A Delta-Opioid Receptor Gene Polymorphism Moderates the Therapeutic Response to Extended-Release Buprenorphine in Opioid Use Disorder

Henry R Kranzler, Kevin G Lynch, Richard C Crist, Emily Hartwell, Anne Le Moigne, Celine M Laffont, Anne C Andorn, Henry R Kranzler, Kevin G Lynch, Richard C Crist, Emily Hartwell, Anne Le Moigne, Celine M Laffont, Anne C Andorn

Abstract

Background: Buprenorphine treatment is not equally effective in all patients with opioid use disorder (OUD). Two retrospective studies showed that, among African Americans (AAs), rs678849, a polymorphism in the delta-opioid receptor gene, moderated the therapeutic effect of sublingual buprenorphine.

Methods: We examined rs678849 as a moderator of the response to an extended-release subcutaneous buprenorphine formulation (BUP-XR) in a 24-week OUD treatment study of 127 AAs and 327 European Americans (EAs). Participants were randomly assigned to receive: (1) BUP-XR as 2 monthly injections of 300 mg followed by either 300 mg monthly or 100 mg monthly for 4 months, or (2) monthly volume-matched placebo injections. Generalized estimating equations logistic regression analyses tested, per population group, the main and interaction effects of treatment (BUP-XR vs placebo) and genotype group (rs678849*CC vs CT/TT) on weekly urine drug screens (UDS).

Results: Among AAs, the placebo group had higher rates of opioid-positive UDS than the BUP-XR group (log odds ratio = 1.67, 95% CI = 0.36, 2.98), but no genotype by treatment effect (P = .80). Among EAs, the placebo group also showed higher rates of opioid-positive UDS than the BUP-XR group (log odds ratio = 1.97, 95% CI = 1.14, 2.79) but a significant genotype by treatment interaction (χ 2(1) = 4.33, P = .04).

Conclusion: We found a moderating effect of rs678849 on the response to buprenorphine treatment of OUD in EAs, but not AAs. These findings require replication in well-powered, prospective studies of both AA and EA OUD patients treated with BUP-XR and stratified on rs678849 genotype.

Trial registration: ClinicalTrials.gov NCT02357901.

Keywords: Buprenorphine; delta-opioid receptor; opioid use disorder; pharmacogenetic; rs678849.

© The Author(s) 2020. Published by Oxford University Press on behalf of CINP.

Figures

Figure 1.
Figure 1.
Proportion of opioid-positive urine drug screens by treatment and genotype groups in African Americans.
Figure 2.
Figure 2.
Proportion of opioid-positive urine drug screens by treatment and genotype groups in European Americans.

