 

  167. Gao B, Hagenbuch B, Kullak-Ublick GA, et al. Organic aniontransporting Polypeptides mediate transport of Opioid peptides across blood-brain barrier. J Pharmacol Exp Ther 2000; 294(1): 73–9

    CAS  PubMed  Google Scholar 

  168. Haga S, Hinoshita E, Ikezaki K, et al. Involvement of the multidrug resistance protein 3 in drug sensitivity and its expression in human glioma. Jpn J Cancer Res 2001; 92(2): 211–9

    Article  CAS  PubMed  Google Scholar 

  169. Sugiyama Y, Kusuhara H, Suzuki H. Kinetic and biochemical analysis of carrier-mediated efflux of drugs through the blood-brain and blood-cerebrospinal fluid barriers: importance in the drug delivery to the brain. J Control Release 1999; 62(1–2): 179–86

    Article  CAS  PubMed  Google Scholar 

  170. Morin RA, Lyness WH. Potentiation of morphine analgesia after pretreatment with probenecid or sulfinpyrazone. Pharmacol Biochem Behav 1983; 18(6): 885–9

    Article  CAS  PubMed  Google Scholar 

  171. Leslie EM, Ito K, Upadhyaya P, et al. Transport of the β-O-glucuronide conjugate of the tobacco-specific carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-l-butanol (NNAL) by the multidrug resistance protein 1 (MRP1): requirement for glutathione or a non-sulfur-containing analog. J Biol Chem 2001; 276(30): 27846–54

    Article  CAS  PubMed  Google Scholar 

  172. Zelcer N, Saeki T, Reid G, et al. Characterization of drug transport by the human multidrug resistance protein 3 (ABCC3). J Biol Chem 2001; 276(49): 46400–7

    Article  CAS  PubMed  Google Scholar 

  173. Sugiyama D, Kusuhara H, Shitara Y, et al. Characterization of the efflux transport of 17β-estradiol-D-17β-glucuronide from the brain across the blood-brain barrier. J Pharmacol Exp Ther 2001; 298(1): 316–22

    CAS  PubMed  Google Scholar 

  174. Huang L, Smit JW, Meijer DK, et al. MRP2 is essential for estradiol-17β (β-D-glucuronide)-induced cholestasis in rats. Hepatology 2000; 32(1): 66–72

    Article  CAS  PubMed  Google Scholar 

  175. Keppler D, Leier I, Jedlitschky G. Transport of glutathione conjugates and glucuronides by the multidrug resistance proteins MRP1 and MRP2. Biol Chem 1997; 378(8): 787–91

    CAS  PubMed  Google Scholar 

  176. Lötsch J, Schmidt R, Vetter G, et al. The influence of inhibition of probenecid sensitive transporters on the central nervous system (CNS) uptake and the antinociceptive activity of morphine-6-glucuronide in rats. Neurosci Lett 2002; 329(2): 145–8

    Article  PubMed  Google Scholar 

  177. Xie R, Bouw MR, Hammarlund-Udenaes M. Modelling of the blood-brain barrier transport of morphine-3-glucuronide studied using microdialysis in the rat: involvement of probenecidsensitive transport. Br J Pharmacol 2000; 131(8): 1784–92

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Dubin IN, Johnson FB. Chronic idiopathic jaundice with unidentified pigment of liver cells: new clinico-pathologic entity with report of 12 cases. Medicine 1954; 33: 155–97

    Article  CAS  PubMed  Google Scholar 

  179. Saito S, Iida A, Sekine A, et al. Identification of 779 genetic variations in eight genes encoding members of the ATP-binding cassette, subfamily C (ABCC/MRP/CFTR). J Hum Genet 2002; 47(4): 147–71

    Article  CAS  PubMed  Google Scholar 

  180. Iida A, Saito S, Sekine A, et al. Catalog of 258 singlenucleotide polymorphisms (SNPs) in genes encoding three organic anion transporters, three organic anion-transporting Polypeptides, and three NADH: ubiquinone oxidoreductase flavoproteins. J Hum Genet 2001; 46(11): 668–83

    Article  CAS  PubMed  Google Scholar 

  181. Wang JB, Johnson PS, Persico AM, et al. Human μ opiate receptor: cDNA and genomic clones, pharmacologic characterization and chromosomal assignment. FEBS Lett 1994; 338(2): 217–22

    Article  CAS  PubMed  Google Scholar 

  182. LaForge KS, Yuferov V, Kreek MJ. Opioid receptor and peptide gene polymorphisms: potential implications for addictions. Eur J Pharmacol 2000; 410(2–3): 249–68

    Article  CAS  PubMed  Google Scholar 

  183. Befort K, Filliol D, Decaillot FM, et al. A single-nucleotide polymorphic mutation in the human μ-opioid receptor severely impairs receptor signaling. J Biol Chem 2001; 276(5): 3130–7

    Article  CAS  PubMed  Google Scholar 

  184. Hoehe MR, Kopke K, Wendel B, et al. Sequence variability and candidate gene analysis in complex disease: association of μ Opioid receptor gene variation with substance dependence. Hum Mol Genet 2000; 9(19): 2895–908

    Article  CAS  PubMed  Google Scholar 

  185. Koch T, Kroslak T, Averbeck M, et al. Allelic variation S268P of the human μ-opioid receptor affects both desensitization and G protein coupling. Mol Pharmacol 2000; 58(2): 328–34

