1
|
Logge WB, Haber PS, Hurzeler T, Gallagher H, Kranzler H, Morley KC. Neural cue reactivity and intrinsic functional connectivity in individuals with alcohol use disorder following treatment with topiramate or naltrexone. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06745-7. [PMID: 39853353 DOI: 10.1007/s00213-025-06745-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 01/10/2025] [Indexed: 01/26/2025]
Abstract
RATIONALE Both topiramate and naltrexone have been shown to affect neural alcohol cue reactivity in alcohol use disorder (AUD). However, their comparative effects on alcohol cue reactivity are unknown. Moreover, while naltrexone has been found to normalize hyperactive localized network connectivity implicated in AUD, no studies have examined the effect of topiramate on intrinsic functional connectivity or compared functional connectivity between these two widely used medications. OBJECTIVE This study compared topiramate versus naltrexone on alcohol cue-elicited brain activation and intrinsic functional connectivity in patients with alcohol use disorder. METHODS Forty-seven participants with alcohol use disorder received daily topiramate (titrating the dose up to 200 mg/day n = 21) or naltrexone (50 mg/day, n = 26) for at least 6 weeks. Using functional magnetic resonance imaging (fMRI), we examined intrinsic functional connectivity during rest and alcohol cue-elicited neural activation during a visual alcohol cue reactivity task 120 min following treatment administration. Functional connectivity and alcohol cue reactivity and percentage of heavy drinking days (% HDD) associations were assessed. RESULTS No differences in either intrinsic functional connectivity or alcohol cue-elicited neural activity were seen between topiramate and naltrexone-treated groups. Overall, participants showed increased alcohol cue-elicited activation in three clusters spanning occipital regions involved in visual recognition of stimuli, and hypoactivation to both alcohol and control cues in three clusters involved in salience attribution and processing of emotional valence of external stimuli. No differences between topiramate versus naltrexone were observed for either functional measure or associations with post-scan % HDD. CONCLUSIONS Topiramate and naltrexone enacted comparable alcohol cue reactivity and intrinsic functional connectivity patterns. Some overall responses of increased brain activation to alcohol cues in visual processing regions coupled with reduced activation to alcohol and control cues were evidenced for both treatments. These activation patterns were in regions expected to show attenuation of brain activity resulting from treatment. Topiramate and naltrexone may thus enact functional effects through similar modulation of functional neural activity in individuals with AUD. TRIAL REGISTRATION ClinicalTrials.gov, NCT03479086 https://www. CLINICALTRIALS gov/study/NCT03479086 .
Collapse
Affiliation(s)
- Warren B Logge
- Edith Collins Centre for Translational Research in Alcohol, Drugs and Toxicology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia.
- Specialty of Addiction Medicine, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
- Specialty of Addiction Medicine, Lv 6, King George V Building 83-117 Missenden Rd, Camperdown, NSW, 2050, Australia.
| | - Paul S Haber
- Edith Collins Centre for Translational Research in Alcohol, Drugs and Toxicology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
- Drug Health Services, Sydney Local Health District, Sydney, NSW, Australia
| | - Tristan Hurzeler
- Edith Collins Centre for Translational Research in Alcohol, Drugs and Toxicology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
- Specialty of Addiction Medicine, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Hugh Gallagher
- Edith Collins Centre for Translational Research in Alcohol, Drugs and Toxicology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
- Specialty of Addiction Medicine, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Henry Kranzler
- Center for Studies of Addiction, Perelman School of Medicine, Education, and Clinical Center, University of Pennsylvania and Mental Illness Research, Crescenz VAMC, Philadelphia, PA, USA
| | - Kirsten C Morley
- Edith Collins Centre for Translational Research in Alcohol, Drugs and Toxicology, Royal Prince Alfred Hospital, Sydney Local Health District, Sydney, NSW, Australia
- Specialty of Addiction Medicine, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
2
|
Schacht JP, Hoffman M, Chen BH, Anton RF. Epigenetic moderators of naltrexone efficacy in reducing heavy drinking in Alcohol Use Disorder: a randomized trial. THE PHARMACOGENOMICS JOURNAL 2022; 22:1-8. [PMID: 34381173 PMCID: PMC8799481 DOI: 10.1038/s41397-021-00250-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023]
Abstract
Polymorphisms in genes associated with opioid signaling and dopamine reuptake and inactivation may moderate naltrexone efficacy in Alcohol Use Disorder (AUD), but the effects of epigenetic modification of these genes on naltrexone response are largely unexplored. This study tested interactions between methylation in the μ-opioid receptor (OPRM1), dopamine transporter (SLC6A3), and catechol-O-methyltransferase (COMT) genes as predictors of naltrexone effects on heavy drinking in a 16-week randomized, placebo-controlled trial among 145 treatment-seeking AUD patients. OPRM1 methylation interacted with both SLC6A3 and COMT methylation to moderate naltrexone efficacy, such that naltrexone-treated individuals with lower methylation of the OPRM1 promoter and the SLC6A3 promoter (p = 0.006), COMT promoter (p = 0.005), or SLC6A3 3' untranslated region (p = 0.004), relative to placebo and to those with higher OPRM1 and SLC6A3 or COMT methylation, had significantly fewer heavy drinking days. Epigenetic modification of opioid- and dopamine-related genes may represent a novel pharmacoepigenetic predictor of naltrexone efficacy in AUD.
Collapse
Affiliation(s)
- Joseph P Schacht
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.
| | - Michaela Hoffman
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Brian H Chen
- FOXO Technologies Inc., Minneapolis, MN, USA
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, USA
| | - Raymond F Anton
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
3
|
Randomized Controlled Trial of an Alcohol-related Sexual Risk Reduction Intervention with Adolescents: The Role of Neurocognitive Activation During Risky Decision-Making. AIDS Behav 2021; 25:265-275. [PMID: 33712986 DOI: 10.1007/s10461-021-03190-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2021] [Indexed: 10/21/2022]
Abstract
Justice-involved youth are at a higher risk of negative outcomes from sexual activity and alcohol use relative to their non-justice involved peers. In the current study, we tested the extent to which variability in neurocognitive response (i.e., activation in the right superior parietal lobule; rSPL) during a risky decision-making task moderated the success of a sexual risk reduction intervention. In a cluster randomized trial blocked by gender, justice-involved adolescents (N = 269) first completed a risky decision-making task during a magnetic resonance imaging (MRI) session, then were assigned to an information-only control (GINFO) or sexual risk reduction intervention incorporating alcohol risk reduction content (GPI + GMET) and then re-contacted every three months for one year. Youth in the GPI + GMET intervention reported less sexual risk behavior 12 months after intervention than those in the control. Although neurocognitive activation was associated with sexual risk behavior, it did not moderate intervention outcomes. This risk-reduction intervention appears to work equally well across a range of neurocognitive responses.
Collapse
|
4
|
Anton RF, Voronin KE, Book SW, Latham PK, Randall PK, Glen WB, Hoffman M, Schacht JP. Opioid and Dopamine Genes Interact to Predict Naltrexone Response in a Randomized Alcohol Use Disorder Clinical Trial. Alcohol Clin Exp Res 2020; 44:2084-2096. [PMID: 32772383 PMCID: PMC8080431 DOI: 10.1111/acer.14431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/27/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND While the opiate antagonist, naltrexone, is approved for treating alcohol use disorder (AUD), not everyone who receives the medication benefits from it. This study evaluated whether the OPRM1 SNP rs1799971 interacts with the dopamine transporter gene DAT1/SLC6A3 VNTR rs28363170 or the catechol-O-methyltransferase (COMT) gene SNP rs4680 in predicting naltrexone response. METHODS Individuals who met DSM-IV alcohol dependence were randomly assigned to naltrexone (50 mg/d) or placebo based on their OPRM1 genotype (75 G-allele carriers and 77 A-allele homozygotes) and also genotyped for DAT1 VNTR (9 vs. 10 repeats) or COMT SNP (val/val vs. met carriers). Heavy drinking days (%HDD) were evaluated over 16 weeks and at the end of treatment. Effect sizes (d) for naltrexone response were calculated based on genotypes. RESULTS Naltrexone, relative to placebo, significantly reduced %HDD among OPRM1 G carriers who also had DAT1 10/10 (p = 0.021, d = 0.72) or COMT val/val genotypes (p = 0.05, d = 0.80), and to a lesser degree in those OPRM1 A homozygotes who were also DAT1 9-repeat carriers (p = 0.09, d = 0.70) or COMT met carriers (p = 0.03, d = 0.63). All other genotype combinations showed no differential response to naltrexone. Diarrhea/abdominal pain was more prominent in OPRM1 A homozygotes who were also DAT 9 or COMT met carriers. CONCLUSIONS These results suggest that individuals with AUD with a more opioid-responsive genotype (OPRM1 G carriers) respond better to naltrexone if they have genotypes indicating normal/less dopamine tone (DAT1 10,10 or COMT val,val), while those with a less responsive opioid-responsive genotype (OPRM1 A homozygotes) respond better to naltrexone if they have genotypes indicating greater dopamine tone (DAT1 9-repeat or COMT met carriers). These results could lead to more personalized AUD treatments.
Collapse
Affiliation(s)
- Raymond F Anton
- From the, Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Konstantin E Voronin
- From the, Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sarah W Book
- From the, Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Patricia K Latham
- From the, Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Patrick K Randall
- From the, Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Willam Bailey Glen
- From the, Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Michaela Hoffman
- From the, Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Joseph P Schacht
- From the, Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
5
|
Lohoff FW. Pharmacotherapies and personalized medicine for alcohol use disorder: a review. Pharmacogenomics 2020; 21:1117-1138. [PMID: 32807012 DOI: 10.2217/pgs-2020-0079] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Alcohol use disorder (AUD) is highly prevalent and among the leading causes of morbidity and mortality in the United States. Pharmacotherapies for AUD are limited, thus making identification of patient subgroups that are most likely to respond favorably crucial. In this article, pharmacogenetic research on US FDA-approved and commonly prescribed off-label medications for the treatment of AUD is comprehensively reviewed. While the field has advanced in understanding pharmacotherapies for AUD and potential genetic moderators of treatment responses, the pharmacogenetic data to guide the prescribing clinician are limited and should be interpreted with caution. Precision medicine for AUD with more beneficial treatment responses and minimal side effects remains a high priority for further research.
Collapse
Affiliation(s)
- Falk W Lohoff
- Section on Clinical Genomics & Experimental Therapeutics, National Institute on Alcohol Abuse & Alcoholism, NIH, Bethesda, MD 20892-1540, USA
| |
Collapse
|
6
|
Hartwell EE, Feinn R, Morris PE, Gelernter J, Krystal J, Arias AJ, Hoffman M, Petrakis I, Gueorguieva R, Schacht JP, Oslin D, Anton RF, Kranzler HR. Systematic review and meta-analysis of the moderating effect of rs1799971 in OPRM1, the mu-opioid receptor gene, on response to naltrexone treatment of alcohol use disorder. Addiction 2020; 115:1426-1437. [PMID: 31961981 PMCID: PMC7340566 DOI: 10.1111/add.14975] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/07/2019] [Accepted: 01/10/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS There is wide inter-individual variability in response to the treatment of alcohol use disorder (AUD) with the opioid receptor antagonist naltrexone. To identify patients who may be most responsive to naltrexone treatment, studies have examined the moderating effect of rs1799971, a single nucleotide polymorphism (SNP) that encodes a non-synonymous substitution (Asn40Asp) in the mu-opioid receptor gene, OPRM1. The aims of this study were to: (1) conduct a systematic review of randomized clinical trials (RCTs); (2) assess the bias of the available studies and gauge publication bias; and (3) meta-analyze the interaction effect of the Asn40Asp SNP on the response to naltrexone treatment. METHODS We searched for placebo-controlled RCTs that examined the effect of Asn40Asp on the response to naltrexone treatment of heavy drinking or AUD. We tested the hypothesis that the minor (Asp40) allele was associated with a greater reduction in five alcohol consumption measures (relapse to heavy drinking, abstinence, percentage of heavy drinking days, percentage of days abstinent and drinks per day) in naltrexone-treated participants by meta-analyzing the interaction effects using a random effects model. RESULTS Seven RCTs met the study criteria. Overall, risk of bias was low and we observed no evidence of publication bias. Of the five alcohol consumption outcomes considered, there was a nominally significant moderating effect of the Asn40Asp SNP only on drinks per day (d = -0.18, P = 0.02). However, the effect was not significant when multiple comparisons were taken into account. CONCLUSIONS From the evidence to date, it remains unclear whether rs1799971, the OPRM1 Asn40Asp single nucleotide polymorphism, predicts naltrexone treatment response in individuals with alcohol use disorder or heavy drinking.
