Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Replication of the pharmacogenetic effect of rs678849 on buprenorphine efficacy in African–Americans with opioid use disorder

Abstract

Many patients with opioid use disorder do not have successful outcomes during treatment but the underlying reasons are not well understood. An OPRD1 variant (rs678849) was previously associated with methadone and buprenorphine efficacy in African–Americans with opioid use disorder. The objective of this study was to determine if the effect of rs678849 on opioid use disorder treatment outcome could be replicated in an independent population. Participants were recruited from African–American patients who had participated in previous studies of methadone or buprenorphine treatment at the outpatient treatment research clinic of the NIDA Intramural Research Program in Baltimore, MD, USA between 2000 and 2017. Rs678849 was genotyped retrospectively, and genotypes were compared with urine drug screen results from the previous studies for opioids other than the one prescribed for treatment. Genotypes were available for 24 methadone patients and 55 buprenorphine patients. After controlling for demographics, the effect of rs678849 genotype was significant in the buprenorphine treatment group (RR = 1.69, 95% confidence interval (CI) 1.59–1.79, p = 0.021). Buprenorphine patients with the C/C genotype were more likely to have opioid-positive drug screens than individuals with the C/T or T/T genotypes, replicating the original pharmacogenetic finding. The effect of genotype was not significant in the methadone group (p = 0.087). Thus, the genotype at rs678849 is associated with buprenorphine efficacy in African–Americans being treated for opioid use disorder. This replication suggests that rs678849 genotype may be a valuable pharmacogenetic marker for deciding which opioid use disorder medication to prescribe in this population.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1

Similar content being viewed by others

References

  1. 2015 National Survey on Drug Use and Health: Detailed Tables. Substance Abuse and Mental Health Services Administration, Rockville, MD.

  2. Mattick RP, Breen C, Kimber J, Davoli M. Buprenorphine maintenance versus placebo or methadone maintenance for opioid dependence. Cochrane Database Syst Rev 2014;CD002207.

  3. Kranzler HR, Covault J, Feinn R, Armeli S, Tennen H, Arias AJ, et al. Topiramate treatment for heavy drinkers: moderation by a GRIK1 polymorphism. Am J Psychiatry. 2014;171:445–52.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Lerman C, Wileyto EP, Patterson F, Rukstalis M, Audrain-McGovern J, Restine S, et al. The functional mu opioid receptor (OPRM1) Asn40Asp variant predicts short-term response to nicotine replacement therapy in a clinical trial. Pharm J. 2004;4:184–92.

    CAS  Google Scholar 

  5. Anton RF, Oroszi G, O’Malley S, Couper D, Swift R, Pettinati H, et al. 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. 2008;65:135–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Chamorro AJ, Marcos M, Miron-Canelo JA, Pastor I, Gonzalez-Sarmiento R, Laso FJ. Association of micro-opioid receptor (OPRM1) gene polymorphism with response to naltrexone in alcohol dependence: a systematic review and meta-analysis. Addict Biol. 2012;17:505–12.

    Article  CAS  PubMed  Google Scholar 

  7. Dennis BB, Bawor M, Thabane L, Sohani Z, Samaan Z. Impact of ABCB1 and CYP2B6 genetic polymorphisms on methadone metabolism, dose and treatment response in patients with opioid addiction: a systematic review and meta-analysis. PLoS ONE. 2014;9:e86114.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Kharasch ED, Stubbert K. Role of cytochrome P4502B6 in methadone metabolism and clearance. J Clin Pharmacol. 2013;53:305–13.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Iribarne C, Picart D, Dreano Y, Bail JP, Berthou F. Involvement of cytochrome P450 3A4 in N-dealkylation of buprenorphine in human liver microsomes. Life Sci. 1997;60:1953–64.

    Article  CAS  PubMed  Google Scholar 

  10. Kobayashi K, Yamamoto T, Chiba K, Tani M, Shimada N, Ishizaki T, et al. Human buprenorphine N-dealkylation is catalyzed by cytochrome P450 3A4. Drug Metab Dispos. 1998;26:818–21.

    CAS  PubMed  Google Scholar 

  11. Totah RA, Sheffels P, Roberts T, Whittington D, Thummel K, Kharasch ED. Role of CYP2B6 in stereoselective human methadone metabolism. Anesthesiology. 2008;108:363–74.