    CAS  PubMed  Google Scholar 

  186. Wang D, Quillan JM, Winans K, et al. Single nucleotide polymorphisms in the human μ Opioid receptor gene alter basal G protein coupling and calmodulin binding. J Biol Chem 2001; 276(37): 34624–30

    Article  CAS  PubMed  Google Scholar 

  187. Uhl GR, Sora I, Wang Z. The μ opiate receptor as a candidate gene for pain: polymorphisms, variations in expression, nociception, and opiate responses. Proc Natl Acad Sci U S A 1999; 96(14): 7752–5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Bond C, LaForge KS, Tian M, et al. Single-nucleotide polymorphism in the human μ opioid receptor gene alters β-endorphin binding and activity: possible implications for opiate addiction. Proc Natl Acad Sci U S A 1998; 95(16): 9608–13

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Li T, Liu X, Zhu HZ, et al. Association analysis of polymorphism in the μ-opioid gene and heroin abuse in Chinese subjects. Addict Biol 2000; 5: 181–6

    Article  CAS  PubMed  Google Scholar 

  190. Gelernter J, Kranzler H, Cubells J. Genetics of two μ Opioid receptor gene (OPRM1) exon I polymorphisms: population studies, and allele frequencies in alcohol- and drug-dependent subjects. Mol Psychiatry 1999; 4(5): 476–83

    Article  CAS  PubMed  Google Scholar 

  191. Town T, Schinka J, Tan J, et al. The Opioid receptor system and alcoholism: a genetic perspective. Eur J Pharmacol 2000; 410(2–3): 243–8

    Article  CAS  PubMed  Google Scholar 

  192. Town T, Abdullah L, Crawford F, et al. Association of a functional μ-opioid receptor allele (+118A) with alcohol dependency. Am J Med Genet 1999; 88(5): 458–61

    Article  CAS  PubMed  Google Scholar 

  193. Bergen AW, Kokoszka J, Peterson R, et al. μ-Opioid receptor gene variants: lack of association with alcohol dependence. Mol Psychiatry 1997; 2(6): 490–4

    Article  CAS  PubMed  Google Scholar 

  194. Szeto CY, Tang NL, Lee DT, et al. Association between μ opioid receptor gene polymorphisms and Chinese heroin addicts. Neuroreport 2001; 12(6): 1103–6

    Article  CAS  PubMed  Google Scholar 

  195. Schinka JA, Town T, Abdullah L, et al. A functional polymorphism within the μ-opioid receptor gene and risk for abuse of alcohol and other substances. Mol Psychiatry 2002; 7(2): 224–8

    Article  CAS  PubMed  Google Scholar 

  196. Ohmori O, Shinkai T, Hori H, et al. Polymorphisms of μ and δ Opioid receptor genes and tardive dyskinesia in patients with schizophrenia. Schizophr Res 2001; 52(1–2): 137–8

    Article  CAS  PubMed  Google Scholar 

  197. Lötsch J, Skarke C, Grösch S, et al. The polymorphism A118G of the human μ-opioid receptor gene decreases the clinical activity of morphine-6-glucuronide but not that of morphine. Pharmacogenetics 2002; 12(1): 3–9

    Article  PubMed  Google Scholar 

  198. Tan EC, Tan CH, Karupathivan U, et al. μ-opioid receptor gene polymorphisms and heroin dependence in Asian populations. Neuroreport 2003; 14(4): 569–72

    Article  CAS  PubMed  Google Scholar 

  199. Shi J, Hui L, Xu Y, et al. Sequence variations in the μ-opioid receptor gene (OPRM1) associated with human addiction to heroin. Hum Mutat 2002; 19(4): 459–60

    Article  PubMed  CAS  Google Scholar 

  200. Mayer P, Rochlitz H, Rauch E, et al. Association between a 6 Opioid receptor gene polymorphism and heroin dependence in man. Neuroreport 1997; 8(11): 2547–50

    Article  CAS  PubMed  Google Scholar 

  201. Gelernter J, Kranzler HR. Variant detection at the 6 Opioid receptor (OPRD1) locus and population genetics of a novel variant affecting protein sequence. Hum Genet 2000; 107(1): 86–8

    Article  CAS  PubMed  Google Scholar 

  202. Beyer A, Koch T, Höllt V. A118G polymorphism does not alter the ligand binding and activity of the human μ-opioid receptor [abstract]. Naunyn Schmiedebergs Arch Pharmacol 2003; 367 Suppl. 1: R17

    Google Scholar 

  203. Angst MS, Bührer M, Lötsch J. Insidious intoxication after morphine treatment in renal failure: delayed onset of morphine-6-glucuronide action. Anesthesiology 2000; 92(5): 1473–6

    Article  CAS  PubMed  Google Scholar 

  204. Hasselström J, Berg U, Lofgren A, et al. Long lasting respiratory depression induced by morphine-6-glucuronide? Br J Clin Pharmacol 1989; 27: 515–8

    Article  PubMed  PubMed Central  Google Scholar 

  205. Bodd E, Jacobsen D, Lund E, et al. Morphine-6-glucuronide might mediate the prolonged Opioid effect of morphine in acute renal failure. Hum Exp Toxicol 1990; 9: 317–21

    Article  CAS  PubMed  Google Scholar 

  206. Caraco Y, Maroz Y, Davidson E. Variability in alfentanil analgesia maybe attributed to polymorphism in the μ-opioid receptor gene [abstract]. Clin Pharmacol Ther 2001; 69(2): 63