Collapse
Affiliation(s)
- Emily E. Hartwell
- Mental Illness Research, Education and Clinical Center, Cpl. Michael J. Crescenz VAMC, Philadelphia, PA 19104
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Richard Feinn
- Department of Medical Sciences, Frank H. Netter School of Medicine at Quinnipiac University, North Haven, CT 06473
| | - Paige E. Morris
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Joel Gelernter
- Departments of Psychiatry, Genetics, and Neuroscience, Yale University School of Medicine, and VA Connecticut Healthcare, West Haven, CT 06516
| | - John Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510
| | - Albert J. Arias
- Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA 23298
| | - Michaela Hoffman
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425
| | - Ismene Petrakis
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510
| | - Ralitza Gueorguieva
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510
| | - Joseph P. Schacht
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425
| | - David Oslin
- Mental Illness Research, Education and Clinical Center, Cpl. Michael J. Crescenz VAMC, Philadelphia, PA 19104
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Raymond F. Anton
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425
| | - Henry R. Kranzler
- Mental Illness Research, Education and Clinical Center, Cpl. Michael J. Crescenz VAMC, Philadelphia, PA 19104
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
7
|
Ray LA, Green R, Roche DJ, Magill M, Bujarski S. Naltrexone effects on subjective responses to alcohol in the human laboratory: A systematic review and meta-analysis. Addict Biol 2019; 24:1138-1152. [PMID: 31148304 DOI: 10.1111/adb.12747] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 02/13/2019] [Accepted: 02/24/2019] [Indexed: 02/03/2023]
Abstract
Naltrexone (NTX) has been widely studied for the treatment of alcohol use disorder with overall support for its efficacy. The mechanisms of action of naltrexone are thought to involve attenuation of the hedonic effects of alcohol and potentiation of its aversive effects. In order to provide a quantitative estimate of the effects of naltrexone on subjective response to alcohol, the aims of this meta-analytic review are to examine the effects of naltrexone across four domains of subjective response. Meta-analyses of naltrexone effects on alcohol craving (k = 16, N = 686), stimulation (k = 15, N = 675), sedation (k = 18, N = 777), and negative mood (k = 9, N = 281) suggested that under laboratory conditions and compared with placebo, naltrexone reduces craving (Hedges g = -0.252; SE = 0.054; 95% CI, -0.375 to -0.130; P < 0.01), reduces stimulation (g = -0.223; SE = 0.067; 95% CI, -0.372 to -0.074; P < 0.01), increases sedation (g = 0.251; SE = 0.064; 95% CI, 0.112-0.389; P < 0.01), and increases negative mood (g = 0.227; SE = 0.047; 95% CI, 0.100-0.354; P < 0.01). Results were robust when drinks per month and alcohol dose were added to the models as covariates. The effects of naltrexone varied by severity of alcohol use with medication effects on craving and stimulation being observed in sample of both heavy drinkers and AUD individuals. These results are consistent with the hypothesized mechanisms of action of NTX, although the effects are of small magnitude. This meta-analysis aggregates across multiple human laboratory studies of NTX's effects on subjective response to alcohol, providing a comprehensive summary of a key mechanism of NTX efficacy, namely, alteration of the subjective experience of alcohol.
Collapse
Affiliation(s)
- Lara A. Ray
- Department of PsychologyUniversity of California, Los Angeles Los Angeles CA USA
- Department of Psychiatry and Biobehavioral SciencesUniversity of California, Los Angeles Los Angeles CA USA
| | - ReJoyce Green
- Department of PsychologyUniversity of California, Los Angeles Los Angeles CA USA
| | | | - Molly Magill
- Center for Alcohol and Addiction StudiesBrown University Providence RI USA
| | - Spencer Bujarski
- Department of PsychologyUniversity of California, Los Angeles Los Angeles CA USA
| |
Collapse
|
8
|
Zastrozhin MS, Skryabin VY, Miroshkin SS, Bryun EA, Sychev DA. Pharmacogenetics of alcohol addiction: current perspectives. APPLICATION OF CLINICAL GENETICS 2019; 12:131-140. [PMID: 31372024 PMCID: PMC6628972 DOI: 10.2147/tacg.s206745] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/10/2019] [Indexed: 12/31/2022]
Abstract
Genetics of alcohol addiction is currently a contradictive and complex field, where data in the most studies reflect methods’ limitations rather than meaningful and complementary results. In our review, we focus on the genetics of alcohol addiction, leaving genetics of acute alcohol intoxication out of the scope. A review of the literature on pharmacogenetic biomarkers development for the pharmacotherapy personalization reveals that today the evidence base concerning these biomarkers is still insufficient. In particular, now the researches with the design of randomized controlled trials and meta-analysis investigating the effect of the SNPs as biomarkers on the therapy efficacy are available for naltrexone only. For other medications, there are only a few studies in small samples. It decreases the possibilities to implement the pharmacogenetic algorithms for the pharmacotherapy personalization in patients with alcohol use disorders (AUD). In view of the importance of the precision approaches development not in addiction medicine only, but in other fields of medicine also to increase the efficacy and safety of the therapy, studies on pharmacogenetic biomarkers development for the medications used in patients with AUD (eg, naltrexone, disulfiram, nalmefene, acamprosate, etc.) remain relevant to this day.
Collapse
Affiliation(s)
- M S Zastrozhin
- Moscow Research and Practical Centre on Addictions of the Moscow Department of Healthcare , Moscow 109390, Russian Federation.,Department of Addictology, Russian Medical Academy of Continuous Professional Education of the Ministry of Health of the Russian Federation, Moscow 123995, Russian Federation
| | - V Yu Skryabin
- Moscow Research and Practical Centre on Addictions of the Moscow Department of Healthcare , Moscow 109390, Russian Federation
| | - S S Miroshkin
- Moscow Research and Practical Centre on Addictions of the Moscow Department of Healthcare , Moscow 109390, Russian Federation.,Department of Addictology, Russian Medical Academy of Continuous Professional Education of the Ministry of Health of the Russian Federation, Moscow 123995, Russian Federation
| | - E A Bryun
- Moscow Research and Practical Centre on Addictions of the Moscow Department of Healthcare , Moscow 109390, Russian Federation.,Department of Addictology, Russian Medical Academy of Continuous Professional Education of the Ministry of Health of the Russian Federation, Moscow 123995, Russian Federation
| | - D A Sychev
- Moscow Research and Practical Centre on Addictions of the Moscow Department of Healthcare , Moscow 109390, Russian Federation.,Department of Addictology, Russian Medical Academy of Continuous Professional Education of the Ministry of Health of the Russian Federation, Moscow 123995, Russian Federation
| |
Collapse
|
9
|
Lim AC, Ghahremani DG, Grodin EN, Green R, Bujarski S, Hartwell EE, Courtney KE, Hutchison K, Miotto K, Ray LA. Neuroimaging findings from an experimental pharmacology trial of naltrexone in heavy drinkers of East Asian descent. Drug Alcohol Depend 2019; 200:181-190. [PMID: 31160146 PMCID: PMC6760244 DOI: 10.1016/j.drugalcdep.2019.02.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/01/2019] [Accepted: 02/22/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Despite known genetic variation across races, studies examining pharmacogenetics of a single nucleotide polymorphism (SNP) of the mu-opioid receptor gene (OPRM1) on clinical response to naltrexone have been conducted in predominantly Caucasian samples. Evidence is mixed for pharmacogenetic OPRM1 and naltrexone effects on neural responses to alcohol cues. The current study tests the pharmacogenetic effects of naltrexone and OPRM1 on neural responses to alcohol taste cues in heavy drinkers of East Asian descent. METHODS Participants (N = 41) completed two double-blinded and counterbalanced functional magnetic resonance imaging (fMRI) sessions: one after taking naltrexone (50 mg/day) for four days and one after taking placebo for four days. Following titration, participants completed an fMRI alcohol taste-cues task. Analyses tested effects of naltrexone, OPRM1, and their interaction in whole-brain and region of interest (ROI) analyses of functional activation and functional connectivity in response to alcohol versus water taste cues. RESULTS We found no effects of naltrexone orOPRM1 on neural activation in whole-brain and ROI analyses, which included left and right ventral striatum (VS), anterior cingulate cortex (ACC), and orbitofrontal cortex (OFC). Naltrexone increased functional connectivity between left VS and clusters in medial prefrontal cortex, posterior cingulate gyrus, as well as right VS and occipital cortex, compared to placebo. CONCLUSIONS Naltrexone treatment enhanced functional connectivity in a key reinforcement-related pathway during alcohol versus water taste cues, corroborating neuroimaging work with other substances. Null medication and pharmacogenetics effects on functional activation add to a mixed naltrexone literature and may underscore the modest size of these effects in East Asians.
Collapse
Affiliation(s)
- Aaron C Lim
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Dara G Ghahremani
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Erica N Grodin
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - ReJoyce Green
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Spencer Bujarski
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Emily E Hartwell
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kelly E Courtney
- Department of Psychology, University of California, San Diego, San Diego, CA, USA
| | - Kent Hutchison
- Department of Psychology, University of Colorado, Boulder, CO, USA
| | - Karen Miotto
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lara A Ray
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
10
|
The OPRM1 A118G polymorphism: converging evidence against associations with alcohol sensitivity and consumption. Neuropsychopharmacology 2018; 43:1530-1538. [PMID: 29497164 PMCID: PMC5983535 DOI: 10.1038/s41386-017-0002-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/28/2017] [Accepted: 12/07/2017] [Indexed: 12/31/2022]
Abstract
The endogenous opioid system may be involved in the development and maintenance of alcohol use disorder (AUD) and is a target for existing AUD pharmacotherapies. A functional polymorphism of the mu-opioid receptor gene (OPRM1 A118G, rs1799971) may alter the risk of developing AUD. Human laboratory studies have demonstrated that minor allele carriers self-administer more alcohol, show greater sensitivity to alcohol's effects, and exhibit increased alcohol-induced dopamine release. On the other hand, large genome-wide association studies and meta-analyses of candidate gene studies have not found an association between this genotype and alcohol dependence diagnosis. Given this discrepancy, the present study sought to verify whether OPRM1 A118G was associated with alcohol self-administration, subjective response to alcohol, and craving in a sample of 106 social drinkers of European ancestry who completed an intravenous alcohol self-administration session. We found no relationship between OPRM1 rs1799971 genotype and subjective response to alcohol or craving. OPRM1 genotype was not associated with total alcohol exposure or likelihood of attaining a binge-level exposure (80 mg%) during the intravenous alcohol self-administration session. Analysis of 90-day Timeline Followback interview data in a larger sample of 965 participants of European ancestry found no relationship between OPRM1 genotype and alcohol consumption in either alcohol dependent or non-dependent participants. These findings suggest that there may not be an association between OPRM1 rs1799971 genotype and alcohol consumption or sensitivity in individuals of European ancestry.
