Skip to main content

Pharmacogenetics of Addiction Therapy

  • Protocol
  • First Online:
Pharmacogenomics in Drug Discovery and Development

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1175))

Abstract

Drug addiction is a serious relapsing disease that has high costs to society and to the individual addicts. Treatment of these addictions is still in its nascency, with only a few examples of successful therapies. Therapeutic response depends upon genetic, biological, social, and environmental components. A role for genetic makeup in the response to treatment has been shown for several addiction pharmacotherapies. For several addiction pharmacotherapies, response to treatment varies based on individual genetic makeup. In this chapter, we discuss the role of genetics in pharmacotherapies, specifically for cocaine, alcohol, and opioid dependences. The elucidation of the role of genetics should aid in the development of new treatments and increase the efficacy of existing treatments.

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

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 89.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 119.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. SAMHSA (2011) Results from the 2010 National Survey on Drug Use and Health (NSDUH). NSDUH Series H-41. Rockville, MD

    Google Scholar 

  2. Wolf BC, Lavezzi WA, Sullivan LM, Flannagan LM (2004) Methadone-related deaths in Palm Beach County. J Forensic Sci 49:375–378

    CAS  PubMed  Google Scholar 

  3. MacGowan RJ, Fichtner RR, Swanson N, Collier C, Kroliczak A, Cole G (1997) Factors associated with client-reported HIV infection among clients entering methadone treatment. AIDS Educ Prev 9:205–217

    CAS  PubMed  Google Scholar 

  4. Meandzija B, O’Connor PG, Fitzgerald B, Rounsaville BJ, Kosten TR (1994) HIV infection and cocaine use in methadone maintained and untreated intravenous drug users. Drug Alcohol Depend 36:109–113

    CAS  PubMed  Google Scholar 

  5. Horgan C, Skwara KC, Strickler G, Andersen L (2001) Substance abuse: the nation’s number one health problem. Schneider Institute for Health Policy, Brandeis University (prepared for the Robert Wood Johnson Foundation), Waltham, MA

    Google Scholar 

  6. Fishbain DA, Cole B, Lewis J, Rosomoff HL, Rosomoff RS (2008) What percentage of chronic nonmalignant pain patients exposed to chronic opioid analgesic therapy develop abuse/addiction and/or aberrant drug-related behaviors? A structured evidence-based review. Pain Med 9:444–459

    PubMed  Google Scholar 

  7. Kreek MJ, Nielsen DA, Butelman ER, LaForge KS (2005) Genetic influences on impulsivity, risk taking, stress responsivity and vulnerability to drug abuse and addiction. Nat Neurosci 8:1450–1457

    CAS  PubMed  Google Scholar 

  8. Haile CN, Kosten TR, Kosten TA (2009) Pharmacogenetic treatments for drug addiction: cocaine, amphetamine and methamphetamine. Am J Drug Alcohol Abuse 35:161–177

    PubMed Central  PubMed  Google Scholar 

  9. Malhotra AK, Murphy GM Jr, Kennedy JL (2004) Pharmacogenetics of psychotropic drug response. Am J Psychiatry 161:780–796

    PubMed  Google Scholar 

  10. Murphy GM Jr, Kremer C, Rodrigues HE, Schatzberg AF (2003) Pharmacogenetics of antidepressant medication intolerance. Am J Psychiatry 160:1830–1835

    PubMed  Google Scholar 

  11. Gupta S, Jain S, Brahmachari SK, Kukreti R (2006) Pharmacogenomics: a path to predictive medicine for schizophrenia. Pharmacogenomics 7:31–47

    CAS  PubMed  Google Scholar 

  12. deLeon J, Armstrong SC, Cozza KL (2006) Clinical guidelines for psychiatrists for the use of pharmacogenetic testing for CYP450 and CYP450 2C19. Psychosomatics 47:75–85

    Google Scholar 

  13. Rogers JF, Nafziger AN, Bertino JS Jr (2002) Pharmacogenetics affects dosing, efficacy, and toxicity of cytochrome P450-metabolized drugs. Am J Med 113:746–750

    CAS  PubMed  Google Scholar 

  14. Consortium EP, Bernstein BE, Birney E, Dunham I, Green ED, Gunter C, Snyder M (2012) An integrated encyclopedia of DNA elements in the human genome. Nature 489:57–74

    Google Scholar 

  15. 1000 Genomes Project Consortium, Abecasis GR, Auton A, Brooks LD, DePristo MA, Durbin RM, Handsaker RE, Kang HM, Marth GT, McVean GA (2012) An integrated map of genetic variation from 1,092 human genomes. Nature 491:56–65

    PubMed  Google Scholar 

  16. 1000 Genomes Project Consortium, Abecasis GR, Altshuler D, Auton A, Brooks LD, Durbin RM, Gibbs RA, Hurles ME, McVean GA (2010) A map of human genome variation from population-scale sequencing. Nature 467:1061–1073

    PubMed  Google Scholar 

  17. Pfaus JG, Damsma G, Nomikos GG, Wenkstern DG, Blaha CD, Phillips AG, Fibiger HC (1990) Sexual behavior enhances central dopamine transmission in the male rat. Brain Res 530:345–348

    CAS  PubMed  Google Scholar 

  18. Small DM, Jones-Gotman M, Dagher A (2003) Feeding-induced dopamine release in dorsal striatum correlates with meal pleasantness ratings in healthy human volunteers. Neuroimage 19:1709–1715

    PubMed  Google Scholar 

  19. Di Chiara G, Imperato A (1988) Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proc Natl Acad Sci U S A 85:5274–5278

    PubMed Central  PubMed  Google Scholar 

  20. Dackis C, O’Brien C (2005) Neurobiology of addiction: treatment and public policy ramifications. Nat Neurosci 8:1431–1436

    CAS  PubMed  Google Scholar 

  21. Han D, Gu H (2006) Comparison of the monoamine transporters from human and mouse in their sensitivities to psychostimulant drugs. BMC Pharmacol 6:6

    PubMed Central  PubMed  Google Scholar 

  22. Nestler EJ (2005) The neurobiology of cocaine addiction. Sci Pract Perspect 3:4–10

    PubMed Central  PubMed  Google Scholar 

  23. Berridge KC, Robinson TE (1998) What is the role of dopamine in reward: hedonic impact, reward learning, or incentive salience? Brain Res Brain Res Rev 28:309–369

    CAS  PubMed  Google Scholar 

  24. Heilig M, Goldman D, Berrettini W, O’Brien CP (2011) Pharmacogenetic approaches to the treatment of alcohol addiction. Nat Rev Neurosci 12:670–684

    CAS  PubMed Central  PubMed  Google Scholar 

  25. Koob GF (1992) Drugs of abuse: anatomy, pharmacology and function of reward pathways. Trends Pharmacol Sci 13:177–184

    CAS  PubMed  Google Scholar 

  26. Weiss F, Lorang MT, Bloom FE, Koob GF (1993) Oral alcohol self-administration stimulates dopamine release in the rat nucleus accumbens: genetic and motivational determinants. J Pharmacol Exp Ther 267:250–258

    CAS  PubMed  Google Scholar 

  27. Imperato A, Di Chiara G (1986) Preferential stimulation of dopamine release in the nucleus accumbens of freely moving rats by ethanol. J Pharmacol Exp Ther 239:219–228

    CAS  PubMed  Google Scholar 

  28. Bourdy R, Barrot M (2012) A new control center for dopaminergic systems: pulling the VTA by the tail. Trends Neurosci 35:681–690

    CAS  PubMed  Google Scholar 

  29. Johnson SW, North RA (1992) Opioids excite dopamine neurons by hyperpolarization of local interneurons. J Neurosci 12:483–488

    CAS  PubMed  Google Scholar 

  30. Comings DE, Comings BG, Muhleman D, Dietz G, Shahbahrami B, Tast D, Knell E, Kocsis P, Baumgarten R, Kovacs BW et al (1991) The dopamine D2 receptor locus as a modifying gene in neuropsychiatric disorders. JAMA 266:1793–1800

    CAS  PubMed  Google Scholar 

  31. Golimbet VE, Aksenova MG, Nosikov VV, Orlova VA, Kaleda VG (2003) Analysis of the linkage of the Taq1A and Taq1B loci of the dopamine D2 receptor gene with schizophrenia in patients and their siblings. Neurosci Behav Physiol 33:223–225

    CAS  PubMed  Google Scholar 

  32. Comings DE, Rosenthal RJ, Lesieur HR, Rugle LJ, Muhleman D, Chiu C, Dietz G, Gade R (1996) A study of the dopamine D2 receptor gene in pathological gambling. Pharmacogenetics 6:223–234

    CAS  PubMed  Google Scholar 

  33. Blum K, Noble EP, Sheridan PJ, Finley O, Montgomery A, Ritchie T, Ozkaragoz T, Fitch RJ, Sadlack F, Sheffield D et al (1991) Association of the A1 allele of the D2 dopamine receptor gene with severe alcoholism. Alcohol 8:409–416

    CAS  PubMed  Google Scholar 

  34. Persico AM, Bird G, Gabbay FH, Uhl GR (1996) D2 dopamine receptor gene TaqI A1 and B1 restriction fragment length polymorphisms: enhanced frequencies in psychostimulant-preferring polysubstance abusers. Biol Psychiatry 40:776–784

    CAS  PubMed  Google Scholar 

  35. Noble EP (1994) Polymorphisms of the D2 dopamine receptor gene and alcoholism and other substance use disorders. Alcohol Alcohol Suppl 2:35–43