    Google Scholar 

  207. Hirota T, Ieiri I, Takane H, et al. Sequence variability and candidate gene analysis in two cancer patients with complex clinical outcomes during morphine therapy. Drug Metab Dispos 2003; 31(5): 677–80

    Article  CAS  PubMed  Google Scholar 

  208. Wand GS, McCaul M, Yang X, et al. The μ-opioid receptor gene polymorphism (A118G) alters HPA axis activation induced by Opioid receptor blockade. Neuropsychopharmacology 2002; 26(1): 106–14

    Article  CAS  PubMed  Google Scholar 

  209. Berrettini WH, Hoehe MR, Ferraro TN, et al. Human μ Opioid receptor gene polymorphisms and vulnerability to substance abuse. Addict Biol 1997; 2(3): 303–8

    Article  CAS  PubMed  Google Scholar 

  210. Franke P, Wang T, Nothen MM, et al. Nonreplication of association between μ-opioid-receptor gene (OPRM1) A118G polymorphism and substance dependence. Am J Med Genet 2001; 105(1): 114–9

    Article  CAS  PubMed  Google Scholar 

  211. Rossi GC, Leventhal L, Pan YX, et al. Antisense mapping of MOR-1 in rats: distinguishing between morphine and morphine-6β-glucuronide antinociception. J Pharmacol Exp Ther 1997; 281(1): 109–14

    CAS  PubMed  Google Scholar 

  212. Thompson TE, Rogan PK, Risinger JI, et al. Splice variants but not mutations of DNA Polymerase β are common in bladder cancer. Cancer Res 2002; 62(11): 3251–6

    CAS  PubMed  Google Scholar 

  213. Beyer KS, Klauck SM, Benner A, et al. Association studies of the HOPA dodecamer duplication variant in different subtypes of autism. Am J Med Genet 2002; 114(1): 110–5

    Article  PubMed  Google Scholar 

  214. De Souza EB, Schmidt WK, Kuhar MJ. Nalbuphine: an autoradiographic Opioid receptor binding profile in the central nervous system of an agonist/antagonist analgesic. J Pharmacol Exp Ther 1988; 244(1): 391–402

    PubMed  Google Scholar 

  215. Nozaki M, Niwa M, Hasegawa J, et al. Opioid receptor interactions of butorphanol, a narcotic antagonist analgesic, and its metabolites [in Japanese]. Nippon Yakurigaku Zasshi 1983; 82(6): 443–50

    Article  CAS  PubMed  Google Scholar 

  216. Goldstein A, Naidu A. Multiple Opioid receptors: ligand selectivity profiles and binding site signatures. Mol Pharmacol 1989; 36(2): 265–72

    CAS  PubMed  Google Scholar 

  217. Ross FB, Smith MT. The intrinsic antinociceptive effects of oxycodone appear to be κ-opioid receptor mediated. Pain 1997; 73(2): 151–7

    Article  CAS  PubMed  Google Scholar 

  218. Monory K, Greiner E, Sartania N, et al. Opioid binding profiles of new hydrazone, oxime, carbazone and semicarbazone derivatives of 14-alkoxymorphinans. Life Sci 1999; 64(22): 2011–20

    Article  CAS  PubMed  Google Scholar 

  219. Yasuda K, Espinosa III R, Takeda J, et al. Localization of the κ Opioid receptor gene to human chromosome band 8q11.2. Genomics 1994; 19(3): 596–7

    Article  CAS  PubMed  Google Scholar 

  220. Simonin F, Gaveriaux-Ruff C, Befort K, et al. κ-Opioid receptor in humans: cDNA and genomic cloning, chromosomal assignment, functional expression, pharmacology, and expression pattern in the central nervous system. Proc Natl Acad Sci U S A 1995; 92(15): 7006–10

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Franke P, Nothen MM, Wang T, et al. Human δ-opioid receptor gene and susceptibility to heroin and alcohol dependence. Am J Med Genet 1999; 88(5): 462–4

    Article  CAS  PubMed  Google Scholar 

  222. Bzdega T, Chin H, Kim H, et al. Regional expression and chromosomal localization of the δ opiate receptor gene. Proc Natl Acad Sci U S A 1993; 90(20): 9305–9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Simonin F, Befort K, Gaveriaux-Ruff C, et al. The human δ-opioid receptor: genomic organization, cDNA cloning, functional expression, and distribution in human brain. Mol Pharmacol 1994; 46(6): 1015–21

    CAS  PubMed  Google Scholar 

  224. Bergen AW, Van Den Bree MB, Yeager M, et al. Candidate genes for anorexia nervosa in the 1p33-36 linkage region: serotonin 1D and δ Opioid receptor loci exhibit significant association to anorexia nervosa. Mol Psychiatry 2003; 8(4): 397–406

    Article  CAS  PubMed  Google Scholar 

  225. Halfpenny DM, Callado LF, Hopwood SE, et al. Effects of tramadol stereoisomers on norepinephrine efflux and uptake in the rat locus coeruleus measured by real time voltammetry. Br J Anaesth 1999; 83(6): 909–15

    Article  CAS  PubMed  Google Scholar 

  226. Oliva P, Aurilio C, Massimo F, 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  CAS  PubMed  Google Scholar 

  227. Garrido MJ, Valle M, Campanero MA, et al. Modeling of the in vivo antinociceptive interaction between an Opioid agonist, (+)-O-desmethyltramadol, and a monoamine reuptake inhibitor, (−)-O-desmethyltramadol, in rats. J Pharmacol Exp Ther 2000; 295(1): 352–9