Collapse
|
11
|
Dopaminergic Genetic Variation Influences Aripiprazole Effects on Alcohol Self-Administration and the Neural Response to Alcohol Cues in a Randomized Trial. Neuropsychopharmacology 2018; 43:1247-1256. [PMID: 29362512 PMCID: PMC5916367 DOI: 10.1038/npp.2017.298] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/27/2017] [Accepted: 12/02/2017] [Indexed: 12/22/2022]
Abstract
Dopamine (DA) signaling regulates many aspects of Alcohol Use Disorder (AUD). However, clinical studies of dopaminergic medications, including the DA partial agonist aripiprazole (APZ), have been inconsistent, suggesting the possibility of a pharmacogenetic interaction. This study examined whether variation in DA-related genes moderated APZ effects on reward-related AUD phenotypes. The interacting effects of APZ and a variable number tandem repeat (VNTR) polymorphism in DAT1/SLC6A3 (the gene encoding the DA transporter (DAT)) were tested. In addition, interactions between APZ and a genetic composite comprising the DAT1 VNTR and functional polymorphisms in catechol-O-methyltransferase (COMT), DRD2, and DRD4 were evaluated. Ninety-four non-treatment-seeking individuals with AUD were genotyped for these polymorphisms, randomized to APZ (titrated to 15 mg) or placebo for 8 days, and underwent an fMRI alcohol cue-reactivity task (day 7; n=81) and a bar lab paradigm (day 8). Primary outcomes were alcohol cue-elicited ventral striatal (VS) activation and the number of drinks consumed in the bar lab. DAT1 genotype significantly moderated medication effects, such that APZ, relative to placebo, reduced VS activation and bar-lab drinking only among carriers of the DAT1 9-repeat allele, previously associated with lower DAT expression and greater reward-related brain activation. The genetic composite further moderated medication effects, such that APZ reduced the primary outcomes more among individuals who carried a larger number of DAT1, COMT, DRD2, and DRD4 alleles associated with higher DA tone. Taken together, these data suggest that APZ may be a promising AUD treatment for individuals with a genetic predisposition to higher synaptic DA tone.
Collapse
|
12
|
Schuckit MA. A Critical Review of Methods and Results in the Search for Genetic Contributors to Alcohol Sensitivity. Alcohol Clin Exp Res 2018; 42:822-835. [PMID: 29623680 PMCID: PMC5916326 DOI: 10.1111/acer.13628] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/06/2018] [Indexed: 02/06/2023]
Abstract
Attributes of alcohol sensitivity are present before alcohol use disorders (AUDs) develop, they predict those adverse alcohol outcomes, are familial in nature, and many are heritable. Whether measured by alcohol challenges or retrospective reports of numbers of drinks required for effects, alcohol sensitivity reflects multiple phenotypes, including low levels of alcohol response and alcohol-related stimulation. Identification of genes that contribute to alcohol sensitivity could help identify individuals carrying risks for AUDs through their alcohol responses for whom early intervention might mitigate their vulnerability. Such genes could also improve understanding of biological underpinnings of AUDs, which could lead to new treatment approaches. However, the existing literature points to a wide range of genetic mechanisms that might contribute to alcohol responses, and few such genetic findings have been widely replicated. This critical review describes the potential impact of the diverse methods used to study sensitivity on the diversity of genetic findings that have been reported, places the genetic variants mentioned in the literature into broader categories rather than isolated results, and offers suggestions regarding how to advance the field by interpreting findings in light of the methods used to select research subjects and to measure alcohol sensitivity. To date, the most promising results have been for GABA, glutamate, opioid, dopamine, serotonin, and cholinergic system genes. The more gene variants that can be identified as contributors to sensitivity the better future gene screening platforms or polygenic scores are likely to be.
Collapse
Affiliation(s)
- Marc A Schuckit
- Department of Psychiatry, University of California, San Diego School of Medicine, La Jolla, California
| |
Collapse
|
13
|
Ray LA, Green R, Roche DJ, Bujarski S, Hartwell EE, Lim AC, Rohrbaugh T, Ghahremani D, Hutchison K, Miotto K. Pharmacogenetic Effects of Naltrexone in Individuals of East Asian Descent: Human Laboratory Findings from a Randomized Trial. Alcohol Clin Exp Res 2018; 42:613-623. [PMID: 29265379 PMCID: PMC6086578 DOI: 10.1111/acer.13586] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Genetic variation in the endogenous opioid system has been identified as 1 potential source of individual variability in naltrexone treatment outcomes. The majority of naltrexone pharmacogenetic studies have focused on a particular single nucleotide polymorphism (SNP) of the mu-opioid receptor gene (OPRM1; rs1799971; commonly known as the Asn40Asp SNP) in Caucasian samples with decidedly mixed results. The goal of this study was to test the pharmacogenetic effects of naltrexone on subjective response to alcohol and self-administration of alcohol in individuals of East Asian descent. We hypothesized that naltrexone, compared with placebo, would potentiate the aversive and sedative effects of alcohol and reduce alcohol self-administration to a greater extent in Asp40 carriers. METHODS Participants (N = 77; Asn40Asn, n = 29; Asn40Asp, n = 34, and Asp40Asp, n = 14) completed 2 double-blinded and counterbalanced experimental sessions: one after taking naltrexone (50 mg/d) for 5 days and one after taking matched placebo for 5 days. In each experimental session, participants received a priming dose of intravenous alcohol up to the breath alcohol concentration target of 0.06 g/dl which was immediately followed by an alcohol self-administration period (1 hour). RESULTS There were no pharmacogenetic effects observed for alcohol-induced stimulation, sedation, craving for alcohol, or alcohol self-administration in the laboratory. During the self-administration period, Asp40 carriers consumed fewer drinks and had a longer latency to first drink as compared to Asn40 homozygotes. CONCLUSIONS These findings in East Asians add to the mixed literature on naltrexone pharmacogenetics from predominantly Caucasian samples and highlight the complexity of these effects and their overall limited replicability. It is plausible that a consistent pharmacogenetic effect in tightly controlled preclinical and experimental medicine models "fades" in more complex and heterogeneous settings and samples.
Collapse
Affiliation(s)
- Lara A. Ray
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - ReJoyce Green
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniel J.O. Roche
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Spencer Bujarski
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Emily E. Hartwell
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Aaron C. Lim
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Taylor Rohrbaugh
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Dara Ghahremani
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kent Hutchison
- Department of Psychology, University of Colorado, Boulder, CO, USA
| | - Karen Miotto
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
14
|
Hendershot CS, Wardell JD, Samokhvalov AV, Rehm J. Effects of naltrexone on alcohol self-administration and craving: meta-analysis of human laboratory studies. Addict Biol 2017; 22:1515-1527. [PMID: 27411969 PMCID: PMC6139429 DOI: 10.1111/adb.12425] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 05/19/2016] [Accepted: 06/21/2016] [Indexed: 12/21/2022]
Abstract
Randomized clinical trials have established the efficacy of naltrexone for reducing quantity of alcohol consumption and incidence of relapse to heavy drinking. To evaluate putative treatment mechanisms, human laboratory studies have examined naltrexone's effects on alcohol responses and self-administration during short-term medication protocols. Results from these studies are inconsistent and have yet to be examined in aggregate. This meta-analysis aimed to quantify naltrexone's effects on alcohol self-administration and craving in the context of placebo-controlled human laboratory trials. Potential moderators of medication effects were also examined. Meta-analyses of alcohol self-administration (k = 9, N = 490) and craving (k = 16, N = 748) confirmed that, under controlled experimental conditions, naltrexone reduces the quantity of consumption (Hedges' g = -.277, SE = .074, 95 percent CI = -.421, -.133, p < .001) and magnitude of self-reported craving (g = -.286, SE = .066, 95 percent CI = -.416, -.156, p < .001) relative to placebo. Subgroup and moderation analyses found no evidence that effect sizes differed by study population (dependent versus non-dependent drinkers), laboratory paradigm or duration of medication exposure. These results substantiate prior evidence for reductions in event-level craving and consumption as potential treatment mediators, also establishing effect sizes to inform future human laboratory trials. From a clinical perspective, these results may provide additional evidence regarding naltrexone's efficacy in the context of acute or subacute dosing regimens.
Collapse
Affiliation(s)
- Christian S. Hendershot
- Campbell Family Mental Health Research InstituteCentre for Addiction and Mental HealthCanada
- Department of Psychiatry, University of TorontoCanada
- Department of PsychologyUniversity of TorontoCanada
- Institute for Mental Health Policy ResearchCentre for Addiction and Mental HealthCanada
| | - Jeffrey D. Wardell
- Campbell Family Mental Health Research InstituteCentre for Addiction and Mental HealthCanada
| | - Andriy V. Samokhvalov
- Department of Psychiatry, University of TorontoCanada
- Institute for Mental Health Policy ResearchCentre for Addiction and Mental HealthCanada
- Institute for Medical SciencesUniversity of TorontoCanada
| | - Jürgen Rehm
- Campbell Family Mental Health Research InstituteCentre for Addiction and Mental HealthCanada
- Department of Psychiatry, University of TorontoCanada
- Institute for Mental Health Policy ResearchCentre for Addiction and Mental HealthCanada
- Dalla Lana School of Public HealthUniversity of TorontoCanada
- Technische UniversitätGermany
| |
Collapse
|
15
|
Hendershot CS, Wardell JD, McPhee MD, Ramchandani VA. A prospective study of genetic factors, human laboratory phenotypes, and heavy drinking in late adolescence. Addict Biol 2017; 22:1343-1354. [PMID: 27046326 PMCID: PMC5293674 DOI: 10.1111/adb.12397] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 02/18/2016] [Accepted: 02/26/2016] [Indexed: 12/16/2022]
Abstract
Subjective responses to alcohol are considered candidate endophenotypes for alcohol use disorder and appear to anticipate future consumption. However, prospective studies have been rare, and laboratory research has typically examined subjective responses absent measures of self-administration. This study examined the association of subjective responses with subsequent laboratory self-administration, also evaluating laboratory phenotypes in relation to putative genetic risk factors [family history (FH) of alcohol dependence and OPRM1 genotype] and subsequent heavy drinking. Participants (N = 61, M = 19.89 years, SD = 0.86) completed laboratory sessions involving intravenous alcohol challenge (Session 1) and free-access intravenous self-administration (Session 2), followed by prospective assessments. Multilevel modeling showed that higher reported stimulation and lower sedation during Session 1 independently predicted greater alcohol self-administration during Session 2. Although self-administration did not differ by FH group, participants with the OPRM1 118G allele evidenced steeper breath alcohol concentration (BrAC) trajectories and greater peak BrAC relative to 118A homozygous participants. Prospective analyses supported significant indirect associations between Session 1 subjective responses and 6-month heavy drinking via peak BrAC in Session 2. Additionally, significant indirect associations of FH (via Session 1 stimulation and Session 2 peak BrAC) and OPRM1 (via peak BrAC) with follow-up heavy drinking were observed. These results further support the utility of human laboratory phenotypes in prospective studies of alcohol use disorder risk and highlight the potential role of self-administration phenotypes in longitudinal research.