    CAS  PubMed  Google Scholar 

  36. Munafo M, Clark T, Johnstone E, Murphy M, Walton R (2004) The genetic basis for smoking behavior: a systematic review and meta-analysis. Nicotine Tob Res 6:583–597

    CAS  PubMed  Google Scholar 

  37. Lawford BR, Young RM, Noble EP, Sargent J, Rowell J, Shadforth S, Zhang X, Ritchie T (2000) The D(2) dopamine receptor A(1) allele and opioid dependence: association with heroin use and response to methadone treatment. Am J Med Genet 96:592–598

    CAS  PubMed  Google Scholar 

  38. Zhang Y, Bertolino A, Fazio L, Blasi G, Rampino A, Romano R, Lee ML, Xiao T, Papp A, Wang D, Sadee W (2007) Polymorphisms in human dopamine D2 receptor gene affect gene expression, splicing, and neuronal activity during working memory. Proc Natl Acad Sci U S A 104:20552–20557

    CAS  PubMed Central  PubMed  Google Scholar 

  39. Moyer RA, Wang D, Papp AC, Smith RM, Duque L, Mash DC, Sadee W (2011) Intronic polymorphisms affecting alternative splicing of human dopamine D2 receptor are associated with cocaine abuse. Neuropsychopharmacology 36:753–762

    CAS  PubMed Central  PubMed  Google Scholar 

  40. Hirvonen MM, Lumme V, Hirvonen J, Pesonen U, Nagren K, Vahlberg T, Scheinin H, Hietala J (2009) C957T polymorphism of the human dopamine D2 receptor gene predicts extrastriatal dopamine receptor availability in vivo. Prog Neuropsychopharmacol Biol Psychiatry 33:630–636

    CAS  PubMed  Google Scholar 

  41. Duan J, Wainwright MS, Comeron JM, Saitou N, Sanders AR, Gelernter J, Gejman PV (2003) Synonymous mutations in the human dopamine receptor D2 (DRD2) affect mRNA stability and synthesis of the receptor. Hum Mol Genet 12:205–216

    CAS  PubMed  Google Scholar 

  42. Gupta M, Chauhan C, Bhatnagar P, Gupta S, Grover S, Singh PK, Purushottam M, Mukherjee O, Jain S, Brahmachari SK, Kukreti R (2009) Genetic susceptibility to schizophrenia: role of dopaminergic pathway gene polymorphisms. Pharmacogenomics 10:277–291

    CAS  PubMed  Google Scholar 

  43. Meyers JL, Nyman E, Loukola A, Rose RJ, Kaprio J, Dick DM (2013) The association between DRD2/ANKK1 and genetically informed measures of alcohol use and problems. Addict Biol 18:523–536

    CAS  PubMed Central  PubMed  Google Scholar 

  44. Zha J, Zhou Q, Xu LG, Chen D, Li L, Zhai Z, Shu HB (2004) RIP5 is a RIP-homologous inducer of cell death. Biochem Biophys Res Commun 319:298–303

    CAS  PubMed  Google Scholar 

  45. Blum K, Noble EP, Sheridan PJ, Montgomery A, Ritchie T, Jagadeeswaran P, Nogami H, Briggs AH, Cohn JB (1990) Allelic association of human dopamine D2 receptor gene in alcoholism. JAMA 263:2055–2060

    CAS  PubMed  Google Scholar 

  46. Jonsson EG, Nothen MM, Grunhage F, Farde L, Nakashima Y, Propping P, Sedvall GC (1999) Polymorphisms in the dopamine D2 receptor gene and their relationships to striatal dopamine receptor density of healthy volunteers. Mol Psychiatry 4:290–296

    CAS  PubMed  Google Scholar 

  47. Ritchie T, Noble EP (1996) [3H]naloxone binding in the human brain: alcoholism and the TaqI A D2 dopamine receptor polymorphism. Brain Res 718:193–197

    CAS  PubMed  Google Scholar 

  48. Volkow ND, Fowler JS, Wolf AP, Schlyer D, Shiue CY, Alpert R, Dewey SL, Logan J, Bendriem B, Christman D et al (1990) Effects of chronic cocaine abuse on postsynaptic dopamine receptors. Am J Psychiatry 147:719–724

    CAS  PubMed  Google Scholar 

  49. Volkow ND, Wang GJ, Fowler JS, Logan J, Hitzemann R, Ding YS, Pappas N, Shea C, Piscani K (1996) Decreases in dopamine receptors but not in dopamine transporters in alcoholics. Alcohol Clin Exp Res 20: 1594–1598

    CAS  PubMed  Google Scholar 

  50. Wang GJ, Volkow ND, Fowler JS, Logan J, Abumrad NN, Hitzemann RJ, Pappas NS, Pascani K (1997) Dopamine D2 receptor availability in opiate-dependent subjects before and after naloxone-precipitated withdrawal. Neuropsychopharmacology 16:174–182

    CAS  PubMed  Google Scholar 

  51. Garrido E, Palomo T, Ponce G, Garcia-Consuegra I, Jimenez-Arriero MA, Hoenicka J (2011) The ANKK1 protein associated with addictions has nuclear and cytoplasmic localization and shows a differential response of Ala239Thr to apomorphine. Neurotox Res 20:32–39

    CAS  PubMed  Google Scholar 

  52. Kosten TR, Wu G, Huang W, Harding MJ, Hamon SC, Lappalainen J, Nielsen DA (2013) Pharmacogenetic randomized trial for cocaine abuse: disulfiram and dopamine beta-hydroxylase. Biol Psychiatry 73:219–224

    CAS  PubMed Central  PubMed  Google Scholar 

  53. Spellicy CJ, Kosten TR, Hamon SC, Harding MJ, Nielsen DA (2013) ANKK1 and DRD2 pharmacogenetics of disulfiram treatment for cocaine abuse. Pharmacogenet Genomics 23:333–340

    CAS  PubMed  Google Scholar 

  54. Balldin JI, Berggren UC, Lindstedt G (1992) Neuroendocrine evidence for reduced dopamine receptor sensitivity in alcoholism. Alcohol Clin Exp Res 16:71–74

    CAS  PubMed  Google Scholar 

  55. Balldin J, Berggren U, Lindstedt G, Sundkler A (1993) Further neuroendocrine evidence for reduced D2 dopamine receptor function in alcoholism. Drug Alcohol Depend 32:159–162

    CAS  PubMed  Google Scholar 

  56. Lawford BR, Young RM, Rowell JA, Qualichefski J, Fletcher BH, Syndulko K, Ritchie T, Noble EP (1995) Bromocriptine in the treatment of alcoholics with the D2 dopamine receptor A1 allele. Nat Med 1:337–341

    CAS  PubMed  Google Scholar 

  57. Lucht MJ, Kuehn KU, Schroeder W, Armbruster J, Abraham G, Schattenberg A, Gaensicke M, Barnow S, Tretzel H, Herrmann FH, Freyberger HJ (2001) Influence of the dopamine D2 receptor (DRD2) exon 8 genotype on efficacy of tiapride and clinical outcome of alcohol withdrawal. Pharmacogenetics 11:647–653

    CAS  PubMed  Google Scholar 

  58. Ooteman W, Naassila M, Koeter MW, Verheul R, Schippers GM, Houchi H, Daoust M, van den Brink W (2009) Predicting the effect of naltrexone and acamprosate in alcohol-dependent patients using genetic indicators. Addict Biol 14:328–337

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  60. Dole VP, Nyswander M (1965) A medical treatment for diacetylmorphine (heroin) addiction. A clinical trial with methadone hydrochloride. JAMA 193:646–650

    CAS  PubMed  Google Scholar 

  61. Kreek MJ, Bart G, Lilly C, LaForge KS, Nielsen DA (2005) Pharmacogenetics and human molecular genetics of opiate and cocaine addictions and their treatments. Pharmacol Rev 57:1–26

    CAS  PubMed  Google Scholar 

  62. Doehring A, Hentig N, Graff J, Salamat S, Schmidt M, Geisslinger G, Harder S, Lotsch J (2009) Genetic variants altering dopamine D2 receptor expression or function modulate the risk of opiate addiction and the dosage requirements of methadone substitution. Pharmacogenet Genomics 19:407–414

    CAS  PubMed  Google Scholar 

  63. Crettol S, Besson J, Croquette-Krokar M, Hammig R, Gothuey I, Monnat M, Deglon JJ, Preisig M, Eap CB (2008) Association of dopamine and opioid receptor genetic polymorphisms with response to methadone maintenance treatment. Prog Neuropsychopharmacol Biol Psychiatry 32:1722–1727

    CAS  PubMed  Google Scholar 

  64. Hung CC, Chiou MH, Huang BH, Hsieh YW, Hsieh TJ, Huang CL, Lane HY (2011) Impact of genetic polymorphisms in ABCB1, CYP2B6, OPRM1, ANKK1 and DRD2 genes on methadone therapy in Han Chinese patients. Pharmacogenomics 12:1525–1533

    CAS  PubMed  Google Scholar 

  65. Van Tol HH, Wu CM, Guan HC, Ohara K, Bunzow JR, Civelli O, Kennedy J, Seeman P, Niznik HB, Jovanovic V (1992) Multiple dopamine D4 receptor variants in the human population. Nature 358:149–152

    PubMed  Google Scholar 

  66. Asghari V, Sanyal S, Buchwaldt S, Paterson A, Jovanovic V, Van Tol HH (1995) Modulation of intracellular cyclic AMP levels by different human dopamine D4 receptor variants. J Neurochem 65:1157–1165