References

    1. American Psychiatric Association (2013) Diagnostic and statistical manual of mental disorders: diagnostic and statistical manual of mental disorders. 5th ed. Arlington, VA: American Psychiatric Association.
    1. Blum K, Han D, Modestino EJ, Saunders S, Roy AK 3rd, Jacobs W, Inaba DS, Baron D, Oscar-Berman M, Hauser M, Badgaiyan RD, Smith DE, Femino J, Gold MS (2018) A systematic, intensive statistical investigation of data from the Comprehensive Analysis of Reported Drugs (CARD) for compliance and illicit opioid abstinence in substance addiction treatment with buprenorphine/naloxone. Subst Use Misuse 53:220–229.
    1. Compton WM, Jones CM, Baldwin GT (2016) Relationship between nonmedical prescription-opioid use and heroin use. N Engl J Med 374:154–163.
    1. Cooper DN (2010) Functional intronic polymorphisms: buried treasure awaiting discovery within our genes. Hum Genomics 4:284–288.
    1. 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 38:2003–2010.
    1. Crist RC, Phillips KA, Furnari MA, Moran LM, Doyle GA, McNicholas LF, Cornish JW, Kampman KM, Preston KL, Berrettini WH (2019) Replication of the pharmacogenetic effect of rs678849 on buprenorphine efficacy in African-Americans with opioid use disorder. Pharmacogenomics J 19:260–268.
    1. FDA Briefing Document RBP-6000 (extended-release buprenorphine) Joint Meeting of Psychopharmacologic Drugs Advisory Committee and Drug Safety and Risk Management Advisory Committee October 31, 2017. . Accessed March 22, 2020.
    1. Fitzmaurice GM, Laird NM, Ware JH (2011) Applied longitudinal analysis. 2nd ed. New York: Wiley Series in Probability and Statistics.
    1. Greenwald M, Johanson CE, Bueller J, Chang Y, Moody DE, Kilbourn M, Koeppe R, Zubieta JK (2007) Buprenorphine duration of action: mu-opioid receptor availability and pharmacokinetic and behavioral indices. Biol Psychiatry 61:101–110.
    1. Haight BR, Learned SM, Laffont CM, Fudala PJ, Zhao Y, Garofalo AS, Greenwald MK, Nadipelli VR, Ling W, Heidbreder C; RB-US-13-0001 Study Investigators (2019) Efficacy and safety of a monthly buprenorphine depot injection for opioid use disorder: a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 393:778–790.
    1. Hancock DB, et al. (2015) Cis-expression quantitative trait loci mapping reveals replicable associations with heroin addiction in OPRM1. Biol Psychiatry 78:474–484.
    1. Handelsman L, Cochrane KJ, Aronson MJ, Ness R, Rubinstein KJ, Kanof PD (1987) Two new rating scales for opiate withdrawal. Am J Drug Alcohol Abuse 13:293–308.
    1. Hedegaard H, Warner M, Miniño AM (2017) Drug overdose deaths in the United States, 1999–2016. NCHS Data Brief 294: 1– 8.
    1. Huang P, Kehner GB, Cowan A, Liu-Chen LY (2001) Comparison of pharmacological activities of buprenorphine and norbuprenorphine: norbuprenorphine is a potent opioid agonist. J Pharmacol Exp Ther 297:688–695.
    1. Manolio TA, et al. (2009) Finding the missing heritability of complex diseases. Nature 461:747–753.
    1. Motsinger-Reif AA, Jorgenson E, Relling MV, Kroetz DL, Weinshilboum R, Cox NJ, Roden DM (2013) Genome-wide association studies in pharmacogenomics: successes and lessons. Pharmacogenet Genomics 23:383–394.
    1. Negus SS, Bidlack JM, Mello NK, Furness MS, Rice KC, Brandt MR (2002) Delta opioid antagonist effects of buprenorphine in rhesus monkeys. Behav Pharmacol 13:557–570.
    1. Paulozzi LJ, Jones C, Mack K, Rudd R (2011) Vital signs: overdoses of prescription opioid pain relievers—United States, 1999–2008. Morb Mortal Wkly Rep 60:1487–1492.
    1. Ross S, Anand SS, Joseph P, Paré G (2012) Promises and challenges of pharmacogenetics: an overview of study design, methodological and statistical issues. JRSM Cardiovasc Dis 1:cvd.2012.012001.
    1. Roux P, Lions C, Michel L, Cohen J, Mora M, Marcellin F, Spire B, Morel A, Carrieri PM, Karila L; ANRS Methaville Study Group (2014) Predictors of non-adherence to methadone maintenance treatment in opioid-dependent individuals: implications for clinicians. Curr Pharm Des 20:4097–4105.
    1. Rudd RA, Seth P, David F, Scholl L (2016) Increases in drug and opioid-involved overdose deaths - United States, 2010-2015. MMWR Morb Mortal Wkly Rep 65:1445–1452.
    1. Substance Abuse and Mental Health Services Administration (2019) Key substance use and mental health indicators in the United States: Results from the 2018 National Survey on Drug Use and Health (HHS Publication No. PEP19-5068, NSDUH Series H-54). Rockville, MD: Center for Behavioral Health Statistics and Quality, Substance Abuse and Mental Health Services Administration; . Accessed April 25, 2020.
    1. Tkacz J, Severt J, Cacciola J, Ruetsch C (2012) Compliance with buprenorphine medication-assisted treatment and relapse to opioid use. Am J Addict 21:55–62.
    1. Valentino RJ, Volkow ND (2018) Untangling the complexity of opioid receptor function. Neuropsychopharmacology 43:2514–2520.
    1. Wesson DR, Ling W (2003) The Clinical Opiate Withdrawal Scale (COWS). J Psychoactive Drugs 35:253–259.

Source: PubMed

3
Předplatit