Collapse
Affiliation(s)
- Christian S. Hendershot
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey D. Wardell
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Matthew D. McPhee
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Vijay A. Ramchandani
- Section on Human Psychopharmacology, Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
16
|
Anton RF, Schacht JP, Voronin KE, Randall PK. Aripiprazole Suppression of Drinking in a Clinical Laboratory Paradigm: Influence of Impulsivity and Self-Control. Alcohol Clin Exp Res 2017; 41:1370-1380. [PMID: 28493623 DOI: 10.1111/acer.13417] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/05/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Aspects of impulsivity have been implicated in the development, or maintenance, of alcohol use disorder (AUD). The brain dopamine system is implicated in both reward processing/memory (typically subcortical) and in brain inhibitory control mechanisms (typically cortical). Using a validated clinical laboratory paradigm, the dopamine/serotonin "stabilizing" drug, aripiprazole was evaluated in non-treatment-seeking AUD individuals based on their level of impulsivity/self-control. METHODS Ninety-nine individuals (77% male; mean age 27; 7.5 drinks per day; 83% heavy drinking days) meeting DSM-IV criteria for alcohol dependence were randomized to aripiprazole (N = 47 evaluable) or placebo (N = 48 evaluable) based on their Barratt Impulsiveness Scale (BIS-11) score (above or below 68). Aripiprazole, or similar placebo, was titrated to 15 mg over 8 days. Drinking was recorded over 6 days under natural conditions. On Day 8, after 1 day of required abstinence, individuals participated in a bar laboratory paradigm that included a priming drink (breath alcohol concentration [BAC] target 0.02 to 0.03 g/dl) and free-choice consumption of up to 8 drinks (max BAC 0.1 g/dl) in exchange for a "bar credit" of $2 per drink (max $16). End points were drinks per day under natural conditions and drinks consumed in the bar laboratory after the priming drink. RESULTS There was no significant main effect of aripiprazole or interaction with BIS-11 score during the natural drinking period. However, there was a main effect of aripiprazole on bar laboratory drinking (p = 0.04) and aripiprazole reduced the total number of drinks consumed more among individuals with low self-control (p = 0.034) and increased latency to consume those drinks (p = 0.045) more among those with high impulsivity. Relative to placebo, aripiprazole caused more side effects and increased alcohol-induced sedation, but neither significantly influenced its interaction with impulsivity/self-control scores on drinking. CONCLUSIONS This paradigm forced a choice between immediate drinking reward and delayed monetary reward. In those with high impulsivity and/or low self-control, aripiprazole shifts the balance away from immediate drinking toward a later reward. Medications targeting cortical dopamine/serotonin balance might show clinical benefit of reduced drinking, among individuals with impulsivity/low self-control.
Collapse
Affiliation(s)
- Raymond F Anton
- Alcohol Research Center, Addictions Science Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Joseph P Schacht
- Alcohol Research Center, Addictions Science Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Konstantin E Voronin
- Alcohol Research Center, Addictions Science Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Patrick K Randall
- Alcohol Research Center, Addictions Science Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
17
|
Wang RS, Hall KT, Giulianini F, Passow D, Kaptchuk TJ, Loscalzo J. Network analysis of the genomic basis of the placebo effect. JCI Insight 2017; 2:93911. [PMID: 28570268 DOI: 10.1172/jci.insight.93911] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/02/2017] [Indexed: 12/13/2022] Open
Abstract
The placebo effect is a phenomenon in which patients who are given an inactive treatment (e.g., inert pill) show a perceived or actual improvement in a medical condition. Placebo effects in clinical trials have been investigated for many years especially because placebo treatments often serve as the control arm of randomized clinical trial designs. Recent observations suggest that placebo effects may be modified by genetics. This observation has given rise to the term "placebome," which refers to a group of genome-related mediators that affect an individual's response to placebo treatments. In this study, we conduct a network analysis of the placebome and identify a placebome module in the comprehensive human interactome using a seed-connector algorithm. The placebome module is significantly enriched with neurotransmitter signaling pathways and brain-specific proteins. We validate the placebome module using a large cohort of the Women's Genome Health Study (WGHS) trial and demonstrate that the placebome module is significantly enriched with genes whose SNPs modify the outcome in the placebo arm of the trial. To gain insights into placebo effects in different diseases and drug treatments, we use a network proximity measure to examine the closeness of the placebome module to different disease modules and drug target modules. The results demonstrate that the network proximity of the placebome module to disease modules in the interactome significantly correlates with the strength of the placebo effect in the corresponding diseases. The proximity of the placebome module to molecular pathways affected by certain drug classes indicates the existence of placebo-drug interactions. This study is helpful for understanding the molecular mechanisms mediating the placebo response, and sets the stage for minimizing its effects in clinical trials and for developing therapeutic strategies that intentionally engage it.
Collapse
Affiliation(s)
| | - Kathryn T Hall
- Department of Medicine and.,Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Franco Giulianini
- Department of Medicine and.,Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dani Passow
- Program in Placebo Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ted J Kaptchuk
- Program in Placebo Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
18
|
Norman H, D'Souza MS. Endogenous opioid system: a promising target for future smoking cessation medications. Psychopharmacology (Berl) 2017; 234:1371-1394. [PMID: 28285326 DOI: 10.1007/s00213-017-4582-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 02/24/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Nicotine addiction continues to be a health challenge across the world. Despite several approved medications, smokers continue to relapse. Several human and animal studies have evaluated the role of the endogenous opioid system as a potential target for smoking cessation medications. METHODS In this review, studies that have elucidated the role of the mu (MORs), delta (DORs), and kappa (KORs) opioid receptors in nicotine reward, nicotine withdrawal, and reinstatement of nicotine seeking will be discussed. Additionally, the review will discuss discrepancies in the literature and therapeutic potential of the endogenous opioid system, and suggest studies to address gaps in knowledge with respect to the role of the opioid receptors in nicotine dependence. RESULTS Data available till date suggest that blockade of the MORs and DORs decreased the rewarding effects of nicotine, while activation of the MORs and DORs decreased nicotine withdrawal-induced aversive effects. In contrast, activation of the KORs decreased the rewarding effects of nicotine, while blockade of the KORs decreased nicotine withdrawal-induced aversive effects. Interestingly, blockade of the MORs and KORs attenuated reinstatement of nicotine seeking. In humans, MOR antagonists have shown benefits in select subpopulations of smokers and further investigation is required to realize their full therapeutic potential. CONCLUSION Future work must assess the influence of polymorphisms in opioid receptor-linked genes in nicotine dependence, which will help in both identifying individuals vulnerable to nicotine addiction and the development of opioid-based smoking cessation medications. Overall, the endogenous opioid system continues to be a promising target for future smoking cessation medications.
Collapse
Affiliation(s)
- Haval Norman
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH, 45810, USA
| | - Manoranjan S D'Souza
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH, 45810, USA.
| |
Collapse
|
19
|
López CM, Barr SC, Reid-Quiñones K, de Arellano MA. Efficacy of Naltrexone for the Treatment of Alcohol Dependence in Latino Populations. J Stud Alcohol Drugs 2017; 78:629-634. [PMID: 28728646 PMCID: PMC5551667 DOI: 10.15288/jsad.2017.78.629] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 01/06/2017] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Naltrexone has been identified as a promising psychopharmacological treatment for alcohol dependence. Previous studies have suggested that its efficacy may vary based on ethnic background. The current study examined the efficacy of naltrexone in the treatment of alcohol dependence in Latino adults, a previously unexplored population. METHOD This was a secondary analysis of the Combined Pharmacotherapies and Behavioral Interventions for Alcohol Dependence (COMBINE) Study. The overall COMBINE sample consisted of 1,383 adult participants who met Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, criteria for alcohol dependence, including 155 Latinos, who are the focus of this report. Consistent with the main trial, primary drinking outcomes, including percentage of days abstinent (PDA) and time to first heavy drinking day (TTHD), were examined. In addition, we examined the effects of naltrexone on a clinically relevant secondary outcome measure, global clinical outcome of alcohol consumption and alcohol-related problems. RESULTS As seen with the subsample of African Americans from the COMBINE Study, results of the present analysis indicated that there were no significant effects of naltrexone on PDA and TTHD despite these significant effects in the original study. However, contrary to findings in the African American subsample, for Latino participants naltrexone was a significant predictor of a good global clinical outcome (i.e., abstinence or moderate drinking without problems). CONCLUSIONS Naltrexone was not significantly associated with improvements in the primary drinking outcomes of PDA or TTHD at the end of treatment or at follow-up. However, Latinos appeared to benefit from naltrexone as demonstrated by improved ratings of global clinical outcome. These results indicate mixed findings for the efficacy of naltrexone among Latinos in the COMBINE Study.
Collapse
Affiliation(s)
- Cristina M. López
- College of Nursing, Medical University of South Carolina, Charleston, South Carolina
| | - Simone C. Barr
- Department of Behavioral Health, Womack Army Medical Center, Fort Bragg, North Carolina
| | | | - Michael A. de Arellano
- Medical University of South Carolina, National Crime Victims Research and Treatment Center, Department of Psychiatry and Behavioral Sciences, Charleston, South Carolina
| |
Collapse
|
20
|
Weerts EM, Wand GS, Maher B, Xu X, Stephens MA, Yang X, McCaul ME. Independent and Interactive Effects of OPRM1 and DAT1 Polymorphisms on Alcohol Consumption and Subjective Responses in Social Drinkers. Alcohol Clin Exp Res 2017; 41:1093-1104. [PMID: 28376280 DOI: 10.1111/acer.13384] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 03/27/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND The current study examined independent and interactive effects of polymorphisms of the mu opioid receptor gene (OPRM1, A118G) and variable number tandem repeats of the dopamine transporter gene (DAT1, SLC6A3) on alcohol consumption and subjective responses to alcohol in 127 young, healthy, social drinkers. METHODS Participants completed an in-person assessment, which included self-reported alcohol drinking patterns and blood sampling for DNA, and in a second visit, a cumulative alcohol dosing procedure with subjective ratings across multiple time points and breath alcohol contents (0.03 to 0.1%). DNA was analyzed for OPRM1 AA versus AG/GG (*G) genotypes, DAT1 10-repeat allele (A10) versus 9 or lesser alleles (A9), and ancestral informative markers. RESULTS There were significant epistatic interactions between OPRM1 and DAT1 genotypes. Subjective High Assessment Scale scores after alcohol consumption were highest in *G and A9 carriers, and lowest in *G and A10 carriers. Negative subjective effects were also highest in *G and A9 carriers. Effects were similar in a sensitivity analysis limited to Caucasian subjects. There were independent and epistatic interactions on drinking. The OPRM1 *G allele was independently associated with fewer heavy drinking days. The A9 allele was associated with a greater number of drinking days, which was attenuated in carriers of the *G allele. CONCLUSIONS These findings highlight the biological importance of interactions between these 2 genes and interactions between brain opioid and dopamine systems.
Collapse
Affiliation(s)
- Elise M Weerts
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gary S Wand
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Brion Maher
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Mental Health, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Xiaoqiang Xu
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mary Ann Stephens
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xiaoju Yang
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mary E McCaul
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
21
|
Rinker JA, Mulholland PJ. Promising pharmacogenetic targets for treating alcohol use disorder: evidence from preclinical models. Pharmacogenomics 2017; 18:555-570. [PMID: 28346058 DOI: 10.2217/pgs-2016-0193] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Inherited genetic variants contribute to risk factors for developing an alcohol use disorder, and polymorphisms may inform precision medicine strategies for treating alcohol addiction. Targeting genetic mutations linked to alcohol phenotypes has provided promising initial evidence for reducing relapse rates in alcoholics. Although successful in some studies, there are conflicting findings and the reports of adverse effects may ultimately limit their clinical utility, suggesting that novel pharmacogenetic targets are necessary to advance precision medicine approaches. Here, we describe promising novel genetic variants derived from preclinical models of alcohol consumption and dependence that may uncover disease mechanisms that drive uncontrolled drinking and identify novel pharmacogenetic targets that facilitate therapeutic intervention for the treatment of alcohol use disorder.
