1
|
Johnson B, Alho H, Addolorato G, Lesch OM, Chick J, Liu L, Schuyler V. Low-dose ondansetron: A candidate prospective precision medicine to treat alcohol use disorder endophenotypes. Eur J Intern Med 2024; 127:50-62. [PMID: 38876929 DOI: 10.1016/j.ejim.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Alcohol use disorder (AUD) is among the leading causes of morbidity and mortality worldwide, and over 95 million people live with alcohol dependence globally. The estimated heritability of AUD is 50-60 %, and multiple genes are thought to contribute to various endophenotypes of the disease. Previous clinical trials support a precision medicine approach using ondansetron (AD04, a 5-HT3 antagonist) by segregating AUD populations by the bio-genetic endophenotype of specific serotonergic genotypes and the bio-psychosocial endophenotype of the severity of drinking or both. By targeting the modulation of biogenetic signaling within the biopsychosocial context of AUD, low-dose AD04 holds promise in reducing alcohol consumption among affected individuals while minimizing adverse effects. METHODS This was a phase III, 6-month, 25-site, randomized, placebo-controlled clinical trial using AD04 to treat DSM-V-categorized AUD individuals who were pre-stratified into the endophenotypes of heavy or very heavy drinking individuals and possessed a pre-defined profile of genetic variants related to the serotonin transporter and serotonin-3AB receptor. Participants (N = 303) presented moderate to severe AUD, >80 % were men, mostly in their fifties, and >95 % were of European descent. Low-dose AD04 (approx. 033 mg twice daily) or a matching placebo was administered twice daily for 6 months. Brief Behavioral Compliance Enhancement Treatment (BBCET [53]) was administered every two weeks to enhance medication compliance and clinic attendance. RESULTS There was a significant reduction in the monthly percentage of heavy drinking days, PHDD (-46·7 % (2·7 %), 95 %CI: -52·1 % to -41·2 % vs. -38·1 % (2·9 %), 95 %CI: -43·8 % to -32·5 %, respectively; LS mean difference=-8·5 %; p = 0.03) among AD04-treated vs. placebo-receiving heavy drinking individuals at month 6. Heavy drinking individuals were also less likely to be diagnosed with AUD [Month 1: -32·0 % (2·8 %), 95 %CI: -37·5 % to -26·5 % vs. -23·2 % (2·9 %), 95 %CI: -28·9 to -17·5 %; LS mean difference= -8·8 %; p = 0·026)], and improved on the WHO quality of life BREF scale with a significant effect for at least a 1-level downward shift (OR = 3.4; 95 % CI: 1·03-11·45, p = 0·044). Importantly, heavy drinking individuals, as distinct from very heavy drinking individuals, were the bio-psychosocial endophenotype more predictive of therapeutic response to AD04. AD04 had an exceptional safety and tolerability profile, like the placebo's. CONCLUSIONS In this Phase 3 clinical trial, AD04 was shown to be a promising treatment for currently drinking heavy drinking individuals with AUD who also possess a specific genotypic profile in the serotonin transporter and serotonin-3AB receptor complex. Using AD04 to reduce the harm of AUD in heavy drinking individuals who are currently drinking, without the necessity of abstinence or detoxification from alcohol use, is an important advance in the field of precision medicine. AD04's adverse events profile, which was like placebo, should enhance accessibility and acceptance of modern medical treatment for AUD by lowering the incorrect but commonly perceived stigma of personal failure.