    CAS  PubMed  Google Scholar 

  67. Hutchison KE, McGeary J, Smolen A, Bryan A, Swift RM (2002) The DRD4 VNTR polymorphism moderates craving after alcohol consumption. Health Psychol 21:139–146

    PubMed  Google Scholar 

  68. Hutchison KE, Wooden A, Swift RM, Smolen A, McGeary J, Adler L, Paris L (2003) Olanzapine reduces craving for alcohol: a DRD4 VNTR polymorphism by pharmacotherapy interaction. Neuropsychopharmacology 28:1882–1888

    CAS  PubMed  Google Scholar 

  69. Hutchison KE, Swift R, Rohsenow DJ, Monti PM, Davidson D, Almeida A (2001) Olanzapine reduces urge to drink after drinking cues and a priming dose of alcohol. Psychopharmacology (Berl) 155:27–34

    CAS  Google Scholar 

  70. Kaufman S, Friedman S (1965) Dopamine-beta-hydroxylase. Pharmacol Rev 17:71–100

    CAS  PubMed  Google Scholar 

  71. Weinshilboum RM (1978) Serum dopamine beta-hydroxylase. Pharmacol Rev 30:133–166

    CAS  PubMed  Google Scholar 

  72. Arnsten AF (2000) Stress impairs prefrontal cortical function in rats and monkeys: role of dopamine D1 and norepinephrine alpha-1 receptor mechanisms. Prog Brain Res 126:183–192

    CAS  PubMed  Google Scholar 

  73. Arnsten AF (2000) Through the looking glass: differential noradenergic modulation of prefrontal cortical function. Neural Plast 7:133–146

    CAS  PubMed Central  PubMed  Google Scholar 

  74. Stewart LC, Klinman JP (1988) Dopamine beta-hydroxylase of adrenal chromaffin granules: structure and function. Annu Rev Biochem 57:551–592

    CAS  PubMed  Google Scholar 

  75. Weinshilboum RM, Raymond FA, Elveback LR, Weidman WH (1973) Serum dopamine-beta-hydroxylase activity: sibling-sibling correlation. Science 181:943–945

    CAS  PubMed  Google Scholar 

  76. Oxenstierna G, Edman G, Iselius L, Oreland L, Ross SB, Sedvall G (1986) Concentrations of monoamine metabolites in the cerebrospinal fluid of twins and unrelated individuals—a genetic study. J Psychiatr Res 20:19–29

    CAS  PubMed  Google Scholar 

  77. Wei J, Ramchand CN, Hemmings GP (1997) Possible control of dopamine beta-hydroxylase via a codominant mechanism associated with the polymorphic (GT)n repeat at its gene locus in healthy individuals. Hum Genet 99:52–55

    CAS  PubMed  Google Scholar 

  78. Wei J, Xu HM, Ramchand CN, Hemmings GP (1997) Is the polymorphic microsatellite repeat of the dopamine beta-hydroxylase gene associated with biochemical variability of the catecholamine pathway in schizophrenia? Biol Psychiatry 41:762–767

    CAS  PubMed  Google Scholar 

  79. Cubells JF, van Kammen DP, Kelley ME, Anderson GM, O’Connor DT, Price LH, Malison R, Rao PA, Kobayashi K, Nagatsu T, Gelernter J (1998) Dopamine beta-hydroxylase: two polymorphisms in linkage disequilibrium at the structural gene DBH associate with biochemical phenotypic variation. Hum Genet 102:533–540

    CAS  PubMed  Google Scholar 

  80. Cubells JF, Kranzler HR, McCance-Katz E, Anderson GM, Malison RT, Price LH, Gelernter J (2000) A haplotype at the DBH locus, associated with low plasma dopamine beta-hydroxylase activity, also associates with cocaine-induced paranoia. Mol Psychiatry 5:56–63

    CAS  PubMed  Google Scholar 

  81. Zabetian CP, Buxbaum SG, Elston RC, Kohnke MD, Anderson GM, Gelernter J, Cubells JF (2003) The structure of linkage disequilibrium at the DBH locus strongly influences the magnitude of association between diallelic markers and plasma dopamine beta-hydroxylase activity. Am J Hum Genet 72:1389–1400

    CAS  PubMed Central  PubMed  Google Scholar 

  82. Zabetian CP, Anderson GM, Buxbaum SG, Elston RC, Ichinose H, Nagatsu T, Kim KS, Kim CH, Malison RT, Gelernter J, Cubells JF (2001) A quantitative-trait analysis of human plasma-dopamine beta-hydroxylase activity: evidence for a major functional polymorphism at the DBH locus. Am J Hum Genet 68: 515–522

    CAS  PubMed Central  PubMed  Google Scholar 

  83. Bhaduri N, Mukhopadhyay K (2008) Correlation of plasma dopamine beta-hydroxylase activity with polymorphisms in DBH gene: a study on eastern Indian populaion. Cell Mol Neurobiol 28:343–350

    CAS  PubMed  Google Scholar 

  84. Preuss UW, Wurst FM, Ridinger M, Rujescu D, Fehr C, Koller G, Bondy B, Wodarz N, Soyka M, Zill P (2013) Association of functional DBH genetic variants with alcohol dependence risk and related depression and suicide attempt phenotypes: results from a large multicenter association study. Drug Alcohol Depend 133(2):459–467

    CAS  PubMed  Google Scholar 

  85. Xie X, Xu L, Liu H, Chen W, Zhuang D, Zhang J, Duan S, Zhou W (2013) Positive association between –1021TT genotype of dopamine beta hydroxylase gene and progressive behavior of injection heroin users. Neurosci Lett 541:258–262

    CAS  PubMed  Google Scholar 

  86. Martell BA, Mitchell E, Poling J, Gonsai K, Kosten TR (2005) Vaccine pharmacotherapy for the treatment of cocaine dependence. Biol Psychiatry 58:158–164

    CAS  PubMed  Google Scholar 

  87. Kosten TR, Domingo CB, Hamon SC, Nielsen DA (2013) DBH gene as predictor of response in a cocaine vaccine clinical trial. Neurosci Lett 541:29–33

    CAS  PubMed  Google Scholar 

  88. Spanagel R, Herz A, Shippenberg TS (1992) Opposing tonically active endogenous opioid systems modulate the mesolimbic dopaminergic pathway. Proc Natl Acad Sci U S A 89:2046–2050

    CAS  PubMed Central  PubMed  Google Scholar 

  89. Di Chiara G, Imperato A (1988) Opposite effects of mu and kappa opiate agonists on dopamine release in the nucleus accumbens and in the dorsal caudate of freely moving rats. J Pharmacol Exp Ther 244:1067–1080

    PubMed  Google Scholar 

  90. Spanagel R, Herz A, Shippenberg TS (1990) The effects of opioid peptides on dopamine release in the nucleus accumbens: an in vivo microdialysis study. J Neurochem 55:1734–1740

    CAS  PubMed  Google Scholar 

  91. Bart G, Heilig M, LaForge KS, Pollak L, Leal SM, Ott J, Kreek MJ (2004) Substantial attributable risk related to a functional mu-opioid receptor gene polymorphism in association with heroin addiction in central Sweden. Mol Psychiatry 9:547–549

    CAS  PubMed  Google Scholar 

  92. Deb I, Chakraborty J, Gangopadhyay PK, Choudhury SR, Das S (2010) Single-nucleotide polymorphism (A118G) in exon 1 of OPRM1 gene causes alteration in downstream signaling by mu-opioid receptor and may contribute to the genetic risk for addiction. J Neurochem 112:486–496

    CAS  PubMed  Google Scholar 

  93. Bergen AW, Kokoszka J, Peterson R, Long JC, Virkkunen M, Linnoila M, Goldman D (1997) Mu opioid receptor gene variants: lack of association with alcohol dependence. Mol Psychiatry 2:490–494

    CAS  PubMed  Google Scholar 

  94. Bond C, LaForge KS, Tian M, Melia D, Zhang S, Borg L, Gong J, Schluger J, Strong JA, Leal SM, Tischfield JA, Kreek MJ, Yu L (1998) Single-nucleotide polymorphism in the human mu opioid receptor gene alters beta-endorphin binding and activity: possible implications for opiate addiction. Proc Natl Acad Sci U S A 95:9608–9613

    CAS  PubMed Central  PubMed  Google Scholar 

  95. Zhang Y, Wang D, Johnson AD, Papp AC, Sadee W (2005) Allelic expression imbalance of human mu opioid receptor (OPRM1) caused by variant A118G. J Biol Chem 280:32618–32624

    CAS  PubMed  Google Scholar 

  96. Kroslak T, Laforge KS, Gianotti RJ, Ho A, Nielsen DA, Kreek MJ (2007) The single nucleotide polymorphism A118G alters functional properties of the human mu opioid receptor. J Neurochem 103:77–87

    CAS  PubMed  Google Scholar 

  97. Wand GS, McCaul M, Yang X, Reynolds J, Gotjen D, Lee S, Ali A (2002) The mu-opioid receptor gene polymorphism (A118G) alters HPA axis activation induced by opioid receptor blockade. Neuropsychopharmacology 26: 106–114

    CAS  PubMed  Google Scholar 

  98. Hernandez-Avila CA, Wand G, Luo X, Gelernter J, Kranzler HR (2003) Association between the cortisol response to opioid blockade and the Asn40Asp polymorphism at the mu-opioid receptor locus (OPRM1). Am J Med Genet B Neuropsychiatr Genet 118B:60–65