    CAS  PubMed  Google Scholar 

  228. Gobbi M, Mennini T. Release studies with rat brain cortical synaptosomes indicate that tramadol is a 5-hydroxytryptamine uptake blocker and not a 5-hydroxytryptamine releaser. Eur J Pharmacol 1999; 370(1): 23–6

    Article  CAS  PubMed  Google Scholar 

  229. Raffa RB, Friderichs E, Reimann W, et al. Complementary and synergistic antinociceptive interaction between the enantiomers of tramadol. J Pharmacol Exp Ther 1993; 267(1): 331–40

    CAS  PubMed  Google Scholar 

  230. Bamigbade TA, Davidson C, Langford RM, et al. Actions of tramadol, its enantiomers and principal metabolite, O-desmethyltramadol, on serotonin (5-HT) efflux and uptake in the rat dorsal raphe nucleus. Br J Anaesth 1997; 79(3): 352–6

    Article  CAS  PubMed  Google Scholar 

  231. Arcioni R, della Rocca M, Romano S, et al. 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

    Article  CAS  PubMed  Google Scholar 

  232. De Witte JL, Schoenmaekers B, Sessler DI, et al. The analgesic efficacy of tramadol is impaired by concurrent administration of ondansetron. Anesth Analg 2001; 92(5): 1319–21

    Article  PubMed  Google Scholar 

  233. Rojas-Corrales MO, Ortega-Alvaro A, Gibert-Rahola J, et al. Pindolol, a β-adrenoceptor blocker/5-hydroxytryptamine (1A/1B) antagonist, enhances the analgesic effect of tramadol. Pain 2000; 88(2): 119–24

    Article  CAS  PubMed  Google Scholar 

  234. Codd EE, Shank RP, Schupsky JJ, et al. Serotonin and norepinephrine uptake inhibiting activity of centrally acting analgesics: structural determinants and role in antinociception. J Pharmacol Exp Ther 1995; 274(3): 1263–70

    CAS  PubMed  Google Scholar 

  235. Nemmani KV, Gullapalli S, Ramarao P. Potentiation of κ-opioid receptor agonist-induced analgesia and hypothermia by fluoxetine. Pharmacol Biochem Behav 2001; 69(1–2): 189–93

    Article  CAS  PubMed  Google Scholar 

  236. Nayebi AR, Hassanpour M, Rezazadeh H. Effect of chronic and acute administration of fluoxetine and its additive effect with morphine on the behavioural response in the formalin test in rats. J Pharm Pharmacol 2001; 53(2): 219–25

    Article  CAS  PubMed  Google Scholar 

  237. Luger TJ, Lorenz IH, Grabner-Weiss C, et al. Effect of fluvoxamine on sufentanil antinociception and tolerance under chronic intravenous infusion in rats. Pharmacol Toxicol 1999; 85(6): 263–8

    Article  CAS  PubMed  Google Scholar 

  238. Wang YX, Bowersox SS, Pettus M, et al. Antinociceptive properties of fenfluramine, a serotonin reuptake inhibitor, in a rat model of neuropathy. J Pharmacol Exp Ther 1999; 291(3): 1008–16

    CAS  PubMed  Google Scholar 

  239. Tulunay FC, Yano I, Takemori AE. The effect of biogenic amine modifiers on morphine analgesia and its antagonism by naloxone. Eur J Pharmacol 1976; 35: 285–92

    Article  CAS  PubMed  Google Scholar 

  240. Tenen SS. Antagonism of the analgesic effect of morphine and other drugs by p-chlorophenylalanine, a serotonin depletor. Psychopharmacologia 1968; 12: 278–85

    Article  CAS  PubMed  Google Scholar 

  241. Tenen SS. The effects of p-chlorophenylalanine, a serotonin depletor, on avoidance acquisition, pain sensitivity and related behavior in the rat. Psychopharmacologia 1967; 10: 204–19

    Article  CAS  PubMed  Google Scholar 

  242. Ho IK, Brase DA, Loh HH, et al. Influence of L-tryptophan on morphine analgesia, tolerance and physical dependence. J Pharmacol Exp Ther 1975; 193: 35–43

    CAS  PubMed  Google Scholar 

  243. Abbott FV, Young SN. Effect of 5-hydroxytryptamine precursors on morphine analgesia in the formalin test. Pharmacol Biochem Behav 1989; 31: 855–60

    Article  Google Scholar 

  244. Coda BA, Hill HF, Schaffer RL, et al. Enhancement of morphine analgesia by fenfluramine in subjects receiving tailored Opioid infusions. Pain 1993; 52(1): 85–91

    Article  CAS  PubMed  Google Scholar 

  245. Erjavec MK, Coda BA, Nguyen Q, et al. Morphine-fluoxetine interactions in healthy volunteers: analgesia and side effects. J Clin Pharmacol 2000; 40(11): 1286–95

    CAS  PubMed  Google Scholar 

  246. Gordon NC, Heller PH, Gear RW, et al. Interactions between fluoxetine and opiate analgesia for postoperative dental pain. Pain 1994; 58: 85–8

    Article  CAS  PubMed  Google Scholar 

  247. Abbott FV, Etienne P, Franklin KB, et al. Acute tryptophan depletion blocks morphine analgesia in the cold-pressor test in humans. Psychopharmacology (Berl) 1992; 108: 60–6