Collapse
Affiliation(s)
- Jennifer A Rinker
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Patrick J Mulholland
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
22
|
McGuier NS, Griffin WC, Gass JT, Padula AE, Chesler EJ, Mulholland PJ. Kv7 channels in the nucleus accumbens are altered by chronic drinking and are targets for reducing alcohol consumption. Addict Biol 2016; 21:1097-1112. [PMID: 26104325 DOI: 10.1111/adb.12279] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Alcohol use disorders (AUDs) are a major public health issue and produce enormous societal and economic burdens. Current Food and Drug Administration (FDA)-approved pharmacotherapies for treating AUDs suffer from deleterious side effects and are only effective in a subset of individuals. It is therefore essential to find improved medications for the management of AUDs. Emerging evidence suggests that anticonvulsants are a promising class of drugs for treating individuals with AUDs. In these studies, we used integrative functional genomics to demonstrate that genes that encode Kv7 channels (i.e. Kcnq2/3) are related to alcohol (ethanol) consumption, preference and acceptance in rodents. We then tested the ability of the FDA-approved anticonvulsant retigabine, a Kv7 channel opener, to reduce voluntary ethanol consumption of Wistar rats in a two-bottle choice intermittent alcohol access paradigm. Systemic administration and microinjections of retigabine into the nucleus accumbens significantly reduced alcohol drinking, and retigabine was more effective at reducing intake in high- versus low-drinking populations of Wistar rats. Prolonged voluntary drinking increased the sensitivity to the proconvulsant effects of pharmacological blockade of Kv7 channels and altered surface trafficking and SUMOylation patterns of Kv7.2 channels in the nucleus accumbens. These data implicate Kcnq2/3 in the regulation of ethanol drinking and demonstrate that long-term drinking produces neuroadaptations in Kv7 channels. In addition, these results have identified retigabine as a potential pharmacotherapy for treating AUDs and Kv7 channels as a novel therapeutic target for reducing heavy drinking.
Collapse
Affiliation(s)
- Natalie S. McGuier
- Department of Neuroscience; Medical University of South Carolina; Charleston SC USA
| | - William C. Griffin
- Department of Psychiatry and Behavioral Sciences; Medical University of South Carolina; Charleston SC USA
| | - Justin T. Gass
- Department of Neuroscience; Medical University of South Carolina; Charleston SC USA
| | - Audrey E. Padula
- Department of Neuroscience; Medical University of South Carolina; Charleston SC USA
| | | | - Patrick J. Mulholland
- Department of Neuroscience; Medical University of South Carolina; Charleston SC USA
- Department of Psychiatry and Behavioral Sciences; Medical University of South Carolina; Charleston SC USA
| |
Collapse
|
23
|
Bujarski S, Ray LA. Experimental psychopathology paradigms for alcohol use disorders: Applications for translational research. Behav Res Ther 2016; 86:11-22. [PMID: 27266992 PMCID: PMC5067182 DOI: 10.1016/j.brat.2016.05.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 05/12/2016] [Accepted: 05/26/2016] [Indexed: 02/05/2023]
Abstract
In spite of high prevalence and disease burden, scientific consensus on the etiology and treatment of Alcohol Use Disorder (AUD) has yet to be reached. The development and utilization of experimental psychopathology paradigms in the human laboratory represents a cornerstone of AUD research. In this review, we describe and critically evaluate the major experimental psychopathology paradigms developed for AUD, with an emphasis on their implications, strengths, weaknesses, and methodological considerations. Specifically we review alcohol administration, self-administration, cue-reactivity, and stress-reactivity paradigms. We also provide an introduction to the application of experimental psychopathology methods to translational research including genetics, neuroimaging, pharmacological and behavioral treatment development, and translational science. Through refining and manipulating key phenotypes of interest, these experimental paradigms have the potential to elucidate AUD etiological factors, improve the efficiency of treatment developments, and refine treatment targets thus advancing precision medicine.
Collapse
Affiliation(s)
- Spencer Bujarski
- Department of Psychology, University of California, Los Angeles, United States.
| | - Lara A Ray
- Department of Psychology, University of California, Los Angeles, United States; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, United States
| |
Collapse
|
24
|
Ray LA, Bujarski S, Roche DJO. Subjective Response to Alcohol as a Research Domain Criterion. Alcohol Clin Exp Res 2016; 40:6-17. [PMID: 26727518 DOI: 10.1111/acer.12927] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 10/12/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND Individual differences in the subjective experience of the pharmacological effects of alcohol have long been implicated in the likelihood that one will drink heavily and develop alcoholism. The theme of this conceptual review and perspective article is to synthesize the literature on subjective responses to alcohol and to set an agenda for the next generation of research in the area. Specifically, we contend that in order for subjective response to alcohol to play a prominent role in alcoholism research, it is critical that it be studied as a multimodal phenotype. METHODS First, we review the human research on subjective response to alcohol measured under controlled laboratory conditions and draw recommendations for the application of these findings to understanding alcoholism neurobiology in humans. Second, we highlight multimodal approaches, including studies of the genetic and neural substrates of individual differences in subjective response to alcohol. Third, we review treatment implications with a focus on subjective response to alcohol as an intervention target. Upon review of the research on subjective response to alcohol across levels of analyses, we provide recommendations for leveraging these phenotypes in a systematic and methodologically rigorous fashion that can address central questions about alcoholism etiology, disease progression, and personalized treatment. DISCUSSION The approach recommended herein is largely consistent with the Research Domain Criteria (RDoC) initiative across the National Institute of Mental Health. The defining feature of such domains is that they inform behavior yet be amenable to examination through multiple units of analysis, such as molecular, genetic, circuit-level, and behavioral measurements. To that end, we contend that subjective response to alcohol represents a behaviorally and biologically plausible phenotype upon which to build using the RDoC framework for understanding alcohol use disorder.
Collapse
Affiliation(s)
- Lara A Ray
- Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - Spencer Bujarski
- Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - Daniel J O Roche
- Department of Psychology, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
25
|
Soyka M, Mutschler J. Treatment-refractory substance use disorder: Focus on alcohol, opioids, and cocaine. Prog Neuropsychopharmacol Biol Psychiatry 2016; 70:148-61. [PMID: 26577297 DOI: 10.1016/j.pnpbp.2015.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/23/2015] [Accepted: 11/11/2015] [Indexed: 12/20/2022]
Abstract
Substance use disorders are common, but only a small minority of patients receive adequate treatment. Although psychosocial therapies are effective, relapse is common. This review focusses on novel pharmacological and other treatments for patients with alcohol, opioid, or cocaine use disorders who do not respond to conventional treatments. Disulfiram, acamprosate, and the opioid antagonist naltrexone have been approved for the treatment of alcoholism. A novel, "as needed" approach is the use of the mu-opioid antagonist and partial kappa agonist nalmefene to reduce alcohol consumption. Other novel pharmacological approaches include the GABA-B receptor agonist baclofen, anticonvulsants such as topiramate and gabapentin, the partial nicotine receptor agonist varenicline, and other drugs. For opioid dependence, opioid agonist therapy with methadone or buprenorphine is the first-line treatment option. Other options include oral or depot naltrexone, morphine sulfate, depot or implant formulations, and heroin (diacetylmorphine) in treatment-refractory patients. To date, no pharmacological treatment has been approved for cocaine addiction; however, 3 potential pharmacological treatments are being studied, disulfiram, methylphenidate, and modafinil. Pharmacogenetic approaches may help to optimize treatment response in otherwise treatment-refractory patients and to identify which patients are more likely to respond to treatment, and neuromodulation techniques such as repeated transcranial magnetic stimulation and deep brain stimulation also may play a role in the treatment of substance use disorders. Although no magic bullet is in sight for treatment-refractory patients, some novel medications and brain stimulation techniques have the potential to enrich treatment options at least for some patients.
Collapse
Affiliation(s)
- Michael Soyka
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University, Nussbaumstrasse 7, 80336 Munich, Germany; Privatklinik Meiringen, Postfach 612, CH-3860 Meiringen, Switzerland.
| | - Jochen Mutschler
- Center for Addictive Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Selnaustrasse 9, 8001 Zurich, Switzerland
| |
Collapse
|
26
|
Ziauddeen H, Nestor LJ, Subramaniam N, Dodds C, Nathan PJ, Miller SR, Sarai BK, Maltby K, Fernando D, Warren L, Hosking LK, Waterworth D, Korzeniowska A, Win B, Richards DB, Vasist Johnson L, Fletcher PC, Bullmore ET. Opioid Antagonists and the A118G Polymorphism in the μ-Opioid Receptor Gene: Effects of GSK1521498 and Naltrexone in Healthy Drinkers Stratified by OPRM1 Genotype. Neuropsychopharmacology 2016; 41:2647-57. [PMID: 27109624 PMCID: PMC5026731 DOI: 10.1038/npp.2016.60] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 03/31/2016] [Accepted: 04/06/2016] [Indexed: 02/02/2023]
Abstract
The A118G single-nucleotide polymorphism (SNP rs1799971) in the μ-opioid receptor gene, OPRM1, has been much studied in relation to alcohol use disorders. The reported effects of allelic variation at this SNP on alcohol-related behaviors, and on opioid receptor antagonist treatments, have been inconsistent. We investigated the pharmacogenetic interaction between A118G variation and the effects of two μ-opioid receptor antagonists in a clinical lab setting. Fifty-six overweight and moderate-heavy drinkers were prospectively stratified by genotype (29 AA homozygotes, 27 carriers of at least 1 G allele) in a double-blind placebo-controlled, three-period crossover design with naltrexone (NTX; 25 mg OD for 2 days, then 50 mg OD for 3 days) and GSK1521498 (10 mg OD for 5 days). The primary end point was regional brain activation by the contrast between alcohol and neutral tastes measured using functional magnetic resonance imaging (fMRI). Secondary end points included other fMRI contrasts, subjective responses to intravenous alcohol challenge, and food intake. GSK1521498 (but not NTX) significantly attenuated fMRI activation by appetitive tastes in the midbrain and amygdala. GSK1521498 (and NTX to a lesser extent) significantly affected self-reported responses to alcohol infusion. Both drugs reduced food intake. Across all end points, there was less robust evidence for significant effects of OPRM1 allelic variation, or for pharmacogenetic interactions between genotype and drug treatment. These results do not support strong modulatory effects of OPRM1 genetic variation on opioid receptor antagonist attenuation of alcohol- and food-related behaviors. However, they do support further investigation of GSK1521498 as a potential therapeutic for alcohol use and eating disorders.
Collapse
Affiliation(s)
- Hisham Ziauddeen
- Department of Psychiatry, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Wellcome Trust MRC Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Fulbourn Hospital, Cambridge, UK
| | - Liam J Nestor
- Department of Psychiatry, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Centre for Neuropsychopharmacology, Imperial College, London, UK
| | - Naresh Subramaniam
- Department of Psychiatry, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Wellcome Trust MRC Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
| | - Chris Dodds
- Department of Psychology, University of Exeter, Exeter, UK
| | - Pradeep J Nathan
- Department of Psychiatry, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Neuroscience Center of Excellence, inVentiv Health Clinical, Maidenhead, UK
- School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| | | | | | - Kay Maltby
- GSK Clinical Unit, Cambridge Biomedical Campus, Cambridge, UK
| | - Disala Fernando
- GSK Clinical Unit, Cambridge Biomedical Campus, Cambridge, UK
| | - Liling Warren
- Acclarogen, St John's Innovation Centre, Cambridge, UK
| | | | - Dawn Waterworth
- Genetics, Target Science, GlaxoSmithKline, King of Prussia, PA, USA
| | | | - Beta Win
- GSK, Global Clinical Safety & Pharmacovigilance, Stockley Park, UK
| | - Duncan B Richards
- Academic Discovery Performance Unit, GlaxoSmithKline R&D, Stevenage, UK
| | | | - Paul C Fletcher
- Department of Psychiatry, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Wellcome Trust MRC Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Fulbourn Hospital, Cambridge, UK
| | - Edward T Bullmore
- Department of Psychiatry, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Fulbourn Hospital, Cambridge, UK
- Academic Discovery Performance Unit, GlaxoSmithKline R&D, Stevenage, UK
| |
Collapse
|
27
|
Ma Y, Fan R, Li MD. Meta-Analysis Reveals Significant Association of the 3'-UTR VNTR in SLC6A3 with Alcohol Dependence. Alcohol Clin Exp Res 2016; 40:1443-53. [PMID: 27219321 DOI: 10.1111/acer.13104] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/12/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Although many studies have analyzed the association of 3'-untranslated region variable-number tandem repeat (VNTR) polymorphism in SLC6A3 with alcohol dependence (AD), the results remain controversial. This study aimed to determine whether this variant indeed has any genetic effect on AD by integrating 17 reported studies with 5,929 participants included. METHODS The A9-dominant genetic model that considers A9-repeat and non-A9 repeat as 2 genotypes and compared their frequencies in alcoholics with that in controls was adopted. Considering the potential influence of ethnicity, differences in diagnostic criteria of AD, and alcoholic subgroups, stratified meta-analyses were conducted. There existed no evidence for the presence of heterogeneity among the studied samples, indicating the results under the fixed-effects model are acceptable. RESULTS We found a significant association of VNTR A9 genotypes with AD in all ethnic populations (pooled odds ratio [OR] 1.12; 95% confidence interval [CI] 1.00, 1.25; p = 0.045) and the Caucasian population (pooled OR 1.15; 95% CI 1.01, 1.31; p = 0.036). We also found VNTR A9 genotypes to be significantly associated with alcoholism as defined by the DSM-IV criteria (pooled OR 1.18; 95% CI 1.03, 1.36; p = 0.02). Further, we found a significant association between VNTR A9 genotypes and alcoholism associated with alcohol withdrawal seizure or delirium tremens (pooled OR 1.55; 95% CI 1.24, 1.92; p = 1.0 × 10(-4) ). In all these meta-analyses, no evidence of publication bias was detected. CONCLUSIONS We concluded that the VNTR polymorphism has an important role in the etiology of AD, and individuals with at least 1 A9 allele are more likely to be dependent on alcohol than persons carrying the non-A9 allele.
