51
|
Goldfine CE, Tom JJ, Im DD, Yudkoff B, Anand A, Taylor JJ, Chai PR, Suzuki J. The therapeutic use and efficacy of ketamine in alcohol use disorder and alcohol withdrawal syndrome: a scoping review. Front Psychiatry 2023; 14:1141836. [PMID: 37181899 PMCID: PMC10172666 DOI: 10.3389/fpsyt.2023.1141836] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/22/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction Alcohol use disorder (AUD) is the most prevalent substance use disorder (SUD) globally. In 2019, AUD affected 14.5 million Americans and contributed to 95,000 deaths, with an annual cost exceeding 250 billion dollars. Current treatment options for AUD have moderate therapeutic effects and high relapse rates. Recent investigations have demonstrated the potential efficacy of intravenous ketamine infusions to increase alcohol abstinence and may be a safe adjunct to the existing alcohol withdrawal syndrome (AWS) management strategies. Methods We followed Preferred Reporting Items for Systematic Reviews (PRISMA) guidelines to conduct a scoping review of two databases (PubMed and Google Scholar) for peer-reviewed manuscripts describing the use of ketamine in AUD and AWS. Studies that evaluated the use of ketamine in AUD and AWS in humans were included. We excluded studies that examined laboratory animals, described alternative uses of ketamine, or discussed other treatments of AUD and AWS. Results We identified 204 research studies in our database search. Of these, 10 articles demonstrated the use of ketamine in AUD or AWS in humans. Seven studies investigated the use of ketamine in AUD and three studies described its use in AWS. Ketamine used in AUD was beneficial in reducing cravings, alcohol consumption and longer abstinence rates when compared to treatment as usual. In AWS, ketamine was used as an adjunct to standard benzodiazepine therapy during severe refractory AWS and at signs of delirium tremens. Adjunctive use of ketamine demonstrated earlier resolution of delirium tremens and AWS, reduced ICU stay, and lowered likelihood of intubation. Oversedation, headache, hypertension, and euphoria were the documented adverse effects after ketamine administration for AUD and AWS. Conclusion The use of sub-dissociative doses of ketamine for the treatment of AUD and AWS is promising but more definitive evidence of its efficacy and safety is required before recommending it for broader clinical use.
Collapse
Affiliation(s)
- Charlotte E. Goldfine
- Department of Emergency Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Jeremiah J. Tom
- Department of Emergency Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Dana D. Im
- Department of Emergency Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Benjamin Yudkoff
- Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA, United States
| | - Amit Anand
- Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA, United States
| | - Joseph J. Taylor
- Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA, United States
- Center for Brain Circuit Therapeutics, Brigham and Women’s Hospital, Boston, MA, United States
| | - Peter R. Chai
- Department of Emergency Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- The Fenway Institute, Fenway Health, Boston, MA, United States
- Koch Institute for Integrated Cancer Research, Massachusetts Institute of Technology, Boston, MA, United States
| | - Joji Suzuki
- Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA, United States
| |
Collapse
|
52
|
Rabiee A, Mahmud N, Falker C, Garcia-Tsao G, Taddei T, Kaplan DE. Medications for alcohol use disorder improve survival in patients with hazardous drinking and alcohol-associated cirrhosis. Hepatol Commun 2023; 7:e0093. [PMID: 36972386 PMCID: PMC10043587 DOI: 10.1097/hc9.0000000000000093] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Medications for alcohol use disorder (MAUD) are highly effective in achieving and maintaining abstinence in patients with alcohol use disorder (AUD). Our aim was to evaluate the effect of MAUD on all-cause mortality in patients with alcohol-associated cirrhosis and active alcohol use. METHODS This was a retrospective cohort study of patients with alcohol-associated cirrhosis and high-risk alcohol use disorder in the Veterans Outcomes and Costs Associated with Liver Disease (VOCAL) database. Propensity score matching for exposure to MAUD (acamprosate or naltrexone) within a year after cirrhosis diagnosis was performed to account for potential confounders, and the association between MAUD and all-cause mortality was subsequently evaluated using Cox regression analysis. RESULTS A total of 9131 patients were included, of whom 886 (9.7%) were exposed to MAUD (naltrexone: 520, acamprosate: 307, both medications: 59). The duration of MAUD exposure was >3 months in 345 patients (39%). The strongest positive predictor of MAUD prescription was an inpatient diagnosis code for AUD, followed by a concurrent diagnosis of depression; the strongest negative predictor was a history of cirrhosis decompensation. After propensity score matching (866 patients in each group) with excellent covariate balance (absolute standardized mean differences <0.1), MAUD exposure was associated with improved survival, with an HR of 0.80 relative to no MAUD exposure (95% CI: 0.67-0.97, p = 0.024). CONCLUSION MAUD are underutilized in patients with alcohol-associated cirrhosis with high-risk alcohol use behavior but are associated with improved survival after adjustment for confounders such as the severity of liver disease, age, and engagement in the healthcare system.
Collapse
Affiliation(s)
- Anahita Rabiee
- Section of Digestive Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nadim Mahmud
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Gastroenterology Section, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| | - Caroline Falker
- VA Connecticut Healthcare System, New Haven, Connecticut, USA
- Department of Internal Medicine & Program in Addiction Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Guadalupe Garcia-Tsao
- Section of Digestive Diseases, Yale School of Medicine, New Haven, Connecticut, USA
- VA Connecticut Healthcare System, New Haven, Connecticut, USA
| | - Tamar Taddei
- Section of Digestive Diseases, Yale School of Medicine, New Haven, Connecticut, USA
- VA Connecticut Healthcare System, New Haven, Connecticut, USA
| | - David E. Kaplan
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Gastroenterology Section, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
53
|
Grigsby KB, Mangieri RA, Roberts AJ, Lopez MF, Firsick EJ, Townsley KG, Beneze A, Bess J, Eisenstein TK, Meissler JJ, Light JM, Miller J, Quello S, Shadan F, Skinner M, Aziz HC, Metten P, Morrisett RA, Crabbe JC, Roberto M, Becker HC, Mason BJ, Ozburn AR. Preclinical and clinical evidence for suppression of alcohol intake by apremilast. J Clin Invest 2023; 133:e159103. [PMID: 36656645 PMCID: PMC10014105 DOI: 10.1172/jci159103] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
Treatment options for alcohol use disorders (AUDs) have minimally advanced since 2004, while the annual deaths and economic toll have increased alarmingly. Phosphodiesterase type 4 (PDE4) is associated with alcohol and nicotine dependence. PDE4 inhibitors were identified as a potential AUD treatment using a bioinformatics approach. We prioritized a newer PDE4 inhibitor, apremilast, as ideal for repurposing (i.e., FDA approved for psoriasis, low incidence of adverse events, excellent safety profile) and tested it using multiple animal strains and models, as well as in a human phase IIa study. We found that apremilast reduced binge-like alcohol intake and behavioral measures of alcohol motivation in mouse models of genetic risk for drinking to intoxication. Apremilast also reduced excessive alcohol drinking in models of stress-facilitated drinking and alcohol dependence. Using site-directed drug infusions and electrophysiology, we uncovered that apremilast may act to lessen drinking in mice by increasing neural activity in the nucleus accumbens, a key brain region in the regulation of alcohol intake. Importantly, apremilast (90 mg/d) reduced excessive drinking in non-treatment-seeking individuals with AUD in a double-blind, placebo-controlled study. These results demonstrate that apremilast suppresses excessive alcohol drinking across the spectrum of AUD severity.
Collapse
Affiliation(s)
- Kolter B. Grigsby
- Portland Alcohol Research Center, Department of Behavioral Neuroscience, Oregon Health & Science University, and VA Portland Health Care System, Portland, Oregon, USA
| | - Regina A. Mangieri
- Waggoner Center for Alcohol and Addiction Research, Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - Amanda J. Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, California, USA
| | - Marcelo F. Lopez
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Evan J. Firsick
- Portland Alcohol Research Center, Department of Behavioral Neuroscience, Oregon Health & Science University, and VA Portland Health Care System, Portland, Oregon, USA
| | - Kayla G. Townsley
- Portland Alcohol Research Center, Department of Behavioral Neuroscience, Oregon Health & Science University, and VA Portland Health Care System, Portland, Oregon, USA
| | - Alan Beneze
- Pearson Center for Alcoholism and Addiction Research, Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Jessica Bess
- Pearson Center for Alcoholism and Addiction Research, Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Toby K. Eisenstein
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Joseph J. Meissler
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | | | - Jenny Miller
- Pearson Center for Alcoholism and Addiction Research, Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Susan Quello
- Pearson Center for Alcoholism and Addiction Research, Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Farhad Shadan
- Pearson Center for Alcoholism and Addiction Research, Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Michael Skinner
- Pearson Center for Alcoholism and Addiction Research, Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Heather C. Aziz
- Waggoner Center for Alcohol and Addiction Research, Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - Pamela Metten
- Portland Alcohol Research Center, Department of Behavioral Neuroscience, Oregon Health & Science University, and VA Portland Health Care System, Portland, Oregon, USA
| | - Richard A. Morrisett
- Waggoner Center for Alcohol and Addiction Research, Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - John C. Crabbe
- Portland Alcohol Research Center, Department of Behavioral Neuroscience, Oregon Health & Science University, and VA Portland Health Care System, Portland, Oregon, USA
| | - Marisa Roberto
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Howard C. Becker
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
- RHJ Department of Veterans Affairs Medical Center, Charleston, South Carolina, USA
| | - Barbara J. Mason
- Pearson Center for Alcoholism and Addiction Research, Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Angela R. Ozburn
- Portland Alcohol Research Center, Department of Behavioral Neuroscience, Oregon Health & Science University, and VA Portland Health Care System, Portland, Oregon, USA
| |
Collapse
|
54
|
Strayer RJ, Friedman BW, Haroz R, Ketcham E, Klein L, LaPietra AM, Motov S, Repanshek Z, Taylor S, Weiner SG, Nelson LS. Emergency Department Management of Patients With Alcohol Intoxication, Alcohol Withdrawal, and Alcohol Use Disorder: A White Paper Prepared for the American Academy of Emergency Medicine. J Emerg Med 2023; 64:517-540. [PMID: 36997435 DOI: 10.1016/j.jemermed.2023.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/06/2023] [Indexed: 03/30/2023]
Affiliation(s)
- Reuben J Strayer
- Department of Emergency Medicine, Maimonides Medical Center, Brooklyn, New York.
| | - Benjamin W Friedman
- Department of Emergency Medicine, Montefiore, Albert Einstein College of Medicine, Bronx, New York
| | - Rachel Haroz
- Cooper Medical School of Rowan University, Cooper University Healthcare, Camden, New Jersey
| | - Eric Ketcham
- Department of Emergency Medicine, Department of Behavioral Health, Addiction Medicine, Presbyterian Healthcare System, Santa Fe & Española, New Mexico
| | - Lauren Klein
- Department of Emergency Medicine, Good Samaritan Hospital, West Islip, New York
| | - Alexis M LaPietra
- Department of Emergency Medicine, Saint Joseph's Regional Medical Center, Paterson, New Jersey
| | - Sergey Motov
- Department of Emergency Medicine, Maimonides Medical Center, Brooklyn, New York
| | - Zachary Repanshek
- Department of Emergency Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Scott Taylor
- Department of Emergency Medicine, Royal Melbourne Hospital, Melbourne, Australia
| | - Scott G Weiner
- Department of Emergency Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lewis S Nelson
- Department of Emergency Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
55
|
Jampel J, Quinn MJ, Catalano JL, Benca-Bachman CB, Brick L, Philip NS, Swift RM, McGeary JE. Synchronised transcranial magnetic stimulation for substance use-disordered Veterans: protocol for the pilot sham-controlled acceptability trial. BMJ Open 2023; 13:e066175. [PMID: 36717148 PMCID: PMC9887721 DOI: 10.1136/bmjopen-2022-066175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
INTRODUCTION Substance use disorders (SUDs) take an enormous toll on US Veterans and civilians alike. Existing empirically supported interventions vary by substance and demonstrate only moderate efficacy. Non-invasive brain stimulation represents an innovative treatment for SUDs, yet aspects of traditional neurostimulation may hinder its implementation in SUD populations. Synchronised transcranial magnetic stimulation (sTMS) uses rotating rare earth magnets to deliver low-field stimulation synchronised to an individual's alpha peak frequency that is safe for at-home administration. The current trial aims to assess the acceptability and feasibility of sTMS, as well as the safety of at-home sTMS administration for substance-disordered Veterans. METHODS AND ANALYSIS Sixty Veterans in substance treatment at the Providence Veterans Affairs will be randomised to receive 6 weeks of active or sham sTMS treatment. Eligibility will be confirmed by meeting Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition criteria for an alcohol, cocaine or opioid use disorder. Daily supervised sTMS treatment will occur either in clinic or at home through video monitoring. Clinical and self-report assessments will be completed at baseline, end of treatment and 1-month follow-up. Urine drug screening will occur once per week during the treatment phase. Primary outcomes include treatment adherence/retention and satisfaction to evaluate sTMS feasibility and acceptability in Veterans with SUDs. The safety of at-home sTMS administration will be assessed via adverse event monitoring. ETHICS AND DISSEMINATION The sTMS device received a significant risk determination for at-home use by the Food and Drug Administration in July 2021. Ethics approval was obtained in August 2021 from the Providence Veterans Affairs institutional review board and research and development committee. Data collection began in September 2021 and is planned to continue through December 2023. Findings will be disseminated at national conferences and in peer-reviewed journals. Results will serve to inform the development of large-scale clinical trials of sTMS efficacy for substance-disordered Veterans. TRIAL REGISTRATION NUMBER ClinicalTrials.gov Registry (NCT04336293).
Collapse
Affiliation(s)
- Jonathan Jampel
- Department of Psychology, Clark University, Worcester, Massachusetts, USA
| | | | - Jamie L Catalano
- Therapeutic Sciences Graduate Program, Division of Biology and Medicine, Brown University, Providence, Rhode Island, USA
| | - Chelsie B Benca-Bachman
- Providence VA Medical Center, Providence, Rhode Island, USA
- Behavioral Genetics of Addition Laboratory, Department of Psychology, Emory University, Atlanta, Georgia, USA
| | - Leslie Brick
- Providence VA Medical Center, Providence, Rhode Island, USA
- Department of Psychiatry and Human Behavior, Quantitative Sciences Program, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Noah S Philip
- The Center of Neurorestoration and Neurotechnology, Providence VA Medical Center, Providence, Rhode Island, USA
| | - Robert M Swift
- The Center of Neurorestoration and Neurotechnology, Providence VA Medical Center, Providence, Rhode Island, USA
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - John E McGeary
- The Center of Neurorestoration and Neurotechnology, Providence VA Medical Center, Providence, Rhode Island, USA
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
56
|
Stokłosa I, Więckiewicz G, Stokłosa M, Piegza M, Pudlo R, Gorczyca P. Medications for the Treatment of Alcohol Dependence-Current State of Knowledge and Future Perspectives from a Public Health Perspective. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1870. [PMID: 36767234 PMCID: PMC9915396 DOI: 10.3390/ijerph20031870] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 06/18/2023]
Abstract
No single effective therapy for alcohol abuse has been found, despite it being a serious sociological and economic problem for hundreds of years. It seems difficult to find a single drug as a panacea for the alcohol problem due to the complexity of the pathophysiology of alcohol dependence. The purpose of this narrative review is to review existing and potentially future pharmaceuticals for the treatment of alcohol dependence in the most affordable way possible. Psychotherapy is the mainstay of treatment for alcoholism, while few drugs approved by legislators are available in the augmentation of this treatment, such as acamprosate, disulfiram, and naltrexone, approved by the FDA, and nalmefene by the EMA. There are recent reports in the literature on the possibility of using baclofen, topiramate, varenicline, and gabapentin in the treatment of alcohol dependence. Moreover, the results of recent clinical trials using psychoactive substances such as psilocybin and MDMA appear to be a breakthrough in the modern treatment of alcohol abuse. Despite this initial optimism, a lot of scientific effort is still needed before new pharmacological methods supporting the treatment of alcohol dependence syndrome will be widely available.
Collapse
Affiliation(s)
- Iga Stokłosa
- Department and Clinic of Psychiatry, Medical University of Silesia, 42-612 Tarnowskie Góry, Poland
| | | | | | | | | | | |
Collapse
|
57
|
Chen H, Feng J, Chen L, Huang J, Zhang P, Chen C, Lu L, Tang C. Acupoint stimulation for alcohol use disorder: A systematic review and meta-analysis. Medicine (Baltimore) 2023; 102:e32614. [PMID: 36607868 PMCID: PMC9829291 DOI: 10.1097/md.0000000000032614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND To assess the effect of acupoint stimulation for Alcohol use disorders (AUD). METHODS AUD is a complex disease that threatens the health of the global population. Acupoint stimulation, a sort of therapy applying stimulation on acupoints to produce a therapeutic effect without side effects, has been widely used in AUD patients, but its efficacy remains controversial. Electronic databases (the Cochrane Library, EMBASE, PubMed, CNKI, VIP, Wan-Fang) were systematically searched for randomized controlled trials (RCTs) on acupoint stimulation for AUD from database inception to September 30, 2022. A meta-analysis was performed using Review Manager 5.4 software. Continuous data (scales) were expressed as mean differences (MDs) or standardized mean difference (SMD) with 95% confidence intervals (95% CI). Study methodological quality was assessed according to the Cochrane risk-of-bias tool for trials. The grading of recommendations assessment, development and evaluation was used to assess the certainty of evidence for outcomes. RESULTS A total of 16 RCTs with 1097 participants were included. Compared to psychotherapy or drug therapy alone, the combination of acupoint stimulation and other sorts of therapies presented advantages in alleviating alcohol craving (SMD = -1.09, 95% CI = -1.40 to -0.77, df = 2, P < .00001, grading of recommendations assessment, development and evaluation very low certainty), (SMD = -2.25, 95% CI = -3.17 to -1.34, df = 3, P < .00001, low certainty) and the severity of alcohol withdrawal symptoms (MD = -1.21, 95% CI = -2.32 to -0.1, df = 2, P = .03, low certainty), as well as improving anxiety (MD = -3.41, 95% CI = -4.06 to -2.76, df = 4, P < .00001, very low certainty) and depression levels (MD = -3.27, 95% CI = -4.92 to -1.62, df = 4, P = .0001, very low certainty) on patients with AUD. In addition, a greater effect was also found with the 4-week treatment courses in reducing craving (SMD = -2.18, 95% CI = -2.61 to -1.75, P < .00001, low certainty). CONCLUSION Acupoint stimulation and its combined therapy may better relieve AUD symptoms effectively and the treatment duration should be set at more than 2 weeks. However, due to the low-quality of the included RCTs, high-quality studies are needed to further confirm it in the future.
Collapse
Affiliation(s)
- Huishan Chen
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, No. 232, Waihuan East Road, University Town, Panyu District, Guangzhou City, Guangdong Province, China
| | - Jiaxin Feng
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, No. 232, Waihuan East Road, University Town, Panyu District, Guangzhou City, Guangdong Province, China
| | - Li Chen
- Fifth Clinical Medical College, Guangzhou University of Chinese Medicine, No. 232, Waihuan East Road, University Town, Panyu District, Guangzhou City, Guangdong Province, China
| | - Jiarong Huang
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Gulou District, Fuzhou City, Fujian Province, China
| | - Peiming Zhang
- Clinical Research and Big Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, No. 232, Waihuan East Road, University Town, Panyu District, Guangzhou City, Guangdong Province, China
| | - Chen Chen
- Clinical Research and Big Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, No. 232, Waihuan East Road, University Town, Panyu District, Guangzhou City, Guangdong Province, China
| | - Liming Lu
- Clinical Research and Big Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, No. 232, Waihuan East Road, University Town, Panyu District, Guangzhou City, Guangdong Province, China
| | - Chunzhi Tang
- Clinical Research and Big Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, No. 232, Waihuan East Road, University Town, Panyu District, Guangzhou City, Guangdong Province, China
| |
Collapse
|
58
|
Konstantelos N, Tourchian N, McCormack D, Lecce P, Tadrous M, Gomes T. Impact of policy changes and drug shortages on acamprosate and naltrexone use in Ontario, Canada. Drug Alcohol Depend 2023; 242:109705. [PMID: 36463765 DOI: 10.1016/j.drugalcdep.2022.109705] [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: 07/11/2022] [Revised: 11/02/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Acamprosate and naltrexone, evidence-based pharmacotherapies for alcohol use disorder (AUD), are publicly covered by the Ontario Drug Benefit (ODB) programs; however, their availability has changed over time, with expanded formulary access in July 2018, followed by an acamprosate shortage in February 2019 and ending in July 2020. We evaluated the impact of these events on the use of these medications in Ontario, Canada. METHODS We conducted a time-series analysis among individuals with AUD dispensed acamprosate or naltrexone through the ODB from July 2016 to December 2020. Outcomes included monthly rates of those with AUD on therapy (primary), and rate of initiation (secondary) overall and by treatment type. We used autoregressive moving average models to evaluate the impact of expanded coverage and the acamprosate shortage on rates of use, and reported characteristics at first dispensation. RESULTS Over the study period, 10,637 individuals (61.0% male) initiated acamprosate or naltrexone. Expanded coverage increased monthly utilization rates of acamprosate (p = 0.0004), naltrexone (p < 0.0001), and either AUD pharmacotherapy (p < 0.0001). The acamprosate shortage led to a 98.1% reduction in acamprosate use (p = 0.0003) but did not impact naltrexone (p = 0.51). Our secondary analysis yielded consistent results with respect to the shortage; however, the expanded formulary listing did not impact the rate of new acamprosate patients (p = 0.3). By December 2020, 5.3% of ODB recipients with AUD were accessing pharmacotherapy. CONCLUSIONS Although coverage expansion increased access to medications that treat AUD, the shortage of acamprosate led to large reductions in its use, with no responsive increase in naltrexone prescribing.
Collapse
Affiliation(s)
| | - Nima Tourchian
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | | | - Pamela Lecce
- Public Health Ontario, Toronto, Canada; Department of Family and Community Medicine, University of Toronto, Toronto, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Mina Tadrous
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada; Women's College Hospital, Toronto, Canada
| | - Tara Gomes
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada; ICES, Toronto, Canada; Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Canada.
| |
Collapse
|
59
|
Flores-Ramirez FJ, Illenberger JM, Pascasio GE, Matzeu A, Mason BJ, Martin-Fardon R. Alternative use of suvorexant (Belsomra ®) for the prevention of alcohol drinking and seeking in rats with a history of alcohol dependence. Front Behav Neurosci 2022; 16:1085882. [PMID: 36620860 PMCID: PMC9813433 DOI: 10.3389/fnbeh.2022.1085882] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Alcohol use disorder (AUD) is one of the most treatment-resistant medical conditions globally. The orexin (Orx) system regulates diverse physiological processes, including stress, and is a system of interest for the development of pharmaceuticals to treat substance use disorders, particularly AUD. The present study tested the ability of the dual orexin receptor antagonist suvorexant (SUV), marketed by Merck as Belsomra®, for the treatment of insomnia, to decrease alcohol self-administration and the stress-induced reinstatement of alcohol-seeking behavior in male Wistar rats with a history of alcohol dependence. Rats were trained to orally self-administer 10% alcohol (30 min/day for 3 weeks) and were either made dependent via chronic intermittent alcohol vapor exposure (14 h ON, 10 h OFF) for 6 weeks or exposed to air (non-dependent). Starting on week 7, the effect of SUV (0-20 mg/kg, p.o.) was tested on alcohol self-administration at acute abstinence (8 h after vapor was turned OFF) twice weekly. A separate cohort of rats that were prepared in parallel was removed from alcohol vapor exposure and then subjected to extinction training for 14 sessions. Once extinction was achieved, the rats received SUV (0 and 5 mg/kg, p.o.) and were tested for the footshock stress-induced reinstatement of alcohol-seeking behavior. Suvorexant at 5, 10, and 20 mg/kg selectively decreased alcohol intake in dependent rats. Furthermore, 5 mg/kg SUV prevented the stress-induced reinstatement of alcohol-seeking behavior in dependent rats only. These results underscore the significance of targeting the Orx system for the treatment of substance use disorders generally and suggest that repurposing SUV could be an alternative approach for the treatment of AUD.
Collapse
|
60
|
Santos GM, Ikeda J, Coffin P, Walker J, Matheson T, Ali A, McLaughlin M, Jain J, Arenander J, Vittinghoff E, Batki S. Targeted Oral Naltrexone for Mild to Moderate Alcohol Use Disorder Among Sexual and Gender Minority Men: A Randomized Trial. Am J Psychiatry 2022; 179:915-926. [PMID: 36285404 PMCID: PMC10072332 DOI: 10.1176/appi.ajp.20220335] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The authors sought to determine the efficacy of targeted naltrexone in sexual and gender minority men (SGM) who binge drink and have mild to moderate alcohol use disorder. METHODS In a double-blind placebo-controlled trial, a total of 120 SGM who binge drink and have mild to moderate alcohol use disorder were randomized in a 1:1 ratio to receive targeted oral naltrexone (50 mg) or placebo with weekly counseling for 12 weeks. The study's primary endpoints were binge-drinking intensity, defined as 1) number of drinks in the past 30 days; 2) any binge drinking in the past week; 3) number of binge-drinking days in the past week; and 4) number of drinking days in the past week. The study also measured changes in alcohol use with two alcohol biomarker measures: ethyl glucuronide in urine samples and phosphatidylethanol (PEth) in dried blood spot samples. RESULTS Ninety-three percent completed the trial, with 85% of weekly follow-up visits completed. In intention-to-treat analyses, naltrexone was associated with a significantly reduced reported number of binge-drinking days (incidence rate ratio [IRR]=0.74, 95% CI=0.56, 0.98; number needed to treat [NNT]=2), weeks with any binge drinking (IRR=0.83, 95% CI=0.72, 0.96; NNT=7.4), number of drinks per month (IRR=0.69, 95% CI=0.52, 0.91; NNT=5.7 for 10 drinks), and alcohol craving scores (coefficient=-9.25, 95% CI=-17.20, -1.31). In as-treated analyses among those who took their medication on average at least 2.5 days per week (the median frequency in the study), naltrexone reduced any binge drinking (IRR=0.84, 95% CI=0.71, 0.99), number of binge-drinking days (IRR=0.67, 95% CI=0.47, 0.96), and PEth concentrations (coefficient=-55.47, 95% CI=-110.75, -0.20). At 6 months posttreatment, naltrexone had sustained effects in number of drinks per month (IRR=0.69, 95% CI=0.50, 0.97), number of binge-drinking days (IRR=0.67, 95% CI=0.47, 0.95), and any binge drinking in the past week (IRR=0.79, 95% CI=0.63, 0.99). CONCLUSIONS Targeted naltrexone significantly reduced drinking outcomes among SGM with mild to moderate alcohol use disorder during treatment, with sustained effects at 6 months posttreatment. Naltrexone may be an important pharmacotherapy to address binge drinking in populations with mild to moderate alcohol use disorder.
Collapse
Affiliation(s)
- Glenn-Milo Santos
- Department of Community Health Systems (Santos, Jain), Division of HIV, Infectious Disease, and Global Medicine (Coffin), Department of Psychiatry and Behavioral Sciences (Jain, Batki), and Department of Epidemiology and Biostatistics (Vittinghoff), University of California, San Francisco; Center on Substance Use and Health, San Francisco Department of Public Health, San Francisco (Santos, Ikeda, Coffin, Walker, Matheson, Ali, McLaughlin); Human Services Department, County of Sonoma, Sonoma, California (Arenander); San Francisco VA Health Care System, San Francisco (Batki)
| | - Janet Ikeda
- Department of Community Health Systems (Santos, Jain), Division of HIV, Infectious Disease, and Global Medicine (Coffin), Department of Psychiatry and Behavioral Sciences (Jain, Batki), and Department of Epidemiology and Biostatistics (Vittinghoff), University of California, San Francisco; Center on Substance Use and Health, San Francisco Department of Public Health, San Francisco (Santos, Ikeda, Coffin, Walker, Matheson, Ali, McLaughlin); Human Services Department, County of Sonoma, Sonoma, California (Arenander); San Francisco VA Health Care System, San Francisco (Batki)
| | - Phillip Coffin
- Department of Community Health Systems (Santos, Jain), Division of HIV, Infectious Disease, and Global Medicine (Coffin), Department of Psychiatry and Behavioral Sciences (Jain, Batki), and Department of Epidemiology and Biostatistics (Vittinghoff), University of California, San Francisco; Center on Substance Use and Health, San Francisco Department of Public Health, San Francisco (Santos, Ikeda, Coffin, Walker, Matheson, Ali, McLaughlin); Human Services Department, County of Sonoma, Sonoma, California (Arenander); San Francisco VA Health Care System, San Francisco (Batki)
| | - John Walker
- Department of Community Health Systems (Santos, Jain), Division of HIV, Infectious Disease, and Global Medicine (Coffin), Department of Psychiatry and Behavioral Sciences (Jain, Batki), and Department of Epidemiology and Biostatistics (Vittinghoff), University of California, San Francisco; Center on Substance Use and Health, San Francisco Department of Public Health, San Francisco (Santos, Ikeda, Coffin, Walker, Matheson, Ali, McLaughlin); Human Services Department, County of Sonoma, Sonoma, California (Arenander); San Francisco VA Health Care System, San Francisco (Batki)
| | - Tim Matheson
- Department of Community Health Systems (Santos, Jain), Division of HIV, Infectious Disease, and Global Medicine (Coffin), Department of Psychiatry and Behavioral Sciences (Jain, Batki), and Department of Epidemiology and Biostatistics (Vittinghoff), University of California, San Francisco; Center on Substance Use and Health, San Francisco Department of Public Health, San Francisco (Santos, Ikeda, Coffin, Walker, Matheson, Ali, McLaughlin); Human Services Department, County of Sonoma, Sonoma, California (Arenander); San Francisco VA Health Care System, San Francisco (Batki)
| | - Arsheen Ali
- Department of Community Health Systems (Santos, Jain), Division of HIV, Infectious Disease, and Global Medicine (Coffin), Department of Psychiatry and Behavioral Sciences (Jain, Batki), and Department of Epidemiology and Biostatistics (Vittinghoff), University of California, San Francisco; Center on Substance Use and Health, San Francisco Department of Public Health, San Francisco (Santos, Ikeda, Coffin, Walker, Matheson, Ali, McLaughlin); Human Services Department, County of Sonoma, Sonoma, California (Arenander); San Francisco VA Health Care System, San Francisco (Batki)
| | - Matthew McLaughlin
- Department of Community Health Systems (Santos, Jain), Division of HIV, Infectious Disease, and Global Medicine (Coffin), Department of Psychiatry and Behavioral Sciences (Jain, Batki), and Department of Epidemiology and Biostatistics (Vittinghoff), University of California, San Francisco; Center on Substance Use and Health, San Francisco Department of Public Health, San Francisco (Santos, Ikeda, Coffin, Walker, Matheson, Ali, McLaughlin); Human Services Department, County of Sonoma, Sonoma, California (Arenander); San Francisco VA Health Care System, San Francisco (Batki)
| | - Jennifer Jain
- Department of Community Health Systems (Santos, Jain), Division of HIV, Infectious Disease, and Global Medicine (Coffin), Department of Psychiatry and Behavioral Sciences (Jain, Batki), and Department of Epidemiology and Biostatistics (Vittinghoff), University of California, San Francisco; Center on Substance Use and Health, San Francisco Department of Public Health, San Francisco (Santos, Ikeda, Coffin, Walker, Matheson, Ali, McLaughlin); Human Services Department, County of Sonoma, Sonoma, California (Arenander); San Francisco VA Health Care System, San Francisco (Batki)
| | - Justine Arenander
- Department of Community Health Systems (Santos, Jain), Division of HIV, Infectious Disease, and Global Medicine (Coffin), Department of Psychiatry and Behavioral Sciences (Jain, Batki), and Department of Epidemiology and Biostatistics (Vittinghoff), University of California, San Francisco; Center on Substance Use and Health, San Francisco Department of Public Health, San Francisco (Santos, Ikeda, Coffin, Walker, Matheson, Ali, McLaughlin); Human Services Department, County of Sonoma, Sonoma, California (Arenander); San Francisco VA Health Care System, San Francisco (Batki)
| | - Eric Vittinghoff
- Department of Community Health Systems (Santos, Jain), Division of HIV, Infectious Disease, and Global Medicine (Coffin), Department of Psychiatry and Behavioral Sciences (Jain, Batki), and Department of Epidemiology and Biostatistics (Vittinghoff), University of California, San Francisco; Center on Substance Use and Health, San Francisco Department of Public Health, San Francisco (Santos, Ikeda, Coffin, Walker, Matheson, Ali, McLaughlin); Human Services Department, County of Sonoma, Sonoma, California (Arenander); San Francisco VA Health Care System, San Francisco (Batki)
| | - Steven Batki
- Department of Community Health Systems (Santos, Jain), Division of HIV, Infectious Disease, and Global Medicine (Coffin), Department of Psychiatry and Behavioral Sciences (Jain, Batki), and Department of Epidemiology and Biostatistics (Vittinghoff), University of California, San Francisco; Center on Substance Use and Health, San Francisco Department of Public Health, San Francisco (Santos, Ikeda, Coffin, Walker, Matheson, Ali, McLaughlin); Human Services Department, County of Sonoma, Sonoma, California (Arenander); San Francisco VA Health Care System, San Francisco (Batki)
| |
Collapse
|
61
|
Mason BJ. Looking Back, Looking Forward: Current Medications and Innovative Potential Medications to Treat Alcohol Use Disorder. Alcohol Res 2022; 42:11. [PMID: 36320345 PMCID: PMC9595448 DOI: 10.35946/arcr.v42.1.11] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
This article is part of a Festschrift commemorating the 50th anniversary of the National Institute on Alcohol Abuse and Alcoholism (NIAAA). Established in 1970, first as part of the National Institute of Mental Health and later as an independent institute of the National Institutes of Health, NIAAA today is the world's largest funding agency for alcohol research. In addition to its own intramural research program, NIAAA supports the entire spectrum of innovative basic, translational, and clinical research to advance the diagnosis, prevention, and treatment of alcohol use disorder and alcohol-related problems. To celebrate the anniversary, NIAAA hosted a 2-day symposium, "Alcohol Across the Lifespan: 50 Years of Evidence-Based Diagnosis, Prevention, and Treatment Research," devoted to key topics within the field of alcohol research. This article is based on Dr. Mason's presentation at the event. NIAAA Director George F. Koob, Ph.D., serves as editor of the Festschrift.
Collapse
Affiliation(s)
- Barbara J Mason
- Pearson Center for Alcoholism and Addiction Research, Department of Molecular Medicine, Scripps Research Institute, La Jolla, California
| |
Collapse
|
62
|
van Amsterdam J, Blanken P, Spijkerman R, van den Brink W, Hendriks V. The Added Value of Pharmacotherapy to Cognitive Behavior Therapy And Vice Versa in the Treatment of Alcohol Use Disorders: A Systematic Review. Alcohol Alcohol 2022; 57:768-775. [PMID: 36085572 DOI: 10.1093/alcalc/agac043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/31/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
AIM To explore whether combined interventions i.e. psychotherapeutic plus psychosocial interventions are more effective than monotherapies in the treatment of alcohol use disorders. METHODS Systematic review of the results of randomized controlled trials that compared combined therapies with monotherapies (either pharmacotherapy or psychotherapy). RESULTS The search resulted in 28 eligible studies. Data from these RCTs showed that 10 out of 19 RCTs (53%) demonstrated an added value of combined therapy (psychotherapy + pharmacotherapy) compared to psychotherapy only, whereas only three out of nine RCTs (33%) comparing combined therapy with pharmacotherapy showed a possible added value for combined therapy. CONCLUSIONS Pharmacotherapy is effective to treat AUD with or without psychotherapy and that psychotherapy can best be offered in combination with pharmacotherapy.
Collapse
Affiliation(s)
- Jan van Amsterdam
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Research Program Compulsivity, Impulsivity & Attention, P.O. Box 22660, 1100 DD, Amsterdam, The Netherlands
| | - Peter Blanken
- Parnassia Addiction Research Centre (PARC), Parnassia Psychiatric Institute, Zoutkeetsingel 40, 2512 HN, The Hague, The Netherlands
| | - Renske Spijkerman
- Parnassia Addiction Research Centre (PARC), Parnassia Psychiatric Institute, Zoutkeetsingel 40, 2512 HN, The Hague, The Netherlands
| | - Wim van den Brink
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Research Program Compulsivity, Impulsivity & Attention, P.O. Box 22660, 1100 DD, Amsterdam, The Netherlands
| | - Vincent Hendriks
- Parnassia Addiction Research Centre (PARC), Parnassia Psychiatric Institute, Zoutkeetsingel 40, 2512 HN, The Hague, The Netherlands.,Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
63
|
Antonelli M, Sestito L, Tarli C, Addolorato G. Perspectives on the pharmacological management of alcohol use disorder: Are the approved medications effective? Eur J Intern Med 2022; 103:13-22. [PMID: 35597734 DOI: 10.1016/j.ejim.2022.05.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/27/2022] [Accepted: 05/13/2022] [Indexed: 12/18/2022]
Abstract
INTRODUCTION In the last decades, many medications have been tested for the treatment of Alcohol Use Disorder (AUD). Among them, disulfiram, acamprosate, naltrexone, nalmefene, sodium oxybate and baclofen have been approved in different countries, with different specific indications. Topiramate is not approved for the treatment of AUD, however, it is suggested as a therapeutic option by the American Psychiatric Association for patients who do not tolerate or respond to approved therapies. AREAS COVERED In this narrative review we have analyzed the main studies available in literature, investigating the efficacy and safety of these medications, distinguishing whether they were oriented towards abstinence or not. Randomized controlled studies, analyzing larger populations for longer periods were the main focus of our analysis. CONCLUSIONS The medications currently available for the treatment of AUD are quite effective, yet further progress can still be achieved through the personalized strategies. Also, these medications are still markedly underutilized in clinical practice and many patients do not have access to specialized treatment.
Collapse
Affiliation(s)
- Mariangela Antonelli
- Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luisa Sestito
- Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudia Tarli
- Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanni Addolorato
- Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; CEMAD Digestive Disease Center, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy.
| |
Collapse
|
64
|
Kirkland AE, Browning BD, Green R, Leggio L, Meyerhoff DJ, Squeglia LM. Brain metabolite alterations related to alcohol use: a meta-analysis of proton magnetic resonance spectroscopy studies. Mol Psychiatry 2022; 27:3223-3236. [PMID: 35508628 PMCID: PMC10578135 DOI: 10.1038/s41380-022-01594-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 11/08/2022]
Abstract
Alcohol misuse and alcohol use disorder (AlUD) have neurobiological consequences. This meta-analysis of proton magnetic resonance spectroscopy (MRS) studies aimed to assess the differences in brain metabolite levels in alcohol misuse and AUD relative to controls (PROSPERO registration: CRD42020209890). Hedge's g with random-effects modeling was used. Sub-group and meta-regression techniques explored potential sources of demographic and MRS parameter heterogeneity. A comprehensive literature review identified 43 studies, resulting in 69 models across gray and white matter (GM, WM). Lower N-acetylaspartate levels were found in frontal, anterior cingulate cortex (ACC), hippocampal, and cerebellar GM, and frontal and parietal WM, suggesting decreased neuronal and axonal viability. Lower choline-containing metabolite levels (all metabolites contributing to choline peak) were found in frontal, temporal, thalamic, and cerebellar GM, and frontal and parietal WM, suggesting membrane alterations related to alcohol misuse. Lower creatine-containing metabolite levels (Cr; all metabolites contributing to Cr peak) were found in temporal and occipital cortical GM, while higher levels were noted in midbrain/brainstem GM; this finding may have implications for using Cr as an internal reference. The lack of significant group differences in glutamate-related levels is possibly related to biological and methodological complexities. The few studies reporting on GABA found lower levels restricted to the ACC. Confounding variables were age, abstinence duration, treatment status, and MRS parameters (echo time, quantification type, data quality). This first meta-analysis of proton MRS studies consolidates the numerous individual studies to identify neurometabolite alterations within alcohol misuse and AUD. Future studies can leverage this new formalized information to investigate treatments that might effectively target the observed disturbances.
Collapse
Affiliation(s)
- Anna E Kirkland
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.
| | - Brittney D Browning
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - ReJoyce Green
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Lorenzo Leggio
- National Institutes of Health, NIDA and NIAAA, Baltimore, MD, USA
| | - Dieter J Meyerhoff
- Department of Radiology and Biomedical Imaging, University of California San Francisco and VA Medical Center, San Francisco, CA, USA
| | - Lindsay M Squeglia
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
65
|
Sorkhou M, Stogios N, Sayrafizadeh N, Hahn MK, Agarwal SM, George TP. Non-invasive neuromodulation of dorsolateral prefrontal cortex to reduce craving in alcohol use disorder: A meta-analysis. DRUG AND ALCOHOL DEPENDENCE REPORTS 2022; 4:100076. [PMID: 36846579 PMCID: PMC9948891 DOI: 10.1016/j.dadr.2022.100076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/16/2022] [Accepted: 06/23/2022] [Indexed: 11/26/2022]
Abstract
Introduction While several pharmacological and behavioral treatments are available for alcohol use disorder (AUD), they may not be effective for all patients. The aim of this systematic review and meta-analysis was to evaluate the efficacy and safety of rTMS and tDCS for craving in AUD. Methods EMBASE, Cochrane Library, PsycINFO, and PubMed databases were searched for original, peer-reviewed research articles in the English language published between January 2000 and January 2022. Randomized controlled trials (RCTs) reporting changes in alcohol craving among patients with AUD were selected. Random-effects meta-analysis was employed to pool data. Results Changes in alcohol craving were extracted from 15 RCTs. Six studies assessed the efficacy of rTMS while nine studies examined tDCS. Results demonstrated that in comparison to sham stimulation, active rTMS to the DLPFC yields small but significant reductions in alcohol craving (standardized mean difference [SMD] = -0.27, p = .03). However, DLPFC stimulation via tDCS was not superior to sham stimulation in producing changes in alcohol craving (SMD = -0.08, p = .59). Conclusions Our meta-analysis suggests that rTMS may be superior to tDCS in reducing alcohol craving in patients with AUD. However, additional research is needed to identify optimal stimulation parameters for both non-invasive neuromodulatory techniques in AUD.
Collapse
Affiliation(s)
- Maryam Sorkhou
- Institute of Medical Sciences (IMS), University of Toronto, Canada,Temerty Faculty of Medicine, University of Toronto, Canada,Centre for Complex Interventions (CCI), Centre for Addiction and Mental Health (CAMH), Canada
| | - Nicolette Stogios
- Institute of Medical Sciences (IMS), University of Toronto, Canada,Schizophrenia Division, Centre for Addiction and Mental Health (CAMH), Canada
| | - Negar Sayrafizadeh
- Institute of Medical Sciences (IMS), University of Toronto, Canada,Temerty Faculty of Medicine, University of Toronto, Canada,Centre for Complex Interventions (CCI), Centre for Addiction and Mental Health (CAMH), Canada
| | - Margaret K. Hahn
- Institute of Medical Sciences (IMS), University of Toronto, Canada,Department of Psychiatry, University of Toronto, Canada,Temerty Faculty of Medicine, University of Toronto, Canada,Schizophrenia Division, Centre for Addiction and Mental Health (CAMH), Canada
| | - Sri Mahavir Agarwal
- Institute of Medical Sciences (IMS), University of Toronto, Canada,Department of Psychiatry, University of Toronto, Canada,Temerty Faculty of Medicine, University of Toronto, Canada,Schizophrenia Division, Centre for Addiction and Mental Health (CAMH), Canada
| | - Tony P. George
- Institute of Medical Sciences (IMS), University of Toronto, Canada,Department of Psychiatry, University of Toronto, Canada,Temerty Faculty of Medicine, University of Toronto, Canada,Centre for Complex Interventions (CCI), Centre for Addiction and Mental Health (CAMH), Canada,Corresponding author at: Addictions Division; Schizophrenia Division, University of Toronto Clinician-Scientist, Centre for Addiction and Mental Health (CAMH), 60 White Squirrel Way, Room 312, Toronto, Ontario M6J 1H4, Canada
| |
Collapse
|
66
|
Stryhn L, Larsen MB, Mejldal A, Sibbersen C, Nielsen DG, Nielsen B, Nielsen AS, Stenager E, Mellentin AI. Relapse prevention for alcohol use disorders: combined acamprosate and cue exposure therapy as aftercare. Nord J Psychiatry 2022; 76:394-402. [PMID: 34622734 DOI: 10.1080/08039488.2021.1985169] [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] [Indexed: 10/20/2022]
Abstract
AIMS Many patients with alcohol use disorders are challenged by cravings leading to repeated relapses. Both cue exposure therapy (CET) and acamprosate target alcohol cravings and are often combined (CET + acamprosate). The main aim of this study was to investigate whether aftercare treatment consisting of CET combined with acamprosate is equivalent to (A) CET as monotherapy, (B) aftercare as usual (AAU) as monotherapy or (C) AAU combined with acamprosate. METHODS Patients were randomized to receive either CET with urge-specific coping skills (USCS) as aftercare or AAU. Acamprosate prescription data were extracted from patient case records. Alcohol consumption, cravings, and USCS were assessed at pre-aftercare, post-aftercare, and 6-month follow-up. RESULTS Overall, patients increased their alcohol consumption during and following aftercare treatment, thereby relapsing despite any treatment. However, CET + acamprosate achieved greater abstinence compared to AAU + acamprosate at follow-up (p=.047). CET + acamprosate also reduced number of drinking days (p=.020) and number of days with excessive drinking (p=.020) at post-aftercare, when compared to AAU monotherapy. CET monotherapy increased sensible drinking at post-aftercare compared to AAU monotherapy (p=.045) and AAU + acamprosate (p=.047). Only CET monotherapy showed improvement in cravings, when compared to AAU at follow-up (mean urge level: p=.032; peak urge level: p=.014). CONCLUSION The study showed that CET both as monotherapy and combined with acamprosate was superior to AAU monotherapy and AAU + acamprosate in reducing alcohol consumption. Only CET + acamprosate was capable of reducing alcohol consumption in the longer term, indicating that anti-craving medication may not impede CET from exerting an effect on alcohol consumption. Trial registration: ClinicalTrials.gov ID: NCT02298751 (24/11-2014).
Collapse
Affiliation(s)
- Lene Stryhn
- Department of Clinical Research, Unit for Clinical Alcohol Research, Unit for Psychiatric Research, University of Southern Denmark, Odense, Denmark.,Unit for Psychiatric Research, Institute of Regional Health Services Research, University of Southern Denmark, Aabenraa, Denmark.,Psychiatric Research Academy, Odense, Denmark
| | - Mathias Bach Larsen
- Department of Clinical Research, Unit for Clinical Alcohol Research, Unit for Psychiatric Research, University of Southern Denmark, Odense, Denmark.,Unit for Psychiatric Research, Institute of Regional Health Services Research, University of Southern Denmark, Aabenraa, Denmark.,Psychiatric Research Academy, Odense, Denmark
| | - Anna Mejldal
- Department of Clinical Research, Unit for Clinical Alcohol Research, Unit for Psychiatric Research, University of Southern Denmark, Odense, Denmark
| | - Christian Sibbersen
- Department of Clinical Research, Unit for Clinical Alcohol Research, Unit for Psychiatric Research, University of Southern Denmark, Odense, Denmark
| | - Dorthe Grüner Nielsen
- Department of Clinical Research, Unit for Clinical Alcohol Research, Unit for Psychiatric Research, University of Southern Denmark, Odense, Denmark
| | - Bent Nielsen
- Department of Clinical Research, Unit for Clinical Alcohol Research, Unit for Psychiatric Research, University of Southern Denmark, Odense, Denmark
| | - Anette Søgaard Nielsen
- Department of Clinical Research, Unit for Clinical Alcohol Research, Unit for Psychiatric Research, University of Southern Denmark, Odense, Denmark.,Department of Clinical Research, Brain Research-Inter-Disciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Odense, Denmark
| | - Elsebeth Stenager
- Unit for Psychiatric Research, Institute of Regional Health Services Research, University of Southern Denmark, Aabenraa, Denmark.,Department of Clinical Research, Brain Research-Inter-Disciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Odense, Denmark
| | - Angelina Isabella Mellentin
- Department of Clinical Research, Unit for Clinical Alcohol Research, Unit for Psychiatric Research, University of Southern Denmark, Odense, Denmark.,Unit for Psychiatric Research, Institute of Regional Health Services Research, University of Southern Denmark, Aabenraa, Denmark.,Department of Clinical Research, Brain Research-Inter-Disciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Odense, Denmark
| |
Collapse
|
67
|
Lohoff FW. Targeting Unmet Clinical Needs in the Treatment of Alcohol Use Disorder. Front Psychiatry 2022; 13:767506. [PMID: 35757224 PMCID: PMC9218222 DOI: 10.3389/fpsyt.2022.767506] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Alcohol Use Disorder (AUD) is a chronic psychiatric disorder marked by impaired control over drinking behavior that poses a significant challenge to the individual, their community, the healthcare system and economy. While the negative consequences of chronic excessive alcohol consumption are well-documented, effective treatment for AUD and alcohol-associated diseases remains challenging. Cognitive and behavioral treatment, with or without pharmaceutical interventions, remain the most commonly used methods; however, their efficacy is limited. The development of new treatment protocols for AUD is challenged by difficulty in accurately measuring patterns of alcohol consumption in AUD patients, a lack of a clear understanding of the neuropsychological basis of the disorder, the high likelihood of AUD patients relapsing after receiving treatment, and the numerous end-organ comorbidities associated with excessive alcohol use. Identification and prediction of patients who may respond well to a certain treatment mechanism as well as clinical measurement of a patient's alcohol exposure are bottlenecks in AUD research which should be further addressed. In addition, greater focus must be placed on the development of novel strategies of drug design aimed at targeting the integrated neural pathways implicated in AUD pathogenesis, so that next-generation AUD treatment protocols can address the broad and systemic effects of AUD and its comorbid conditions.
Collapse
Affiliation(s)
- Falk W. Lohoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, United States
| |
Collapse
|
68
|
Serra YA, Barros-Santos T, Anjos-Santos A, Kisaki ND, Jovita-Farias C, Leite JPC, Santana MCE, Coimbra JPSA, de Jesus NMS, Sulima A, Barbosa PCR, Malpezzi-Marinho ELA, Rice KC, Oliveira-Lima AJ, Berro LF, Marinho EAV. Role of 5-HT 2A receptors in the effects of ayahuasca on ethanol self-administration using a two-bottle choice paradigm in male mice. Psychopharmacology (Berl) 2022; 239:1679-1687. [PMID: 35253069 PMCID: PMC10750423 DOI: 10.1007/s00213-022-06104-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/23/2022] [Indexed: 01/21/2023]
Abstract
RATIONALE Ayahuasca has been proposed as a potential treatment of alcohol (ethanol) use disorder (AUD). The serotonin 5-HT2A receptor agonist N,N-dimethyltryptamine (DMT) is the main psychoactive component of ayahuasca, suggesting that its therapeutic effects may be mediated by 5-HT2A receptors. OBJECTIVES The aim of the present study was to investigate the effects of ayahuasca on the expression of ethanol self-administration using a two-bottle choice procedure and the role of 5-HT2A receptors in those effects. METHODS Male mice had intermittent access to ethanol (10% v/v) in a two-bottle choice procedure for 30 days. Animals were then submitted to 3 treatment phases, each followed by ethanol re-exposure tests. During the treatment phase, every 3 days, animals received i.p. injections of either vehicle or the 5-HT2A receptor antagonist M100907 (M100, 1 mg/kg) followed by an i.g. (gavage) administration of vehicle or ayahuasca (100 mg/kg) and were exposed to the self-administration apparatus with no ethanol availability. During re-exposure tests, animals were submitted to the same conditions as during acquisition, with no treatments prior to those sessions. RESULTS Treatment with ayahuasca blocked the expression of ethanol self-administration, decreasing ethanol intake and preference during re-exposure tests. Pretreatment with M100 blocked the effects of ayahuasca on ethanol drinking without significantly attenuating ethanol self-administration. CONCLUSIONS Treatment with ayahuasca during alcohol abstinence blocked the expression of alcohol self-administration in mice, and 5-HT2A receptor activation is critical for those effects to emerge. Our findings support a potential for ayahuasca and other 5-HT2A receptor agonists as adjunctive pharmacotherapies for the treatment of AUD.
Collapse
Affiliation(s)
- Yasmim A Serra
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Thaísa Barros-Santos
- Department of Biological Sciences, Universidade Estadual de Santa Cruz, Ilhéus, BA, Brazil
| | - Alexia Anjos-Santos
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, National Institute On Drug Abuse and the National Institute On Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Natali D Kisaki
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Caio Jovita-Farias
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - João P C Leite
- Department of Biological Sciences, Universidade Estadual de Santa Cruz, Ilhéus, BA, Brazil
| | - Maria C E Santana
- Department of Biological Sciences, Universidade Estadual de Santa Cruz, Ilhéus, BA, Brazil
| | - João P S A Coimbra
- Department of Biological Sciences, Universidade Estadual de Santa Cruz, Ilhéus, BA, Brazil
| | - Nailton M S de Jesus
- Department of Biological Sciences, Universidade Estadual de Santa Cruz, Ilhéus, BA, Brazil
| | - Agnieszka Sulima
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, National Institute On Drug Abuse and the National Institute On Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Paulo C R Barbosa
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | | | - Kenner C Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, National Institute On Drug Abuse and the National Institute On Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Alexandre J Oliveira-Lima
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil
| | - Laís F Berro
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil.
- Department of Psychiatry and Human Behavior, Center for Innovation and Discovery in Addictions, University of Mississippi Medical Center, 2500 N State St, Jackson, MS, 39216, USA.
| | - Eduardo A V Marinho
- Department of Health Sciences, Universidade Estadual de Santa Cruz, Rod. Ilhéus/Itabuna, Km 16, Ilhéus, BA, 45662-0, Brazil.
| |
Collapse
|
69
|
Wizła M, Kraus SW, Lewczuk K. Perspective: Can psychedelic-assisted therapy be a promising aid in compulsive sexual behavior disorder treatment? Compr Psychiatry 2022; 115:152303. [PMID: 35334305 DOI: 10.1016/j.comppsych.2022.152303] [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: 10/14/2021] [Revised: 11/24/2021] [Accepted: 03/01/2022] [Indexed: 11/03/2022] Open
Abstract
Recently, there has been an increase in studies yielding evidence for psychedelics' anxiolytic and anti-depressive qualities. Preliminary evidence for treatment in substance addiction is also available. In our manuscript, we present a perspective on the possible effectiveness and mechanisms of action of psychedelics' introduction in the treatment of Compulsive Sexual Behavior Disorder (CSBD) and other p roblematic sexual behaviors, which are considered representative of the so-called "behavioral addiction" category. Evidence for the efficacy of Mindfulness Based Interventions in CSBD treatment is promising. Psychedelics- and mindfulness-induced states share common characteristics on both a subjective and objective level. One of the proposed mechanisms regards reduction of experiential avoidance through the promotion of exposure and acceptance. On the neurophysiological level, a shift from higher- to lower-level association regions and an impact on 5- HT2A receptors is observed. Elaborated mechanisms explain the possible enhancement of therapeutic processes by psychedelics. Psychedelics' relative safety and low addictive potential support their introduction into traditional forms of therapy for CSBD and other out of control behaviors.
Collapse
Affiliation(s)
- Magdalena Wizła
- Institute of Psychology, Cardinal Stefan Wyszynski University, Warsaw, Poland.
| | - Shane W Kraus
- Department of Psychology, University of Nevada, Las Vegas, NV, USA.
| | - Karol Lewczuk
- Institute of Psychology, Cardinal Stefan Wyszynski University, Warsaw, Poland.
| |
Collapse
|
70
|
Dhital R, Coleman R, Day E, Drummond C, Lingford-Hughes A, Marsden J, Phillips T, Sinclair J, Strang J, Weinman J, Whittlesea C, Widyaratna K, Donoghue K. Service Users' Views and Experiences of Alcohol Relapse Prevention Treatment and Adherence: New Role for Pharmacists? Alcohol Alcohol 2022; 57:602-608. [PMID: 35292814 PMCID: PMC9465522 DOI: 10.1093/alcalc/agac011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 02/08/2022] [Accepted: 02/15/2022] [Indexed: 12/01/2022] Open
Abstract
AIMS To understand service users' views and experiences of alcohol relapse prevention medication, views of a telephone behavioural modification intervention delivered by pharmacists and the use of Contingency Management (CM) to support acamprosate adherence following assisted alcohol withdrawal. METHODS Four focus groups were conducted within four alcohol treatment and recovery groups across England (UK), with service users with lived experience of alcohol dependence (26 participants). Semi-structured topic guide was used to explore participants' views and experiences of alcohol relapse prevention medication, a telephone behavioural modification medication intervention delivered by pharmacists, and the use of CM to support acamprosate adherence. These were audio-recorded, transcribed verbatim and thematically analysed inductively and deductively. RESULTS Four themes were identified: concerns about support and availability of alcohol relapse prevention medication; lack of knowledge and understanding about acamprosate treatment; positive perceptions of acamprosate adherence telephone support from pharmacists; and negative perceptions of CM to support acamprosate adherence. There were misunderstandings about acamprosate's mode of action and strong negative beliefs about CM. However, most were positive about pharmacists' new role to support acamprosate adherence. CONCLUSION This study highlighted challenges service users face to commence alcohol relapse prevention medication. It appears service users could benefit from a pharmacist-led telephone intervention to improve understanding about acamprosate medication, particularly, if delivered in an engaging and motivating way.
Collapse
Affiliation(s)
- Ranjita Dhital
- Corresponding author: Dr Ranjita Dhital, UCL Arts and Sciences Department, University College London, 33-35 Torrington Place, London, WC1E 7LA, UK.
| | - Rachel Coleman
- Addictions Department, National Addiction Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK,Institute for Clinical and Applied Health Research (ICAHR), Faculty of Health Sciences, University of Hull, Hull, UK
| | - Ed Day
- Institute for Mental Health, School of Psychology, University of Birmingham and Solihull Integrated Addiction Service, UK
| | - Colin Drummond
- Addictions Department, National Addiction Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Anne Lingford-Hughes
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, UK
| | - John Marsden
- Addictions Department, National Addiction Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Tom Phillips
- Addictions Department, National Addiction Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK,Institute for Clinical and Applied Health Research (ICAHR), Faculty of Health Sciences, University of Hull, Hull, UK
| | - Julia Sinclair
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - John Strang
- Addictions Department, National Addiction Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - John Weinman
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Cate Whittlesea
- UCL School of Pharmacy, University College London, London, UK
| | - Kideshini Widyaratna
- Addictions Department, National Addiction Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Kim Donoghue
- Addictions Department, National Addiction Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK,Department of Clinical, Educational and Health Psychology, University College London, London, UK
| |
Collapse
|
71
|
Waddell JT, Corbin WR, MacKinnon DP, Leeman RF, DeMartini KS, Fucito LM, Kranzler HR, O’Malley SS. Within- and between-person effects of naltrexone on the subjective response to alcohol and craving: A daily diary investigation. Alcohol Clin Exp Res 2022; 46:477-491. [PMID: 35076087 PMCID: PMC9679805 DOI: 10.1111/acer.14780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Naltrexone is an effective treatment for heavy drinking among young adults. Laboratory-based studies have shown that naltrexone dampens the subjective response to alcohol and craving. However, few studies have tested naltrexone's dynamic, within-person effects on subjective response and craving among young adults in natural drinking environments. METHODS Using daily diary data from a randomized, placebo-controlled study of naltrexone's efficacy in young adults, we examined the between-person effects of treatment condition (i.e., naltrexone vs. placebo) and medication dosage (i.e., daily, targeted, and daily + targeted) on the subjective response to alcohol and craving on drinking days. Multilevel mediation models predicted subjective response and craving from treatment condition (between-person) and medication dosage (within-person), accounting for drinking levels. All effects were disaggregated within and between persons. RESULTS At the between-person level, naltrexone directly blunted intense subjective effects (i.e., "impaired", "drunk") and indirectly blunted subjective effects through reduced drinking. Naltrexone was not associated with craving. Between-person effects were not significant after alpha correction, but their effect sizes (bs = 0.14 to 0.17) exceeded the smallest effect size of interest. At the within-person level, taking two (vs. 1) pills was associated with heavier drinking, and taking one (vs. 0) pill was associated with lighter drinking, and lighter drinking was associated with a lower subjective response and craving. Treatment condition did not moderate the within-person effects of dosing on outcomes. CONCLUSIONS Our findings suggest that the direct between-person effect of naltrexone was largest on intense subjective responses, blunting feelings of being "drunk" and "impaired". Future research using momentary (rather than daily) assessments could confirm and extend these findings.
Collapse
Affiliation(s)
| | | | | | - Robert F. Leeman
- Yale School of Medicine, Department of Psychiatry,University of Florida, Department of Health Education and Behavior
| | | | | | - Henry R. Kranzler
- University of Pennsylvania Perelman School of Medicine, Department of Psychiatry
| | | |
Collapse
|
72
|
Votaw VR, Mann K, Kranzler HR, Roos CR, Nakovics H, Witkiewitz K. Examining a brief measure and observed cutoff scores to identify reward and relief drinking profiles: Psychometric properties and pharmacotherapy response. Drug Alcohol Depend 2022; 232:109257. [PMID: 35042096 PMCID: PMC8885897 DOI: 10.1016/j.drugalcdep.2021.109257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND Precision medicine approaches attempt to reduce variability in alcohol use disorder (AUD) outcomes by identifying patient characteristics that predict response to a particular treatment. Recent work has examined the extent to which individuals with AUD may seek alcohol to enhance positive experiences (reward drinking) or relieve negative states (relief drinking) and shown that a high reward/low relief phenotype predicts naltrexone treatment response. Yet, limitations of reward/relief drinking measures may hamper efforts to translate findings to clinical practice. We sought to refine a brief measure of reward/relief drinking and develop cutoff scores to identify reward/relief subgroups that predict pharmacotherapy response. METHODS The Inventory of Drinking Situations (IDS), used in previous studies to measure reward/relief drinking, was administered to 426 participants (77% male; average age = 45.3) in a clinical trial examining naltrexone and acamprosate. RESULTS Item response theory and tests of differential item functioning across sex, age, and alcohol dependence severity were used to create a 10-item measure, titled the Reward and Relief IDS (RR-IDS). Cutoff scores on the RR-IDS for the reward/relief drinking subgroups were identified using latent profile and area under the curve analyses. The cutoff scores demonstrated good construct validity. Individuals in the high reward/low relief subgroup who received naltrexone or acamprosate had a decreased likelihood of heavy drinking (large effect sizes) versus those who received placebo. CONCLUSIONS The RR-IDS is a practical measure for identifying reward/relief subgroups and predicting pharmacotherapy response. Pending replication of these findings, the RR-IDS could be a critical precision medicine tool for prescribing AUD medications.
Collapse
Affiliation(s)
- Victoria R. Votaw
- Department of Psychology, University of New Mexico, Albuquerque, NM,Center on Alcohol, Substance use, & Addictions, University of New Mexico, Albuquerque, NM
| | - Karl Mann
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Henry R. Kranzler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Corey R. Roos
- Department of Psychiatry, Yale School of Medicine, New Haven, CT
| | - Helmut Nakovics
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Katie Witkiewitz
- Department of Psychology, University of New Mexico, Albuquerque, NM,Center on Alcohol, Substance use, & Addictions, University of New Mexico, Albuquerque, NM
| |
Collapse
|
73
|
Murphy CE, Wang RC, Montoy JC, Whittaker E, Raven M. Effect of extended-release naltrexone on alcohol consumption: a systematic review and meta-analysis. Addiction 2022; 117:271-281. [PMID: 34033183 DOI: 10.1111/add.15572] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/07/2020] [Accepted: 05/05/2021] [Indexed: 12/29/2022]
Abstract
AIMS The aims of this study were to (1) estimate the effect of extended-release naltrexone compared with placebo on alcohol consumption in patients with alcohol use disorder (AUD) and (2) conduct pre-planned subgroup analyses to test whether being abstinent when initiating treatment (lead-in abstinence) or the duration of treatment improves treatment efficacy. DESIGN Systematic review and random-effects meta-analysis of blinded randomized placebo-controlled trials reporting the effect extended-release naltrexone on alcohol consumption. SETTING Outpatient clinics. PARTICIPANTS Seven trials evaluating a total of 1500 adults with AUD receiving monthly injections of either placebo or extended-release naltrexone at doses of 150-400 mg for 2-6 months and some form of behavioral therapy. MEASUREMENTS Pooled weighted mean difference (WMD) in drinking days per month and heavy drinking days per month. FINDINGS The WMD was -2.0 [95% confidence interval (CI) = -3.4, -0.6; P = 0.03] in favor of extended-release naltrexone for drinking days per month and -1.2 (95% CI = -0.2, -2.1; P = 0.02) for heavy drinking days per month, indicating that treatment resulted in two fewer drinking days per month and 1.2 fewer heavy drinking days per month compared with placebo. Trials not requiring lead-in abstinence and those lasting longer than 3 months reported larger reductions in heavy drinking days per month; WMD -2.0 (95% CI = -3.52, -0.48; P = 0.01) and -1.9 (95% CI = -3.2, -0.5; P = 0.01), respectively. In all cases, the I2 statistics (0-7.2%) did not suggest substantial heterogeneity. CONCLUSIONS Extended-release naltrexone reduces drinking days and heavy drinking days per month compared with placebo. Reductions are larger with a longer duration of treatment.
Collapse
Affiliation(s)
- Charles E Murphy
- Department of Emergency Medicine, University of California, San Francisco, CA, USA
| | - Ralph C Wang
- Department of Emergency Medicine, University of California, San Francisco, CA, USA
| | - Juan Carlos Montoy
- Department of Emergency Medicine, University of California, San Francisco, CA, USA
| | - Evans Whittaker
- UCSF Health Sciences Library, University of California, San Francisco, CA, USA
| | - Maria Raven
- Department of Emergency Medicine, University of California, San Francisco, CA, USA.,Philip R. Lee Institute for Health Policy Studies, University of California, San Francisco, CA, USA
| |
Collapse
|
74
|
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
|
75
|
Carpenter RW, Emery NN, Meisel SN, Miranda R. Naltrexone moderates the association of alcohol use and affect among adolescent drinkers in daily life. Alcohol Clin Exp Res 2022; 46:326-337. [PMID: 34959253 PMCID: PMC8858857 DOI: 10.1111/acer.14768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Naltrexone is an efficacious medication for the treatment of alcohol use disorder in adults. As an opioid receptor antagonist, naltrexone blocks activation of the endogenous opioid system, which is involved in the affectively reinforcing properties of substance use. Few studies, however, have examined the moderating effect of naltrexone on the association between affect and alcohol use. Additionally, most existing research on naltrexone has been with adults in the human laboratory. METHOD We conducted a secondary analysis of ecological momentary assessment data from a randomized, double-blinded, placebo-controlled cross-over study that compared naltrexone (50 mg/daily) and placebo in 26 adolescents (15 to 19 years old) who exhibited problematic drinking patterns. Multilevel models tested whether naltrexone moderated associations of alcohol use with both positive and negative affect (PA, NA). RESULTS Results indicated that, during naltrexone treatment, greater estimated blood alcohol concentration (eBAC) levels were associated with greater NA further into drinking episodes. In turn, greater NA after the first drink of an episode was associated with reduced subsequent eBAC values during naltrexone treatment. Low PA was also associated with lower subsequent eBAC levels in the naltrexone condition after the first drink. CONCLUSIONS These findings support the idea that naltrexone can disrupt the association between affect and alcohol use, effects that emerge later in drinking episodes. Greater attention to the effects of naltrexone on affect and reinforcement may help to tailor psychotherapy to maximize the benefits of naltrexone. However, in the present study, as most drink reports were in the first 2 h of the drinking episode and participants reported affect only at the first three end-drink reports of a drinking episode (limiting the number of drinks reported), we had reduced power to detect effects in the continuation phase. Thus, replication of the findings is needed using a design that assesses the impact of naltrexone across the entire episode.
Collapse
Affiliation(s)
- Ryan W. Carpenter
- Department of Psychological Sciences, University of Missouri, St. Louis
| | | | - Samuel N. Meisel
- Center for Alcohol and Addiction Studies, Brown University,E. P. Bradley Hospital, Riverside, RI
| | - Robert Miranda
- Center for Alcohol and Addiction Studies, Brown University,E. P. Bradley Hospital, Riverside, RI
| |
Collapse
|
76
|
Tufvesson-Alm M, Shevchouk OT, Jerlhag E. Insight into the role of the gut-brain axis in alcohol-related responses: Emphasis on GLP-1, amylin, and ghrelin. Front Psychiatry 2022; 13:1092828. [PMID: 36699502 PMCID: PMC9868418 DOI: 10.3389/fpsyt.2022.1092828] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Alcohol use disorder (AUD) contributes substantially to global morbidity and mortality. Given the heterogenicity of this brain disease, available pharmacological treatments only display efficacy in sub-set of individuals. The need for additional treatment options is thus substantial and is the goal of preclinical studies unraveling neurobiological mechanisms underlying AUD. Although these neurobiological processes are complex and numerous, one system gaining recent attention is the gut-brain axis. Peptides of the gut-brain axis include anorexigenic peptide like glucagon-like peptide-1 (GLP-1) and amylin as well as the orexigenic peptide ghrelin. In animal models, agonists of the GLP-1 or amylin receptor and ghrelin receptor (GHSR) antagonists reduce alcohol drinking, relapse drinking, and alcohol-seeking. Moreover, these three gut-brain peptides modulate alcohol-related responses (behavioral and neurochemical) in rodents, suggesting that the alcohol reduction may involve a suppression of alcohol's rewarding properties. Brain areas participating in the ability of these gut-brain peptides to reduce alcohol-mediated behaviors/neurochemistry involve those important for reward. Human studies support these preclinical studies as polymorphisms of the genes encoding for GLP-1 receptor or the ghrelin pathway are associated with AUD. Moreover, a GLP-1 receptor agonist decreases alcohol drinking in overweight patients with AUD and an inverse GHSR agonist reduces alcohol craving. Although preclinical and clinical studies reveal an interaction between the gut-brain axis and AUD, additional studies should explore this in more detail.
Collapse
Affiliation(s)
- Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
77
|
Goodyear K, Miranda R, MacKillop J. Behavioral economic analysis of topiramate pharmacotherapy for alcohol: a placebo-controlled investigation of effects on alcohol reinforcing value and delayed reward discounting. Psychopharmacology (Berl) 2022; 239:153-161. [PMID: 34981180 PMCID: PMC8776590 DOI: 10.1007/s00213-021-06034-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/22/2021] [Indexed: 01/07/2023]
Abstract
RATIONALE Pharmacotherapies are an important clinical strategy for treating alcohol use disorder and an understanding of their functional mechanisms can inform optimal use. Behavioral economics provides a translational platform that may advance our understanding of the motivational impacts of pharmacotherapies. OBJECTIVES This secondary analysis study examined the effect of topiramate, a promising pharmacotherapy for treating alcohol use disorder, on two behavioral economic domains, the reinforcing value of alcohol (alcohol demand and alcohol-specific monetary expenditures) and delayed reward discounting (preference for smaller immediate rewards over larger delayed rewards). METHODS A double-blind randomized placebo-controlled study (n = 99) was conducted with non-treatment seeking heavy drinkers, comparing topiramate (target dose of 200 mg/day titrated for 3 weeks and remained at the target dose for 2 weeks) to matched placebo. RESULTS We found that compared to placebo, topiramate reduced the reinforcing value of alcohol, as shown by a reduction in two alcohol demand indices (intensity and Omax), money spent per week on alcohol and an almost a 50% increase in days without expenditures on alcohol from baseline. Directionally consistent patterns were also present for breakpoint and elasticity (ps = .08). No significant effects were found for delayed reward discounting. CONCLUSIONS This study provides evidence that topiramate reduces alcohol's reinforcing value as measured by alcohol demand and alcohol expenditure. More broadly, these findings support the utility of behavioral economics for understanding how medications reduce alcohol use.
Collapse
Affiliation(s)
- Kimberly Goodyear
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA.
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI, USA.
| | - Robert Miranda
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI, USA
| | - James MacKillop
- Peter Boris Centre for Addictions Research, McMaster University & St. Joseph's Healthcare Hamilton, Hamilton, ON, Canada
- Homewood Research Institute, Guelph, ON, Canada
| |
Collapse
|
78
|
Patterson Silver Wolf DA, Dulmus CN, Wilding GE, Yu J, Barczykowski AL, Shi T, Diebold JR, Harvey SJ, Tomasello NM, Linn BK. Treatment Resistant Alcohol Use Disorder. ALCOHOLISM TREATMENT QUARTERLY 2021. [DOI: 10.1080/07347324.2021.1989994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
| | | | | | - Jihnhee Yu
- Biostatistics, University at Buffalo, Buffalo, New York, USA
| | | | - Tiange Shi
- Biostatistics, University at Buffalo, Buffalo, New York, USA
| | - Josal R. Diebold
- School of Social Work, University at Buffalo, Buffalo, New York, USA
| | - Steven J. Harvey
- Integrity Partners for Behavioral Health, Ipa, Inc, Batavia, New York, USA
| | | | - Braden K. Linn
- School of Social Work, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
79
|
Holbrook OT, Molligoda B, Bushell KN, Gobrogge KL. Behavioral consequences of the downstream products of ethanol metabolism involved in alcohol use disorder. Neurosci Biobehav Rev 2021; 133:104501. [PMID: 34942269 DOI: 10.1016/j.neubiorev.2021.12.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/08/2021] [Accepted: 12/12/2021] [Indexed: 01/04/2023]
Abstract
Research concerning Alcohol Use Disorder (AUD) has previously focused primarily on either the behavioral or chemical consequences experienced following ethanol intake, but these areas of research have rarely been considered in tandem. Compared with other drugs of abuse, ethanol has been shown to have a unique metabolic pathway once it enters the body, which leads to the formation of downstream metabolites which can go on to form biologically active products. These metabolites can mediate a variety of behavioral responses that are commonly observed with AUD, such as ethanol intake, reinforcement, and vulnerability to relapse. The following review considers the preclinical and chemical research implicating these downstream products in AUD and proposes a chemobehavioral model of AUD.
Collapse
Affiliation(s)
- Otto T Holbrook
- Program in Neuroscience, Boston University, Boston, MA, 02215-2425, USA.
| | - Brandon Molligoda
- Program in Neuroscience, Boston University, Boston, MA, 02215-2425, USA.
| | - Kristen N Bushell
- Program in Neuroscience, Boston University, Boston, MA, 02215-2425, USA
| | - Kyle L Gobrogge
- Program in Neuroscience, Boston University, Boston, MA, 02215-2425, USA
| |
Collapse
|
80
|
Davy-Mendez T, Sarovar V, Levine-Hall T, Lea AN, Sterling SA, Chi FW, Palzes VA, Bryant KJ, Weisner CM, Silverberg MJ, Satre DD. Treatment for alcohol use disorder among persons with and without HIV in a clinical care setting in the United States. Drug Alcohol Depend 2021; 229:109110. [PMID: 34700145 PMCID: PMC8671330 DOI: 10.1016/j.drugalcdep.2021.109110] [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: 04/23/2021] [Revised: 09/10/2021] [Accepted: 09/23/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Alcohol use disorders (AUD) can lead to poor health outcomes. Little is known about AUD treatment among persons with HIV (PWH). In an integrated health system in Northern California, 2014-2017, we compared AUD treatment rates between PWH with AUD and persons without HIV (PWoH) with AUD. METHODS Using Poisson regression with GEE, we estimated prevalence ratios (PRs) comparing the annual probability of receiving AUD treatment (behavioral intervention or dispensed medication), adjusted for sociodemographics, psychiatric comorbidities, insurance type, and calendar year. Among PWH, we examined independent AUD treatment predictors using PRs adjusted for calendar year only. RESULTS PWH with AUD (N = 633; 93% men, median age 49) were likelier than PWoH with AUD (N = 7006; 95% men, median age 52) to have depression (38% vs. 21%) and a non-alcohol substance use disorder (SUD, 48% vs. 25%) (both P < 0.01). Annual probabilities of receiving AUD treatment were 45.4% for PWH and 34.4% for PWoH. After adjusting, there was no difference by HIV status (PR 1.02 [95% CI 0.94-1.11]; P = 0.61). Of treated PWH, 59% received only a behavioral intervention, 5% only a medication, and 36% both, vs. 67%, 4%, 30% for treated PWoH, respectively. Irrespective of HIV status, the most common medication was gabapentin. Among PWH, receiving AUD treatment was associated with having depression (PR 1.78 [1.51-2.10]; P < 0.01) and another SUD (PR 2.68 [2.20-3.27]; P < 0.01). CONCLUSIONS PWH with AUD had higher AUD treatment rates than PWoH with AUD in unadjusted but not adjusted analyses, which may be explained by higher psychiatric comorbidity burden among PWH.
Collapse
Affiliation(s)
- Thibaut Davy-Mendez
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California, 401 Parnassus Ave, San Francisco, CA 94143, USA; Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA.
| | - Varada Sarovar
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
| | - Tory Levine-Hall
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
| | - Alexandra N. Lea
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
| | - Stacy A. Sterling
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
| | - Felicia W. Chi
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
| | - Vanessa A. Palzes
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
| | - Kendall J. Bryant
- National Institute on Alcohol Abuse and Alcoholism HIV/AIDS Program, Bethesda, MD, 20892-7003, USA
| | - Constance M. Weisner
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
| | - Michael J. Silverberg
- Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
| | - Derek D. Satre
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California, San Francisco, 401 Parnassus Ave, San Francisco, CA 94143, USA,Division of Research, Kaiser Permanente Northern California, 2000 Broadway, Oakland, CA 94612, USA
| |
Collapse
|
81
|
Johansson M, Berman AH, Sinadinovic K, Lindner P, Hermansson U, Andréasson S. Effects of Internet-Based Cognitive Behavioral Therapy for Harmful Alcohol Use and Alcohol Dependence as Self-help or With Therapist Guidance: Three-Armed Randomized Trial. J Med Internet Res 2021; 23:e29666. [PMID: 34821563 PMCID: PMC8663526 DOI: 10.2196/29666] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/23/2021] [Accepted: 07/27/2021] [Indexed: 11/25/2022] Open
Abstract
Background Alcohol use is a major contributor to health loss. Many persons with harmful use or alcohol dependence do not obtain treatment because of limited availability or stigma. They may use internet-based interventions as an alternative way of obtaining support. Internet-based interventions have previously been shown to be effective in reducing alcohol consumption in studies that included hazardous use; however, few studies have been conducted with a specific focus on harmful use or alcohol dependence. The importance of therapist guidance in internet-based cognitive behavioral therapy (ICBT) programs is still unclear. Objective This trial aims to investigate the effects of a web-based alcohol program with or without therapist guidance among anonymous adult help-seekers. Methods A three-armed randomized controlled trial was conducted to compare therapist-guided ICBT and self-help ICBT with an information-only control condition. Swedish-speaking adult internet users with alcohol dependence (3 or more International Classification of Diseases, Tenth Revision criteria) or harmful alcohol use (alcohol use disorder identification test>15) were included in the study. Participants in the therapist-guided ICBT and self-help ICBT groups had 12-week access to a program consisting of 5 main modules, as well as a drinking calendar with automatic feedback. Guidance was given by experienced therapists trained in motivational interviewing. The primary outcome measure was weekly alcohol consumption in standard drinks (12 g of ethanol). Secondary outcomes were alcohol-related problems measured using the total alcohol use disorder identification test-score, diagnostic criteria for alcohol dependence and alcohol use disorder, depression, anxiety, health, readiness to change, and access to other treatments or support. Follow-up was conducted 3 (posttreatment) and 6 months after recruitment. Results During the recruitment period, from March 2015 to March 2017, 1169 participants were included. Participants had a mean age of 45 (SD 13) years, and 56.72% (663/1169) were women. At the 3-month follow-up, the therapist-guided ICBT and control groups differed significantly in weekly alcohol consumption (−3.84, 95% Cl −6.53 to −1.16; t417=2.81; P=.005; Cohen d=0.27). No significant differences were found in weekly alcohol consumption between the self-help ICBT group and the therapist-guided ICBT at 3 months, between the self-help ICBT and the control group at 3 months, or between any of the groups at the 6-month follow-up. A limitation of the study was the large number of participants who were completely lost to follow-up (477/1169, 40.8%). Conclusions In this study, a therapist-guided ICBT program was not found to be more effective than the same program in a self-help ICBT version for reducing alcohol consumption or other alcohol-related outcomes. In the short run, therapist-guided ICBT was more effective than information. Only some internet help-seekers may need a multisession program and therapist guidance to change their drinking when they use internet-based interventions. Trial Registration ClinicalTrials.gov NCT02377726; https://clinicaltrials.gov/ct2/show/NCT02377726
Collapse
Affiliation(s)
- Magnus Johansson
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Anne H Berman
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden.,Department of Psychology, Uppsala University, Uppsala, Sweden
| | - Kristina Sinadinovic
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Philip Lindner
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Ulric Hermansson
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Sven Andréasson
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
82
|
Chen G, Ghazal M, Rahman S, Lutfy K. The impact of adolescent nicotine exposure on alcohol use during adulthood: The role of neuropeptides. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:53-93. [PMID: 34801174 DOI: 10.1016/bs.irn.2021.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Nicotine and alcohol abuse and co-dependence represent major public health crises. Indeed, previous research has shown that the prevalence of alcoholism is higher in smokers than in non-smokers. Adolescence is a susceptible period of life for the initiation of nicotine and alcohol use and the development of nicotine-alcohol codependence. However, there is a limited number of pharmacotherapeutic agents to treat addiction to nicotine or alcohol alone. Notably, there is no effective medication to treat this comorbid disorder. This chapter aims to review the early nicotine use and its impact on subsequent alcohol abuse during adolescence and adulthood as well as the role of neuropeptides in this comorbid disorder. The preclinical and clinical findings discussed in this chapter will advance our understanding of this comorbid disorder's neurobiology and lay a foundation for developing novel pharmacotherapies to treat nicotine and alcohol codependence.
Collapse
Affiliation(s)
- G Chen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States; Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - M Ghazal
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - S Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, United States
| | - K Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States.
| |
Collapse
|
83
|
Biernacka JM, Coombes BJ, Batzler A, Ho AMC, Geske JR, Frank J, Hodgkinson C, Skime M, Colby C, Zillich L, Pozsonyiova S, Ho MF, Kiefer F, Rietschel M, Weinshilboum R, O'Malley SS, Mann K, Anton R, Goldman D, Karpyak VM. Genetic contributions to alcohol use disorder treatment outcomes: a genome-wide pharmacogenomics study. Neuropsychopharmacology 2021; 46:2132-2139. [PMID: 34302059 PMCID: PMC8505452 DOI: 10.1038/s41386-021-01097-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/23/2021] [Accepted: 07/08/2021] [Indexed: 01/09/2023]
Abstract
Naltrexone can aid in reducing alcohol consumption, while acamprosate supports abstinence; however, not all patients with alcohol use disorder (AUD) benefit from these treatments. Here we present the first genome-wide association study of AUD treatment outcomes based on data from the COMBINE and PREDICT studies of acamprosate and naltrexone, and the Mayo Clinic CITA study of acamprosate. Primary analyses focused on treatment outcomes regardless of pharmacological intervention and were followed by drug-stratified analyses to identify treatment-specific pharmacogenomic predictors of acamprosate and naltrexone response. Treatment outcomes were defined as: (1) time until relapse to any drinking (TR) and (2) time until relapse to heavy drinking (THR; ≥ 5 drinks for men, ≥4 drinks for women in a day), during the first 3 months of treatment. Analyses were performed within each dataset, followed by meta-analysis across the studies (N = 1083 European ancestry participants). Single nucleotide polymorphisms (SNPs) in the BRE gene were associated with THR (min p = 1.6E-8) in the entire sample, while two intergenic SNPs were associated with medication-specific outcomes (naltrexone THR: rs12749274, p = 3.9E-8; acamprosate TR: rs77583603, p = 3.1E-9). The top association signal for TR (p = 7.7E-8) and second strongest signal in the THR (p = 6.1E-8) analysis of naltrexone-treated patients maps to PTPRD, a gene previously implicated in addiction phenotypes in human and animal studies. Leave-one-out polygenic risk score analyses showed significant associations with TR (p = 3.7E-4) and THR (p = 2.6E-4). This study provides the first evidence of a polygenic effect on AUD treatment response, and identifies genetic variants associated with potentially medication-specific effects on AUD treatment response.
Collapse
Affiliation(s)
- Joanna M Biernacka
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA.
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA.
| | - Brandon J Coombes
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Anthony Batzler
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Ada Man-Choi Ho
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Jennifer R Geske
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Colin Hodgkinson
- National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, USA
| | - Michelle Skime
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Colin Colby
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Lea Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sofia Pozsonyiova
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Ming-Fen Ho
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Falk Kiefer
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | | | - Karl Mann
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ray Anton
- Medical University of South Carolina, Charleston, SC, USA
| | - David Goldman
- National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, USA
| | - Victor M Karpyak
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
84
|
Roos CR, Bold KW, Witkiewitz K, Leeman RF, DeMartini KS, Fucito LM, Corbin WR, Mann K, Kranzler HR, O’Malley SS. Reward drinking and naltrexone treatment response among young adult heavy drinkers. Addiction 2021; 116:2360-2371. [PMID: 33620746 PMCID: PMC8328878 DOI: 10.1111/add.15453] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/11/2020] [Accepted: 02/10/2021] [Indexed: 12/13/2022]
Abstract
AIMS Theory-driven, exploratory study to: (i) identify a reward drinking phenotype in young adults; (ii) evaluate this phenotype as a predictor of naltrexone response; and (iii) examine mechanisms of naltrexone in reward drinkers. DESIGN Secondary analysis of a randomized controlled trial. SETTING USA. PARTICIPANTS A total of 128 young adult (ages 18-25) heavy drinkers. INTERVENTIONS Naltrexone versus placebo. MEASUREMENTS Daily surveys assessed affect, urge, drinking, and context. The Drinking Motives Questionnaire was used to identify phenotypes based on reward (enhancement motives) and relief (coping motives) drinking. FINDINGS We identified three profiles: "Low reward/Low relief" (14.1%; low enhancement/low coping motives); "Reward drinkers" (62.2%; high enhancement/low coping motives); and "High reward/High relief" (22.7%; high enhancement/high coping motives). Among reward drinkers (versus low profile), naltrexone significantly reduced percent days drinking to intoxication (blood alcohol concentration [BAC] ≥0.08) (PDI) (d = 0.56; 95% CI [0.17, 0.96]) and percent high intensity drinking days (PHID) (8/10 drinks for women/men) (d = 0.32; 95% CI [0.01, 0.68]). Among the high reward/high relief profile drinkers (versus low profile), naltrexone reduced PHID (d = 0.69; 95% CI [0.02, 1.50]). Using profile-informed cutoffs and observed scores (for clinical applicability): (i) among cutoff-derived reward drinkers, we found a medium-to-large (d = 0.66; 95% CI [0.24, 1.16]) and small effect (d = 0.28; 95% CI [0.04, 0.72]) of naltrexone in reducing PDI and PHID, respectively; and (ii) among the cutoff-derived high reward/high relief subgroup, we found a medium-to-large effect (d = 0.63; 95% CI [0.05, 1.1]) of naltrexone in reducing PHID. Among reward drinkers (not other profiles), naltrexone reduced drinking on days a drinking event occurred by weakening the within-day association between positive affect and urges (P < 0.05). CONCLUSIONS Naltrexone has pronounced effects in reducing risky drinking among young adult reward drinkers (high reward/low relief) by reducing urges on days when individuals have higher positive affect and are exposed to a drinking event. Naltrexone also appears to reduce risky drinking among young adult high reward/high relief drinkers, but not via the same mechanism.
Collapse
Affiliation(s)
- Corey R. Roos
- Yale School of Medicine, Department of Psychiatry, New Haven, CT
| | - Krysten W. Bold
- Yale School of Medicine, Department of Psychiatry, New Haven, CT
| | - Katie Witkiewitz
- University of New Mexico, Department of Psychology, Albuquerque, NM
| | - Robert F. Leeman
- Yale School of Medicine, Department of Psychiatry, New Haven, CT,University of Florida, Department of Health Education and Behavior, Gainesville, FL
| | | | - Lisa M. Fucito
- Yale School of Medicine, Department of Psychiatry, New Haven, CT
| | | | - Karl Mann
- Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim Germany
| | - Henry R. Kranzler
- Perelman School of Medicine, University of Pennsylvania and Crescenz VAMC, Philadelphia, PA
| | | |
Collapse
|
85
|
Theory and Practice of Treatment of Concurrent Major Depressive and Alcohol Use Disorders: 7 Lessons from Clinical Practice and Research. CANADIAN JOURNAL OF ADDICTION 2021. [DOI: 10.1097/cxa.0000000000000125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
86
|
Trends in the Use of Naltrexone for Addiction Treatment among Alcohol Use Disorder Admissions in U.S. Substance Use Treatment Facilities. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18168884. [PMID: 34444639 PMCID: PMC8394149 DOI: 10.3390/ijerph18168884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 01/12/2023]
Abstract
Background: Naltrexone, a medication for addiction treatment (MAT), is an FDA-approved medication recommended for the treatment of alcohol use disorder (AUD). Despite the high prevalence of AUD and efficacy of naltrexone, only a small percentage of individuals with AUD receive treatment. Objectives: To identify trends for the prescription of naltrexone in AUD admissions in substance use treatment centers across the U.S. Methods: Data from the 2000–2018 U.S. Treatment Episode Data Set: Admissions (TEDS-A) were used in temporal trend analysis of naltrexone prescription in admissions that only used alcohol. Data from the 2019 National Survey of Substance Abuse Treatment Services (N-SSATS) were also used to characterize medication use among AUD clients across different treatment service settings. Results: Treatment of AUD with naltrexone was 0.49% in 2000 and tripled from 0.53% in 2015 to 1.64% in 2018 in AUD admissions (p < 0.0001 for the Cochran–Armitage trend test). Women, middle-aged adults, and admissions for clients living in the Northeast U.S. were more likely to be prescribed naltrexone than their respective counterparts, as were admissions with prior treatment episodes and referrals through alcohol/drug use care providers, who paid for treatment primarily through private insurance, used alcohol daily in the month prior to admission, and waited 1–7 days to enter treatment. Naltrexone was more commonly prescribed by AUD admissions compared to acamprosate and disulfiram and was more frequently prescribed in residential and outpatient services as opposed to hospital inpatient services. Conclusions: Naltrexone remains underutilized for AUD, and factors that influence prescription of medication are multifaceted. This study may contribute to the creation of effective interventions aimed at reducing naltrexone disparities for AUD.
Collapse
|
87
|
Ting PS, Gurakar A, Wheatley J, Chander G, Cameron AM, Chen PH. Approaching Alcohol Use Disorder After Liver Transplantation for Acute Alcoholic Hepatitis. Clin Liver Dis 2021; 25:645-671. [PMID: 34229846 PMCID: PMC8264137 DOI: 10.1016/j.cld.2021.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Severe alcoholic hepatitis portends a high risk of mortality without liver transplantation. Transplant outcomes in patients with severe alcoholic hepatitis exhibit a strong inverse association with post-transplant alcohol relapse. The ingredients most central to ameliorating alcohol relapse risk may include destigmatized post-transplant alcohol monitoring, a nonpunitive clinician-patient partnership, and multimodal therapies to maintain abstinence and mitigate high-risk drinking. We here review the core principles of post-liver transplant management specific to alcohol use disorder.
Collapse
Affiliation(s)
- Peng-Sheng Ting
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, 1830 East Monument Street, Suite 431, Baltimore, MD 21287, USA
| | - Ahmet Gurakar
- Liver Transplant, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Suite 918, Baltimore, MD 21205, USA.
| | - Jason Wheatley
- Department of Social Work, Johns Hopkins Hospital, 600 North Wolfe Street, Carnegie Suite 100, Baltimore, MD 21287, USA
| | - Geetanjali Chander
- Division of General Internal Medicine, Johns Hopkins University School of Medicine, 1830 East Monument Street, Room 8047A, Baltimore, MD 21287, USA
| | - Andrew M Cameron
- Division of Liver Transplant Surgery, Department of Surgery, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 765, Baltimore, MD 21205, USA
| | - Po-Hung Chen
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, 1830 East Monument Street, Suite 429, Baltimore, MD 21287, USA
| |
Collapse
|
88
|
Heikkinen M, Taipale H, Tanskanen A, Mittendorfer-Rutz E, Lähteenvuo M, Tiihonen J. Real-world effectiveness of pharmacological treatments of alcohol use disorders in a Swedish nation-wide cohort of 125 556 patients. Addiction 2021; 116:1990-1998. [PMID: 33394527 PMCID: PMC8359433 DOI: 10.1111/add.15384] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/06/2020] [Accepted: 12/16/2020] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND AIM Pharmacotherapy for alcohol use disorder (AUD) is recommendable, but under-used, possibly due to deficient knowledge of medications. This study aimed to investigate the real-world effectiveness of approved pharmacological treatments (disulfiram, acamprosate, naltrexone and nalmefene) of AUD. DESIGN A nation-wide, register-based cohort study. SETTING Sweden. PARTICIPANTS All residents aged 16-64 years living in Sweden with registered first-time treatment contact due to AUD from July 2006 to December 2016 (n = 125 556, 62.5% men) were identified from nation-wide registers. MEASUREMENTS The main outcome was hospitalization due to AUD. The secondary outcomes were hospitalization due to any cause, alcohol-related somatic causes, as well as work disability (sickness absence or disability pension), and death. Mortality was analysed with between-individual analysis using a traditional multivariate-adjusted Cox hazards regression model. Recurrent outcomes, such as hospitalization-based events and work disability, were analysed with within-individual analyses to eliminate selection bias. FINDINGS Naltrexone combined with acamprosate [hazard ratio (HR) = 0.74; 95% confidence interval (CI) = 0.61-0.89], combined with disulfiram (HR = 0.76, 95% CI = 0.60-0.96) and as monotherapy (HR = 0.89, 95% CI = 0.81-0.97) was associated with a significantly lower risk of AUD-hospitalization compared with no use of AUD medication. Similar results were found for risk of hospitalization due to any cause. Benzodiazepine use and acamprosate monotherapy were associated with an increased risk of AUD-hospitalization (HR = 1.18, 95% CI = 1.14-1.22 and HR = 1.10, 95% CI = 1.04-1.17, respectively). No statistically significant effects were found for work disability or mortality. CONCLUSIONS Naltrexone as monotherapy and when combined with disulfiram and acamprosate appears to be associated with lower risk of hospitalization due to any and alcohol-related causes, compared with no use of alcohol use disorder (AUD) medication. Acamprosate monotherapy and benzodiazepine use appear to be associated with increased risk of AUD-associated hospitalization.
Collapse
Affiliation(s)
- Milja Heikkinen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland.,Department of Clinical Neuroscience, Division of Insurance Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Heidi Taipale
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland.,Department of Clinical Neuroscience, Division of Insurance Medicine, Karolinska Institutet, Stockholm, Sweden.,School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Antti Tanskanen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland.,Department of Clinical Neuroscience, Division of Insurance Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ellenor Mittendorfer-Rutz
- Department of Clinical Neuroscience, Division of Insurance Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Markku Lähteenvuo
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Jari Tiihonen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland.,Department of Clinical Neuroscience, Karolinska Institutet and Centre for Psychiatry Research, Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| |
Collapse
|
89
|
Abstract
The misuse of alcohol in the United States continues to take a large toll on society, resulting in the deaths of about 88,000 Americans per year. Moreover, it is estimated that nearly 14.6 million Americans currently meet diagnostic criteria for current alcohol use disorder (AUD). However, very few individuals receive treatment, with an even smaller portion receiving medications approved by the U.S. Food and Drug Administration (FDA) for the treatment of AUD, despite scientifically rigorous evidence showing the benefits of combining medication approved for treating AUD with evidence-based behavioral therapy. These benefits include higher rates of abstinence and less risk of relapse to heavy drinking, with associated improvements in medical and mental health and in quality of life. This review provides an overview of FDA-approved medications and "off-label" drugs for the treatment of AUD. The article emphasizes that AUD medical advice and prescription recommendations should come from professionals with training in the treatment of AUD and that treatment plans should consider medication in conjunction with evidence-based behavioral therapy. Finally, this review notes the limited number of medications available and the continued need for the development of new pharmacotherapies to optimize AUD recovery goals.
Collapse
Affiliation(s)
- Barbara J Mason
- Pearson Center for Alcoholism and Addiction Research, Department of Molecular Medicine, Scripps Research Institute, La Jolla, California. Center for Human Development, University of California, San Diego, La Jolla, California
| | - Charles J Heyser
- Pearson Center for Alcoholism and Addiction Research, Department of Molecular Medicine, Scripps Research Institute, La Jolla, California. Center for Human Development, University of California, San Diego, La Jolla, California
| |
Collapse
|
90
|
Abstract
Alcohol use disorder (AUD) and family functioning are inextricably bound, and families are impacted negatively by AUD, but families show substantial improvements with AUD recovery. Family members can successfully motivate a person with AUD to initiate changes in drinking or to seek AUD treatment. During recovery, family members can provide active support for recovery. Several couple- or family-involved treatments for AUD have been developed and tested in rigorous efficacy trials. Efficacious treatments based in family systems theory or cognitive behavioral approaches focus on the concerned family member alone, or they engage the couple or family as a unit in the treatment. However, most treatments have been studied in fairly homogeneous, heterosexual, White, non-Hispanic populations, limiting the potential generalizability of these treatments. Substantial gaps remain in our understanding of family processes associated with the initiation and maintenance of AUD recovery among adults. This review outlines the existing literature and describes opportunities for future research to address knowledge gaps in understanding the mechanisms by which these treatments are efficacious, use of family-based treatments with diverse populations, integration of pharmacotherapies with family-involved treatment, role of families in recovery-oriented systems of care, and how to improve treatment development and dissemination.
Collapse
Affiliation(s)
- Barbara S. McCrady
- Department of Psychology and Center on Alcoholism, Substance Abuse, and Addictions, University of New Mexico, Albuquerque, New Mexico
| | - Julianne C. Flanagan
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, South Carolina,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
91
|
Affiliation(s)
- Barbara J Mason
- Pearson Center for Alcoholism and Addiction Research, Department of Molecular Medicine, Scripps Research Institute, La Jolla, Calif
| |
Collapse
|
92
|
Balhara YPS, Tarwani J. Is there a case for inclusion of medicines used to manage alcohol use disorder in the World Health Organization Model List of Essential Medicines? Indian J Psychiatry 2021; 63:297-298. [PMID: 34211226 PMCID: PMC8221208 DOI: 10.4103/psychiatry.indianjpsychiatry_305_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/06/2020] [Accepted: 02/17/2021] [Indexed: 11/04/2022] Open
Affiliation(s)
- Yatan Pal Singh Balhara
- Department of Psychiatry, National Drug Dependence Treatment Centre, All India Institute of Medical Sciences, New Delhi, India. E-mail:
| | - Jatin Tarwani
- Department of Psychiatry, National Drug Dependence Treatment Centre, All India Institute of Medical Sciences, New Delhi, India. E-mail:
| |
Collapse
|
93
|
Johansson M, Sinadinovic K, Gajecki M, Lindner P, Berman AH, Hermansson U, Andréasson S. Internet-based therapy versus face-to-face therapy for alcohol use disorder, a randomized controlled non-inferiority trial. Addiction 2021; 116:1088-1100. [PMID: 32969541 PMCID: PMC8247312 DOI: 10.1111/add.15270] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/08/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Most people with alcohol use disorder (AUD) are never treated. Internet-based interventions are effective in reducing alcohol consumption and could help to overcome some of the barriers to people not seeking or receiving treatment. The aim of the current study was to compare internet-delivered and face-to-face treatment among adult users with AUD. DESIGN Randomized controlled non-inferiority trial with a parallel design, comparing internet-delivered cognitive-behavioural therapy (ICBT) (n = 150) with face-to-face CBT (n = 151), at 3- and 6-month follow-ups. SETTING A specialized clinic for people with AUD in Stockholm, Sweden. Participants were recruited between 8 December 2015 and 5 January 2018. PARTICIPANTS A total of 301 patients [mean age 50 years, standard deviation (SD) = 12.3] with AUD, of whom 115 (38%) were female and 186 (62%) were male. INTERVENTION AND COMPARATOR Participants were randomized in blocks of 20 at a ratio of 1 : 1 to five modules of therapist-guided ICBT or to five modules of face-to-face CBT, delivered over a 3-month period. The same treatment material and the same therapists were used in both groups. MEASUREMENTS The primary outcome was standard drinks of alcohol consumed during the previous week at 6-month follow-up, analysed according to intention-to-treat. The pre-specified non-inferiority limit was five standard drinks of alcohol and d = 0.32 for secondary outcomes. RESULTS The difference in alcohol consumption between the internet and the face-to-face group was non-inferior in the intention-to-treat analysis of data from the 6-month follow-up [internet = 12.33 and face-to-face = 11.43, difference = 0.89, 95% confidence interval (CI) = -1.10 to 2.88]. The secondary outcome, Alcohol Use Disorder Identification Test score, failed to show non-inferiority of internet compared with face-to-face in the intention-to-treat analysis at 6-month follow-up (internet = 12.26 and face-to-face = 11.57, d = 0.11, 95% CI = -0.11 to 0.34). CONCLUSIONS Internet-delivered treatment was non-inferior to face-to-face treatment in reducing alcohol consumption among help-seeking patients with alcohol use disorder but failed to show non-inferiority on some secondary outcomes.
Collapse
Affiliation(s)
- Magnus Johansson
- Department of Global Public HealthKarolinska InstitutetStockholmSweden
| | - Kristina Sinadinovic
- Centre for Psychiatry Research, Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
- Stockholm Health Care ServicesStockholm County CouncilStockholmSweden
| | - Mikael Gajecki
- Centre for Psychiatry Research, Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
- Stockholm Health Care ServicesStockholm County CouncilStockholmSweden
| | - Philip Lindner
- Centre for Psychiatry Research, Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
- Stockholm Health Care ServicesStockholm County CouncilStockholmSweden
| | - Anne H. Berman
- Centre for Psychiatry Research, Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
- Stockholm Health Care ServicesStockholm County CouncilStockholmSweden
- Department of PsychologyUppsala UniversityUppsalaSweden
| | - Ulric Hermansson
- Centre for Psychiatry Research, Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
- Stockholm Health Care ServicesStockholm County CouncilStockholmSweden
| | - Sven Andréasson
- Department of Global Public HealthKarolinska InstitutetStockholmSweden
| |
Collapse
|
94
|
Villavicencio-Tejo F, Flores-Bastías O, Marambio-Ruiz L, Pérez-Reytor D, Karahanian E. Fenofibrate (a PPAR-α Agonist) Administered During Ethanol Withdrawal Reverts Ethanol-Induced Astrogliosis and Restores the Levels of Glutamate Transporter in Ethanol-Administered Adolescent Rats. Front Pharmacol 2021; 12:653175. [PMID: 33959021 PMCID: PMC8093785 DOI: 10.3389/fphar.2021.653175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/31/2021] [Indexed: 01/11/2023] Open
Abstract
High-ethanol intake induces a neuroinflammatory response, which has been proposed as responsible for the maintenance of chronic ethanol consumption. Neuroinflammation decreases glutamate transporter (GLT-1) expression, increasing levels of glutamate that trigger dopamine release at the corticolimbic reward areas, driving long-term drinking behavior. The activation of peroxisome proliferator-activated receptor alpha (PPARα) by fibrates inhibits neuroinflammation, in models other than ethanol consumption. However, the effect of fibrates on ethanol-induced neuroinflammation has not yet been studied. We previously reported that the administration of fenofibrate to ethanol-drinking rats decreased ethanol consumption. Here, we studied whether fenofibrate effects are related to a decrease in ethanol-induced neuroinflammation and to the normalization of the levels of GLT-1. Rats were administered ethanol on alternate days for 4 weeks (2 g/kg/day). After ethanol withdrawal, fenofibrate was administered for 14 days (50 mg/kg/day) and the levels of glial fibrillary acidic protein (GFAP), phosphorylated NF-κB-inhibitory protein (pIκBα) and GLT-1, were quantified in the prefrontal cortex, hippocampus, and hypothalamus. Ethanol treatment increased the levels of GFAP in the hippocampus and hypothalamus, indicating a clear astrocytic activation. Similarly, ethanol increased the levels of pIκBα in the three areas. The administration of fenofibrate decreased the expression of GFAP and pIκBα in the three areas. These results indicate that fenofibrate reverts both astrogliosis and NF-κB activation. Finally, ethanol decreased GLT-1 expression in the prefrontal cortex and hippocampus. Fenofibrate normalized the levels of GLT-1 in both areas, suggesting that its effect in reducing ethanol consumption could be due to the normalization of glutamatergic tone.
Collapse
Affiliation(s)
| | - Osvaldo Flores-Bastías
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Lucas Marambio-Ruiz
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Diliana Pérez-Reytor
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Eduardo Karahanian
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
95
|
Pharmacotherapy of substance use disorders in the neuroscience-based nomenclature (NbN). Therapie 2021; 76:127-136. [DOI: 10.1016/j.therap.2020.12.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022]
|
96
|
Ray LA, Grodin EN, Leggio L, Bechtholt AJ, Becker H, Ewing SWF, Jentsch JD, King AC, Mason BJ, O’Malley S, MacKillop J, Heilig M, Koob GF. The future of translational research on alcohol use disorder. Addict Biol 2021; 26:e12903. [PMID: 32286721 PMCID: PMC7554164 DOI: 10.1111/adb.12903] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 03/06/2020] [Accepted: 03/13/2020] [Indexed: 12/13/2022]
Abstract
In March 2019, a scientific meeting was held at the University of California, Los Angeles (UCLA) Luskin Center to discuss approaches to expedite the translation of neurobiological insights to advances in the treatment of alcohol use disorder (AUD). A guiding theme that emerged was that while translational research in AUD is clearly a challenge, it is also a field ripe with opportunities. Herein, we seek to summarize and disseminate the recommendations for the future of translational AUD research using four sections. First, we briefly review the current landscape of AUD treatment including the available evidence-based treatments and their uptake in clinical settings. Second, we discuss AUD treatment development efforts from a translational science viewpoint. We review current hurdles to treatment development as well as opportunities for mechanism-informed treatment. Third, we consider models of translational science and public health impact. Together, these critical insights serve as the bases for a series of recommendations and future directions. Towards the goal of improving clinical care and population health for AUD, scientists are tasked with bolstering the clinical applicability of their research findings so as to expedite the translation of knowledge into patient care.
Collapse
Affiliation(s)
- Lara A. Ray
- University of California Los Angeles, Department of Psychology, Los Angeles, CA, USA
| | - Erica N. Grodin
- University of California Los Angeles, Department of Psychology, Los Angeles, CA, USA
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research and National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Bethesda, MD; Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD; Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
| | - Anita J. Bechtholt
- National Institute on Alcohol Abuse and Alcoholism, Division of Treatment and Recovery Research, National Institutes of Health, Bethesda, MD, USA
| | - Howard Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Science; Department of Neuroscience, Medical University of South Carolina; Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Sarah W. Feldstein Ewing
- Oregon Health and Science University, Department of Child and Adolescent Psychiatry, Portland, Oregon, USA
| | - J. David Jentsch
- Binghamton University, Department of Psychology, Binghamton, NY, USA
| | - Andrea C. King
- University of Chicago, Department of Psychiatry and Behavioral Neuroscience, Chicago, IL, USA
| | - Barbara J. Mason
- The Scripps Research Institute, Pearson Center for Alcoholism and Addiction Research, La Jolla, CA, USA
| | | | - James MacKillop
- McMaster University and St. Joseph’s Healthcare Hamilton, Peter Boris Center for Addictions Research, Hamilton, ON, CAN
| | - Markus Heilig
- Linkoping University, Center for Social and Affective Neuroscience, Linkoping, Sweden
| | - George F. Koob
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA; National Institute on Drug Abuse, Baltimore, MD, USA
| |
Collapse
|
97
|
Ray LA, Du H, Green R, Roche DJO, Bujarski S. Do behavioral pharmacology findings predict clinical trial outcomes? A proof-of-concept in medication development for alcohol use disorder. Neuropsychopharmacology 2021; 46:519-527. [PMID: 33235284 PMCID: PMC8026961 DOI: 10.1038/s41386-020-00913-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 12/21/2022]
Abstract
Behavioral pharmacology paradigms have been used for early efficacy testing of novel compounds for alcohol use disorder (AUD). However, the degree to which early efficacy in the human laboratory predicts clinical efficacy remains unclear. To address this gap in the literature we employed a novel meta-analytic approach. We searched the literature for medications tested for AUD using both behavioral pharmacology (i.e., alcohol administration) and randomized clinical trials (RCTs). For behavioral pharmacology, we computed medication effects on alcohol-induced stimulation, sedation, and craving during the alcohol administration (k = 51 studies, 24 medications). For RCTs, we computed medication effects on any drinking and heavy drinking (k = 118 studies, 17 medications). We used medication as the unit of analysis and applied the Williamson-York bivariate weighted least squares estimation to preserve the errors in both the independent and dependent variables. Results, with correction for publication bias, revealed a significant and positive relationship between medication effects on alcohol-induced stimulation (β = 1.18 p < 0.05), sedation (β = 2.38, p < 0.05), and craving (β = 3.28, p < 0.001) in the laboratory, and drinking outcomes in RCTs, such that medications that reduced stimulation, sedation, and craving during the alcohol administration were associated with better clinical outcomes. A leave-one-out Monte Carlo analysis examined the predictive utility of these laboratory endpoints for each medication. The observed clinical effect size was within one standard deviation of the mean predicted effect size for all but three pharmacotherapies. This proof-of-concept study demonstrates that behavioral pharmacology endpoints of alcohol-induced stimulation, sedation, and craving track medication effects from the human laboratory to clinical trial outcomes. These results apply to alcohol administration phenotypes and may be especially useful to medications for which the mechanisms of action involve alterations in subjective responses to alcohol (e.g., antagonist medication). These methods and results can be applied to a host of clinical questions and can streamline the process of screening novel compounds for AUD. For instance, this approach can be used to quantify the predictive utility of cue-reactivity screening models and even preclinical models of medication development.
Collapse
Affiliation(s)
- Lara A. Ray
- grid.19006.3e0000 0000 9632 6718University of California, Department of Psychology, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718University of California, Department of Psychiatry and Biobehavioral Sciences, Los Angeles, CA USA
| | - Han Du
- grid.19006.3e0000 0000 9632 6718University of California, Department of Psychology, Los Angeles, CA USA
| | - ReJoyce Green
- grid.19006.3e0000 0000 9632 6718University of California, Department of Psychology, Los Angeles, CA USA
| | - Daniel J. O. Roche
- grid.19006.3e0000 0000 9632 6718University of California, Department of Psychology, Los Angeles, CA USA
| | - Spencer Bujarski
- grid.19006.3e0000 0000 9632 6718University of California, Department of Psychology, Los Angeles, CA USA
| |
Collapse
|
98
|
Abstract
Bislang sind nur wenige Medikamente zur pharmakologischen Rückfallprophylaxe der Alkoholabhängigkeit zugelassen. Neben dem in Deutschland nicht mehr vertriebenen Disulfiram sind es die Opioidantagonisten Naltrexon und Nalmefen sowie das vermutlich über glutamaterge Neurone wirkende Acamprosat. Baclofen und γ‑Hydroxybutyrat (GHB) sind in einzelnen Ländern zugelassen. Wirkstoffe wie z. B. Vareniclin, Gabapentin und Topiramat können für die Rückfallprophylaxe der Alkoholabhängigkeit von Interesse sein, jedoch ist bislang keine Zulassung erfolgt. Vor dem Hintergrund der zur Revision anstehenden S3-Leitlinie zur Diagnose und Behandlung alkoholbezogener Störungen wird der heutige Kenntnisstand zur Pharmakotherapie der Alkoholabhängigkeit dargestellt.
Collapse
|
99
|
Choudhary NS, Saraf N, Mehrotra S, Saigal S, Soin AS. Recidivism in Liver Transplant Recipients for Alcohol-related Liver Disease. J Clin Exp Hepatol 2021; 11:387-396. [PMID: 33994719 PMCID: PMC8103326 DOI: 10.1016/j.jceh.2020.08.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/30/2020] [Indexed: 12/12/2022] Open
Abstract
Liver transplantation (LT) is the only cure for patients with end-stage liver disease, which offers good long-term survival. The long-term issues after LT affecting survival are cardiovascular disease, chronic kidney disease, de novo malignancies, recurrence of original disease and immunological causes. Alcoholic-related liver disease (ALD) is one of the most common indications for LT worldwide including India. LT for ALD is associated with several unique challenges as compared with other etiologies. Long-term survival after LT in patients with ALD is affected by recidivism. Various studies have shown different predictors of relapse; the main predictors of relapse are pretransplant abstinence, psychiatric comorbidities, and lack of social support. Although several risk scores have been proposed, these scores are not validated. Studies with active involvement of psychiatrist have shown lower relapse rates. The relapse prevention strategy for reducing likelihood and severity of relapse after initial cessation of alcohol uses a combination of pharmacotherapy and cognitive behavioral approach (identifying and addressing high-risk situations for relapse).
Collapse
Affiliation(s)
- Narendra S. Choudhary
- Institute of Liver Transplantation and Regenerative Medicine, Medanta the Medicity, Gurgaon, Delhi (NCR), India
| | - Neeraj Saraf
- Institute of Liver Transplantation and Regenerative Medicine, Medanta the Medicity, Gurgaon, Delhi (NCR), India,Address for correspondence: Medanta Institute of Liver Transplantation and Regenerative Medicine, Medanta The Medicity hospital, sector 38, Gurgaon, Delhi (NCR), India.
| | - Saurabh Mehrotra
- Department of Mental Health, Medanta the Medicity, Gurgaon, Delhi (NCR), India
| | - Sanjiv Saigal
- Institute of Liver Transplantation and Regenerative Medicine, Medanta the Medicity, Gurgaon, Delhi (NCR), India
| | - Arvinder S. Soin
- Institute of Liver Transplantation and Regenerative Medicine, Medanta the Medicity, Gurgaon, Delhi (NCR), India
| |
Collapse
|
100
|
Koob GF. Drug Addiction: Hyperkatifeia/Negative Reinforcement as a Framework for Medications Development. Pharmacol Rev 2021; 73:163-201. [PMID: 33318153 PMCID: PMC7770492 DOI: 10.1124/pharmrev.120.000083] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Compulsive drug seeking that is associated with addiction is hypothesized to follow a heuristic framework that involves three stages (binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation) and three domains of dysfunction (incentive salience/pathologic habits, negative emotional states, and executive function, respectively) via changes in the basal ganglia, extended amygdala/habenula, and frontal cortex, respectively. This review focuses on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the addiction cycle. Hyperkatifeia provides an additional source of motivation for compulsive drug seeking via negative reinforcement. Negative reinforcement reflects an increase in the probability of a response to remove an aversive stimulus or drug seeking to remove hyperkatifeia that is augmented by genetic/epigenetic vulnerability, environmental trauma, and psychiatric comorbidity. Neurobiological targets for hyperkatifeia in addiction involve neurocircuitry of the extended amygdala and its connections via within-system neuroadaptations in dopamine, enkephalin/endorphin opioid peptide, and γ-aminobutyric acid/glutamate systems and between-system neuroadaptations in prostress corticotropin-releasing factor, norepinephrine, glucocorticoid, dynorphin, hypocretin, and neuroimmune systems and antistress neuropeptide Y, nociceptin, endocannabinoid, and oxytocin systems. Such neurochemical/neurocircuitry dysregulations are hypothesized to mediate a negative hedonic set point that gradually gains allostatic load and shifts from a homeostatic hedonic state to an allostatic hedonic state. Based on preclinical studies and translational studies to date, medications and behavioral therapies that reset brain stress, antistress, and emotional pain systems and return them to homeostasis would be promising new targets for medication development. SIGNIFICANCE STATEMENT: The focus of this review is on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the drug addiction cycle and a driving force for negative reinforcement in addiction. Medications and behavioral therapies that reverse hyperkatifeia by resetting brain stress, antistress, and emotional pain systems and returning them to homeostasis would be promising new targets for medication development.
Collapse
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|