    PubMed  Google Scholar 

  99. Bart G, Kreek MJ, Ott J, LaForge KS, Proudnikov D, Pollak L, Heilig M (2005) Increased attributable risk related to a functional mu-opioid receptor gene polymorphism in association with alcohol dependence in central Sweden. Neuropsychopharmacology 30:417–422

    CAS  PubMed  Google Scholar 

  100. Rouvinen-Lagerstrom N, Lahti J, Alho H, Kovanen L, Aalto M, Partonen T, Silander K, Sinclair D, Raikkonen K, Eriksson JG, Palotie A, Koskinen S, Saarikoski ST (2013) mu-Opioid receptor gene (OPRM1) polymorphism A118G: lack of association in Finnish populations with alcohol dependence or alcohol consumption. Alcohol Alcohol 48:519–525

    PubMed  Google Scholar 

  101. Cupic B, Stefulj J, Zapletal E, Matosic A, Bordukalo-Niksic T, Cicin-Sain L, Gabrilovac J (2013) Opioid system genes in alcoholism: a case-control study in Croatian population. Neuropeptides 47(5):315–319

    CAS  PubMed  Google Scholar 

  102. Ray LA, Hutchison KE (2004) A polymorphism of the mu-opioid receptor gene (OPRM1) and sensitivity to the effects of alcohol in humans. Alcohol Clin Exp Res 28:1789–1795

    CAS  PubMed  Google Scholar 

  103. Tan EC, Tan CH, Karupathivan U, Yap EP (2003) Mu opioid receptor gene polymorphisms and heroin dependence in Asian populations. Neuroreport 14:569–572

    CAS  PubMed  Google Scholar 

  104. Filbey FM, Claus E, Audette AR, Niculescu M, Banich MT, Tanabe J, Du YP, Hutchison KE (2008) Exposure to the taste of alcohol elicits activation of the mesocorticolimbic neurocircuitry. Neuropsychopharmacology 33:1391–1401

    CAS  PubMed Central  PubMed  Google Scholar 

  105. Ramchandani VA, Umhau J, Pavon FJ, Ruiz-Velasco V, Margas W, Sun H, Damadzic R, Eskay R, Schoor M, Thorsell A, Schwandt ML, Sommer WH, George DT, Parsons LH, Herscovitch P, Hommer D, Heilig M (2011) A genetic determinant of the striatal dopamine response to alcohol in men. Mol Psychiatry 16:809–817

    CAS  PubMed Central  PubMed  Google Scholar 

  106. Baron SA, Testa FM, Gintzler AR (1985) Simultaneous quantitation of norepinephrine, dopamine and serotonin in brain during and following chronic naltrexone administration. Brain Res 340:192–198

    CAS  PubMed  Google Scholar 

  107. Volpicelli JR, O’Brien CP, Alterman AI, Hayashida M (1990) Naltrexone and the treatment of alcohol dependence: initial observations. In: Reid LD (ed) Opioids, bulimia, and alcohol abuse & addiction. Springer, New York, NY, pp 195–214

    Google Scholar 

  108. Volpicelli JR, Alterman AI, Hayashida M, O’Brien CP (1992) Naltrexone in the treatment of alcohol dependence. Arch Gen Psychiatry 49:876–880

    CAS  PubMed  Google Scholar 

  109. O’Malley SS, Jaffe AJ, Chang G, Schottenfeld RS, Meyer RE, Rounsaville B (1992) Naltrexone and coping skills therapy for alcohol dependence. A controlled study. Arch Gen Psychiatry 49:881–887

    PubMed  Google Scholar 

  110. Anton RF, Moak DH, Waid LR, Latham PK, Malcolm RJ, Dias JK (1999) Naltrexone and cognitive behavioral therapy for the treatment of outpatient alcoholics: results of a placebo-controlled trial. Am J Psychiatry 156: 1758–1764

    CAS  PubMed  Google Scholar 

  111. Balldin J, Berglund M, Borg S, Mansson M, Bendtsen P, Franck J, Gustafsson L, Halldin J, Nilsson LH, Stolt G, Willander A (2003) A 6-month controlled naltrexone study: combined effect with cognitive behavioral therapy in outpatient treatment of alcohol dependence. Alcohol Clin Exp Res 27:1142–1149

    CAS  PubMed  Google Scholar 

  112. Chick J, Anton R, Checinski K, Croop R, Drummond DC, Farmer R, Labriola D, Marshall J, Moncrieff J, Morgan MY, Peters T, Ritson B (2000) A multicentre, randomized, double-blind, placebo-controlled trial of naltrexone in the treatment of alcohol dependence or abuse. Alcohol Alcohol 35:587–593

    CAS  PubMed  Google Scholar 

  113. Davidson D, Swift R, Fitz E (1996) Naltrexone increases the latency to drink alcohol in social drinkers. Alcohol Clin Exp Res 20:732–739

    CAS  PubMed  Google Scholar 

  114. Guardia J, Caso C, Arias F, Gual A, Sanahuja J, Ramirez M, Mengual I, Gonzalvo B, Segura L, Trujols J, Casas M (2002) A double-blind, placebo-controlled study of naltrexone in the treatment of alcohol-dependence disorder: results from a multicenter clinical trial. Alcohol Clin Exp Res 26:1381–1387

    CAS  PubMed  Google Scholar 

  115. Heinala P, Alho H, Kiianmaa K, Lonnqvist J, Kuoppasalmi K, Sinclair JD (2001) Targeted use of naltrexone without prior detoxification in the treatment of alcohol dependence: a factorial double-blind, placebo-controlled trial. J Clin Psychopharmacol 21:287–292

    CAS  PubMed  Google Scholar 

  116. Kiefer F, Jahn H, Tarnaske T, Helwig H, Briken P, Holzbach R, Kampf P, Stracke R, Baehr M, Naber D, Wiedemann K (2003) Comparing and combining naltrexone and acamprosate in relapse prevention of alcoholism: a double-blind, placebo-controlled study. Arch Gen Psychiatry 60:92–99

    CAS  PubMed  Google Scholar 

  117. Latt NC, Jurd S, Houseman J, Wutzke SE (2002) Naltrexone in alcohol dependence: a randomised controlled trial of effectiveness in a standard clinical setting. Med J Aust 176:530–534

    PubMed  Google Scholar 

  118. Monti PM, Rohsenow DJ, Swift RM, Gulliver SB, Colby SM, Mueller TI, Brown RA, Gordon A, Abrams DB, Niaura RS, Asher MK (2001) Naltrexone and cue exposure with coping and communication skills training for alcoholics: treatment process and 1-year outcomes. Alcohol Clin Exp Res 25:1634–1647

    CAS  PubMed  Google Scholar 

  119. Oslin D, Liberto JG, O’Brien J, Krois S, Norbeck J (1997) Naltrexone as an adjunctive treatment for older patients with alcohol dependence. Am J Geriatr Psychiatry 5:324–332

    CAS  PubMed  Google Scholar 

  120. Kranzler HR, Modesto-Lowe V, Van Kirk J (2000) Naltrexone vs. nefazodone for treatment of alcohol dependence. A placebo-controlled trial. Neuropsychopharmacology 22:493–503

    CAS  PubMed  Google Scholar 

  121. Krystal JH, Cramer JA, Krol WF, Kirk GF, Rosenheck RA, Veterans Affairs Naltrexone Cooperative Study 425 Group (2001) Naltrexone in the treatment of alcohol dependence. N Engl J Med 345:1734–1739

    CAS  PubMed  Google Scholar 

  122. Volpicelli JR, Watson NT, King AC, Sherman CE, O’Brien CP (1995) Effect of naltrexone on alcohol “high” in alcoholics. Am J Psychiatry 152:613–615

    CAS  PubMed  Google Scholar 

  123. Drobes DJ, Anton RF, Thomas SE, Voronin K (2004) Effects of naltrexone and nalmefene on subjective response to alcohol among non-treatment-seeking alcoholics and social drinkers. Alcohol Clin Exp Res 28:1362–1370

    CAS  PubMed  Google Scholar 

  124. Swift RM, Whelihan W, Kuznetsov O, Buongiorno G, Hsuing H (1994) Naltrexone-induced alterations in human ethanol intoxication. Am J Psychiatry 151:1463–1467

    CAS  PubMed  Google Scholar 

  125. Anton RF, Drobes DJ, Voronin K, Durazo-Avizu R, Moak D (2004) Naltrexone effects on alcohol consumption in a clinical laboratory paradigm: temporal effects of drinking. Psychopharmacology (Berl) 173:32–40

    CAS  Google Scholar 

  126. Jaffe SB, Sobieszczyk S, Wardlaw SL (1994) Effect of opioid antagonism on beta-endorphin processing and proopiomelanocortin-peptide release in the hypothalamus. Brain Res 648:24–31

    CAS  PubMed  Google Scholar 

  127. O’Malley SS, Krishnan-Sarin S, Farren C, Sinha R, Kreek MJ (2002) Naltrexone decreases craving and alcohol self-administration in alcohol-dependent subjects and activates the hypothalamo-pituitary-adrenocortical axis. Psychopharmacology (Berl) 160:19–29

    Google Scholar 

  128. Ray LA, Hutchison KE (2007) Effects of naltrexone on alcohol sensitivity and genetic moderators of medication response: a double-blind placebo-controlled study. Arch Gen Psychiatry 64:1069–1077

    CAS  PubMed  Google Scholar 

  129. McGeary JE, Monti PM, Rohsenow DJ, Tidey J, Swift R, Miranda R Jr (2006) Genetic moderators of naltrexone’s effects on alcohol cue reactivity. Alcohol Clin Exp Res 30: 1288–1296

