1
|
Schacht JP, Ray LA, Miranda R, Falk DE, Ryan ML, Sakai JT, Miotto K, Chun T, Scott C, Ransom J, Alsharif N, Ito M, Litten RZ. Effects of a novel GABA-B positive allosteric modulator, ASP8062, on alcohol cue-elicited craving and naturalistic alcohol consumption in a multisite randomized, double-blind, placebo-controlled trial. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:2352-2363. [PMID: 39623527 DOI: 10.1111/acer.15468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/22/2024] [Accepted: 09/30/2024] [Indexed: 12/11/2024]
Abstract
BACKGROUND The γ-aminobutyric acid-B (GABAB) receptor is a promising target for the development of new medications to treat alcohol use disorder (AUD). The GABAB agonist baclofen has been reported to reduce alcohol consumption but is associated with some undesirable side effects, including sedation. ASP8062 is a novel compound that acts as a positive allosteric modulator at the GABAB receptor and may be more tolerable than baclofen. This proof-of-concept human laboratory clinical trial evaluated the safety profile of ASP8062 and tested its effects on cue-elicited alcohol craving and alcohol use among treatment-seeking individuals with AUD. METHODS This double-blind, randomized, multisite trial tested the effect of ASP8062 (25 mg once daily), relative to placebo, on alcohol cue-elicited craving in a laboratory setting and alcohol consumption, craving, mood, sleep, cigarette smoking, and alcohol-related consequences in the natural environment over a 6-week treatment period. Participants were 60 individuals (26 females and 34 males) with moderate or severe AUD. RESULTS ASP8062, relative to placebo, was well tolerated, and there were no adverse events (AEs) that significantly differed between treatment groups. Most AEs were mild/moderate, and there were no serious AEs among individuals treated with ASP8062. However, ASP8062 did not attenuate alcohol cue-elicited craving compared with placebo. Moreover, exploratory analyses indicated that ASP8062, relative to placebo, did not significantly affect alcohol consumption, naturalistic alcohol craving, mood, sleep, cigarette smoking, or alcohol-related negative consequences during the 6-week treatment period. CONCLUSIONS Although ASP8062 was well tolerated with no serious AEs, the novel compound did not significantly dampen alcohol cue-elicited craving or improve other AUD-related outcome measures. These data indicate positive allosteric modulation of the GABAB receptor at the dose evaluated here may not blunt alcohol cue-elicited craving, and preliminary drinking outcome data suggest it may not be an efficacious treatment strategy for AUD.
Collapse
Affiliation(s)
- Joseph P Schacht
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Lara A Ray
- Department of Psychology, University of California, Los Angeles, California, USA
| | - Robert Miranda
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Daniel E Falk
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Megan L Ryan
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Joseph T Sakai
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Karen Miotto
- Department of Psychology, University of California, Los Angeles, California, USA
| | - Thomas Chun
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - Charles Scott
- Fast-Track Drugs & Biologics, LLC, Poolesville, Maryland, USA
| | - Janet Ransom
- Fast-Track Drugs & Biologics, LLC, Poolesville, Maryland, USA
| | - Nour Alsharif
- Astellas Pharma Global Development Inc., Northbrook, Illinois, USA
| | - Mototsugu Ito
- Astellas Pharma Global Development Inc., Northbrook, Illinois, USA
| | - Raye Z Litten
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Tareen K, Clifton EG, Perumalswami P, Mellinger JL, Winder GS. Treatment of Alcohol Use Disorder: Behavioral and Pharmacologic Therapies. Clin Liver Dis 2024; 28:761-778. [PMID: 39362720 PMCID: PMC11450263 DOI: 10.1016/j.cld.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The prevalence of alcohol use disorder (AUD) has significantly increased over the last decade, leading to an increase in alcohol-associated liver disease (ALD) rates worldwide. Despite this prominence, AUD in ALD remains undertreated and carries significant implications in the progression to end-stage ALD and increased mortality. In efforts to bridge this gap, interprofessional and integrated AUD treatment is necessary for patients with ALD to ensure early detection and an appropriately targeted level of care. Although pharmacotherapy, psychotherapy, and psychosocial interventions independently play a role in treating AUD, a combination of these evidence-based modalities often results in lasting change.
Collapse
Affiliation(s)
- Kinza Tareen
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA.
| | - Erin G Clifton
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Ponni Perumalswami
- Gastroenterology Section, Veterans Affairs, Ann Arbor Healthcare System, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jessica L Mellinger
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Gerald Scott Winder
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA; Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
3
|
Lei J, Zhang P, Li T, Cui C, Li M, Yang X, Peng X, Ren K, Yang J, Shi Y, Luo G, Yao Y, Tian B. Alternating bilateral sensory stimulation alleviates alcohol-induced conditioned place preference via a superior colliculus-VTA circuit. Cell Rep 2024; 43:114383. [PMID: 38923461 DOI: 10.1016/j.celrep.2024.114383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/01/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Alcohol is the most widely used addictive substance, potentially leading to brain damage and genetic abnormalities. Despite its prevalence and associated risks, current treatments have yet to identify effective methods for reducing cravings and preventing relapse. In this study, we find that 4-Hz alternating bilateral sensory stimulation (ABS) effectively reduces ethanol-induced conditioned place preference (CPP) in male mice, while 4-Hz flash light does not exhibit therapeutic effects. Whole-brain c-Fos mapping demonstrates that 4-Hz ABS triggers notable activation in superior colliculus GABAergic neurons (SCGABA). SCGABA forms monosynaptic connections with ventral tegmental area dopaminergic neurons (VTADA), which is implicated in ethanol-induced CPP. Bidirectional chemogenetic manipulation of SC-VTA circuit either replicates or blocks the therapeutic effects of 4-Hz ABS on ethanol-induced CPP. These findings elucidate the role of SC-VTA circuit for alleviating ethanol-related CPP by 4-Hz ABS and point to a non-drug and non-invasive approach that might have potential for treating alcohol use disorder.