    Article  CAS  PubMed  Google Scholar 

  12. Shiran MR, Lennard MS, Iqbal MZ, Lagundoye O, Seivewright N, Tucker GT, et al. Pharmacokinetic-pharmacodynamic modeling of mood and withdrawal symptoms in relation to plasma concentrations of methadone in patients undergoing methadone maintenance treatment. J Clin Psychopharmacol. 2012;32:666–71.

    Article  CAS  PubMed  Google Scholar 

  13. Tsai HJ, Wang SC, Liu SW, Ho IK, Chang YS, Tsai YT, et al. Assessment of CYP450 genetic variability effect on methadone dose and tolerance. Pharmacogenomics. 2014;15:977–86.

    Article  CAS  PubMed  Google Scholar 

  14. Levran O, Peles E, Hamon S, Randesi M, Adelson M, Kreek MJ. CYP2B6 SNPs are associated with methadone dose required for effective treatment of opioid addiction. Addict Biol. 2013;18:709–16.

    Article  CAS  PubMed  Google Scholar 

  15. Levran O, O’Hara K, Peles E, Li D, Barral S, Ray B, et al. ABCB1 (MDR1) genetic variants are associated with methadone doses required for effective treatment of heroin dependence. Hum Mol Genet. 2008;17:2219–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Barratt DT, Coller JK, Hallinan R, Byrne A, White JM, Foster DJ, et al. ABCB1 haplotype and OPRM1 118A > G genotype interaction in methadone maintenance treatment pharmacogenetics. Pharmgenomics Pers Med. 2012;5:53–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Lee HY, Li JH, Sheu YL, Tang HP, Chang WC, Tang TC, et al. Moving toward personalized medicine in the methadone maintenance treatment program: a pilot study on the evaluation of treatment responses in Taiwan. Biomed Res Int. 2013;2013:741403.

    PubMed  PubMed Central  Google Scholar 

  18. Yang HC, Chu SK, Huang CL, Kuo HW, Wang SC, Liu SW, et al. Genome-wide pharmacogenomic study on methadone maintenance treatment identifies SNP rs17180299 and multiple haplotypes on CYP2B6, SPON1, and GSG1L associated with plasma concentrations of methadone R- and S-enantiomers in heroin-dependent patients. PLoS Genet. 2016;12:e1005910.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Smith AH, Jensen KP, Li J, Nunez Y, Farrer LA, Hakonarson H, et al. Genome-wide association study of therapeutic opioid dosing identifies a novel locus upstream of OPRM1. Mol Psychiatry. 2017;22:346–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Oneda B, Crettol S, Bochud M, Besson J, Croquette-Krokar M, Hammig R, et al. Beta-Arrestin2 influences the response to methadone in opioid-dependent patients. Pharm J. 2011;11:258–66.

    CAS  Google Scholar 

  21. Crettol S, Besson J, Croquette-Krokar M, Hammig R, Gothuey I, Monnat M, et al. Association of dopamine and opioid receptor genetic polymorphisms with response to methadone maintenance treatment. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:1722–7.

    Article  CAS  PubMed  Google Scholar 

  22. de Cid R, Fonseca F, Gratacos M, Gutierrez F, Martin-Santos R, Estivill X, et al. BDNF variability in opioid addicts and response to methadone treatment: preliminary findings. Genes Brain Behav. 2008;7:515–22.

    Article  PubMed  Google Scholar 

  23. Eap CB, Broly F, Mino A, Hammig R, Deglon JJ, Uehlinger C, et al. Cytochrome P450 2D6 genotype and methadone steady-state concentrations. J Clin Psychopharmacol. 2001;21:229–34.

    Article  CAS  PubMed  Google Scholar 

  24. Saxon AJ, Ling W, Hillhouse M, Thomas C, Hasson A, Ang A, et al. Buprenorphine/Naloxone and methadone effects on laboratory indices of liver health: a randomized trial. Drug Alcohol Depend. 2013;128:71–76.

    Article  CAS  PubMed  Google Scholar 

  25. Clarke TK, Crist RC, Ang A, Ambrose-Lanci LM, Lohoff FW, Saxon AJ, et al. Genetic variation in OPRD1 and the response to treatment for opioid dependence with buprenorphine in European-American females. Pharm J. 2014;14:303–8.