    CAS  PubMed  Google Scholar 

  130. Oslin DW, Berrettini W, Kranzler HR, Pettinati H, Gelernter J, Volpicelli JR, O’Brien CP (2003) A functional polymorphism of the mu-opioid receptor gene is associated with naltrexone response in alcohol-dependent patients. Neuropsychopharmacology 28:1546–1552

    CAS  PubMed  Google Scholar 

  131. Anton RF, Oroszi G, O’Malley S, Couper D, Swift R, Pettinati H, Goldman D (2008) An evaluation of mu-opioid receptor (OPRM1) as a predictor of naltrexone response in the treatment of alcohol dependence: results from the Combined Pharmacotherapies and Behavioral Interventions for Alcohol Dependence (COMBINE) study. Arch Gen Psychiatry 65:135–144

    CAS  PubMed Central  PubMed  Google Scholar 

  132. Ray LA, Bujarski S, Chin PF, Miotto K (2012) Pharmacogenetics of naltrexone in asian americans: a randomized placebo-controlled laboratory study. Neuropsychopharmacology 37:445–455

    CAS  PubMed Central  PubMed  Google Scholar 

  133. Setiawan E, Pihl RO, Cox SM, Gianoulakis C, Palmour RM, Benkelfat C, Leyton M (2011) The effect of naltrexone on alcohol’s stimulant properties and self-administration behavior in social drinkers: influence of gender and genotype. Alcohol Clin Exp Res 35:1134–1141

    CAS  PubMed  Google Scholar 

  134. Kim SG, Kim CM, Choi SW, Jae YM, Lee HG, Son BK, Kim JG, Choi YS, Kim HO, Kim SY, Oslin DW (2009) A micro opioid receptor gene polymorphism (A118G) and naltrexone treatment response in adherent Korean alcohol-dependent patients. Psychopharmacology (Berl) 201:611–618

    CAS  Google Scholar 

  135. Coller JK, Cahill S, Edmonds C, Farquharson AL, Longo M, Minniti R, Sullivan T, Somogyi AA, White JM (2011) OPRM1 A118G genotype fails to predict the effectiveness of naltrexone treatment for alcohol dependence. Pharmacogenet Genomics 21:902–905

    CAS  PubMed  Google Scholar 

  136. Oroszi G, Anton RF, O’Malley S, Swift R, Pettinati H, Couper D, Yuan Q, Goldman D (2009) OPRM1 Asn40Asp predicts response to naltrexone treatment: a haplotype-based approach. Alcohol Clin Exp Res 33:383–393

    CAS  PubMed Central  PubMed  Google Scholar 

  137. Gomes I, Jordan BA, Gupta A, Trapaidze N, Nagy V, Devi LA (2000) Heterodimerization of mu and delta opioid receptors: a role in opiate synergy. J Neurosci 20:RC110

    CAS  PubMed Central  PubMed  Google Scholar 

  138. Gupta A, Mulder J, Gomes I, Rozenfeld R, Bushlin I, Ong E, Lim M, Maillet E, Junek M, Cahill CM, Harkany T, Devi LA (2010) Increased abundance of opioid receptor heteromers after chronic morphine administration. Sci Signal 3:ra54

    PubMed Central  PubMed  Google Scholar 

  139. Hirose N, Murakawa K, Takada K, Oi Y, Suzuki T, Nagase H, Cools AR, Koshikawa N (2005) Interactions among mu- and delta-opioid receptors, especially putative delta1- and delta2-opioid receptors, promote dopamine release in the nucleus accumbens. Neuroscience 135:213–225

    CAS  PubMed  Google Scholar 

  140. Zhang H, Kranzler HR, Yang BZ, Luo X, Gelernter J (2008) The OPRD1 and OPRK1 loci in alcohol or drug dependence: OPRD1 variation modulates substance dependence risk. Mol Psychiatry 13:531–543

    CAS  PubMed Central  PubMed  Google Scholar 

  141. Mayer P, Rochlitz H, Rauch E, Rommelspacher H, Hasse HE, Schmidt S, Hollt V (1997) Association between a delta opioid receptor gene polymorphism and heroin dependence in man. Neuroreport 8:2547–2550

    CAS  PubMed  Google Scholar 

  142. Levran O, Londono D, O’Hara K, Nielsen DA, Peles E, Rotrosen J, Casadonte P, Linzy S, Randesi M, Ott J, Adelson M, Kreek MJ (2008) Genetic susceptibility to heroin addiction: a candidate gene association study. Genes Brain Behav 7:720–729

    CAS  PubMed Central  PubMed  Google Scholar 

  143. Crist RC, Clarke TK, Ang A, Ambrose-Lanci LM, Lohoff FW, Saxon AJ, Ling W, Hillhouse MP, Bruce RD, Woody G, Berrettini WH (2013) An intronic variant in OPRD1 predicts treatment outcome for opioid dependence in African-Americans. Neuropsychopharmacology 38(10):2003–2010

    CAS  PubMed  Google Scholar 

  144. Filliol D, Ghozland S, Chluba J, Martin M, Matthes HW, Simonin F, Befort K, Gaveriaux-Ruff C, Dierich A, LeMeur M, Valverde O, Maldonado R, Kieffer BL (2000) Mice deficient for delta- and mu-opioid receptors exhibit opposing alterations of emotional responses. Nat Genet 25:195–200

    CAS  PubMed  Google Scholar 

  145. Zhu Y, King MA, Schuller AG, Nitsche JF, Reidl M, Elde RP, Unterwald E, Pasternak GW, Pintar JE (1999) Retention of supraspinal delta-like analgesia and loss of morphine tolerance in delta opioid receptor knockout mice. Neuron 24:243–252

    CAS  PubMed  Google Scholar 

  146. Nitsche JF, Schuller AG, King MA, Zengh M, Pasternak GW, Pintar JE (2002) Genetic dissociation of opiate tolerance and physical dependence in delta-opioid receptor-1 and preproenkephalin knock-out mice. J Neurosci 22:10906–10913

    CAS  PubMed  Google Scholar 

  147. Unterwald EM, Cox BM, Kreek MJ, Cote TE, Izenwasser S (1993) Chronic repeated cocaine administration alters basal and opioid-regulated adenylyl cyclase activity. Synapse 15:33–38

    CAS  PubMed  Google Scholar 

  148. Tejeda HA, Shippenberg TS, Henriksson R (2012) The dynorphin/kappa-opioid receptor system and its role in psychiatric disorders. Cell Mol Life Sci 69:857–896

    CAS  PubMed  Google Scholar 

  149. Claye LH, Maisonneuve IM, Yu J, Ho A, Kreek MJ (1997) Local perfusion of dynorphin A 1-17 reduces extracellular dopamine levels in the nucleus accumbens. NIDA Res Monogr 174:113

    Google Scholar 

  150. Zhang Y, Butelman ER, Schlussman SD, Ho A, Kreek MJ (2004) Effect of the kappa opioid agonist R-84760 on cocaine-induced increases in striatal dopamine levels and cocaine-induced place preference in C57BL/6J mice. Psychopharmacology (Berl) 173:146–152

    CAS  Google Scholar 

  151. Unterwald EM, Rubenfeld JM, Kreek MJ (1994) Repeated cocaine administration upregulates kappa and mu, but not delta, opioid receptors. Neuroreport 5:1613–1616

    CAS  PubMed  Google Scholar 

  152. Simonin F, Valverde O, Smadja C, Slowe S, Kitchen I, Dierich A, Le Meur M, Roques BP, Maldonado R, Kieffer BL (1998) Disruption of the kappa-opioid receptor gene in mice enhances sensitivity to chemical visceral pain, impairs pharmacological actions of the selective kappa-agonist U-50,488H and attenuates morphine withdrawal. EMBO J 17:886–897

    CAS  PubMed Central  PubMed  Google Scholar 

  153. McLaughlin JP, Marton-Popovici M, Chavkin C (2003) Kappa opioid receptor antagonism and prodynorphin gene disruption block stress-induced behavioral responses. J Neurosci 23:5674–5683

    CAS  PubMed Central  PubMed  Google Scholar 

  154. Haile CN, Kosten TA, Kosten TR (2008) Pharmacogenetic treatments for drug addiction: alcohol and opiates. Am J Drug Alcohol Abuse 34:355–381

    PubMed  Google Scholar 

  155. Xuei X, Dick D, Flury-Wetherill L, Tian HJ, Agrawal A, Bierut L, Goate A, Bucholz K, Schuckit M, Nurnberger J Jr, Tischfield J, Kuperman S, Porjesz B, Begleiter H, Foroud T, Edenberg HJ (2006) Association of the kappa-opioid system with alcohol dependence. Mol Psychiatry 11:1016–1024

    CAS  PubMed  Google Scholar 

  156. Nielsen DA, Hamon S, Kosten TR (2013) The K-opioid receptor gene as a predictor of response in a cocaine vaccine trial. Psychiatr Genet 23:225–232

    CAS  PubMed  Google Scholar 

  157. Lesch KP, Bengel D, Heils A, Sabol SZ, Greenberg BD, Petri S, Benjamin J, Muller CR, Hamer DH, Murphy DL (1996) Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region. Science 274:1527–1531

    CAS  PubMed  Google Scholar 

  158. Heils A, Teufel A, Petri S, Stober G, Riederer P, Bengel D, Lesch KP (1996) Allelic variation of human serotonin transporter gene expression. J Neurochem 66:2621–2624