    Article  CAS  Google Scholar 

  248. Hosobuchi Y, Lamb S, Baskin D. Tryptophan loading may reverse tolerance to opiate analgesics in humans: a preliminary report. Pain 1980; 9: 161–9

    Article  CAS  PubMed  Google Scholar 

  249. Staner L, Uyanik G, Correa H, et al. A dimensional impulsiveaggressive phenotype is associated with the A218C polymorphism of the tryptophan hydroxylase gene: a pilot study in well-characterized impulsive inpatients. Am J Med Genet 2002; 114(5): 553–7

    Article  PubMed  Google Scholar 

  250. Ebstein RP, Benjamin J, Belmaker RH. Personality and polymorphisms of genes involved in aminergic neurotransmission. Eur J Pharmacol 2000; 410(2–3): 205–14

    Article  CAS  PubMed  Google Scholar 

  251. Arinami T, Ishiguro H, Onaivi ES. Polymorphisms in genes involved in neurotransmission in relation to smoking. Eur J Pharmacol 2000; 410(2–3): 215–26

    Article  CAS  PubMed  Google Scholar 

  252. Weizman A, Weizman R. Serotonin transporter polymorphism and response to SSRIs in major depression and relevance to anxiety disorders and substance abuse. Pharmacogenomics 2000; 1(3): 335–41

    Article  CAS  PubMed  Google Scholar 

  253. Blakely RD. Physiological genomics of antidepressant targets: keeping the periphery in mind. J Neurosci 2001; 21(21): 8319–23

    CAS  PubMed  Google Scholar 

  254. Catalano M. Functionally gene-linked polymorphic regions and genetically controlled neurotransmitters metabolism. Eur Neuropsychopharmacol 2001; 11(6): 431–9

    Article  CAS  PubMed  Google Scholar 

  255. Du L, Faludi G, Palkovits M, et al. Serotonergic genes and suicidality. Crisis 2001; 22(2): 54–60

    Article  CAS  PubMed  Google Scholar 

  256. Arranz MJ, Munro J, Owen MJ, et al. Evidence for association between polymorphisms in the promoter and coding regions of the 5-HT2A receptor gene and response to Clozapine. Mol Psychiatry 1998; 3(1): 61–6

    Article  CAS  PubMed  Google Scholar 

  257. Arranz M, Collier D, Sodhi M, et al. Association between Clozapine response and allelic variation in 5-HT2A receptor gene. Lancet 1995; 346(8970): 281–2

    Article  CAS  PubMed  Google Scholar 

  258. Arranz MJ, Collier DA, Munro J, et al. Analysis of a structural polymorphism in the 5-HT2A receptor and clinical response to Clozapine. Neurosci Lett 1996; 217(2–3): 177–8

    Article  CAS  PubMed  Google Scholar 

  259. Reynolds GP, Zhang ZJ, Zhang XB. Association of antipsychotic drug-induced weight gain with a 5-HT2C receptor gene polymorphism. Lancet 2002; 359(9323): 2086–7

    Article  CAS  PubMed  Google Scholar 

  260. Ramamoorthy S, Bauman AL, Moore KR, et al. Antidepressant- and cocaine-sensitive human serotonin transporter: molecular cloning, expression, and chromosomal localization. Proc Natl Acad Sci U S A 1993; 90(6): 2542–6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  261. Herken H, Erdal E, Mutlu N, et al. Possible association of temporomandibular joint pain and dysfunction with a polymorphism in the serotonin transporter gene. Am J Orthod Dentofacial Orthop 2001; 120(3): 308–13

    Article  CAS  PubMed  Google Scholar 

  262. Heils A, Teufel A, Petri S, et al. Allelic variation of human serotonin transporter gene expression. J Neurochem 1996; 66(6): 2621–4

    Article  CAS  PubMed  Google Scholar 

  263. Offenbaecher M, Bondy B, de Jonge S, et al. Possible association of fibromyalgia with a polymorphism in the serotonin transporter gene regulatory region. Arthritis Rheum 1999; 42(11): 2482–8

    Article  CAS  PubMed  Google Scholar 

  264. Sparkes RS, Lan N, Klisak I, et al. Assignment of a serotonin 5HT-2 receptor gene (HTR2) to human chromosome 13q14-q21 and mouse chromosome 14. Genomics 1991; 9(3): 461–5

    Article  CAS  PubMed  Google Scholar 

  265. Bondy B, Spaeth M, Offenbaecher M, et al. The T102C polymorphism of the 5-HT2A-receptor gene in fibromyalgia. Neurobiol Dis 1999; 6(5): 433–9

    Article  CAS  PubMed  Google Scholar 

  266. Lubov U, Alfimova M, Golimbet V. Pain thresholds and serotonin receptor 2A gene polymorphism in schizophrenic families [abstract]. Eur Psychiatry 2002; 17 Suppl. 1: 172

    Article  Google Scholar 

  267. Desmeules JA, Piguet V, Collart L, et al. Contribution of monoaminergic modulation to the analgesic effect of tramadol. Br J Clin Pharmacol 1996; 41(1): 7–12

    Article  CAS  PubMed  Google Scholar 

  268. Small KM, Liggett SB. Identification and functional characterization of α (2)-adrenoceptor polymorphisms. Trends Pharmacol Sci 2001; 22(9): 471–7

    Article  CAS  PubMed  Google Scholar 

  269. Pacholczyk T, Blakely RD, Amara SG. Expression cloning of a cocaine- and antidepressant-sensitive human noradrenaline transporter. Nature 1991; 350(6316): 350–4