Collapse
Affiliation(s)
- Yunlong Ma
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Center for Air Pollution and Health, Zhejiang University, Hangzhou, China
| | - Rongli Fan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Center for Air Pollution and Health, Zhejiang University, Hangzhou, China
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Center for Air Pollution and Health, Zhejiang University, Hangzhou, China.,Institute for NeuroImmune Pharmacology, Seton Hall University, South Orange, New Jersey
| |
Collapse
|
28
|
Boyd SJ, Schacht JP, Prisciandaro JJ, Voronin K, Anton RF. Alcohol-Induced Stimulation Mediates the Effect of a GABRA2 SNP on Alcohol Self-Administrated among Alcohol-Dependent Individuals. Alcohol Alcohol 2016; 51:549-54. [PMID: 27117237 DOI: 10.1093/alcalc/agw024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 04/05/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND A single-nucleotide polymorphism (SNP) in GABRA2 (rs279858) may moderate subjective response (SR) to alcohol. Results of studies in non-dependent drinkers examining this GABRA2 SNP on SR have been equivocal. This study examined this SNP's direct and indirect effects on alcohol self-administration in dependent drinkers. METHOD The sample consisted of 63 Caucasian, non-treatment-seeking individuals with alcohol dependence. Subjective stimulation was assessed using the Biphasic Alcohol Effects Scale following consumption of an alcoholic priming drink (target breath alcohol content = 0.02 g%). Participants were subsequently offered the opportunity to self-administer up to eight additional drinks. RESULTS Controlling for baseline stimulation, T-allele homozygotes, relative to individuals with at least one copy of the C-allele, reported greater initial stimulation, t(58) = 2.011, p = 0.049. Greater stimulation predicted greater subsequent alcohol self-administration, t(57) = 2.522, p = 0.015. Although rs279858 genotype did not directly impact self-administration (t(57) = -0.674, p = 0.503), it did have an indirect effect (95% confidence interval [0.068, 1.576]), such that T-allele homozygotes reported greater stimulation, which in turn predicted greater self-administration. CONCLUSION These results suggest that the influence of this SNP on SR differs depending on dose or stage of dependence. This study is the first to demonstrate an indirect effect of rs279858 genotype on drinking through SR. Although C-allele carriers have been shown to have an increased risk for alcohol dependence, in our dependent sample, greater stimulation was found among T-allele homozygotes, suggesting that the influence of SR on developing and maintaining dependence differs based on rs279858 genotype.This study demonstrates an indirect effect of rs279858 genotype on drinking through SR. Although C-allele carriers have an increased risk for alcohol dependence, in our dependent sample, greater stimulation was found among T-allele homozygotes, suggesting that the influence of SR on developing dependence differs based on rs279858 genotype.
Collapse
Affiliation(s)
- Stephen J Boyd
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Joseph P Schacht
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - James J Prisciandaro
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Konstantin Voronin
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Raymond F Anton
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
29
|
Hendershot CS, Claus ED, Ramchandani VA. Associations of OPRM1 A118G and alcohol sensitivity with intravenous alcohol self-administration in young adults. Addict Biol 2016; 21:125-35. [PMID: 25039301 PMCID: PMC4300289 DOI: 10.1111/adb.12165] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human laboratory and animal models implicate variation in the μ-opioid receptor gene (OPRM1) as relevant for alcohol-related reward. OPRM1 is associated with alcohol self-administration in non-human primate studies, but the relevance of this finding to human models is unclear. This study used computer-assisted self-infusion of ethanol (CASE) to examine associations among OPRM1 A118G genotype, subjective responses to alcohol and intravenous alcohol self-administration in young heavy drinkers (n = 40, mean age = 19.95 years, SD = 0.82). Participants completed a 2-hour CASE session comprising a priming phase followed by ad libitum self-administration in a free-access paradigm. Participants achieved a mean peak breath alcohol concentration (BrAC) of 81.18 mg% (SD = 24.96). Those with the OPRM1 118G variant (GA or GG genotypes) achieved significantly higher peak BrAC (M = 94.90 mg%, SD = 16.56) than those with the AA genotype (M = 74.46 mg%, SD = 25.36), reflecting a significantly greater number of alcohol requests among GA/GG participants. Eighty percent of GA/GG participants surpassed a threshold defining a laboratory analog of heavy alcohol exposure (80 mg%) compared with 46 percent of AA participants. Results indicated significant associations between subjective measures of alcohol sensitivity and CASE outcomes, although the pattern of findings differed across self-report measures. Subjective responses did not differ by OPRM1 status. These results offer further support for the feasibility of the CASE paradigm and provide initial evidence for an association of OPRM1 with alcohol self-administration in a human laboratory context.
Collapse
Affiliation(s)
- Christian S Hendershot
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Canada
- Department of Psychiatry, University of Toronto, Canada
| | - Eric D Claus
- The Mind Research Network and Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, USA
| | - Vijay A Ramchandani
- Section on Human Psychopharmacology, Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
30
|
Pharmacogenetics of alcohol use disorders and comorbid psychiatric disorders. Psychiatry Res 2015; 230:121-9. [PMID: 26455758 DOI: 10.1016/j.psychres.2015.09.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/04/2015] [Accepted: 09/10/2015] [Indexed: 11/21/2022]
Abstract
Alcohol use disorders (AUDs) represent a significant health burden worldwide. Currently, there are three medications approved by the U.S. Food and Drug Administration for the treatment of AUDs, and other drugs are being prescribed off-label for this purpose. However, response rates for pharmacologic treatment are low, and extant research suggests that treatment effects may partially depend on genetic factors. Personalized medicine, or using a patient's genetics and/or personal history to determine efficacy of treatment prior to prescription, is an emerging tool that will help clinicians treat their patients more effectively and safely. This review systematically discusses current findings from AUD pharmacotherapy trials examining disulfiram, acamprosate, naltrexone, the injectable naltrexone, and topiramate. Furthermore, it presents pharmacogenetics findings associated with these medications in an attempt to further the field of personalized medicine. Research from trials examining AUDs and comorbid major depressive disorder and anxiety disorders is also presented, and pharmacogenetic findings for these treatments are discussed. Lastly, the authors comment on the present and future states of the field of personalized medicine for AUD.
Collapse
|
31
|
Patriquin MA, Bauer IE, Soares JC, Graham DP, Nielsen DA. Addiction pharmacogenetics: a systematic review of the genetic variation of the dopaminergic system. Psychiatr Genet 2015; 25:181-93. [PMID: 26146874 PMCID: PMC4549168 DOI: 10.1097/ypg.0000000000000095] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Substance use disorders have significant personal, familial, and societal consequences. Despite the serious consequences of substance use, only a few therapies are effective in treating substance use disorders, thus highlighting a need for improved treatment practices. Substance use treatment response depends on multiple factors such as genetic, biological, and social factors. It is essential that each component is represented in treatment plans. The dopaminergic system plays a critical role in the pharmacotherapy for addictions, and an understanding of the role of variation of genes involved in this system is essential for its success. This review adheres to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Statement guidelines. A computerized literature search was conducted using PubMed and Scopus (all databases). Articles published up to April 2015 that examined the role of dopaminergic gene variation in the pharmacotherapy of alcohol, opioid, and cocaine use disorders were reviewed. Search terms were dopamine, gene, polymorphism, substance abuse, treatment, and response. Polymorphisms of the DRD2, ANKK1, DAT1, DBH, and DRD4 genes have been found to moderate the effects of pharmacotherapy of alcohol, opioid, and cocaine use disorders. The integration of genetic information with clinical data will inform health professionals of the most efficacious pharmacotherapeutic intervention for substance use disorders. More studies are needed to confirm and extend these findings.
Collapse
Affiliation(s)
- Michelle A. Patriquin
- The Menninger Clinic, Baylor College of Medicine, Houston, TX USA
- The Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
| | - Isabelle E. Bauer
- Department of Psychiatry and Behavioral Science, University of Texas Health Sciences Center, Houston, TX USA
| | - Jair C. Soares
- Department of Psychiatry and Behavioral Science, University of Texas Health Sciences Center, Houston, TX USA
| | - David P. Graham
- The Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
- Michael E. DeBakey Veterans Affairs Medical Center; Houston, TX USA
| | - David A. Nielsen
- The Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
- Michael E. DeBakey Veterans Affairs Medical Center; Houston, TX USA
| |
Collapse
|
32
|
KCNN Genes that Encode Small-Conductance Ca2+-Activated K+ Channels Influence Alcohol and Drug Addiction. Neuropsychopharmacology 2015; 40:1928-39. [PMID: 25662840 PMCID: PMC4839516 DOI: 10.1038/npp.2015.42] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 01/17/2015] [Accepted: 01/26/2015] [Indexed: 02/02/2023]
Abstract
Small-conductance Ca(2+)-activated K(+) (KCa2) channels control neuronal excitability and synaptic plasticity, and have been implicated in substance abuse. However, it is unknown if genes that encode KCa2 channels (KCNN1-3) influence alcohol and drug addiction. In the present study, an integrative functional genomics approach shows that genetic datasets for alcohol, nicotine, and illicit drugs contain the family of KCNN genes. Alcohol preference and dependence QTLs contain KCNN2 and KCNN3, and Kcnn3 transcript levels in the nucleus accumbens (NAc) of genetically diverse BXD strains of mice predicted voluntary alcohol consumption. Transcript levels of Kcnn3 in the NAc negatively correlated with alcohol intake levels in BXD strains, and alcohol dependence enhanced the strength of this association. Microinjections of the KCa2 channel inhibitor apamin into the NAc increased alcohol intake in control C57BL/6J mice, while spontaneous seizures developed in alcohol-dependent mice following apamin injection. Consistent with this finding, alcohol dependence enhanced the intrinsic excitability of medium spiny neurons in the NAc core and reduced the function and protein expression of KCa2 channels in the NAc. Altogether, these data implicate the family of KCNN genes in alcohol, nicotine, and drug addiction, and identify KCNN3 as a mediator of voluntary and excessive alcohol consumption. KCa2.3 channels represent a promising novel target in the pharmacogenetic treatment of alcohol and drug addiction.