    CAS  PubMed  Google Scholar 

  159. Hu XZ, Lipsky RH, Zhu G, Akhtar LA, Taubman J, Greenberg BD, Xu K, Arnold PD, Richter MA, Kennedy JL, Murphy DL, Goldman D (2006) Serotonin transporter promoter gain-of-function genotypes are linked to obsessive-compulsive disorder. Am J Hum Genet 78:815–826

    CAS  PubMed Central  PubMed  Google Scholar 

  160. Praschak-Rieder N, Kennedy J, Wilson AA, Hussey D, Boovariwala A, Willeit M, Ginovart N, Tharmalingam S, Masellis M, Houle S, Meyer JH (2007) Novel 5-HTTLPR allele associates with higher serotonin transporter binding in putamen: a [(11)C] DASB positron emission tomography study. Biol Psychiatry 62:327–331

    CAS  PubMed  Google Scholar 

  161. Fox E, Zougkou K, Ridgewell A, Garner K (2011) The serotonin transporter gene alters sensitivity to attention bias modification: evidence for a plasticity gene. Biol Psychiatry 70:1049–1054

    CAS  PubMed Central  PubMed  Google Scholar 

  162. Graham DP, Helmer DA, Harding MJ, Kosten TR, Petersen NJ, Nielsen DA (2013) Serotonin transporter genotype and mild traumatic brain injury independently influence resilience and perception of limitations in Veterans. J Psychiatr Res 47:835–842

    PubMed Central  PubMed  Google Scholar 

  163. Caspi A, Moffitt TE (2006) Gene-environment interactions in psychiatry: joining forces with neuroscience. Nat Rev Neurosci 7:583–590

    CAS  PubMed  Google Scholar 

  164. Hariri AR (2009) The neurobiology of individual differences in complex behavioral traits. Annu Rev Neurosci 32:225–247

    CAS  PubMed Central  PubMed  Google Scholar 

  165. Uher R (2009) The role of genetic variation in the causation of mental illness: an evolution-informed framework. Mol Psychiatry 14: 1072–1082

    CAS  PubMed  Google Scholar 

  166. Torrens M, Fonseca F, Mateu G, Farre M (2005) Efficacy of antidepressants in substance use disorders with and without comorbid depression. A systematic review and meta-analysis. Drug Alcohol Depend 78:1–22

    CAS  PubMed  Google Scholar 

  167. Mannelli P, Patkar AA, Murray HW, Certa K, Peindl K, Mattila-Evenden M, Berrettini WH (2005) Polymorphism in the serotonin transporter gene and response to treatment in African American cocaine and alcohol-abusing individuals. Addict Biol 10:261–268

    CAS  PubMed  Google Scholar 

  168. Nielsen DA, Harding MJ, Hamon SC, Huang W, Kosten TR (2012) Modifying the role of serotonergic 5-HTTLPR and TPH2 variants on disulfiram treatment of cocaine addiction: a preliminary study. Genes Brain Behav 11:1001–1008

    CAS  Google Scholar 

  169. Nagendra SN, Shetty KT, Subhash MN, Udaya HB, Pradhan N (1993) Effect of disulfiram administration on brain tryptophan, serotonin and peripheral tryptophan content. Neurochem Int 22:31–36

    CAS  PubMed  Google Scholar 

  170. Nilsson GE, Tottmar O (1989) Effects of disulfiram and coprine on rat brain tryptophan hydroxylation in vivo. Neurochem Res 14:537–540

    CAS  PubMed  Google Scholar 

  171. Fukumori R, Minegishi A, Satoh T, Kitagawa H, Yanaura S (1980) Changes in the serotonin and 5-hydroxyindoleacetic acid contents in rat brain after ethanol and disulfiram treatments. Eur J Pharmacol 61:199–202

    CAS  PubMed  Google Scholar 

  172. Johnson BA, Roache JD, Javors MA, DiClemente CC, Cloninger CR, Prihoda TJ, Bordnick PS, Ait-Daoud N, Hensler J (2000) Ondansetron for reduction of drinking among biologically predisposed alcoholic patients: a randomized controlled trial. JAMA 284:963–971

    CAS  PubMed  Google Scholar 

  173. Johnson BA, Ait-Daoud N, Seneviratne C, Roache JD, Javors MA, Wang XQ, Liu L, Penberthy JK, DiClemente CC, Li MD (2011) Pharmacogenetic approach at the serotonin transporter gene as a method of reducing the severity of alcohol drinking. Am J Psychiatry 168:265–275

    PubMed Central  PubMed  Google Scholar 

  174. Kranzler HR, Armeli S, Tennen H, Covault J, Feinn R, Arias AJ, Pettinati H, Oncken C (2011) A double-blind, randomized trial of sertraline for alcohol dependence: moderation by age of onset [corrected] and 5-hydroxytryptamine transporter-linked promoter region genotype. J Clin Psychopharmacol 31:22–30

    CAS  PubMed Central  PubMed  Google Scholar 

  175. Kranzler HR, McKay JR (2012) Personalized treatment of alcohol dependence. Curr Psychiatry Rep 14:486–493

    PubMed  Google Scholar 

  176. Kenna GA, Zywiak WH, McGeary JE, Leggio L, McGeary C, Wang S, Grenga A, Swift RM (2009) A within-group design of nontreatment seeking 5-HTTLPR genotyped alcohol-dependent subjects receiving ondansetron and sertraline. Alcohol Clin Exp Res 33:315–323

    CAS  PubMed  Google Scholar 

  177. Cooper JR, Melcer I (1961) The enzymatic oxidation of tryptophan to 5-hydroxytryptophan in the biosynthesis of serotonin. J Pharmacol Exp Ther 132: 265–268

    CAS  PubMed  Google Scholar 

  178. Zill P, Buttner A, Eisenmenger W, Moller H-J, Ackenheil M, Bondy B (2007) Analysis of tryptophan hydroxylase I and II mRNA expression in the human brain: a post-mortem study. J Psychiatr Res 41:168–173

    PubMed  Google Scholar 

  179. Waider J, Araragi N, Gutknecht L, Lesch KP (2011) Tryptophan hydroxylase-2 (TPH2) in disorders of cognitive control and emotion regulation: a perspective. Psychoneuroendocrinology 36:393–405

    CAS  PubMed  Google Scholar 

  180. Nielsen DA, Barral S, Proudnikov D, Kellogg S, Ho A, Ott J, Kreek MJ (2008) TPH2 and TPH1: association of variants and interactions with heroin addiction. Behav Genet 38: 133–150

    PubMed  Google Scholar 

  181. Lim JE, Pinsonneault J, Sadee W, Saffen D (2007) Tryptophan hydroxylase 2 (TPH2) haplotypes predict levels of TPH2 mRNA expression in human pons. Mol Psychiatry 12:491–501

    CAS  PubMed  Google Scholar 

  182. Crettol S, Digon P, Powell Golay K, Brawand M, Eap CB (2007) In vitro P-glycoprotein-mediated transport of (R)-, (S)-, (R, S)-methadone, LAAM and their main metabolites. Pharmacology 80:304–311

    CAS  PubMed  Google Scholar 

  183. Dagenais C, Graff CL, Pollack GM (2004) Variable modulation of opioid brain uptake by P-glycoprotein in mice. Biochem Pharmacol 67:269–276

    CAS  PubMed  Google Scholar 

  184. Levran O, O’Hara K, Peles E, Li D, Barral S, Ray B, Borg L, Ott J, Adelson M, Kreek MJ (2008) ABCB1 (MDR1) genetic variants are associated with methadone doses required for effective treatment of heroin dependence. Hum Mol Genet 17:2219–2227

    CAS  PubMed Central  PubMed  Google Scholar 

  185. Coller JK, Barratt DT, Dahlen K, Loennechen MH, Somogyi AA (2006) ABCB1 genetic variability and methadone dosage requirements in opioid-dependent individuals. Clin Pharmacol Ther 80:682–690

    CAS  PubMed  Google Scholar 

  186. Barratt DT, Coller JK, Hallinan R, Byrne A, White JM, Foster DJ, Somogyi AA (2012) ABCB1 haplotype and OPRM1 118A > G genotype interaction in methadone maintenance treatment pharmacogenetics. Pharmacogenomics Pers Med 5:53–62

    CAS  Google Scholar 

  187. Guindalini C, Laranjeira R, Collier D, Messas G, Vallada H, Breen G (2008) Dopamine-beta hydroxylase polymorphism and cocaine addiction. Behav Brain Funct 4:1

    PubMed Central  PubMed  Google Scholar 

  188. Shorter D, Nielsen DA, Huang W, Harding MJ, Hamon SC, Kosten TR (2013) Pharmacogenetic randomized trial for cocaine abuse: disulfiram and alpha-adrenoceptor gene variation. Eur Neuropsychopharmacol 23(11):1401–1407

    CAS  PubMed  Google Scholar 

  189. Lu B, Nagappan G, Guan X, Nathan PJ, Wren P (2013) BDNF-based synaptic repair as a disease-modifying strategy for neurodegenerative diseases. Nat Rev Neurosci 14:401–416

    CAS  PubMed  Google Scholar 

  190. Rostami E, Krueger F, Zoubak S, Dal Monte O, Raymont V, Pardini M, Hodgkinson CA, Goldman D, Risling M, Grafman J (2011) BDNF polymorphism predicts general intelligence after penetrating traumatic brain injury. PLoS One 6:e27389