    Article  CAS  PubMed  Google Scholar 

  270. Gorman AL, Elliott KJ, Inturrisi CE. The d- and 1-isomers of methadone bind to the non-competitive site on the N-methyl-D-aspartate (NMDA) receptor in rat forebrain and spinal cord. Neurosci Lett 1997; 223(1): 5–8

    Article  CAS  PubMed  Google Scholar 

  271. Choi DW, Viseskul V. Opioids and non-opioid enantiomers selectively attenuate N-methyl-D-aspartate neurotoxicity on cortical neurons. Eur J Pharmacol 1988; 155(1–2): 27–35

    Article  CAS  PubMed  Google Scholar 

  272. Bulka A, Wiesenfeld-Hallin Z, Xu XJ. Differential antinociception by morphine and methadone in two sub-strains of Sprague-Dawley rats and its potentiation by dextromethorphan. Brain Res 2002; 942(1–2): 95–100

    Article  CAS  PubMed  Google Scholar 

  273. Oxenham D, Farrer K. Methadone: Opioid, N-methyl-D-aspartate antagonist or both [letter]. Palliat Med 1998; 12(4): 302

    Article  CAS  PubMed  Google Scholar 

  274. Yamakura T, Sakimura K, Shimoji K. N-methyl-D-aspartate receptor channel block by meperidine is dependent on extracellular pH. Anesth Analg 2000; 90(4): 928–32

    Article  CAS  PubMed  Google Scholar 

  275. Ebert B, Andersen S, Krogsgaard-Larsen P. Ketobemidone, methadone and pethidine are non-competitive N-methyl-D-aspartate (NMDA) antagonists in the rat cortex and spinal cord. Neurosci Lett 1995; 187(3): 165–8

    Article  CAS  PubMed  Google Scholar 

  276. Davis AM, Inturrisi CE. d-Methadone blocks morphine tolerance and N-methyl-D-aspartate-induced hyperalgesia. J Pharmacol Exp Ther 1999; 289(2): 1048–53

    CAS  PubMed  Google Scholar 

  277. Chizh BA, Schlutz H, Scheede M, et al. The N-methyl-D-aspartate antagonistic and Opioid components of d-methadone antinociception in the rat spinal cord. Neurosci Lett 2000; 296(2–3): 117–20

    Article  CAS  PubMed  Google Scholar 

  278. Stringer M, Makin MK, Miles J, et al. d-Morphine, but not 1-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 2000; 295(1–2): 21–4

    Article  CAS  PubMed  Google Scholar 

  279. Ebert B, Thorkildsen C, Andersen S, et al. Opioid analgesics as noncompetitive N-methyl-D-aspartate (NMDA) antagonists. Biochem Pharmacol 1998; 56(5): 553–9

    Article  CAS  PubMed  Google Scholar 

  280. Tsai SJ, Liu HC, Liu TY, et al. Association analysis for genetic variants of the NMDA receptor 2b subunit (GRIN2B) and Parkinson’s disease. J Neural Transm 2002; 109(4): 483–8

    Article  CAS  PubMed  Google Scholar 

  281. Rice SR, Niu N, Berman DB, et al. Identification of single nucleotide polymorphisms (SNPs) and other sequence changes and estimation of nucleotide diversity in coding and flanking regions of the NMDAR1 receptor gene in schizophrenic patients. Mol Psychiatry 2001; 6(3): 274–84

    Article  CAS  PubMed  Google Scholar 

  282. Sakurai K, Toru M, Yamakawa-Kobayashi K, et al. Mutation analysis of the N-methyl-D-aspartate receptor NR1 subunit gene (GRIN1) in schizophrenia. Neurosci Lett 2000; 296(2–3): 168–70

    Article  CAS  PubMed  Google Scholar 

  283. Nishiguchi N, Shirakawa O, Ono H, et al. Novel polymorphism in the gene region encoding the carboxyl-terminal intracellular domain of the NMDA receptor 2B subunit: analysis of association with schizophrenia. Am J Psychiatry 2000; 157(8): 1329–31

    Article  CAS  PubMed  Google Scholar 

  284. Williams NM, Bowen T, Spurlock G, et al. Determination of the genomic structure and mutation screening in schizophrenic individuals for five subunits of the N-methyl-D-aspartate glutamate receptor. Mol Psychiatry 2002; 7(5): 508–14

    Article  CAS  PubMed  Google Scholar 

  285. Monyer H, Sprengel R, Schoepfer R, et al. Heteromeric NMDA receptors: molecular and functional distinction of subtypes. Science 1992; 256(5060): 1217–21

    Article  CAS  PubMed  Google Scholar 

  286. Ohtsuki T, Sakurai K, Dou H, et al. Mutation analysis of the NMDAR2B (GRIN2B) gene in schizophrenia. Mol Psychiatry 2001; 6(2): 211–6

    Article  CAS  PubMed  Google Scholar 

  287. Sander T, Ostapowicz A, Samochowiec J, et al. Genetic variation of the glutamate transporter EAAT2 gene and vulnerability to alcohol dependence. Psychiatr Genet 2000; 10(3): 103–7

    Article  CAS  PubMed  Google Scholar 

  288. Mogil JS, Wilson SG, Bon K, et al. Heritability of nociception I: responses of 11 inbred mouse strains on 12 measures of nociception. Pain 1999; 80(1–2): 67–82