Collapse
|
33
|
Pharmacotherapy for alcohol dependence: A stratified approach. Pharmacol Ther 2015; 153:10-24. [PMID: 25985735 DOI: 10.1016/j.pharmthera.2015.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 12/20/2022]
Abstract
Alcohol dependence is a common disorder in many societies worldwide, and remains difficult to identify and treat. It is also a risk factor for many secondary non-communicable diseases. Pharmacotherapy is one available treatment option, but appears to be underutilised in practice. Major barriers to use of medications in this area include lack of clinical guidance and questionable efficacy. However, for each medication there appears to be a subpopulation that responds positively, and understanding the moderating factors to treatment efficacy is an important research goal. Thus, this review provides a narrative regarding potential stratification techniques in pharmacological treatment of alcohol dependence, with a specific focus on typologies and pharmacogenetics. In addition, we discuss the basic background of stratified medicine and recent studies on genetic predisposition to alcohol dependence. A growing repository of data exists for both approved and non-approved pharmacotherapies, but failure to replicate findings, inadequate sample sizes, and insufficient funding has resulted in a translational gap. Implementing evidence-based stratified/personalised therapy and identifying new therapeutic agents may lead to improved clinical outcomes and reduced financial burden. Despite some promising findings to date, much work is still required.
Collapse
|
34
|
Roche DJ, Ray LA. Subjective response as a consideration in the pharmacogenetics of alcoholism treatment. Pharmacogenomics 2015; 16:721-36. [PMID: 25950242 DOI: 10.2217/pgs.14.143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Currently available pharmacological treatments for alcoholism have modest efficacy and high individual variability in treatment outcomes, both of which have been partially attributed to genetic factors. One path to reducing the variability and improving the efficacy associated with these pharmacotherapies may be to identify overlapping genetic contributions to individual differences in both subjective responses to alcohol and alcoholism pharmacotherapy outcomes. As acute subjective response to alcohol is highly predictive of future alcohol related problems, identifying such shared genetic mechanisms may inform the development of personalized treatments that can effectively target converging pathophysiological mechanisms that convey risk for alcoholism. The focus of this review is to revisit the association between subjective response to alcohol and the etiology of alcoholism while also describing genetic contributions to this relationship, discuss potential pharmacogenetic approaches to target subjective response to alcohol in order to improve the treatment of alcoholism and examine conceptual and methodological issues associated with these topics, and outline future approaches to overcome these challenges.
Collapse
Affiliation(s)
- Daniel Jo Roche
- 1Department of Psychology, University of California, 1285 Franz Hall, Box 951563, Los Angeles, CA 90095-1563, USA
| | - Lara A Ray
- 1Department of Psychology, University of California, 1285 Franz Hall, Box 951563, Los Angeles, CA 90095-1563, USA
| |
Collapse
|
35
|
Garbutt JC, Greenblatt AM, West SL, Morgan LC, Kampov-Polevoy A, Jordan HS, Bobashev GV. Clinical and biological moderators of response to naltrexone in alcohol dependence: a systematic review of the evidence. Addiction 2014; 109:1274-84. [PMID: 24661324 DOI: 10.1111/add.12557] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 12/19/2013] [Accepted: 03/18/2014] [Indexed: 01/29/2023]
Abstract
AIM The goal of this systematic review was to identify moderators of naltrexone efficacy in the treatment of alcohol dependence. METHODS We searched Pubmed, CINHAL, Embase, PsycINFO and the Cochrane Library from 1990 to April 2012 and reference lists of pertinent review articles, which yielded 622 trial, pooled analysis and review articles. Using pre-established eligibility criteria, two reviewers independently determined whether abstracts contained evidence of demographic or biological characteristics, i.e. moderators, influencing naltrexone response in alcohol dependence. We assessed each publication for risk of bias and evaluated the strength of the body of evidence for each moderator. RESULTS Twenty-eight publications (on 20 studies) met criteria for data synthesis. These included 26 publications from 12 randomized, placebo-controlled trials, three non-randomized, non-placebo studies and one randomized, non-placebo study. In addition, there were two publications from pooled analyses of four randomized, placebo-controlled trials. Family history of alcohol problems and the Asn40Asp polymorphism of the μ-opioid receptor gene showed a positive association with efficacy in four of five and three of five studies, respectively. Other moderators reported to be associated with efficacy included male sex (two of five studies), pre-treatment drinking (two of two studies) and high craving (two of five studies). However, the overall risk of bias in the published literature is high. CONCLUSIONS The identification of naltrexone-responsive alcohol-dependent patients is still in development. Studies to date point to two potential moderators-family history and presence of the OPRM1 Asn40Asp polymorphism-as having the strongest evidence. However, the data to date is still insufficient to recommend that any moderator be used in determining clinical treatment.
Collapse
Affiliation(s)
- James C Garbutt
- Department of Psychiatry and Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
To date, few pharmacotherapies have been established for the treatment of alcoholism. There is a plethora of research concerning the involvement of the opioid-endorphin system in mediating the reinforcing effects of alcohol. The opioid antagonist naltrexone has been found to be effective in alcohol treatment. In addition, the mu-opioid antagonist and partial kappa agonist nalmefene was recently approved by the European Medicines Agency for the treatment of alcoholism. The relevant studies followed a harm-reduction, 'as needed' approach and showed a reduction in alcohol consumption with nalmefene 20 mg rather than increased abstinence rates, (which was not the primary goal of the relevant studies). The available literature is reviewed and discussed. Nalmefene appears to be a safe and effective treatment for alcohol dependence.
Collapse
|
37
|
Ray LA, Bujarski S, Squeglia LM, Ashenhurst JR, Anton RF. Interactive effects of OPRM1 and DAT1 genetic variation on subjective responses to alcohol. Alcohol Alcohol 2014; 49:261-70. [PMID: 24421289 DOI: 10.1093/alcalc/agt183] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
AIMS Subjective response to alcohol represents a marker of alcoholism risk. The A118G single-nucleotide polymorphism (SNP) of the mu opioid receptor (OPRM1) gene has been associated with subjective response to alcohol. Recently, the dopamine transporter (DAT1) variable number of tandem repeat (VNTR; SLC6A3) has been found to interact with the OPRM1 A118G SNP in predicting neural and behavioral responses to naltrexone and to alcohol. This exploratory study examines the OPRM1 × DAT1 interaction on subjective responses to alcohol. METHODS Non-treatment-seeking problem drinkers (n = 295) were assessed in the laboratory for alcohol dependence. Following prospective genotyping for the OPRM1 gene, 43 alcohol-dependent individuals were randomized to two intravenous infusion sessions, one of alcohol (target BrAC = 0.06 g/dl) and one of saline. Measures of subjective responses to alcohol were administered in both infusion sessions. RESULTS Analyses revealed significant Alcohol × OPRM1 × DAT1 interactions for alcohol-induced stimulation, vigor and positive mood as well as significant Alcohol × OPRM1 × DAT1 × Time interactions for stimulation and positive mood. These effects were such that, compared with other genotype groups, OPRM1 G-allele carriers + DAT1 A10 homozygotes reported steeper increases in stimulation and positive mood across rising BrAC, when compared with placebo. All Alcohol × OPRM1 × DAT1 interactions remained significant when analyses were restricted to a subsample of Caucasian participants (n = 34); however, 4-way interactions did not reach statistical significance in this subsample. CONCLUSIONS This study suggests that the contribution of OPRM1 genotype to alcohol-induced stimulation, vigor and positive mood is moderated by DAT1 genotype. These findings are consistent with the purported interaction between opioidergic and dopaminergic systems in determining the reinforcing properties of alcohol.
Collapse
Affiliation(s)
- Lara A Ray
- Corresponding author: Department of Psychology, University of California, 1285 Franz Hall, Box 951563, Los Angeles, CA 90095-1563, USA.
| | | | | | | | | |
Collapse
|
38
|
Anton RF, Schacht JP, Book SW. Pharmacologic treatment of alcoholism. HANDBOOK OF CLINICAL NEUROLOGY 2014; 125:527-42. [PMID: 25307594 DOI: 10.1016/b978-0-444-62619-6.00030-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Progress in understanding the neuroscience of addiction has significantly advanced the development of more efficacious medications for the treatment of alcohol use disorders (AUD). While several medications have been approved by regulatory bodies around the world for the treatment of AUD, they are not universally efficacious. Recent research has yielded improved understanding of the genetics and brain circuits that underlie alcohol reward and its habitual use. This research has contributed to pharmacogenetic studies of medication response, and will ultimately lead to a more "personalized medicine" approach to AUD pharmacotherapy. This chapter summarizes work on clinically available medications (both approved by regulatory bodies and investigational) for the treatment of alcohol dependence, as well as the psychiatric disorders that are commonly comorbid with AUD. Studies that have evaluated genetic influences on medication response and those that have employed neuroimaging to probe mechanisms of medication action or response are highlighted. Finally, new targets discovered in animal models for possible pharmacologic intervention in humans are overviewed and future directions in medications development provided.
Collapse
Affiliation(s)
- Raymond F Anton
- Center for Drug and Alcohol Programs, Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.
| | - Joseph P Schacht
- Center for Drug and Alcohol Programs, Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Sarah W Book
- Center for Drug and Alcohol Programs, Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
39
|
Enoch MA. Genetic influences on response to alcohol and response to pharmacotherapies for alcoholism. Pharmacol Biochem Behav 2013; 123:17-24. [PMID: 24220019 DOI: 10.1016/j.pbb.2013.11.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 09/24/2013] [Accepted: 11/04/2013] [Indexed: 12/16/2022]
Abstract
Although very many individuals drink alcohol at safe levels, a significant proportion escalates their consumption with addiction as the end result. Alcoholism is a common, moderately heritable, psychiatric disorder that is accompanied by considerable morbidity and mortality. Variation in clinical presentation suggests inter-individual variation in mechanisms of vulnerability including genetic risk factors. The development of addiction is likely to involve numerous functional genetic variants of small effects. The first part of this review will focus on genetic factors underlying inter-individual variability in response to alcohol consumption, including variants in alcohol metabolizing genes that produce an aversive response (the flushing syndrome) and variants that predict the level of subjective and physiological response to alcohol. The second part of this review will report on genetic variants that identify subgroups of alcoholics who are more likely to respond to pharmacotherapy to reduce levels of drinking or maintain abstinence. Genetic analyses of the level of response to alcohol, particularly of the functional OPRM1 A118G polymorphism and 5' and 3' functional polymorphisms in SLC6A4, are beginning to provide insights into the etiology of alcoholism and also genotype-stratified subgroup responses to naltrexone and SSRIs/ondansetron respectively. Because of large inter-ethnic variation in allele frequencies, the relevance of these functional polymorphisms will vary between ethnic groups. However there are relatively few published studies in this field, particularly with large sample sizes in pharmacogenetic studies, therefore it is premature to draw any conclusions at this stage.
Collapse
Affiliation(s)
- Mary-Anne Enoch
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
40
|
Crist RC, Berrettini WH. Pharmacogenetics of OPRM1. Pharmacol Biochem Behav 2013; 123:25-33. [PMID: 24201053 DOI: 10.1016/j.pbb.2013.10.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 09/19/2013] [Accepted: 10/23/2013] [Indexed: 12/23/2022]
Abstract
Pharmacogenetic research has the potential to explain the variation in treatment efficacy within patient populations. Understanding the interaction between genetic variation and medications may provide a method for matching patients to the most effective therapeutic options and improving overall patient outcomes. The OPRM1 gene has been a target of interest in a large number of pharmacogenetic studies due to its genetic and structural variation, as well as the role of opioid receptors in a variety of disorders. The mu-opioid receptor (MOR), encoded by OPRM1, naturally regulates the analgesic response to pain and also controls the rewarding effects of many drugs of abuse, including opioids, nicotine, and alcohol. Genetic variants in OPRM1, particularly the non-synonymous polymorphism A118G, have been repeatedly associated with the efficacy of treatments for pain and various types of dependence. This review focuses on the current understanding of the pharmacogenetic impact of OPRM1, primarily with regard to the treatment of pain and addiction.