    CAS  PubMed Central  PubMed  Google Scholar 

  191. Li W, Zhou N, Yu Q, Li X, Yu Y, Sun S, Kou C, da Chen C, Xiu MH, Kosten TR, Zhang XY (2013) Association of BDNF gene polymorphisms with schizophrenia and clinical symptoms in a Chinese population. Am J Med Genet B Neuropsychiatr Genet 162: 538–545

    CAS  Google Scholar 

  192. de Cid R, Fonseca F, Gratacos M, Gutierrez F, Martin-Santos R, Estivill X, Torrens M (2008) BDNF variability in opioid addicts and response to methadone treatment: preliminary findings. Genes Brain Behav 7:515–522

    PubMed  Google Scholar 

  193. Pezet S, McMahon SB (2006) Neurotrophins: mediators and modulators of pain. Annu Rev Neurosci 29:507–538

    CAS  PubMed  Google Scholar 

  194. Levran O, Peles E, Hamon S, Randesi M, Zhao C, Zhang B, Adelson M, Kreek MJ (2012) Nerve growth factor beta polypeptide (NGFB) genetic variability: association with the methadone dose required for effective maintenance treatment. Pharmacogenomics J 12:319–327

    CAS  PubMed Central  PubMed  Google Scholar 

  195. Gainetdinov RR, Premont RT, Bohn LM, Lefkowitz RJ, Caron MG (2004) Desensitization of G protein-coupled receptors and neuronal functions. Annu Rev Neurosci 27:107–144

    CAS  PubMed  Google Scholar 

  196. Oneda B, Crettol S, Bochud M, Besson J, Croquette-Krokar M, Hammig R, Monnat M, Preisig M, Eap CB (2011) beta-Arrestin2 influences the response to methadone in opioid-dependent patients. Pharmacogenomics J 11:258–266

    CAS  PubMed  Google Scholar 

  197. Ferrari A, Coccia CP, Bertolini A, Sternieri E (2004) Methadone—metabolism, pharmacokinetics and interactions. Pharmacol Res 50: 551–559

    CAS  PubMed  Google Scholar 

  198. Reynolds KK, Ramey-Hartung B, Jortani SA (2008) The value of CYP2D6 and OPRM1 pharmacogenetic testing for opioid therapy. Clin Lab Med 28:581–598

    PubMed  Google Scholar 

  199. Perez de los Cobos J, Sinol N, Trujols J, del Rio E, Banuls E, Luquero E, Menoyo A, Queralto JM, Baiget M, Alvarez E (2007) Association of CYP2D6 ultrarapid metabolizer genotype with deficient patient satisfaction regarding methadone maintenance treatment. Drug Alcohol Depend 89: 190–194

    CAS  PubMed  Google Scholar 

  200. Crettol S, Deglon JJ, Besson J, Croquette-Krokar M, Hammig R, Gothuey I, Monnat M, Eap CB (2006) ABCB1 and cytochrome P450 genotypes and phenotypes: influence on methadone plasma levels and response to treatment. Clin Pharmacol Ther 80:668–681

    CAS  PubMed  Google Scholar 

  201. Levran O, Peles E, Hamon S, Randesi M, Adelson M, Kreek MJ (2013) CYP2B6 SNPs are associated with methadone dose required for effective treatment of opioid addiction. Addict Biol 18:709–716

    CAS  PubMed Central  PubMed  Google Scholar 

  202. Crettol S, Deglon JJ, Besson J, Croquette-Krokkar M, Gothuey I, Hammig R, Monnat M, Huttemann H, Baumann P, Eap CB (2005) Methadone enantiomer plasma levels, CYP2B6, CYP2C19, and CYP2C9 genotypes, and response to treatment. Clin Pharmacol Ther 78:593–604

    CAS  PubMed  Google Scholar 

  203. Dobrinas M, Crettol S, Oneda B, Lahyani R, Rotger M, Choong E, Lubomirov R, Csajka C, Eap CB (2013) Contribution of CYP2B6 alleles in explaining extreme (S)-methadone plasma levels: a CYP2B6 gene resequencing study. Pharmacogenet Genomics 23:84–93

    CAS  PubMed  Google Scholar 

  204. He Q, Mendez M, LaPointe MC (2002) Regulation of the human brain natriuretic peptide gene by GATA-4. Am J Physiol Endocrinol Metab 283:E50–E57

    CAS  PubMed  Google Scholar 

  205. Kovacs GL (2003) Natriuretic peptides in alcohol withdrawal: central and peripheral mechanisms. Curr Med Chem 10:2559–2576

    CAS  PubMed  Google Scholar 

  206. Kiefer F, Witt SH, Frank J, Richter A, Treutlein J, Lemenager T, Nothen MM, Cichon S, Batra A, Berner M, Wodarz N, Zimmermann US, Spanagel R, Wiedemann K, Smolka MN, Heinz A, Rietschel M, Mann K (2011) Involvement of the atrial natriuretic peptide transcription factor GATA4 in alcohol dependence, relapse risk and treatment response to acamprosate. Pharmacogenomics J 11:368–374

    CAS  PubMed  Google Scholar 

  207. Karpyak VM, Winham SJ, Biernacka JM, Cunningham JM, Lewis KA, Geske JR, Colby CL, Abulseoud OA, Hall-Flavin DK, Loukianova LL, Schneekloth TD, Frye MA, Heit JA, Mrazek DA (2012) Association of GATA4 sequence variation with alcohol dependence. Addict Biol 19(2):312–315

    PubMed  Google Scholar 

  208. Treutlein J, Cichon S, Ridinger M, Wodarz N, Soyka M, Zill P, Maier W, Moessner R, Gaebel W, Dahmen N, Fehr C, Scherbaum N, Steffens M, Ludwig KU, Frank J, Wichmann HE, Schreiber S, Dragano N, Sommer WH, Leonardi-Essmann F, Lourdusamy A, Gebicke-Haerter P, Wienker TF, Sullivan PF, Nothen MM, Kiefer F, Spanagel R, Mann K, Rietschel M (2009) Genome-wide association study of alcohol dependence. Arch Gen Psychiatry 66:773–784

    CAS  PubMed  Google Scholar 

  209. Frosst P, Blom HJ, Milos R, Goyette P, Sheppard CA, Matthews RG, Boers GJ, den Heijer M, Kluijtmans LA, van den Heuvel LP et al (1995) A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet 10:111–113

    CAS  PubMed  Google Scholar 

  210. van der Put NM, Steegers-Theunissen RP, Frosst P, Trijbels FJ, Eskes TK, van den Heuvel LP, Mariman EC, den Heyer M, Rozen R, Blom HJ (1995) Mutated methylenetetrahydrofolate reductase as a risk factor for spina bifida. Lancet 346:1070–1071

    PubMed  Google Scholar 

  211. Harisha PN, Devi BI, Christopher R, Kruthika-Vinod TP (2010) Impact of 5,10-methylenetetrahydrofolate reductase gene polymorphism on neural tube defects. J Neurosurg Pediatr 6:364–367

    PubMed  Google Scholar 

  212. Spellicy CJ, Kosten TR, Hamon SC, Harding MJ, Nielsen DA (2012) The MTHFR C677T variant is associated with responsiveness to disulfiram treatment for cocaine dependency. Front Psychiatry 3:109

    PubMed Central  PubMed  Google Scholar 

  213. Lin J, Haffner MC, Zhang Y, Lee BH, Brennen WN, Britton J, Kachhap SK, Shim JS, Liu JO, Nelson WG, Yegnasubramanian S, Carducci MA (2011) Disulfiram is a DNA demethylating agent and inhibits prostate cancer cell growth. Prostate 71:333–343

    CAS  PubMed Central  PubMed  Google Scholar 

  214. Anier K, Malinovskaja K, Aonurm-Helm A, Zharkovsky A, Kalda A (2010) DNA methylation regulates cocaine-induced behavioral sensitization in mice. Neuropsychopharmacology 35:2450–2461

    CAS  PubMed Central  PubMed  Google Scholar 

  215. Pipes GC, Creemers EE, Olson EN (2006) The myocardin family of transcriptional coactivators: versatile regulators of cell growth, migration, and myogenesis. Genes Dev 20:1545–1556

    CAS  PubMed  Google Scholar 

  216. Nielsen DA, Ji F, Yuferov V, Ho A, Chen A, Levran O, Ott J, Kreek MJ (2008) Genotype patterns that contribute to increased risk for or protection from developing heroin addiction. Mol Psychiatry 13:417–428

    CAS  PubMed  Google Scholar 

  217. Fonseca F, Gratacos M, Escaramis G, De Cid R, Martin-Santos R, Fernandez-Espejo E, Estivill X, Torrens M (2010) Response to methadone maintenance treatment is associated with the MYOCD and GRM6 genes. Mol Diagn Ther 14:171–178

    CAS  PubMed  Google Scholar 

  218. Mitrovic I, Margeta-Mitrovic M, Bader S, Stoffel M, Jan LY, Basbaum AI (2003) Contribution of GIRK2-mediated postsynaptic signaling to opiate and alpha 2-adrenergic analgesia and analgesic sex differences. Proc Natl Acad Sci U S A 100:271–276

    CAS  PubMed Central  PubMed  Google Scholar 

  219. Reyes S, Fu Y, Double K, Thompson L, Kirik D, Paxinos G, Halliday GM (2012) GIRK2 expression in dopamine neurons of the substantia nigra and ventral tegmental area. J Comp Neurol 520:2591–2607

    CAS  PubMed  Google Scholar 

  220. Clarke TK, Laucht M, Ridinger M, Wodarz N, Rietschel M, Maier W, Lathrop M, Lourdusamy A, Zimmermann US, Desrivieres S, Schumann G (2011) KCNJ6 is associated with adult alcohol dependence and involved in gene x early life stress interactions in adolescent alcohol drinking. Neuropsychopharmacology 36:1142–1148