    Article  CAS  PubMed  Google Scholar 

  289. Mogil JS, Wilson SG, Bon K, et al. Heritability of nociception II; ‘types’ of nociception revealed by genetic correlation analysis. Pain 1999; 80(1–2): 83–93

    Article  CAS  PubMed  Google Scholar 

  290. Mogil JS. The genetic mediation of individual differences in sensitivity to pain and its inhibition. Proc Natl Acad Sci U S A 1999; 96(14): 7744–51

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  291. Desta Z, Zhao X, Shin JG, et al. Clinical significance of the cytochrome P450 2C19 genetic polymorphism. Clin Pharmacokinet 2002; 41(12): 913–58

    Article  CAS  PubMed  Google Scholar 

  292. Xie HG, Kim RB, Wood AJ, et al. Molecular basis of ethnic differences in drug disposition and response. Annu Rev Pharmacol Toxicol 2001; 41: 815–50

    Article  CAS  PubMed  Google Scholar 

  293. Wood AJ. Ethnic differences in drug disposition and response. Ther Drug Monit 1998; 20(5): 525–6

    Article  CAS  PubMed  Google Scholar 

  294. Llerena A, Cobaleda J, Martinez C, et al. Interethnic differences in drug metabolism: influence of genetic and environmental factors on debrisoquine hydroxylation phenotype. Eur J Drug Metab Pharmacokinet 1996; 21(2): 129–38

    Article  CAS  PubMed  Google Scholar 

  295. Gaedigk A. Interethnic differences of drug-metabolizing enzymes. Int J Clin Pharmacol Ther 2000; 38(2): 61–8

    Article  CAS  PubMed  Google Scholar 

  296. Lemmens HJ, Bovill JG, Hennis PJ, et al. Alcohol consumption alters the pharmacodynamics of alfentanil. Anesthesiology 1989; 71(5): 669–74

    Article  CAS  PubMed  Google Scholar 

  297. Harada S, Agarwal DP, Nomura F, et al. Metabolic and ethnic determinants of alcohol drinking habits and vulnerability to alcohol-related disorder. Alcohol Clin Exp Res 2001; 25(5 Suppl. ISBRA): 71S–5S

    Article  CAS  PubMed  Google Scholar 

  298. Dahmani S, Dupont H, Mantz J, et al. Predictive factors of early morphine requirements in the post-anaesthesia care unit (PACU). Br J Anaesth 2001; 87(3): 385–9

    Article  CAS  PubMed  Google Scholar 

  299. Faucett J, Gordon N, Levine J. Differences in postoperative pain severity among four ethnic groups. J Pain Symptom Manage 1994; 9(6): 383–9

    Article  CAS  PubMed  Google Scholar 

  300. Lipton JA, Marbach JJ. Ethnicity and the pain experience. Soc Sci Med 1984; 19(12): 1279–98

    Article  CAS  PubMed  Google Scholar 

  301. Greenwald HP. Interethnic differences in pain perception. Pain 1991; 44(2): 157–63

    Article  CAS  PubMed  Google Scholar 

  302. Weisenberg M, Caspi Z. Cultural and educational influences on pain of childbirth. J Pain Symptom Manage 1989; 4(1): 13–9

    Article  CAS  PubMed  Google Scholar 

  303. Mogil JS, Kest B. Sex differences in Opioid analgesia: of mice and women. Pain Forum 1999; 8(1): 48–50

    Article  Google Scholar 

  304. Kest B, Sarton E, Dahan A. Gender differences in opioidmediated analgesia: animal and human studies. Anesthesiology 2000; 93(2): 539–47

    Article  CAS  PubMed  Google Scholar 

  305. Schwartz JB. Gender differences in response to drugs: pain medications. J Gend Specif Med 1999; 2(5): 28–30

    CAS  PubMed  Google Scholar 

  306. Miaskowski C, Levine JD. Does Opioid analgesia show a gender preference for females? Pain Forum 1999; 8: 34–44

    Article  Google Scholar 

  307. Gear RW, Miaskowski C, Gordon NC, et al. The κ Opioid nalbuphine produces gender- and dose-dependent analgesia and anti-analgesia in patients with postoperative pain. Pain 1999; 83(2): 339–45

    Article  CAS  PubMed  Google Scholar 

  308. Gear RW, Miaskowski C, Gordon NC, et al. κ-Opioids produce significantly greater analgesia in women than in men. Nat Med 1996; 2(11): 1248–50

    Article  CAS  PubMed  Google Scholar 

  309. Sarton E, Olofsen E, Romberg R, et al. Sex differences in morphine analgesia: an experimental study in healthy volunteers. Anesthesiology 2000; 93(5): 1245–54

    Article  CAS  PubMed  Google Scholar 

  310. Pleuvry BJ, Maddison SE. A sex difference in the effects of oral codeine and Promethazine on the ventilatory response to carbon dioxide in human volunteers. Br J Clin Pharmacol 1980; 9(2): 159–64

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  311. Dahan A, Sarton E, Teppema L, et al. Sex-related differences in the influence of morphine on ventilatory control in humans. Anesthesiology 1998; 88(4): 903–13

    Article  CAS  PubMed  Google Scholar 

  312. Zubieta JK, Dannals RF, Frost JJ. Gender and age influences on human brain μ-opioid receptor binding measured by PET. Am J Psychiatry 1999; 156(6): 842–8