Collapse
Affiliation(s)
- Bankole Johnson
- Adial Pharmaceuticals Inc., Division of Biomedical Sciences, Larkin University, Miami, USA.
| | - Hannu Alho
- Addiction Medicine, Faculty of Medicine, University of Helsinki, Finland; Addictum Helsinki, Finland
| | - Giovanni Addolorato
- Department of Medical and Surgical Sciences, Catholic University of Rome, Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | | | - Lei Liu
- Division of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Vinzant Schuyler
- Adial Pharmaceuticals Inc., Division of Biomedical Sciences, Larkin University, Miami, USA
| |
Collapse
|
2
|
Cornell J, Conchas A, Wang XQ, Fink JC, Chen H, Kane MA, Pilli N, Ait-Daoud N, Gorelick DA, Li MD, Johnson BA, Seneviratne C. Validation of serotonin transporter mRNA as a quantitative biomarker of heavy drinking and its comparison to ethyl glucuronide/ethyl sulfate: A randomized, double-blind, crossover trial. Alcohol Clin Exp Res 2022; 46:1888-1899. [PMID: 36031718 PMCID: PMC9588643 DOI: 10.1111/acer.14931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 08/02/2022] [Accepted: 08/22/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND The serotonin transporter (SERT) mRNA was previously reported to be a quantitative and pathophysiology-based biomarker of heavy drinking in 5HTTLPR:LL genotype-carriers treated with ondansetron. Here, we validated the potential use of SERT mRNA for quantitative prediction of recent alcohol consumption (in the absence of treatment) and compared it with the known biomarkers ethyl glucuronide (EtG) and ethyl sulfate (EtS). METHODS Binge drinking men and women of European ancestry aged 21 to 65 years were enrolled in a 12-day, in-patient, randomized, double-blind, crossover study, where they were administered three beverage doses (placebo, 0.5 g/kg [0.4 g/kg] ethanol, and 1 g/kg [0.9 g/kg] ethanol for men [women]) individually in three 4-day periods (experiments), separated by minimum 7-day washout period. Diet, sleep, and physical activity were controlled throughout the inpatient experiments. Twenty-nine participants were randomized to receive beverage doses counterbalancing the sequence of treatment and gender within subgroups stratified by SERT genotypes 5HTTLPR:LL+rs25531:AA (LA LA ) versus 5HTTLPR:LS/SS. Peripheral venous blood was collected daily for (1) quantification of SERT mRNA (the primary outcome measure) using qRT-PCR and (2) plasma EtG and EtS levels using tandem mass-spectrometry. RESULTS The association between administered beverage dose and SERT mRNA from completers of at least one 4-day experiment (N = 18) assessed by a linear mixed model was not statistically significant. Significant positive associations were found with beverage dose and plasma EtG, EtS and EtG/EtS ratio (β = 5.8, SE = 1.2, p < 0.0001; β = 1.3, SE = 0.6, p = 0.023; and β = 3.0, SE = 0.7, p < 0.0001, respectively; the C-statistics for discriminating outcomes were 0.97, 0.8, and 0.92, respectively). Additionally, we observed a sequence effect with a greater placebo effect on SERT mRNA when it was administered during the first experiment (p = 0.0009), but not on EtG/EtS measures. CONCLUSION The findings do not validate the use of SERT as a biomarker of heavy drinking. Larger and more innovative studies addressing the effects of placebo, race, gender, and response to treatment with serotonergic agents are needed to fully assess the utility of SERT as a biomarker of heavy and binge drinking.