    CAS  Google Scholar 

  26. Crist RC, Doyle GA, Nelson EC, Degenhardt L, Martin NG, Montgomery GW, et al. A polymorphism in the OPRM1 3’-untranslated region is associated with methadone efficacy in treating opioid dependence. Pharmacogenomics J. 2018; 18:173-9.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Crist RC, Clarke TK, Ang A, Ambrose-Lanci LM, Lohoff FW, Saxon AJ, et al. An intronic variant in OPRD1 predicts treatment outcome for opioid dependence in African-Americans. Neuropsychopharmacology. 2013;38:2003–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhang H, Kranzler HR, Yang BZ, Luo X, Gelernter J. The OPRD1 and OPRK1 loci in alcohol or drug dependence: OPRD1 variation modulates substance dependence risk. Mol Psychiatry. 2008;13:531–43.

    Article  CAS  PubMed  Google Scholar 

  29. Epstein DH, Schmittner J, Umbricht A, Schroeder JR, Moolchan ET, Preston KL. Promoting abstinence from cocaine and heroin with a methadone dose increase and a novel contingency. Drug Alcohol Depend. 2009;101:92–100.

    Article  PubMed  Google Scholar 

  30. Epstein DH, Willner-Reid J, Vahabzadeh M, Mezghanni M, Lin JL, Preston KL. Real-time electronic diary reports of cue exposure and mood in the hours before cocaine and heroin craving and use. Arch Gen Psychiatry. 2009;66:88–94.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Kowalczyk WJ, Phillips KA, Jobes ML, Kennedy AP, Ghitza UE, Agage DA, et al. Clonidine maintenance prolongs opioid abstinence and decouples stress from craving in daily life: a randomized controlled trial with ecological momentary assessment. Am J Psychiatry. 2015;172:760–7.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Preston KL, Ghitza UE, Schmittner JP, Schroeder JR, Epstein DH. Randomized trial comparing two treatment strategies using prize-based reinforcement of abstinence in cocaine and opiate users. J Appl Behav Anal. 2008;41:551–63.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Preston KL, Kowalczyk WJ, Phillips KA, Jobes ML, Vahabzadeh M, Lin JL, et al. Context and craving during stressful events in the daily lives of drug-dependent patients. Psychopharmacology (Berl). 2017; 234:2631-42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Crist RC, Ambrose-Lanci LM, Vaswani M, Clarke TK, Zeng A, Yuan C, et al. Case-control association analysis of polymorphisms in the delta-opioid receptor, OPRD1, with cocaine and opioid addicted populations. Drug Alcohol Depend. 2013;127:122–8.

    Article  CAS  PubMed  Google Scholar 

  35. Brewer DD, Catalano RF, Haggerty K, Gainey RR, Fleming CB. A meta-analysis of predictors of continued drug use during and after treatment for opiate addiction. Addiction. 1998;93:73–92.

    Article  CAS  PubMed  Google Scholar 

  36. Birnbaum HG, White AG, Schiller M, Waldman T, Cleveland JM, Roland CL. Societal costs of prescription opioid abuse, dependence, and misuse in the United States. Pain Med. 2011;12:657–67.

    Article  PubMed  Google Scholar 

  37. Sharafshah A, Fazel H, Albonaim A, Omarmeli V, Rezaei S, Mirzajani E, et al. Association of OPRD1 gene variants with opioid dependence in addicted male individuals undergoing methadone treatment in the north of Iran. J Psychoact Drugs. 2017;49:242–51.

    Article  Google Scholar 

  38. Roussotte FF, Jahanshad N, Hibar DP, Sowell ER, Kohannim O, Barysheva M, et al. A commonly carried genetic variant in the delta opioid receptor gene, OPRD1, is associated with smaller regional brain volumes: replication in elderly and young populations. Hum Brain Mapp. 2014;35:1226–36.

    Article  PubMed  Google Scholar 

  39. Chadwick LH. The NIH Roadmap Epigenomics Program data resource. Epigenomics. 2012;4:317–24.

    Article  CAS  PubMed  Google Scholar 

  40. Ward LD, Kellis M. HaploReg v4: systematic mining of putative causal variants, cell types, regulators and target genes for human complex traits and disease. Nucleic Acids Res. 2016;44(D1):D877–81.

    Article  CAS  PubMed  Google Scholar 

  41. The GTEx Consortium. Human genomics. The Genotype-tissue expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015;348:648–60.