Collapse
Affiliation(s)
- Jie Lei
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Pei Zhang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China; Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei, P.R. China.
| | - Tongxia Li
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Chi Cui
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Ming Li
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Xueke Yang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Xiang Peng
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Kun Ren
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Jian Yang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Yulong Shi
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Gangan Luo
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Yibo Yao
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Bo Tian
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China; School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, P.R. China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China; Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei, P.R. China.
| |
Collapse
|
4
|
Cieslik-Starkiewicz A, Noworyta K, Solich J, Korlatowicz A, Faron-Górecka A, Rygula R. Trait sensitivity to positive feedback is a predisposing factor for several aspects of compulsive alcohol drinking in male rats: behavioural, physiological, and molecular correlates. Psychopharmacology (Berl) 2024; 241:33-47. [PMID: 37682294 PMCID: PMC10774643 DOI: 10.1007/s00213-023-06460-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/27/2023] [Indexed: 09/09/2023]
Abstract
INTRODUCTION Alcohol use disorder (AUD) is one of the most common psychiatric disorders and a leading cause of mortality worldwide. While the pathophysiology underlying AUD is relatively well known, the cognitive mechanisms of an individual's susceptibility to the development of alcohol dependence remain poorly understood. In this study, we investigated the theoretical claim that sensitivity to positive feedback (PF), as a stable and enduring behavioural trait, can predict individual susceptibility to the acquisition and maintenance of alcohol-seeking behaviour in rats. METHODS Trait sensitivity to PF was assessed using a series of probabilistic reversal learning tests. The escalation of alcohol intake in rats was achieved by applying a mix of intermittent free access and instrumental paradigms of alcohol drinking. The next steps included testing the influence of sensitivity to PF on the acquisition of compulsive alcohol-seeking behaviour in the seeking-taking punishment task, measuring motivation to seek alcohol, and comparing the speed of extinction and reinstatement of alcohol-seeking after a period of abstinence between rats expressing trait insensitivity and sensitivity to PF. Finally, trait differences in the level of stress hormones and in the expression of genes and proteins in several brain regions of interest were measured to identify potential physiological and neuromolecular mechanisms of the observed interactions. RESULTS We showed that trait sensitivity to PF in rats determines the level of motivation to seek alcohol following the experience of its negative consequences. They also revealed significant differences between animals classified as insensitive and sensitive to PF in their propensity to reinstate alcohol-seeking behaviours after a period of forced abstinence. The abovementioned effects were accompanied by differences in blood levels of stress hormones and differences in the cortical and subcortical expression of genes and proteins related to dopaminergic, serotonergic, and GABAergic neurotransmission. CONCLUSION Trait sensitivity to PF can determine the trajectory of alcohol addiction in rats. This effect is, at least partially, mediated via distributed physiological and molecular changes within cortical and subcortical regions of the brain.
Collapse
Affiliation(s)
- Agata Cieslik-Starkiewicz
- Department of Pharmacology, Affective Cognitive Neuroscience Laboratory, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Street, 31-343, Krakow, Poland
| | - Karolina Noworyta
- Department of Pharmacology, Affective Cognitive Neuroscience Laboratory, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Street, 31-343, Krakow, Poland
| | - Joanna Solich
- Department of Pharmacology, Biochemical Pharmacology Laboratory, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Street, 31-343, Krakow, Poland
| | - Agata Korlatowicz
- Department of Pharmacology, Biochemical Pharmacology Laboratory, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Street, 31-343, Krakow, Poland
| | - Agata Faron-Górecka
- Department of Pharmacology, Biochemical Pharmacology Laboratory, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Street, 31-343, Krakow, Poland
| | - Rafal Rygula
- Department of Pharmacology, Affective Cognitive Neuroscience Laboratory, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Street, 31-343, Krakow, Poland.
| |
Collapse
|
5
|
Huang J, Wang C, Zhang HB, Zheng H, Huang T, Di JZ. Neuroimaging and neuroendocrine insights into food cravings and appetite interventions in obesity. PSYCHORADIOLOGY 2023; 3:kkad023. [PMID: 38666104 PMCID: PMC10917384 DOI: 10.1093/psyrad/kkad023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/30/2023] [Accepted: 10/13/2023] [Indexed: 04/28/2024]
Abstract
This article reviews the previous studies on the distinction between food cravings and appetite, and how they are regulated by hormones and reflected in brain activity. Based on existing research, food cravings are defined as individual preferences influenced by hormones and psychological factors, which differ from appetite, as they are not necessarily related to hunger or nutritional needs. The article also evaluates the neuroimaging findings about food cravings, and interventions to reduce food cravings, such as mindfulness training, alternative sweeteners, non-invasive brain stimulation techniques, cognitive-behavioral therapy, and imaginal retraining, and points out their advantages, disadvantages, and limitations. Furthermore, the article delves into the potential future directions in the field, emphasizing the need for a neuroendocrine perspective, considerations for associated psychiatric disorders, innovative clinical interventions, and emerging therapeutic frontiers in obesity management. The article outlines the neuro-endocrine basis of food cravings, including ghrelin, leptin, melanocortin, oxytocin, glucagon-like peptide-1, baclofen, and other hormones and their brain regions of action. The article argues that food cravings are an important target for obesity, and more research is needed to explore their complex characteristics and mechanisms, and how to effectively interact with their neuro-endocrine pathways. The article provides a new perspective and approach to the prevention and treatment of obesity.
Collapse
Affiliation(s)
- Jin Huang
- Department of Metabolic & Bariatric Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chen Wang
- Department of Metabolic & Bariatric Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Hang-Bin Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Centre for Mental Disorders, Shanghai Mental Health Centre, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Hui Zheng
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Centre for Mental Disorders, Shanghai Mental Health Centre, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Tao Huang
- Xuhui Health Care Commission, Shanghai 200030, China
| | - Jian-Zhong Di
- Department of Metabolic & Bariatric Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
6
|
Jeanblanc J, Sauton P, Houdant C, Fernandez Rodriguez S, de Sousa SV, Jeanblanc V, Bodeau S, Labat L, Soichot M, Vorspan F, Naassila M. Sex-related differences in the efficacy of Baclofen enantiomers on self-administered alcohol in a binge drinking pattern and dopamine release in the core of the nucleus accumbens. Front Pharmacol 2023; 14:1146848. [PMID: 37007041 PMCID: PMC10060511 DOI: 10.3389/fphar.2023.1146848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction: Clinical studies on the effectiveness of Baclofen in alcohol use disorder (AUD) yielded mixed results possibly because of differential effects of the enantiomers and sex-related differences. Here we examined the effect of the different Baclofen enantiomers on alcohol intake and on evoked dopamine release in the core of the nucleus accumbens (NAcc) in male and female Long Evans rats.Methods: Rats were trained to chronically self-administer 20% alcohol solution in daily binge drinking sessions and were treated with the different forms of Baclofen [RS(±), R(+) and S(−)]. The effects on the evoked dopamine release within the core of the nucleus accumbens were measured in brain slices from the same animals and the alcohol naïve animals using the fast scan cyclic voltammetry technique.Results: RS(±)-Baclofen reduced alcohol intake regardless of sex but more females were non-responders to the treatment. R(+)-Baclofen also reduced alcohol intake regardless of sex but females were less sensitive than males. S(−)-Baclofen did not have any effect on average but in some individuals, especially in the females, it did increase alcohol intake by at least 100%. There were no sex differences in Baclofen pharmacokinetic but a strong negative correlation was found in females with a paradoxical effect of increased alcohol intake with higher blood Baclofen concentration. Chronic alcohol intake reduced the sensitivity to the effect of Baclofen on evoked dopamine release and S(−)-Baclofen increased dopamine release specifically in females.Discussion: Our results demonstrate a sex-dependent effect of the different forms of Baclofen with no or negative effects (meaning an increase in alcohol self-administration) in subgroup of females that could be linked to a differential effect on dopamine release and should warrant future clinical studies on alcohol use disorder pharmacotherapy that will deeply analyze sex difference.
Collapse
Affiliation(s)
- Jérôme Jeanblanc
- INSERM UMR-S 1247, Research Group on Alcohol and Pharmacodependences (GRAP), Université de Picardie Jules Verne, Amiens, France
- GDR3557 Psychiatrie-Addictions, Institut de Psychiatrie, University Hospital Federation (FHU A2M2P), Caen, France
| | - Pierre Sauton
- INSERM UMR-S 1247, Research Group on Alcohol and Pharmacodependences (GRAP), Université de Picardie Jules Verne, Amiens, France
- GDR3557 Psychiatrie-Addictions, Institut de Psychiatrie, University Hospital Federation (FHU A2M2P), Caen, France
| | - Charles Houdant
- INSERM UMR-S 1247, Research Group on Alcohol and Pharmacodependences (GRAP), Université de Picardie Jules Verne, Amiens, France
- GDR3557 Psychiatrie-Addictions, Institut de Psychiatrie, University Hospital Federation (FHU A2M2P), Caen, France
| | - Sandra Fernandez Rodriguez
- INSERM UMR-S 1247, Research Group on Alcohol and Pharmacodependences (GRAP), Université de Picardie Jules Verne, Amiens, France
| | - Sofia Vilelas de Sousa
- INSERM UMR-S 1247, Research Group on Alcohol and Pharmacodependences (GRAP), Université de Picardie Jules Verne, Amiens, France
| | - Virginie Jeanblanc
- Animal Facility of the Université de Picardie Jules Verne, Amiens, France
| | - Sandra Bodeau
- MP3CV Laboratory, Department of Clinical Pharmacology, Amiens University Hospital, University of Picardie Jules Verne, Amiens, France
| | - Laurence Labat
- INSERM UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Université de Paris, Paris, France
- Faculté de Médecine, Université de Paris Cité, Paris, France
- Département de Psychiatrie et de Médecine Addictologique, Hôpital Fernand Widal, Assistance Publique—Hôpitaux de Paris, Paris, France
| | - Marion Soichot
- Laboratoire de Toxicologie Biologique, Hôpital Lariboisière, Paris, France
| | - Florence Vorspan
- INSERM UMRS1144, Département de Psychiatrie et de Médecine Addictologique, Assistance Publique—Hôpitaux de Paris, GH Lariboisière—Fernand Widal, GHU NORD, Université de Paris, Paris, France
| | - Mickael Naassila
- INSERM UMR-S 1247, Research Group on Alcohol and Pharmacodependences (GRAP), Université de Picardie Jules Verne, Amiens, France
- GDR3557 Psychiatrie-Addictions, Institut de Psychiatrie, University Hospital Federation (FHU A2M2P), Caen, France
- *Correspondence: Mickael Naassila,
| |
Collapse
|
7
|
Halsall L, Jones A, Roberts C, Knibb G, Rose AK. The impact of alcohol priming on craving and motivation to drink: a meta-analysis. Addiction 2022; 117:2986-3003. [PMID: 35638379 PMCID: PMC9796461 DOI: 10.1111/add.15962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/08/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS An initial dose of alcohol can motivate-or prime-further drinking and may precipitate (re)lapse and bingeing. Lab-based studies have investigated the alcohol priming effect; however, heterogeneity in designs has resulted in some inconsistent findings. The aims of this meta-analysis were to (i) determine the pooled effect size for motivation to drink following priming, measured by alcohol consumption and craving, and (ii) examine whether design characteristics influenced any priming effect. METHODS Literature searches of PsycINFO, PubMed and Scopus in October 2020 (updated October 2021) identified lab-based alcohol priming studies that assessed effect of priming on motivation to drink. A tailored risk-of-bias tool assessed quality of lab-based studies. Random effects meta-analyses were computed on outcome data from 38 studies comparing the effect of a priming dose of alcohol against control on subsequent alcohol consumption/self-reported craving. Study characteristics that might have affected outcomes were design type (within/between-participant), dose of prime, time of motivation assessment, type of control drink (placebo alcohol/soft drink). RESULTS Relative to control, alcohol had a small-to-moderate priming effect on subsequent alcohol consumption (standardised mean difference [SMD] = 0.336 [95% CI, 0.171, 0.500]) and craving (SMD = 0.431 [95% CI, 0.306, 0.555]). Aspects of study design differentially affected consumption and craving. The size of the priming dose had no effect on consumption, but larger doses were sometimes associated with greater craving (with craving generally following the blood alcohol curve). Alcohol priming effects for consumption, but not craving, were smaller when compared with placebo, relative to soft drink, control. CONCLUSIONS Lab-based alcohol priming studies are a valid paradigm from which to investigate the impact of acute intoxication on alcohol motivation. Designs are needed that assess the impact of acute consumption on motivation to drink in more varied and realistic ways.
Collapse
Affiliation(s)
- Lauren Halsall
- Department of Psychology, Institute of Population HealthUniversity of LiverpoolLiverpoolUnited Kingdom
| | - Andrew Jones
- Department of Psychology, Institute of Population HealthUniversity of LiverpoolLiverpoolUnited Kingdom,Liverpool Centre for Alcohol ResearchLiverpoolUnited Kingdom
| | - Carl Roberts
- Department of Psychology, Institute of Population HealthUniversity of LiverpoolLiverpoolUnited Kingdom
| | - Graeme Knibb
- Department of PsychologyEdge Hill UniversityOrmskirkUnited Kingdom
| | - Abigail K. Rose
- Liverpool Centre for Alcohol ResearchLiverpoolUnited Kingdom,School of PsychologyLiverpool John Moore's UniversityLiverpoolUnited Kingdom
| |
Collapse
|
8
|
Gatta E, Camussi D, Auta J, Guidotti A, Pandey SC. Neurosteroids (allopregnanolone) and alcohol use disorder: From mechanisms to potential pharmacotherapy. Pharmacol Ther 2022; 240:108299. [PMID: 36323379 PMCID: PMC9810076 DOI: 10.1016/j.pharmthera.2022.108299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Alcohol Use Disorder (AUD) is a multifaceted relapsing disorder that is commonly comorbid with psychiatric disorders, including anxiety. Alcohol exposure produces a plethora of effects on neurobiology. Currently, therapeutic strategies are limited, and only a few treatments - disulfiram, acamprosate, and naltrexone - are available. Given the complexity of this disorder, there is a great need for the identification of novel targets to develop new pharmacotherapy. The GABAergic system, the primary inhibitory system in the brain, is one of the well-known targets for alcohol and is responsible for the anxiolytic effects of alcohol. Interestingly, GABAergic neurotransmission is fine-tuned by neuroactive steroids that exert a regulatory role on several endocrine systems involved in neuropsychiatric disorders including AUD. Mounting evidence indicates that alcohol alters the biosynthesis of neurosteroids, whereas acute alcohol increases and chronic alcohol decreases allopregnanolone levels. Our recent work highlighted that chronic alcohol-induced changes in neurosteroid levels are mediated by epigenetic modifications, e.g., DNA methylation, affecting key enzymes involved in neurosteroid biosynthesis. These changes were associated with changes in GABAA receptor subunit expression, suggesting an imbalance between excitatory and inhibitory signaling in AUD. This review will recapitulate the role of neurosteroids in the regulation of the neuroendocrine system, highlight their role in the observed allostatic load in AUD, and develop a framework from mechanisms to potential pharmacotherapy.
Collapse
Affiliation(s)
- Eleonora Gatta
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - Diletta Camussi
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - James Auta
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - Alessandro Guidotti
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA; Jesse Brown Veterans Affairs Medical Center Chicago, IL 60612, USA.
| |
Collapse
|
9
|
Martins B, Rutland W, De Aquino JP, Kazer BL, Funaro M, Potenza MN, Angarita GA. Helpful or Harmful? The Therapeutic Potential of Medications with Varying Degrees of Abuse Liability in the Treatment of Substance Use Disorders. CURRENT ADDICTION REPORTS 2022; 9:647-659. [PMID: 35990796 PMCID: PMC9376579 DOI: 10.1007/s40429-022-00432-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 11/06/2022]
Abstract
Purpose of Review This review summarizes recent clinical trial research on pharmacological treatments for substance use disorders, with a specific focus on agents with potential abuse liability. Recent Findings Pharmacological treatments for substance use disorders may include gabapentinoids, baclofen, modafinil, ketamine, cannabinoids, gamma-hydroxybutyrate, and psychedelics. Gabapentinoids may decrease negative subjective effects of withdrawal in alcohol and cannabis use disorders. Cannabinoids similarly appear to decrease use and withdrawal symptoms in cannabis use disorder, while research shows stimulant medications may reduce cravings and increase abstinence in cocaine use disorder. Ketamine and psychedelics may help treat multiple substance use disorders. Ketamine may reduce withdrawal symptoms, promote abstinence, and diminish cravings in alcohol and cocaine use disorders and psychedelics may promote remission, decrease use, and reduce cravings in alcohol and opioid use disorders. Summary Regardless of current regulatory approval statuses and potentials for abuse, multiple agents should not be dismissed prematurely as possible treatments for substance use disorders. However, further clinical research is needed before effective implementation can begin in practice. Supplementary Information The online version contains supplementary material available at 10.1007/s40429-022-00432-9.
Collapse
Affiliation(s)
- Bradford Martins
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
| | - Will Rutland
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
| | - Joao P. De Aquino
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
| | - Benjamin L. Kazer
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
| | - Melissa Funaro
- Harvey Cushing/John Hay Whitney Medical Library, Yale University, New Haven, CT 06510 USA
| | - Marc N. Potenza
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Child Study Center, Yale University School of Medicine, New Haven, CT 06510 USA
- Department of Neuroscience, Yale University, New Haven, CT 06510 USA
- Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
- Connecticut Council On Problem Gambling, Wethersfield, CT 06109 USA
- Wu Tsai Institute, Yale University, New Haven, CT 06510 USA
| | - Gustavo A. Angarita
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511 USA
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519 USA
| |
Collapse
|
10
|
Ito M, Spence A, Blauwet MB, Heo N, Goldwater R, Maruff P, Marek GJ. A phase 1 study to assess potential interaction between ASP8062 and alcohol in healthy adult subjects. J Psychopharmacol 2022; 36:756-767. [PMID: 34994232 DOI: 10.1177/02698811211058967] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND ASP8062 is a novel orally active GABAB receptor positive allosteric modulator in clinical development for the treatment of alcohol use disorder (AUD) and opioid use disorder (OUD). AIMS This study assessed the potential pharmacokinetic/pharmacodynamic interaction between ASP8062 and alcohol under single-dose conditions in healthy adults. METHODS A double-blind, placebo-controlled, crossover phase 1 study was conducted in which 20 subjects were randomly assigned to four treatment sequences (ASP8062 + alcohol; ASP8062 + placebo alcohol; placebo + alcohol; placebo + placebo alcohol) each consisting of four treatment periods, separated by washout periods of at least 14 days. An analysis of variance was used to assess pharmacokinetic interaction and a mixed-effects analysis of covariance was used to assess pharmacodynamic interaction. RESULTS/OUTCOMES After administration of alcohol, a mild to minimal increase in plasma exposure (AUCinf and Cmax) of ASP8062 was observed, but tmax and t½ for ASP8062 remained unchanged after administration of alcohol. In contrast, ASP8062 did not affect the AUClast and Cmax of ethanol. No clinically relevant differences in cognition measurements were observed with ASP8062 compared with placebo, but there were expected impairments in psychomotor and executive function with alcohol alone. ASP8062 in combination with alcohol resulted in worse scores in cognition measurements than alcohol alone, but this potentiation was not consistent. ASP8062 administered alone was safe and well-tolerated and safety findings in subjects administered alcohol alone were not augmented when ASP8062 was administered in combination with alcohol. CONCLUSION/INTERPRETATION The data support further clinical studies investigating ASP8062 in patients with AUD.
Collapse
Affiliation(s)
- Mototsugu Ito
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Anna Spence
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | | | - Nakyo Heo
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | | | | | - Gerard J Marek
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| |
Collapse
|
11
|
Abstract
Alcohol use disorder (AUD) is a highly prevalent but severely under-treated disorder, with only three widely-approved pharmacotherapies. Given that AUD is a very heterogeneous disorder, it is unlikely that one single medication will be effective for all individuals with an AUD. As such, there is a need to develop new, more effective, and diverse pharmacological treatment options for AUD with the hopes of increasing utilization and improving care. In this qualitative literature review, we discuss the efficacy, mechanism of action, and tolerability of approved, repurposed, and novel pharmacotherapies for the treatment of AUD with a clinical perspective. Pharmacotherapies discussed include: disulfiram, acamprosate, naltrexone, nalmefene, topiramate, gabapentin, varenicline, baclofen, sodium oxybate, aripiprazole, ondansetron, mifepristone, ibudilast, suvorexant, prazosin, doxazosin, N-acetylcysteine, GET73, ASP8062, ABT-436, PF-5190457, and cannabidiol. Overall, many repurposed and novel agents discussed in this review demonstrate clinical effectiveness and promise for the future of AUD treatment. Importantly, these medications also offer potential improvements towards the advancement of precision medicine and personalized treatment for the heterogeneous AUD population. However, there remains a great need to improve access to treatment, increase the menu of approved pharmacological treatments, and de-stigmatize and increase treatment-seeking for AUD.
Collapse
|
12
|
Logge WB, Morris RW, Baillie AJ, Haber PS, Morley KC. Baclofen attenuates fMRI alcohol cue reactivity in treatment-seeking alcohol dependent individuals. Psychopharmacology (Berl) 2021; 238:1291-1302. [PMID: 30788529 DOI: 10.1007/s00213-019-05192-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/04/2019] [Indexed: 12/20/2022]
Abstract
RATIONALE Baclofen has been shown to effect fMRI alcohol cue reactivity in alcohol dependence, but potential varying effects related to baclofen dose levels have not been examined. OBJECTIVE This study investigated whether baclofen attenuates craving and alcohol cue-elicited activation in alcohol-dependent treatment seekers, and the relationship between this response and clinical outcomes (Morley et al. 2018; Morley et al. 2013). METHODS Participants included 30 alcohol-dependent individuals who had received daily baclofen 30 mg (n = 11), 75 mg (n = 8) or placebo (n = 11) for at least 2 weeks. Using functional magnetic resonance imaging (fMRI), we examined alcohol cue-elicited neural activation during a visual alcohol cue reactivity task 120 min following treatment administration, and alcohol cue reactivity and percentage of heavy drinking days (% HDD) associations were assessed. RESULTS Both baclofen-treated groups reported fewer post-scan % HDD when compared to the placebo-treated group, but no subjective craving group differences were found. Increased alcohol cue-elicited activation was seen in placebo compared to the 75 mg/day baclofen participants in two clusters spanning prefrontal regions implicated in cue reactivity, chiefly frontal regions (i.e., frontal and precentral gyri, anterior cingulate cortex), but no observed alcohol cue reactivity differences between placebo and 30 mg/day baclofen groups. Post-scan % HDD was positively correlated with increased alcohol cue-elicited activation in a cluster encompassing the bilateral caudate nucleus and dorsal anterior cingulate cortex when comparing placebo versus 75 mg/day baclofen groups, and several clusters including prefrontal and mesolimbic regions when comparing placebo versus 30 mg/day baclofen groups. CONCLUSIONS Baclofen administration attenuates alcohol cue-elicited activation and reduced the association in baclofen-treated participants between increased activity in key drug cue reactivity regions and higher post-scan % HDD observed in placebo-treated participants, suggesting a dose-specific response effect that may lead to reduced heavy drinking in chronic alcohol-dependent individuals. TRIAL REGISTRATION ClinicalTrials.gov , NCT01711125, https://clinicaltrials.gov/ct2/show /NCT01711125.
Collapse
Affiliation(s)
- Warren B Logge
- Discipline of Addiction Medicine, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Lv 6, King George V Building, 83-117 Missenden Rd, Camperdown, NSW, 2050, Australia.
| | - Richard W Morris
- Central Clinical School, Faculty of Medicine and Health, & Centre for Translational Data Science, University of Sydney, Sydney, NSW, Australia
| | - Andrew J Baillie
- Faculty of Health Sciences, University of Sydney, Sydney, NSW, Australia
| | - Paul S Haber
- Discipline of Addiction Medicine, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Lv 6, King George V Building, 83-117 Missenden Rd, Camperdown, NSW, 2050, Australia
- Drug Health Services, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Kirsten C Morley
- Discipline of Addiction Medicine, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Lv 6, King George V Building, 83-117 Missenden Rd, Camperdown, NSW, 2050, Australia
| |
Collapse
|
13
|
Domi E, Domi A, Adermark L, Heilig M, Augier E. Neurobiology of alcohol seeking behavior. J Neurochem 2021; 157:1585-1614. [PMID: 33704789 DOI: 10.1111/jnc.15343] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/29/2022]
Abstract
Alcohol addiction is a chronic relapsing brain disease characterized by an impaired ability to stop or control alcohol use despite adverse consequences. A main challenge of addiction treatment is to prevent relapse, which occurs in more than >50% of newly abstinent patients with alcohol disorder within 3 months. In people suffering from alcohol addiction, stressful events, drug-associated cues and contexts, or re-exposure to a small amount of alcohol trigger a chain of behaviors that frequently culminates in relapse. In this review, we first present the preclinical models that were developed for the study of alcohol seeking behavior, namely the reinstatement model of alcohol relapse and compulsive alcohol seeking under a chained schedule of reinforcement. We then provide an overview of the neurobiological findings obtained using these animal models, focusing on the role of opioids systems, corticotropin-release hormone and neurokinins, followed by dopaminergic, glutamatergic, and GABAergic neurotransmissions in alcohol seeking behavior.
Collapse
Affiliation(s)
- Esi Domi
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| | - Ana Domi
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Markus Heilig
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| | - Eric Augier
- Center for Social and Affective Neuroscience, BKV, Linköping University, Linköping, Sweden
| |
Collapse
|
14
|
Gatta E, Guidotti A, Saudagar V, Grayson DR, Aspesi D, Pandey SC, Pinna G. Epigenetic Regulation of GABAergic Neurotransmission and Neurosteroid Biosynthesis in Alcohol Use Disorder. Int J Neuropsychopharmacol 2020; 24:130-141. [PMID: 32968808 PMCID: PMC7883893 DOI: 10.1093/ijnp/pyaa073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alcohol use disorder (AUD) is a chronic relapsing brain disorder. GABAA receptor (GABAAR) subunits are a target for the pharmacological effects of alcohol. Neurosteroids play an important role in the fine-tuning of GABAAR function in the brain. Recently, we have shown that AUD is associated with changes in DNA methylation mechanisms. However, the role of DNA methylation in the regulation of neurosteroid biosynthesis and GABAergic neurotransmission in AUD patients remains under-investigated. METHODS In a cohort of postmortem brains from 20 male controls and AUD patients, we investigated the expression of GABAAR subunits and neurosteroid biosynthetic enzymes and their regulation by DNA methylation mechanisms. Neurosteroid levels were quantified by gas chromatography-mass spectrometry. RESULTS The α 2 subunit expression was reduced due to increased DNA methylation at the gene promoter region in the cerebellum of AUD patients, a brain area particularly sensitive to the effects of alcohol. Alcohol-induced alteration in GABAAR subunits was also observed in the prefrontal cortex. Neurosteroid biosynthesis was also affected with reduced cerebellar expression of the 18kDa translocator protein and 3α-hydroxysteroid dehydrogenase mRNAs. Notably, increased DNA methylation levels were observed at the promoter region of 3α-hydroxysteroid dehydrogenase. These changes were associated with markedly reduced levels of allopregnanolone and pregnanolone in the cerebellum. CONCLUSION Given the key role of neurosteroids in modulating the strength of GABAAR-mediated inhibition, our data suggest that alcohol-induced impairments in GABAergic neurotransmission might be profoundly impacted by reduced neurosteroid biosynthesis most likely via DNA hypermethylation.
Collapse
Affiliation(s)
- Eleonora Gatta
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois
| | - Alessandro Guidotti
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois,Correspondence: Dr Alessandro Guidotti, Center for Alcohol Research in Epigenetics, Psychiatric Institute - Department of Psychiatry, University of Illinois at Chicago, 1601 West Taylor Street, Chicago, IL 60612 ()
| | - Vikram Saudagar
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois
| | - Dennis R Grayson
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois
| | - Dario Aspesi
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Graziano Pinna
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
15
|
Haass-Koffler CL, Perciballi R. Alcohol Tolerance in Human Laboratory Studies for Development of Medications to treat Alcohol Use Disorder. Alcohol Alcohol 2020; 55:129-135. [PMID: 31950152 PMCID: PMC7082491 DOI: 10.1093/alcalc/agz103] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/17/2019] [Accepted: 11/26/2019] [Indexed: 01/28/2023] Open
Abstract
AIMS Human laboratory studies have contributed extensively in the research and development of novel medications to treat alcohol use disorder (AUD). Alcohol tolerance may represent one additional variable that can be utilized to expand the understanding of the AUD wide phenotypic profile and provide support to the medication development process. Tolerance is characterized as an individual's subjective response to alcohol and has been recognized as a predictor of AUD progression. Tolerance can be evaluated both by self-reported response (e.g. assessments) and objective measurements (e.g. motor impairment); as such, it represents an exploitable variable in the field of alcohol research. METHODS This Narrative Review focuses on the use of alcohol tolerance, specifically within alcohol laboratory studies, for medication development. It seeks to identify a research gap and a research opportunity in clinical studies to evaluate biobehavioral responses captured in order to develop medications to treat AUD. RESULTS Alcohol tolerance may provide additional information on the safety and tolerability of medications to treat AUD, in particular, when novel medications are co-administered with alcohol within the AUD population. CONCLUSIONS As such, alcohol tolerance represents an additional outcome that may be included in randomized clinical trial (RCT) protocols designed for developing AUD pharmacotherapies.
Collapse
Affiliation(s)
- Carolina L Haass-Koffler
- Department of Psychiatry and Human Behavior, Center for Alcohol and Addiction Studies, Brown University, 121 South Main Street, Providence, RI 02912, USA
- Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, School of Public Health, Brown University, 121 South Main Street, Providence, RI 02912, USA
- 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, 10 Center Drive, Bethesda, MD 20892-1108, USA
| | - Roberta Perciballi
- Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, School of Public Health, Brown University, 121 South Main Street, Providence, RI 02912, USA
- Sapienza University, Rome, Italy, Piazzale Aldo Moro, 5, 00185 Roma RM, Italy
| |
Collapse
|
16
|
Abstract
Harmful alcohol use and alcohol use disorders (AUD) result in major health and community burden worldwide, yet treatment options are limited. Novel pharmacotherapies are urgently required, and treatments involving GABAB receptors have been used in treating alcohol-related disorders. This chapter will review the clinical evidence of GABAB pharmacotherapies, such as baclofen and γ-hydroxybutyric acid. This includes the use of these treatments in individuals experiencing alcohol withdrawal symptoms and outlining the outcomes of studies of alcohol relapse prevention relapse including case studies, comparative studies and randomised controlled trials. Laboratory research investigating biobehavioural effects of baclofen will also be summarised and polymorphisms associated with baclofen treatment, and safety concerns of GABAB treatments will be addressed. In summary, pharmacological treatments targeting GABAB receptors such as baclofen may be modestly effective in the management of alcohol use disorder, but safety concerns limit the widespread applicability of the currently available agents.
Collapse
|
17
|
Witkiewitz K, Litten RZ, Leggio L. Advances in the science and treatment of alcohol use disorder. SCIENCE ADVANCES 2019; 5:eaax4043. [PMID: 31579824 PMCID: PMC6760932 DOI: 10.1126/sciadv.aax4043] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/28/2019] [Indexed: 05/03/2023]
Abstract
Alcohol is a major contributor to global disease and a leading cause of preventable death, causing approximately 88,000 deaths annually in the United States alone. Alcohol use disorder is one of the most common psychiatric disorders, with nearly one-third of U.S. adults experiencing alcohol use disorder at some point during their lives. Alcohol use disorder also has economic consequences, costing the United States at least $249 billion annually. Current pharmaceutical and behavioral treatments may assist patients in reducing alcohol use or facilitating alcohol abstinence. Although recent research has expanded understanding of alcohol use disorder, more research is needed to identify the neurobiological, genetic and epigenetic, psychological, social, and environmental factors most critical in the etiology and treatment of this disease. Implementation of this knowledge in clinical practice and training of health care providers is also needed to ensure appropriate diagnosis and treatment of individuals suffering from alcohol use disorder.
Collapse
Affiliation(s)
- K. Witkiewitz
- Department of Psychology and Center on Alcoholism, Substance Abuse, and Addictions, University of New Mexico, 2650 Yale Blvd. SE, Albuquerque, NM 87106, USA
| | - R. Z. Litten
- Division of Medications Development and Division of Treatment and Recovery Research, National Institute on Alcohol Abuse and Alcoholism, 6700B Rockledge Drive, Bethesda, MD 20892-6902, USA
| | - L. 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, 10 Center Drive (10CRC/15330), Bethesda, MD 21224, USA
- Medication Development Program, National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd., Baltimore, MD 21224, USA
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI 02912, USA
- Corresponding author.
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW The aim of this study was to provide an update on medication development efforts for alcohol use disorder (AUD) by reviewing recently published (past 2 years) human studies that evaluated medications' effects on alcohol-related outcomes. RECENT FINDINGS Forty-five publications were found suitable for this review. A variety of compounds have been tested in the past 2 years as potential pharmacological options for AUD, including medications that act on multiple targets (topiramate, aripiprazole, quetiapine), calcium channels (gabapentin), gamma-Aminobutyric acid receptors (baclofen, diazepam), glutamate receptors (ifenprodil, memantine, glycine), nicotinic acetylcholine receptors (varenicline, mecamylamine), α1 adrenergic receptors (prazosin, doxazosin), neuroendocrine pathways (oxytocin, a vasopressin receptor 1b antagonist, a ghrelin receptor inverse agonist) and others (samidorphan, ibudilast, N-acetylcysteine, citoline). Important findings and limitations regarding the effects of these medications on alcohol-related outcomes are discussed. SUMMARY There is a critical need to increase the armamentarium of medications for AUD. Human laboratory studies may help screen and prioritize promising targets and compounds before running large clinical trials. Given the complexity of AUD and the heterogeneity of afflicted patients, future studies should also investigate potential moderators and predictors of response to each pharmacological intervention.
Collapse
|
19
|
Leggio L, Falk DE, Ryan ML, Fertig J, Litten RZ. Medication Development for Alcohol Use Disorder: A Focus on Clinical Studies. Handb Exp Pharmacol 2019; 258:443-462. [PMID: 31628604 DOI: 10.1007/164_2019_295] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Compared to other medical disorders, including other brain diseases, the number of medications approved for alcohol use disorder (AUD) is very small. Disulfiram, naltrexone (oral and long-acting), and acamprosate are approved by the US Food and Drug Administration (FDA) to treat patients with AUD. These medications are also approved in other countries, including in Europe, where the European Medicines Agency (EMA) also approved nalmefene for AUD. Furthermore, baclofen was recently approved for AUD in France. These approved medications have small effect sizes, which are probably the consequence of the fact that they only work for some patients, yet a personalized approach to match the right medication with the right patient is still in its infancy. Therefore, research is needed to expand the armamentarium of medications that clinicians can use to treat their patients, as well as to better develop personalized approaches. This book chapter reviews other medications, beyond those approved by the FDA, that have shown efficacy in clinical trials, as well as medications which are still in the early stages of evaluation in human studies.
Collapse
Affiliation(s)
- 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, Bethesda, MD, USA. .,Medication Development Program, National Institute on Drug Abuse Intramural Research Program , Baltimore, MD, USA. .,Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA.
| | - Daniel E Falk
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Megan L Ryan
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Joanne Fertig
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Raye Z Litten
- Division of Medications Development, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| |
Collapse
|
20
|
Béracochéa D, Mons N, David V. Targeting the Glucocorticoid Receptors During Alcohol Withdrawal to Reduce Protracted Neurocognitive Disorders. Front Psychiatry 2019; 10:580. [PMID: 31620025 PMCID: PMC6759466 DOI: 10.3389/fpsyt.2019.00580] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 07/23/2019] [Indexed: 11/13/2022] Open
Abstract
Persistent regional glucocorticoid (GC) dysregulation in alcohol-withdrawn subjects emerges as a key factor responsible for protracted molecular and neural alterations associated with long-term cognitive dysfunction. Regional brain concentrations of corticosterone vary independently from plasma concentrations in alcohol-withdrawn subjects, which may account for the treatment of alcohol withdrawal-induced persistent pathology. Thus, from a pharmacological point of view, a main issue remains to determine the relative efficacy of compounds targeting the GC receptors to attenuate or suppress the long-lasting persistence of brain regional GC dysfunctions in abstinent alcoholics, as well as persistent changes of neural plasticity. Data from animal research show that acting directly on GC receptors during the withdrawal period, via selective antagonists, can significantly counteract the development and persistence of cognitive and neural plasticity disorders during protracted abstinence. A critical remaining issue is to better assess the relative long-term efficacy of GC antagonists and other compounds targeting the corticotropic axis activity such as gamma-aminobutyric acid A (GABAA) and GABAB agonists. Indeed, benzodiazepines (acting indirectly on GABAA receptors) and baclofen (agonist of the GABAB receptor) are the compounds most widely used to reduce alcohol dependence. Clinical and preclinical data suggest that baclofen exerts an effective and more powerful counteracting action on such persistent cognitive and endocrine dysfunctions as compared to diazepam, even though its potential negative effects on memory processes, particularly at high doses, should be better taken into account.
Collapse
Affiliation(s)
- Daniel Béracochéa
- Université de Bordeaux, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Pessac, France.,CNRS UMR 5287, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Pessac, France
| | - Nicole Mons
- Université de Bordeaux, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Pessac, France.,CNRS UMR 5287, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Pessac, France
| | - Vincent David
- Université de Bordeaux, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Pessac, France.,CNRS UMR 5287, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Pessac, France
| |
Collapse
|
21
|
de Beaurepaire R, Sinclair JMA, Heydtmann M, Addolorato G, Aubin HJ, Beraha EM, Caputo F, Chick JD, de La Selle P, Franchitto N, Garbutt JC, Haber PS, Jaury P, Lingford-Hughes AR, Morley KC, Müller CA, Owens L, Pastor A, Paterson LM, Pélissier F, Rolland B, Stafford A, Thompson A, van den Brink W, Leggio L, Agabio R. The Use of Baclofen as a Treatment for Alcohol Use Disorder: A Clinical Practice Perspective. Front Psychiatry 2018; 9:708. [PMID: 30662411 PMCID: PMC6328471 DOI: 10.3389/fpsyt.2018.00708] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022] Open
Abstract
Alcohol use disorder (AUD) is a brain disorder associated with high rates of mortality and morbidity worldwide. Baclofen, a selective gamma-aminobutyric acid-B (GABA-B) receptor agonist, has emerged as a promising drug for AUD. The use of this drug remains controversial, in part due to uncertainty regarding dosing and efficacy, alongside concerns about safety. To date there have been 15 randomized controlled trials (RCTs) investigating the use of baclofen in AUD; three using doses over 100 mg/day. Two additional RCTs have been completed but have not yet been published. Most trials used fixed dosing of 30-80 mg/day. The other approach involved titration until the desired clinical effect was achieved, or unwanted effects emerged. The maintenance dose varies widely from 30 to more than 300 mg/day. Baclofen may be particularly advantageous in those with liver disease, due to its limited hepatic metabolism and safe profile in this population. Patients should be informed that the use of baclofen for AUD is as an "off-label" prescription, that no optimal fixed dose has been established, and that existing clinical evidence on efficacy is inconsistent. Baclofen therapy requires careful medical monitoring due to safety considerations, particularly at higher doses and in those with comorbid physical and/or psychiatric conditions. Baclofen is mostly used in some European countries and Australia, and in particular, for patients who have not benefitted from the currently used and approved medications for AUD.
Collapse
Affiliation(s)
| | - Julia M A Sinclair
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Mathis Heydtmann
- Department of Gastroenterology, Royal Alexandra Hospital Paisley, Paisley, United Kingdom
| | - Giovanni Addolorato
- AUD and Alcohol Related Diseases Unit, Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario A Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Department of Internal Medicine and Gastroenterology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Henri-Jean Aubin
- Faculté de Médecine, Centre de Recherche en Epidémiologie et Santé des Populations, Université Paris-Sud, Paris, France.,Faculté de Médecine, Université de Versailles Saint-Quentin-en-Yvelines, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, Paris, France.,Hôpitaux Universitaires Paris-Sud, Paris, France
| | - Esther M Beraha
- Department of Psychology, University of Amsterdam, Amsterdam, Netherlands
| | - Fabio Caputo
- Department of Internal Medicine, SS. Annunziata Hospital, Cento, Italy
| | - Jonathan D Chick
- Castle Craig Hospital, Blyth Bridge, United Kingdom.,School of Health and Social Care, Edinburgh Napier University, Edinburgh, United Kingdom
| | | | - Nicolas Franchitto
- Department of Addiction Medicine, Poisons and Substance Abuse Treatment Centre, Toulouse-Purpan University Hospital, Toulouse, France
| | - James C Garbutt
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Paul S Haber
- National Health Medical Research Council, Centre of Research Excellence in Mental Health and Substance Use, Central Clinical School, Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Drug Health Services, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Philippe Jaury
- Département de Médecine Générale, Faculté de Médecine, Université Paris Descartes, Paris, France
| | - Anne R Lingford-Hughes
- Neuropsychopharmacology Unit, Division of Brain Sciences, Centre for Psychiatry, Imperial College London, London, United Kingdom
| | - Kirsten C Morley
- Discipline of Addiction Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Christian A Müller
- Department of Psychiatry, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lynn Owens
- Wolfson Centre for Personalised Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Adam Pastor
- Department Addiction Medicine, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Louise M Paterson
- Neuropsychopharmacology Unit, Division of Brain Sciences, Centre for Psychiatry, Imperial College London, London, United Kingdom
| | - Fanny Pélissier
- Poison Control Center, Toulouse-Purpan University Hospital, Toulouse, France
| | - Benjamin Rolland
- Service Universitaire d'Addictologie de Lyon, Lyon, France.,University of Lyon, Lyon, France
| | | | - Andrew Thompson
- Wolfson Centre for Personalised Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Wim van den Brink
- Department of Psychiatry, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam University, Amsterdam, Netherlands
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, Division of Intramural Clinical and Basic Research, National Institute on Alcohol Abuse and Alcoholism, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Bethesda, MD, United States.,Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States.,Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, United States
| | - Roberta Agabio
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|