Collapse
Affiliation(s)
- Jessica Cornell
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
| | - Andrew Conchas
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD (former affiliation)
| | - Xin-Qun Wang
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - Jeffrey C. Fink
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Hegang Chen
- Department of Epidemiology, University of Maryland School of Medicine, Baltimore, MD
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD
| | - Nageswara Pilli
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD
| | - Nassima Ait-Daoud
- Department of Psychiatry, University of Virginia, Charlottesville, VA
| | - David A. Gorelick
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
| | | | - Bankole A. Johnson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD (former affiliation)
| | - Chamindi Seneviratne
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD
- The Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
3
|
Seneviratne C, Gorelick DA, Lynch KG, Brown C, Romer D, Pond T, Kampman K, Kranzler HR. A randomized, double-blind, placebo-controlled, pharmacogenetic study of ondansetron for treating alcohol use disorder. Alcohol Clin Exp Res 2022; 46:1900-1912. [PMID: 36055978 PMCID: PMC9901168 DOI: 10.1111/acer.14932] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND In a previous study, ondansetron, a serotonin 5-HT3 receptor antagonist, reduced drinking intensity (drinks/drinking day [DPDD]) among European-ancestry (EA) participants with moderate-to-severe alcohol use disorder (AUD) and variants in genes encoding the serotonin transporter (SLC6A4) and 5-HT3A (HTR3A), and 5-HT3B (HTR3B) receptors. We tested whether (1) ondansetron reduces DPDD among individuals of either European or African ancestry (AA), and (2) that reductions in DPDD are greatest among ondansetron-treated individuals with population-specific combinations of genotypes at SLC6A4, HTR3A, and HTR3B. METHODS In this 16-week, double-blind, placebo-controlled, parallel-group clinical trial, adults with AUD were randomized to receive low-dose oral ondansetron (0.33 mg twice daily) or placebo stratified by "responsive" versus "nonresponsive" genotype defined using population-specific genotypes at the three genetic loci. Generalized estimating equation regression models and a modified intent-to-treat analysis were used to compare the treatment groups on the primary outcome-DPDD-and two secondary outcomes-heavy drinking days per week [HDD] and drinks per day [DPD] across the 16 weeks of treatment. RESULTS Of 296 prospective participants screened, 95 (58 EA and 37 AA) were randomized and received at least one dose of study medication. In the modified intent-to-treat analysis, the ondansetron group averaged 0.40 more DPDD (p = 0.51), 1.35 times as many HDD (p = 0.16), and 1.06 times as many DPD (p = 0.59) as the placebo group. There were no significant interactions with genotype. There were no study-related serious adverse events (AEs) and similar proportions of participants in the two treatment groups experienced AEs across organ systems. CONCLUSIONS We found no evidence that low-dose oral ondansetron is beneficial in the treatment of AUD, irrespective of genotype, thus failing to confirm prior study findings. However, the study was underpowered to identify medication by genotype interactions.
Collapse
Affiliation(s)
- Chamindi Seneviratne
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201
| | - David A. Gorelick
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201
| | - Kevin G. Lynch
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104
- Mental Illness Research, Education and Clinical Center, Crezcenz VAMC, Philadelphia, PA 19104
| | - Clayton Brown
- Department of Epidemiology, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201
| | - Danielle Romer
- Mental Illness Research, Education and Clinical Center, Crezcenz VAMC, Philadelphia, PA 19104
| | - Timothy Pond
- Mental Illness Research, Education and Clinical Center, Crezcenz VAMC, Philadelphia, PA 19104
- Department of Epidemiology, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201
| | - Kyle Kampman
- Mental Illness Research, Education and Clinical Center, Crezcenz VAMC, Philadelphia, PA 19104
- Department of Epidemiology, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201
| | - Henry R. Kranzler
- Mental Illness Research, Education and Clinical Center, Crezcenz VAMC, Philadelphia, PA 19104
- Department of Epidemiology, University of Maryland School of Medicine, Baltimore, Maryland, MD 21201
| |
Collapse
|
4
|
Drögemöller BI, Wright GEB, Trueman J, Shaw K, Staub M, Chaudhry S, Miao F, Higginson M, Groeneweg GSS, Brown J, Magee LA, Whyte SD, West N, Brodie SM, Jong G', Israels S, Berger H, Ito S, Rassekh SR, Sanatani S, Ross CJD, Carleton BC. A pharmacogenomic investigation of the cardiac safety profile of ondansetron in children and pregnant women. Pharmacotherapy 2022; 148:112684. [PMID: 35149390 DOI: 10.1016/j.biopha.2022.112684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/22/2022] [Accepted: 02/01/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Ondansetron is a highly effective antiemetic for the treatment of nausea and vomiting. However, this medication has also been associated with QT prolongation. Pharmacogenomic information on therapeutic response to ondansetron exists, but no investigation has been performed on genetic factors that influence the cardiac safety of this medication. METHODS Three patient groups receiving ondansetron were recruited and followed prospectively (pediatric post-surgical patients n = 101; pediatric oncology patients n = 98; pregnant women n = 62). Electrocardiograms were conducted at baseline, and 5- and 30-min post-ondansetron administration, to determine the effect of ondansetron treatment on QT interval. Pharmacogenomic associations were assessed via analyses of comprehensive CYP2D6 genotyping and genome-wide association study data. RESULTS In the entire cohort, 62 patients (24.1%) met the criteria for prolonged QT, with 1.2% of the cohort exhibiting unsafe QT prolongation. The most significant shift from baseline occurred at five minutes post-ondansetron administration (P = 9.8 × 10-4). CYP2D6 activity score was not associated with prolonged QT. Genome-wide analyses identified novel associations with a missense variant in TLR3 (rs3775291; P = 2.00 × 10-7) and a variant linked to the expression of SLC36A1 (rs34124313; P = 1.97 × 10-7). CONCLUSIONS This study has provided insight into the genomic basis of ondansetron-induced cardiac changes and has emphasized the importance of genes that have been implicated in serotonin-related traits. These biologically-relevant findings represent the first step towards understanding this adverse event with the overall goal to improve the safety of this commonly used antiemetic medication.
Collapse
Affiliation(s)
- Britt I Drögemöller
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Canada; Faculty of Pharmaceutical Sciences, University of British Columbia, Canada; British Columbia Children's Hospital Research Institute, Canada; Pharmaceutical Outcomes Programme, British Columbia Children's Hospital, Canada
| | - Galen E B Wright
- British Columbia Children's Hospital Research Institute, Canada; Pharmaceutical Outcomes Programme, British Columbia Children's Hospital, Canada; Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Canada; Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Jessica Trueman
- British Columbia Children's Hospital Research Institute, Canada; Pharmaceutical Outcomes Programme, British Columbia Children's Hospital, Canada
| | - Kaitlyn Shaw
- British Columbia Children's Hospital Research Institute, Canada; Pharmaceutical Outcomes Programme, British Columbia Children's Hospital, Canada
| | - Michelle Staub
- Clinical Research Unit, Children's Hospital Research Institute of Manitoba, Canada
| | - Shahnaz Chaudhry
- Division of Clinical Pharmacology and Toxicology, The Hospital for Sick Children, Canada
| | - Fudan Miao
- British Columbia Children's Hospital Research Institute, Canada
| | | | - Gabriella S S Groeneweg
- British Columbia Children's Hospital Research Institute, Canada; Pharmaceutical Outcomes Programme, British Columbia Children's Hospital, Canada
| | - James Brown
- British Columbia Women's Hospital and Health Centre, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Canada
| | - Laura A Magee
- School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College, UK
| | - Simon D Whyte
- British Columbia Children's Hospital Research Institute, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Canada; Department of Pediatric Anesthesia, British Columbia Children's Hospital, Canada
| | - Nicholas West
- British Columbia Children's Hospital Research Institute, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Canada; Department of Pediatric Anesthesia, British Columbia Children's Hospital, Canada
| | - Sonia M Brodie
- British Columbia Children's Hospital Research Institute, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Canada
| | - Geert 't Jong
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Canada; Clinical Research Unit, Children's Hospital Research Institute of Manitoba, Canada; Department of Pediatrics and Child Health, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Sara Israels
- Department of Pediatrics and Child Health, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Howard Berger
- Department of Obstetrics and Gynecology, St. Michael's Hospital, Canada; Epi Methods Consulting, Canada
| | - Shinya Ito
- Division of Clinical Pharmacology and Toxicology, The Hospital for Sick Children, Canada
| | - Shahrad R Rassekh
- British Columbia Children's Hospital Research Institute, Canada; Pharmaceutical Outcomes Programme, British Columbia Children's Hospital, Canada; Division of Pediatric Hematology/Oncology/BMT, Department of Pediatrics, Children's Heart Centre, BC Children's Hospital, University of British Columbia, Canada
| | - Shubhayan Sanatani
- Division of Cardiology, Department of Pediatrics, Children's Heart Centre, BC Children's Hospital, University of British Columbia, Canada
| | - Colin J D Ross
- Faculty of Pharmaceutical Sciences, University of British Columbia, Canada; British Columbia Children's Hospital Research Institute, Canada; Pharmaceutical Outcomes Programme, British Columbia Children's Hospital, Canada
| | - Bruce C Carleton
- Faculty of Pharmaceutical Sciences, University of British Columbia, Canada; British Columbia Children's Hospital Research Institute, Canada; Pharmaceutical Outcomes Programme, British Columbia Children's Hospital, Canada; Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Canada.
| |
Collapse
|
5
|
Thompson MD, Kenna GA. Variation in the Serotonin Transporter Gene and Alcoholism: Risk and Response to Pharmacotherapy. Alcohol Alcohol 2015; 51:164-71. [PMID: 26311211 DOI: 10.1093/alcalc/agv090] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 07/15/2015] [Indexed: 01/06/2023] Open
Abstract
SLC6A4, the gene encoding the serotonin transporter protein (5-HTT), has been extensively examined as a risk factor for alcohol dependence (AD). More recently, variability in the transporter gene was identified to be a potential moderator of treatment response to serotonergic medications such as ondansetron and sertraline. There is an insertion-deletion polymorphism in the promoter region (5-HTTLPR) of the SLC6A4, with the most common alleles being a 14-repeat short (S) allele and a 16-repeat long (L) allele. The S allele has often been associated with AD. By contrast, the L allele has been associated with pharmacological responsiveness in some individuals with AD. Differences in clinical phenotype may determine the utility of the 5-HTTLPR polymorphism as a moderator of pharmacological interventions for AD. We review the AD typology and disease onset in the context of pharmacogenetic and genomic studies that examine the utility of 5-HTTLPR in improving treatment outcomes.
Collapse
Affiliation(s)
- Miles D Thompson
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - George A Kenna
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
| |
Collapse
|
6
|
Bauer IE, Graham DP, Soares JC, Nielsen DA. Serotonergic gene variation in substance use pharmacotherapy: a systematic review. Pharmacogenomics 2015; 16:1307-14. [PMID: 26265436 DOI: 10.2217/pgs.15.72] [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: 12/16/2022] Open
Abstract
Drug addiction is a serious disease with damaging effects on the brain and physical health. Despite the increase in the number of affected individuals, there are few effective pharmacological treatment options for substance use disorders. The study of the influence of an individual's genetic features on the treatment response may help to identify more efficacious treatment options. This systematic review focuses on the serotonergic system because of its relevant role in mood and impulse control disorders, and its contribution to the development and maintenance of drug use disorders. In particular, we examine the role of serotonergic genes in the response to pharmacotherapy for alcohol, cocaine and nicotine addiction. Current evidence suggests that genetic variability of the serotonergic biosynthesis enzyme tryptophan hydroxylase 2 (TPH2) and the serotonin transporter (SLC6A4) genes mediates the efficacy of several addiction treatments, such as ondansetron and disulfiram, and the antidepressants bupropion, nortriptyline and sertraline.
Collapse
Affiliation(s)
- Isabelle E Bauer
- Department of Psychiatry & Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - David P Graham
- Michael E DeBakey VA Medical Center, Baylor College of Medicine, 2002 Holcombe Boulevard, Research 151, Building 110, Suite 227, Houston, TX 77030, USA
| | - Jair C Soares
- Department of Psychiatry & Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - David A Nielsen
- Michael E DeBakey VA Medical Center, Baylor College of Medicine, 2002 Holcombe Boulevard, Research 151, Building 110, Suite 227, Houston, TX 77030, USA
| |
Collapse
|
7
|
Seneviratne C. Advances in Medications and Tailoring Treatment for Alcohol Use Disorder. Alcohol Res 2015; 37:15-28. [PMID: 26259086 PMCID: PMC4476601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Alcohol use disorder (AUD) is a chronic heritable brain disorder with a variable clinical presentation. This variability, or heterogeneity, in clinical presentation suggests complex interactions between environmental and biological factors, resulting in several underlying pathophysiological mechanisms in the development and progression of AUD. Classifying AUD into subgroups of common clinical or pathological characteristics would ease the complexity of teasing apart underlying molecular mechanisms. Genetic association analyses have revealed several polymorphisms-small differences in DNA-that increase a person's vulnerability to develop AUD and other alcohol-related intermediate characteristics, such as severity of drinking, age of AUD onset, or measures of craving. They also have identified polymorphisms associated with reduced drinking. Researchers have begun utilizing these genetic polymorphisms to identify alcoholics who might respond best to various treatments, thereby enhancing the effectiveness of currently tested medications for treating AUD. This review compares the efficacy of medications tested for treatment of AUD with and without incorporating genetics. It then discusses advances in pre-clinical genetic and genomic studies that potentially could be adapted to clinical trials to improve treatment efficacy. Although a pharmacogenetic approach is promising, it is relatively new and will need to overcome many challenges, including inadequate scientific knowledge and social and logistic constraints, to be utilized in clinical practice.
Collapse
|
8
|
Abstract
Addictions are prevalent psychiatric disorders that confer remarkable personal and social burden. Despite substantial evidence for their moderate, yet robust, heritability (approx. 50%), specific genetic mechanisms underlying their development and maintenance remain unclear. The goal of this selective review is to highlight progress in unveiling the genetic underpinnings of addiction. First, we revisit the basis for heritable variation in addiction before reviewing the most replicable candidate gene findings and emerging signals from genomewide association studies for alcohol, nicotine and cannabis addictions. Second, we survey the modest but growing field of neurogenetics examining how genetic variation influences corticostriatal structure, function, and connectivity to identify neural mechanisms that may underlie associations between genetic variation and addiction. Third, we outline how extant genomic findings are being used to develop and refine pharmacotherapies. Finally, as sample sizes for genetically informed studies of addiction approach critical mass, we posit five exciting possibilities that may propel further discovery (improved phenotyping, rare variant discovery, gene-environment interplay, epigenetics, and novel neuroimaging designs).
Collapse
|
9
|
Gass JT, Olive MF. Neurochemical and neurostructural plasticity in alcoholism. ACS Chem Neurosci 2012; 3:494-504. [PMID: 22896799 DOI: 10.1021/cn300013p] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 04/16/2012] [Indexed: 01/06/2023] Open
Abstract
The behavioral manifestations of alcoholism are primarily attributable to the numerous and lasting adaptations that occur in the brain as a result of chronic heavy alcohol consumption. As will be reviewed here, these adaptations include alcohol-induced plasticity in chemical neurotransmission, density and morphology of dendritic spines, as well as neurodegeneration and cerebral atrophy. Within the context of these neuroadaptations that have been observed in both human and animal studies, we will discuss how these changes potentially contribute to the cognitive and behavioral dysfunctions that are hallmark features of alcoholism, as well as how they reveal novel potential pharmacological targets for the treatment of this disorder.
Collapse
Affiliation(s)
- Justin T. Gass
- Center for
Drug and Alcohol
Programs, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South
Carolina 29425, United States
| | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, Arizona 85287, United
States
- Interdisciplinary
Graduate Program
in Neuroscience, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|