    Article  Google Scholar 

  42. Piltonen M, Parisien M, Gregoire S, Chabot-Dore AJ, Jafarnejad SM, Berube P, et al. Alternative Splicing of the delta-opioid receptor gene suggests existence of new functional isoforms. Mol Neurobiol 2018 https://doi.org/10.1007/s12035-018-1253-z.

    Article  PubMed  Google Scholar 

  43. Negus SS, Bidlack JM, Mello NK, Furness MS, Rice KC, Brandt MR. Delta opioid antagonist effects of buprenorphine in rhesus monkeys. Behav Pharmacol. 2002;13:557–70.

    Article  CAS  PubMed  Google Scholar 

  44. Kong H, Raynor K, Yasuda K, Moe ST, Portoghese PS, Bell GI, et al. A single residue, aspartic acid 95, in the delta opioid receptor specifies selective high affinity agonist binding. J Biol Chem. 1993;268:23055–8.

    CAS  PubMed  Google Scholar 

  45. Belcheva MM, Barg J, McHale RJ, Dawn S, Ho MT, Ignatova E, et al. Differential down- and up-regulation of rat brain opioid receptor types and subtypes by buprenorphine. Mol Pharmacol. 1993;44:173–9.

    CAS  PubMed  Google Scholar 

  46. Belcheva MM, Ho MT, Ignatova EG, Jefcoat LB, Barg J, Vogel Z, et al. Buprenorphine differentially alters opioid receptor adaptation in rat brain regions. J Pharmacol Exp Ther. 1996;277:1322–7.

    CAS  PubMed  Google Scholar 

  47. Law PY, Erickson-Herbrandson LJ, Zha QQ, Solberg J, Chu J, Sarre A, et al. Heterodimerization of mu- and delta-opioid receptors occurs at the cell surface only and requires receptor-G protein interactions. J Biol Chem. 2005;280:11152–64.

    Article  CAS  PubMed  Google Scholar 

  48. Liu JG, Liao XP, Gong ZH, Qin BY. Methadone-induced desensitization of the delta-opioid receptor is mediated by uncoupling of receptor from G protein. Eur J Pharmacol. 1999;374:301–8.

    Article  CAS  PubMed  Google Scholar 

  49. Damian AJ, Mendelson T, Agus D. Predictors of buprenorphine treatment success of opioid dependence in two Baltimore City grassroots recovery programs. Addict Behav. 2017;73:129–32.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Mertens JR, Weisner CM. Predictors of substance abuse treatment retention among women and men in an HMO. Alcohol Clin Exp Res. 2000;24:1525–33.

    Article  CAS  PubMed  Google Scholar 

  51. McCaul ME, Svikis DS, Moore RD. Predictors of outpatient treatment retention: patient versus substance use characteristics. Drug Alcohol Depend. 2001;62:9–17.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

START study funding came from the National Institute on Drug Abuse (NIDA) through the Clinical Trials Network (CTN) through a series of grants provided to each participating node: the Pacific Northwest Node (U10 DA01714), the Oregon Hawaii Node (U10 DA013036), the California/Arizona Node (U10 DA015815), the New England Node (U10 DA13038), the Delaware Valley Node (U10 DA13043), the Pacific Region Node (U10 DA13045), and the New York Node (U10 DA013046). Protocol 326, Protocol 385, Protocol 407, and Protocol 020 were supported by the Intramural Research Program of the NIH NIDA. Dr. Berrettini was supported by the Delaware Valley Node of the CTN (U10 DA13043) and by NIDA grant R21 DA036808. Dr. Crist was supported by NIDA grant K01 DA036751. Drs. Crist, Berrettini, and Doyle were supported by NIDA grant R01 DA044015. The funders had no role in design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Richard C. Crist.

Ethics declarations

Conflict of interest

KMK has received consulting fees from Alkermes and Opiant Pharmaceuticals. He has also received grant funding from Alkermes, Opiant Pharmaceuticals, and Indivior. WHB has received consulting fees from Mundipharma and Geisinger Health Systems and grant support from Saniona. The remaining authors declare no conflict of interests.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Crist, R.C., Phillips, K.A., Furnari, M.A. et al. Replication of the pharmacogenetic effect of rs678849 on buprenorphine efficacy in African–Americans with opioid use disorder. Pharmacogenomics J 19, 260–268 (2019). https://doi.org/10.1038/s41397-018-0065-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41397-018-0065-x

This article is cited by

Search

Quick links