Collapse
Affiliation(s)
- Richard C Crist
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, 125 South 31st St., Philadelphia, PA 19104, United States.
| | - Wade H Berrettini
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, 125 South 31st St., Philadelphia, PA 19104, United States
| |
Collapse
|
41
|
Hulse GK. Improving clinical outcomes for naltrexone as a management of problem alcohol use. Br J Clin Pharmacol 2013; 76:632-41. [PMID: 22946873 PMCID: PMC3853523 DOI: 10.1111/j.1365-2125.2012.04452.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 08/30/2012] [Indexed: 12/13/2022] Open
Abstract
Despite being a relatively effective and safe treatment, the clinical management of alcohol abuse/dependence by oral naltrexone can be compromised due to the patient's non-compliance with daily use of this medication. Over the past decade an increasing body of research has suggested that the use of sustained release depot naltrexone preparations can overcome this issue and deliver improved clinical outcomes. However, at the same time, research findings from diverse areas of pharmacogenetics, neurobiology and behavioural psychology have also been converging to identify variables including genetic markers, patient psychosocial characteristics and drug use history differences, or clusters of these variables that play a major role in mediating the response of alcohol abuse/dependent persons to treatment by naltrexone. While this article does not attempt to review all available data pertaining to an individual alcohol dependent patient's response to treatment by naltrexone, it does identify relevant research areas and highlights the importance of data arising from them. The characterization of clinical markers, to identify those patients who are most likely to benefit from naltrexone and to tailor a more individual naltrexone treatment, will ultimately provide significant benefit to both patients and clinicians by optimizing treatment outcome.
Collapse
Affiliation(s)
- Gary K Hulse
- Addiction Medicine, School of Psychiatry & Clinical Neurosciences (M521), The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
42
|
Abstract
Functional polymorphisms in the dopamine transporter gene (DAT1 or SLC6A3) modulate responsiveness to salient stimuli, such that carriers of one 9R-allele of DAT1 (compared with homozygote carriers of the 10R-allele) show heightened reactivity to drug-related reinforcement in addiction. Here, using multimodal neuroimaging and behavioral dependent variables in 73 human cocaine-addicted individuals and 47 healthy controls, we hypothesized and found that cocaine-addicted carriers of a 9R-allele exhibited higher responses to drug cues, but only among individuals who had used cocaine within 72 h of the study as verified by positive cocaine urine screens (a state characterized by intense craving). Importantly, this responsiveness to drug cues was reliably preserved across multimodal imaging and behavioral probes: psychophysiological event-related potentials, self-report, simulated cocaine choice, and fMRI. Because drug cues contribute to relapse, our results identify the DAT1R 9R-allele as a vulnerability allele for relapse especially during early abstinence (e.g., detoxification).
Collapse
|
43
|
Ziauddeen H, Nathan PJ, Dodds C, Maltby K, Miller SR, Waterworth D, Song K, Warren L, Hosking L, Zucchetto M, Bush M, Johnson LV, Sarai B, Mogg K, Bradley BP, Richards DB, Fletcher PC, Bullmore ET. The effects of alcohol on the pharmacokinetics and pharmacodynamics of the selective mu-opioid receptor antagonist GSK1521498 in healthy subjects. J Clin Pharmacol 2013; 53:1078-90. [PMID: 23934621 PMCID: PMC4282435 DOI: 10.1002/jcph.110] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 05/11/2013] [Indexed: 11/27/2022]
Abstract
The mu-opioid system has a key role in hedonic and motivational processes critical to substance addiction. However, existing mu-opioid antagonists have had limited success as anti-addiction treatments. GSK1521498 is a selective and potent mu-opioid antagonist being developed for the treatment of overeating and substance addictions. In this study, 28 healthy participants were administered single doses of GSK1521498 20 mg, ethanol 0.5 g/kg body weight, or both in combination, in a double blind placebo controlled four-way crossover design. The primary objective was to determine the risk of significant adverse pharmacodynamic and pharmacokinetic (PK) interactions. The effects of GSK1521498 on hedonic and consummatory responses to alcohol and the attentional processing of alcohol-related stimuli, and their modulation by the OPRM1 A118G polymorphism were also explored. GSK1521498 20 mg was well tolerated alone and in combination with ethanol. There were mild transient effects of GSK1521498 on alertness and mood that were greater when it was combined with ethanol. These effects were not of clinical significance. There were no effects of GSK1521498 on reaction time, hedonic or consummatory responses. These findings provide encouraging safety and PK data to support continued development of GSK1521498 for the treatment of alcohol addiction.
Collapse
Affiliation(s)
- Hisham Ziauddeen
- Medicines Discovery and Development, GlaxoSmithKline, Clinical Unit Cambridge, Addenbrooke's Centre for Clinical Investigations, Cambridge, UK; Department of Psychiatry, Behavioural and Clinical Neuroscience Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK; Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK; Cambridgeshire and Peterborough NHS Foundation Trust (CPFT), Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
AIMS To evaluate the pharmacogenetic evidence relating to the use of opioid antagonists (in particular naltrexone) in treating patients with alcohol abuse problems. METHODS Narrative review of pre-clinical and clinical published research regarding genetic modulation of psychotropic effects produced by alcohol and the therapeutic effects of opioid antagonists. RESULTS Alcohol activates brain reward pathways, leading to positive reinforcement of alcohol seeking and consumption. Thus, the underlying biological mechanisms may be targets for treatment, particularly in the early stages of addiction development. Alcohol reward is in part mediated by endogenous opioids. A single-nucleotide polymorphism (SNP) within the OPRM1 gene, A118G, leading to an amino acid change (Asn40Asp) in the extracellular portion of the receptor, has been implicated in alcoholism as well as in drug addiction, pain sensitivity and stress response, and in animal and human studies relates to the alcohol-dependent phenotype as well as to the treatment response to the µ-opioid antagonist naltrexone. CONCLUSION The effect size reported in naltrexone clinical studies is often small, which may be due to heterogeneity among patients. Pharmacogenetic approaches may help guide us in the search for the appropriate treatment optimal for one patient's need.
Collapse
Affiliation(s)
- Annika Thorsell
- Department of Clinical and Experimental Medicine, Linköping University, SE-581 83 Linköping, Sweden.
| |
Collapse
|
45
|
Schacht JP, Anton RF, Voronin KE, Randall PK, Li X, Henderson S, Myrick H. Interacting effects of naltrexone and OPRM1 and DAT1 variation on the neural response to alcohol cues. Neuropsychopharmacology 2013; 38:414-22. [PMID: 23032071 PMCID: PMC3547192 DOI: 10.1038/npp.2012.195] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Variation at a single nucleotide polymorphism in the μ-opioid receptor gene (OPRM1), A118G (Asn40Asp), may moderate naltrexone (NTX) effects in alcohol dependence. Both NTX and A118G variation have also been reported to affect alcohol cue-elicited brain activation. This study investigated whether sub-acute NTX treatment and A118G genotype interacted in their effects on cue-elicited activation of the ventral striatum (VS), medial prefrontal cortex (mPFC), and orbitofrontal cortex (OFC). Secondarily, variation at a variable number tandem repeat polymorphism in the dopamine transporter gene (DAT1/SLC6A3), which has been associated with increased reward-related activation in VS, was analyzed as a moderator of medication and A118G effects. Seventy-four non-treatment-seeking alcohol-dependent individuals, half preselected to carry at least one copy of the A118G G (Asp) allele, were randomized to NTX (50 mg) or placebo for 7 days, and performed an fMRI alcohol cue reactivity task on day 6. Region-of-interest analyses indicated no main effects of medication or A118G genotype. However, these factors interacted in their effects on OFC activation, such that, among NTX-treated individuals, G-allele carriers had less activation than A-allele homozygotes. DAT1 variation also moderated medication/A118G effects. There was a three-way interaction between medication and A118G and DAT1 genotypes on VS activation, such that, among G-allele carriers who received NTX, DAT1 10-repeat-allele (10R) homozygotes had less activation than 9-repeat-allele (9R) carriers. Further, 10R homozygotes who received NTX had less mPFC activation than 9R carriers. Polymorphic variation in OPRM1 and DAT1 should be considered in future studies of NTX, particularly regarding its effects on reward processing.
Collapse
Affiliation(s)
- Joseph P Schacht
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Raymond F Anton
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Konstantin E Voronin
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Patrick K Randall
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Xingbao Li
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Scott Henderson
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Hugh Myrick
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA,Ralph H Johnson VA Medical Center, Charleston, SC, USA
| |
Collapse
|
46
|
Xu QW, Zhao W, Wang Y, Sartor MA, Han DM, Deng J, Ponnala R, Yang JY, Zhang QY, Liao GQ, Qu YM, Li L, Liu FF, Zhao HM, Yin YH, Chen WF, Zhang Y, Wang XS. An integrated genome-wide approach to discover tumor-specific antigens as potential immunologic and clinical targets in cancer. Cancer Res 2012; 72:6351-61. [PMID: 23135912 PMCID: PMC3525759 DOI: 10.1158/0008-5472.can-12-1656] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tumor-specific antigens (TSA) are central elements in the immune control of cancers. To systematically explore the TSA genome, we developed a computational technology called heterogeneous expression profile analysis (HEPA), which can identify genes relatively uniquely expressed in cancer cells in contrast to normal somatic tissues. Rating human genes by their HEPA score enriched for clinically useful TSA genes, nominating candidate targets whose tumor-specific expression was verified by reverse transcription PCR (RT-PCR). Coupled with HEPA, we designed a novel assay termed protein A/G-based reverse serological evaluation (PARSE) for quick detection of serum autoantibodies against an array of putative TSA genes. Remarkably, highly tumor-specific autoantibody responses against seven candidate targets were detected in 4% to 11% of patients, resulting in distinctive autoantibody signatures in lung and stomach cancers. Interrogation of a larger cohort of 149 patients and 123 healthy individuals validated the predictive value of the autoantibody signature for lung cancer. Together, our results establish an integrated technology to uncover a cancer-specific antigen genome offering a reservoir of novel immunologic and clinical targets.
Collapse
Affiliation(s)
- Qing-Wen Xu
- Department of Immunology, Peking University Health Science Center, Beijing 100191, China
| | - Wei Zhao
- Department of Immunology, Peking University Health Science Center, Beijing 100191, China
| | - Yue Wang
- Lester & Sue Smith Breast Center and Dan L, Duncan Cancer Center, Baylor College of Medicine, CCMB, University of Michigan, MI, 48109, USA
| | - Maureen A. Sartor
- National Center for Integrative Biomedical Informatics, CCMB, University of Michigan, MI, 48109, USA
| | - Dong-Mei Han
- Department of Hematology, PLA Air Force General Hospital, Beijing 100036, China
| | - Jixin Deng
- Human Genome Sequencing Center, Baylor College of Medicine
| | - Rakesh Ponnala
- Lester & Sue Smith Breast Center and Dan L, Duncan Cancer Center, Baylor College of Medicine, CCMB, University of Michigan, MI, 48109, USA
| | - Jiang-Ying Yang
- Department of Clinical Laboratory, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Qing-Yun Zhang
- Department of Clinical Laboratory, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Guo-Qing Liao
- Department of Oncology, PLA 309 Hospital, Beijing, China
| | - Yi-Mei Qu
- Department of Oncology, PLA 309 Hospital, Beijing, China
| | - Lu Li
- Department of Cardiothoracic Surgery, the 306th Hospital of PLA, Beijing, China
| | - Fang-Fang Liu
- Department of Pathology, Peking University People’s Hospital, Beijing 100044, China
| | - Hong-Mei Zhao
- Department of Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Yan-Hui Yin
- Department of Immunology, Peking University Health Science Center, Beijing 100191, China
| | - Wei-Feng Chen
- Department of Immunology, Peking University Health Science Center, Beijing 100191, China
| | - Yu Zhang
- Department of Immunology, Peking University Health Science Center, Beijing 100191, China
| | - Xiao-Song Wang
- Lester & Sue Smith Breast Center and Dan L, Duncan Cancer Center, Baylor College of Medicine, CCMB, University of Michigan, MI, 48109, USA
| |
Collapse
|