    Article  CAS  PubMed  Google Scholar 

  313. Zubieta JK, Smith YR, Bueller JA, et al. Mu-opioid receptormediated antinociceptive responses differ in men and women. J Neurosci 2002; 22(12): 5100–7

    CAS  PubMed  Google Scholar 

  314. Unruh AM. Gender variations in clinical pain experience. Pain 1996; 65(2–3): 123–67

    Article  CAS  PubMed  Google Scholar 

  315. Fernandez-Real JM, Penarroja G, Richart C, et al. G protein β3 gene variant, vascular function, and insulin sensitivity in type 2 diabetes. Hypertension 2003; 41(1): 124–9

    Article  CAS  PubMed  Google Scholar 

  316. Stitham J, Stojanovic A, Hwa J. Impaired receptor binding and activation associated with a human prostacyclin receptor polymorphism. J Biol Chem 2002; 277(18): 15439–44

    Article  CAS  PubMed  Google Scholar 

  317. Chang AC, Cochet M, Cohen SN. Structural organization of human genomic DNA encoding the pro-opiomelanocortin peptide. Proc Natl Acad Sci U S A 1980; 77(8): 4890–4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  318. Comb M, Seeburg PH, Adelman J, et al. Primary structure of the human Met- and Leu-enkephalin precursor and its mRNA. Nature 1982; 295(5851): 663–6

    Article  CAS  PubMed  Google Scholar 

  319. Noda M, Teranishi Y, Takahashi H, et al. Isolation and structural organization of the human preproenkephalin gene. Nature 1982; 297(5865): 431–4

    Article  CAS  PubMed  Google Scholar 

  320. Esapa CT, Harris PE. Mutation analysis of protein kinase A catalytic subunit in thyroid adenomas and pituitary tumours. Eur J Endocrinol 1999; 141(4): 409–12

    Article  CAS  PubMed  Google Scholar 

  321. Ulrich CM, Bigler J, Sibert J, et al. Cyclooxygenase 1 (COX1) polymorphisms in African-American and Caucasian populations. Hum Mutat 2002; 20(5): 409–10

    Article  PubMed  CAS  Google Scholar 

  322. Cipollone F, Patrono C. Cyclooxygenase-2 polymorphism: putting a brake on the inflammatory response to vascular injury? Arterioscler Thromb Vasc Biol 2002; 22(10): 1516–8

    Article  PubMed  Google Scholar 

  323. Halushka MK, Walker LP, Halushka PV. Genetic variation in cyclooxygenase 1: effects on response to aspirin. Clin Pharmacol Ther 2003; 73(1): 122–30

    Article  CAS  PubMed  Google Scholar 

  324. Kanai N, Lu R, Satriano JA, et al. Identification and characterization of a Prostaglandin transporter. Science 1995; 268(5212): 866–9

    Article  CAS  PubMed  Google Scholar 

  325. Samad TA, Sapirstein A, Woolf CJ. Prostanoids and pain: unraveling mechanisms and revealing therapeutic targets. Trends Mol Med 2002; 8(8): 390–6

    Article  CAS  PubMed  Google Scholar 

  326. Sander T, Berlin W, Ostapowicz A, et al. Variation of the genes encoding the human glutamate EAAT2, serotonin and dopamine transporters and susceptibility to idiopathic generalized epilepsy. Epilepsy Res 2000; 41(1): 75–81

    Article  CAS  PubMed  Google Scholar 

  327. Wu D, Otton SV, Sproule BA, et al. Inhibition of human cytochrome P450 2D6 (CYP2D6) by methadone. Br J Clin Pharmacol 1993; 35(1): 30–4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  328. Pauli-Magnus C, Rekersbrink S, Klotz U, et al. Interaction of omeprazole, lansoprazole and pantoprazole with P-glycoprotein. Naunyn Schmiedebergs Arch Pharmacol 2001; 364(6): 551–7

    Article  CAS  PubMed  Google Scholar 

  329. Perault MC, Bouquet S, Bertschy G, et al. Debrisoquine and dextromethorphan phenotyping and antidepressant treatment. Therapie 1991; 46(1): 1–3

    CAS  PubMed  Google Scholar 

  330. Weinbroum AA. Dextromethorphan reduces immediate and late postoperative analgesic requirements and improves patients’ subjective scorings after epidural lidocaine and general anesthesia. Anesth Analg 2002; 94(6): 1547–52

    Article  CAS  PubMed  Google Scholar 

  331. Ilkjaer S, Bach LF, Nielsen PA, et al. Effect of preoperative oral dextromethorphan on immediate and late postoperative pain and hyperalgesia after total abdominal hysterectomy. Pain 2000; 86(1–2): 19–24

    Article  CAS  PubMed  Google Scholar 

  332. Henderson DJ, Withington BS, Wilson JA, et al. Perioperative dextromethorphan reduces postoperative pain after hysterectomy. Anesth Analg 1999; 89(2): 399–402

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge funding from Deutsche Forschungsgemeinschaft, Bonn, Germany (DFG Lo 612/3-3) and Dr Robert Pfleger Stiftung, Bamberg, Germany. The authors have no conflicts of interest directly relevant to the content of this review.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jörn Lötsch.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lötsch, J., Skarke, C., Liefhold, J. et al. Genetic Predictors of the Clinical Response to Opioid Analgesics. Clin Pharmacokinet 43, 983–1013 (2004). https://doi.org/10.2165/00003088-200443140-00003

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003088-200443140-00003

Keywords

Navigation