    CAS  PubMed Central  PubMed  Google Scholar 

  221. Webb CK, McCudden CR, Willard FS, Kimple RJ, Siderovski DP, Oxford GS (2005) D2 dopamine receptor activation of potassium channels is selectively decoupled by Galpha-specific GoLoco motif peptides. J Neurochem 92:1408–1418

    CAS  PubMed  Google Scholar 

  222. Minami M, Satoh M (1995) Molecular biology of the opioid receptors: structures, functions and distributions. Neurosci Res 23:121–145

    CAS  PubMed  Google Scholar 

  223. Ikeda K, Kobayashi T, Kumanishi T, Niki H, Yano R (2000) Involvement of G-protein-activated inwardly rectifying K (GIRK) channels in opioid-induced analgesia. Neurosci Res 38:113–116

    CAS  PubMed  Google Scholar 

  224. Nishizawa D, Nagashima M, Katoh R, Satoh Y, Tagami M, Kasai S, Ogai Y, Han W, Hasegawa J, Shimoyama N, Sora I, Hayashida M, Ikeda K (2009) Association between KCNJ6 (GIRK2) gene polymorphisms and postoperative analgesic requirements after major abdominal surgery. PLoS One 4:e7060

    PubMed Central  PubMed  Google Scholar 

  225. Lotsch J, Pruss H, Veh RW, Doehring A (2010) A KCNJ6 (Kir3.2, GIRK2) gene polymorphism modulates opioid effects on analgesia and addiction but not on pupil size. Pharmacogenet Genomics 20:291–297

    PubMed  Google Scholar 

  226. Kreek MJ, Nielsen DA, LaForge KS (2004) Genes associated with addiction: alcoholism, opiate, and cocaine addiction. Neuromolecular Med 5:85–108

    CAS  PubMed  Google Scholar 

  227. Mason BJ, Heyser CJ (2010) Acamprosate: a prototypic neuromodulator in the treatment of alcohol dependence. CNS Neurol Disord Drug Targets 9:23–32

    CAS  PubMed Central  PubMed  Google Scholar 

  228. Jarvik ME, Caskey NH, Wirshing WC, Madsen DC, Iwamoto-Schaap PN, Elins JL, Eisenberger NI, Olmstead RE (2000) Bromocriptine reduces cigarette smoking. Addiction 95:1173–1183

    CAS  PubMed  Google Scholar 

  229. Bentley KW, Hardy DG (1967) Novel analgesics and molecular rearrangements in the morphine-thebaine group. 3. Alcohols of the 6,14-endo-ethenotetrahydrooripavine series and derived analogs of N-allylnormorphine and -norcodeine. J Am Chem Soc 89: 3281–3292

    CAS  PubMed  Google Scholar 

  230. Rolly G, Versichelen L (1976) Buprenorphine as postoperative analgesic. Acta Anaesthesiol Belg 27:183–186

    CAS  PubMed  Google Scholar 

  231. Jasinski DR, Pevnick JS, Griffith JD (1978) Human pharmacology and abuse potential of the analgesic buprenorphine: a potential agent for treating narcotic addiction. Arch Gen Psychiatry 35:501–516

    CAS  PubMed  Google Scholar 

  232. Barth KS, Malcolm RJ (2010) Disulfiram: an old therapeutic with new applications. CNS Neurol Disord Drug Targets 9:5–12

    CAS  PubMed  Google Scholar 

  233. Shorter D, Kosten TR (2011) Novel pharmacotherapeutic treatments for cocaine addiction. BMC Med 9:119

    CAS  PubMed Central  PubMed  Google Scholar 

  234. Bourdelat-Parks BN, Anderson GM, Donaldson ZR, Weiss JM, Bonsall RW, Emery MS, Liles LC, Weinshenker D (2005) Effects of dopamine beta-hydroxylase genotype and disulfiram inhibition on catecholamine homeostasis in mice. Psychopharmacology (Berl) 183:72–80

    CAS  Google Scholar 

  235. Schank JR, Ventura R, Puglisi-Allegra S, Alcaro A, Cole CD, Liles LC, Seeman P, Weinshenker D (2006) Dopamine beta-hydroxylase knockout mice have alterations in dopamine signaling and are hypersensitive to cocaine. Neuropsychopharmacology 31: 2221–2230

    CAS  PubMed  Google Scholar 

  236. Carroll KM, Nich C, Ball SA, McCance E, Rounsavile BJ (1998) Treatment of cocaine and alcohol dependence with psychotherapy and disulfiram. Addiction 93: 713–727

    CAS  PubMed  Google Scholar 

  237. Carroll KM, Fenton LR, Ball SA, Nich C, Frankforter TL, Shi J, Rounsaville BJ (2004) Efficacy of disulfiram and cognitive behavior therapy in cocaine-dependent outpatients: a randomized placebo-controlled trial. Arch Gen Psychiatry 61:264–272

    CAS  PubMed Central  PubMed  Google Scholar 

  238. Petrakis IL, Carroll KM, Nich C, Gordon LT, McCance-Katz EF, Frankforter T, Rounsaville BJ (2000) Disulfiram treatment for cocaine dependence in methadone-maintained opioid addicts. Addiction 95:219–228

    CAS  PubMed  Google Scholar 

  239. McCance-Katz EF, Kosten TR, Jatlow P (1998) Disulfiram effects on acute cocaine administration. Drug Alcohol Depend 52: 27–39

    CAS  PubMed  Google Scholar 

  240. Hameedi FA, Rosen MI, McCance-Katz EF, McMahon TJ, Price LH, Jatlow PI, Woods SW, Kosten TR (1995) Behavioral, physiological, and pharmacological interaction of cocaine and disulfiram in humans. Biol Psychiatry 37:560–563

    CAS  PubMed  Google Scholar 

  241. Baker JR, Jatlow P, McCance-Katz EF (2007) Disulfiram effects on responses to intravenous cocaine administration. Drug Alcohol Depend 87:202–209

    CAS  PubMed Central  PubMed  Google Scholar 

  242. Carroll KM, Ziedonis D, O’Malley S, McCance-Katz E, Gordon L, Rounsaville B (1993) Pharmacologic interventions for alcohol- and cocaine-abusing individuals. Am J Addict 2:77–79

    Google Scholar 

  243. Higgins ST, Budney AJ, Bickel WK, Hughes JR, Foerg F (1993) Disulfiram therapy in patients abusing cocaine and alcohol. Am J Psychiatry 150:675–676

    CAS  PubMed  Google Scholar 

  244. Kleiderer E, Rice J, Conquest V, Williams J (1945) Pharmaceutical activities at the I.G. Farbenindustrie plant, Hochst am Main. Office of Publication Board, Department of Commerce, Washington, DC

    Google Scholar 

  245. Isbell H, Wilker A et al (1948) Liability of addiction to 6-dimethylamino-4-4-diphenyl-3-heptanone (methadon, amidone or 10820) in man; experimental addiction to methadon. Arch Intern Med (Chic) 82:362–392

    CAS  Google Scholar 

  246. Dole VP, Nyswander ME, Kreek MJ (1966) Narcotic blockade–a medical technique for stopping heroin use by addicts. Trans Assoc Am Physicians 79:122–136

    CAS  PubMed  Google Scholar 

  247. Thorsell A (2013) The mu-opioid receptor and treatment response to naltrexone. Alcohol Alcohol 48:402–408

    CAS  PubMed  Google Scholar 

  248. Bart G (2012) Maintenance medication for opiate addiction: the foundation of recovery. J Addict Dis 31:207–225

    PubMed Central  PubMed  Google Scholar 

  249. Bymaster FP, Calligaro DO, Falcone JF, Marsh RD, Moore NA, Tye NC, Seeman P, Wong DT (1996) Radioreceptor binding profile of the atypical antipsychotic olanzapine. Neuropsychopharmacology 14:87–96

    CAS  PubMed  Google Scholar 

  250. Bymaster FP, Rasmussen K, Calligaro DO, Nelson DL, DeLapp NW, Wong DT, Moore NA (1997) In vitro and in vivo biochemistry of olanzapine: a novel, atypical antipsychotic drug. J Clin Psychiatry 58(Suppl 10):28–36

    CAS  PubMed  Google Scholar 

  251. Constenla M (2004) 5-HT3 receptor antagonists for prevention of late acute-onset emesis. Ann Pharmacother 38:1683–1691

    CAS  PubMed  Google Scholar 

  252. Peters DH, Faulds D (1994) Tiapride. A review of its pharmacology and therapeutic potential in the management of alcohol dependence syndrome. Drugs 47:1010–1032

    CAS  PubMed  Google Scholar 

  253. Sachse C, Brockmoller J, Bauer S, Roots I (1997) Cytochrome P450 2D6 variants in a Caucasian population: allele frequencies and phenotypic consequences. Am J Hum Genet 60:284–295

    CAS  PubMed Central  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to David A. Nielsen .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2014 Springer Science+Business Media New York

About this protocol

Cite this protocol

Nielsen, D.A., Nielsen, E.M., Dasari, T., Spellicy, C.J. (2014). Pharmacogenetics of Addiction Therapy. In: Yan, Q. (eds) Pharmacogenomics in Drug Discovery and Development. Methods in Molecular Biology, vol 1175. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-0956-8_15

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-0956-8_15

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-0955-1

  • Online ISBN: 978-1-4939-0956-8

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics