1
|
Faulkner ML, Farokhnia M, Lee MR, Farinelli L, Browning BD, Abshire K, Daurio AM, Munjal V, Deschaine SL, Boukabara SR, Fortney C, Sherman G, Schwandt M, Akhlaghi F, Momenan R, Ross TJ, Persky S, Leggio L. A randomized, double-blind, placebo-controlled study of a GHSR blocker in people with alcohol use disorder. JCI Insight 2024; 9:e182331. [PMID: 39704175 PMCID: PMC11665556 DOI: 10.1172/jci.insight.182331] [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] [Received: 04/23/2024] [Accepted: 10/30/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUNDStudies have demonstrated the role of ghrelin in alcohol-related behaviors and consumption. Blockade of the growth hormone secretagogue receptor (GHSR), which is the ghrelin receptor, has been shown to decrease alcohol drinking and reward-related behaviors across several animal models. We previously conducted a human study testing a GHSR inverse agonist/competitive antagonist, PF-5190457, in individuals who are heavy drinkers and showed its safety when coadministered with alcohol. Here, we conducted a phase IIa experimental medicine study in patients with alcohol use disorder (AUD) to investigate the effects of PF-5190457 on alcohol- and food-related outcomes.METHODSForty-two individuals with AUD (n = 29 completers) participated in a randomized, double-blind, placebo-controlled study where they received PF-5190457 100mg b.i.d. (or placebo) in 2 counterbalanced, within-subject stages. Participants completed an alcohol cue-reactivity (CR) experiment in a bar-like laboratory and a virtual food choice experiment in a cafeteria-like virtual reality (VR) environment. A subset of participants (n = 12) performed a CR task during a brain functional MRI (fMRI) experiment.RESULTSPF-5190457 did not reduce cue-elicited alcohol craving. PF-5190457 reduced virtual calories selected (P = 0.04) in the VR environment. PF-5190457 did not influence neural activation during CR task in the fMRI experiment.CONCLUSIONThis study provides human evidence of the role of GHSR blockade in behaviors related to food selection and highlights the need for future investigations into targeting the ghrelin system in AUD.TRIAL REGISTRATIONClinicalTrials.gov (accession no. NCT02707055).FUNDINGNIDA and NIAAA ZIA-DA000635; National Center for Advancing Translational Sciences UH2/UH3-TR000963.
Collapse
Affiliation(s)
- Monica L. Faulkner
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Mary R. Lee
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Lisa Farinelli
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Brittney D. Browning
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Kelly Abshire
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Allison M. Daurio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Vikas Munjal
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Sara L. Deschaine
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Selim R. Boukabara
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| | - Christopher Fortney
- Immersive Simulation Program, Social and Behavioral Research Branch, National Human Genome Research Institute (NHGRI), NIH, Bethesda, Maryland, USA
| | - Garrick Sherman
- Office of the Clinical Director, NIDA, Intramural Research Program, NIH, Baltimore, Maryland, USA
| | - Melanie Schwandt
- Office of the Clinical Director, NIAAA Division of Intramural Clinical and Biological Research, NIH, Bethesda, Maryland, USA
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Reza Momenan
- Clinical NeuroImaging Research Core, NIAAA, NIH, Bethesda, Maryland, USA
| | - Thomas J. Ross
- Neuroimaging Core, NIDA Intramural Research Program, NIH, Baltimore, Maryland, USA
| | - Susan Persky
- Immersive Simulation Program, Social and Behavioral Research Branch, National Human Genome Research Institute (NHGRI), NIH, Bethesda, Maryland, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse (NIDA) Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism (NIAAA) Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island, USA
- Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Neuroscience, Georgetown University Medical Center, Washington DC, USA
| |
Collapse
|
2
|
Spencer CN, Elton A, Dove S, Faulkner ML, Robinson DL, Boettiger CA. Naltrexone engages a brain reward network in the presence of reward-predictive distractor stimuli in males. ADDICTION NEUROSCIENCE 2023; 7:100085. [PMID: 37424633 PMCID: PMC10328541 DOI: 10.1016/j.addicn.2023.100085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The non-selective opioid receptor antagonist, naltrexone is one of the most prescribed medications for treating alcohol and opioid addiction. Despite decades of clinical use, the mechanism(s) by which naltrexone reduces addictive behavior remains unclear. Pharmaco-fMRI studies to date have largely focused on naltrexone's impact on brain and behavioral responses to drug or alcohol cues or on decision-making circuitry. We hypothesized that naltrexone's effects on reward-associated brain regions would associate with reduced attentional bias (AB) to non-drug, reward-conditioned cues. Twenty-three adult males, including heavy and light drinkers, completed a two-session, placebo-controlled, double-blind study testing the effects of acute naltrexone (50 mg) on AB to reward-conditioned cues and neural correlates of such bias measured via fMRI during a reward-driven AB task. While we detected significant AB to reward-conditioned cues, naltrexone did not reduce this bias in all participants. A whole-brain analysis found that naltrexone significantly altered activity in regions associated with visuomotor control regardless of whether a reward-conditioned distractor was present. A region-of-interest analysis of reward-associated areas found that acute naltrexone increased BOLD signal in the striatum and pallidum. Moreover, naltrexone effects in the pallidum and putamen predicted individual reduction in AB to reward-conditioned distractors. These findings suggest that naltrexone's effects on AB primarily reflect not reward processing per se, but rather top-down control of attention. Our results suggest that the therapeutic actions of endogenous opioid blockade may reflect changes in basal ganglia function enabling resistance to distraction by attractive environmental cues, which could explain some variance in naltrexone's therapeutic efficacy.
Collapse
Affiliation(s)
- Cory N. Spencer
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Amanda Elton
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, North Carolina, USA
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Samantha Dove
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Monica L. Faulkner
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Donita L. Robinson
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Charlotte A. Boettiger
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, North Carolina, USA
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
3
|
Meredith LR, Burnette EM, Nieto SJ, Du H, Donato S, Grodin EN, Green R, Magill M, Baskerville WA, Ray LA. Testing pharmacotherapies for alcohol use disorder with cue exposure paradigms: A systematic review and quantitative synthesis of human laboratory trial methodology. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1629-1645. [PMID: 37423771 DOI: 10.1111/acer.15143] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/30/2023] [Accepted: 06/24/2023] [Indexed: 07/11/2023]
Abstract
Alcohol cue exposure is a widely used experimental paradigm for screening pharmacotherapies for alcohol use disorder (AUD). Medication-related reductions in cue-reactivity signal early efficacy and inform medications development. Yet, across trials, the design of cue exposure, parameter testing, and outcome reporting is heterogeneous. This systematic review is a quantitative synthesis of trial methodologies and effect size estimation for AUD medication-related craving and psychophysiological outcomes under the cue exposure paradigm. A PubMed search was conducted on January 3, 2022 based on identified pharmacotherapies for peer-reviewed articles reported in English. Study-level characteristics, including sample descriptors, paradigm design, analytic approach, and Cochrane Risk of Bias, along with descriptive statistics for cue-exposure outcomes, were coded by two independent raters. Study-level effect sizes were estimated for craving and psychophysiological outcomes separately and sample-level effect sizes were calculated for each medication. Thirty-six trials, comprising 1640 participants and testing 19 different medications satisfied eligibility criteria. All studies reported on biological sex (71% male participants on average). The exposure paradigms implemented used in vivo (n = 26), visual (n = 8), and audio script (n = 2) cues. Some trials included means for craving by medication condition in text (k = 7) or figures (k = 18). The quantitative synthesis included 63 effect sizes (craving kes = 47; psychophysiological kes = 16) from 28 unique randomized trials testing 15 medications for effects on cue reactivity. For cue-induced craving, eight medications (kes range: 1-12) demonstrated small-to-medium effects (Cohen's d range: |0.24-0.64|) compared to placebo, with individuals randomized to receive medication reporting lower craving following cue exposure. Recommendations are provided to promote further consilience, so that the utility of cue exposure paradigms can be maximized in the development of effective AUD pharmacotherapies. Future work should explore the predictive utility of medication-related reductions in cue-reactivity on clinical outcomes.
Collapse
Affiliation(s)
- Lindsay R Meredith
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
| | - Elizabeth M Burnette
- Brain Research Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Steven J Nieto
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
| | - Han Du
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
| | - Suzanna Donato
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
| | - Erica N Grodin
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | - ReJoyce Green
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Molly Magill
- Center for Alcohol and Addiction Studies, Brown University School of Public Health, Providence, Rhode Island, USA
| | - Wave-Ananda Baskerville
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
| | - Lara A Ray
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
- Brain Research Institute, University of California, Los Angeles, Los Angeles, California, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
4
|
Robinson JD, Cui Y, Karam-Hage M, Kypriotakis G, Versace F, Ait-Daoud Tiouririne N, Anthenelli RM, Cinciripini PM. Topiramate decreases the salience of motivationally relevant visual cues among smokers with alcohol use disorder. Alcohol Clin Exp Res 2022; 46:384-395. [PMID: 35037278 PMCID: PMC8920769 DOI: 10.1111/acer.14771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/17/2021] [Accepted: 01/03/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND There is preliminary evidence that the anticonvulsant topiramate increases the likelihood of both smoking and alcohol abstinence among smokers with alcohol use disorder (AUD), but its therapeutic mechanism has not been determined. We used event-related potentials (ERPs) to evaluate topiramate's effect on the salience of drug-related, emotional, and neutral pictorial cues to identify whether one of its potential therapeutic mechanisms involves reduction of the salience of motivationally relevant cues. METHODS Participants enrolled in a multisite clinical trial treating smokers with AUD were randomly assigned to receive placebo, low-dose topiramate (up to 125 mg/day), or high-dose topiramate (up to 250 mg/day), along with brief behavioral compliance enhancement treatment. A subsample (n = 101) completed ERP assessments at baseline (1 week pre-medication) and week 5 (5 weeks on medication; 1 week pre-quit). We assessed the salience of pleasant, unpleasant, cigarette-related, alcohol-related, and neutral pictorial cues using the late positive potential (LPP) ERP component and measured self-reported substance use, reinforcement, craving, and withdrawal. RESULTS Five weeks of high-dose topiramate treatment decreased LPP amplitudes in response to both emotional (pleasant and unpleasant) and drug-related cues (alcohol and cigarette), but not to neutral cues. However, results showed that the LPPs were not significant mediators of the relationship between topiramate dose and post-quit measures of substance use, reinforcement, craving, or withdrawal. CONCLUSIONS These findings suggest that high-dose topiramate (up to 250 mg/day) decreases the motivational salience of both drug-related and emotional cues among smokers with AUD. However, the nonsignificant mediation analyses preclude any firm conclusions about whether this effect represents one of topiramate's therapeutic mechanisms of action.
Collapse
Affiliation(s)
- Jason D. Robinson
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yong Cui
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Maher Karam-Hage
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - George Kypriotakis
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Francesco Versace
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nassima Ait-Daoud Tiouririne
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Robert M. Anthenelli
- Pacific Treatment and Research Center, Department of Psychiatry, University of California, San Diego, Health Sciences, La Jolla, California, USA
| | - Paul M. Cinciripini
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
5
|
Schick MR, Spillane NS, Hostetler KL. A Call to Action: A Systematic Review Examining the Failure to Include Females and Members of Minoritized Racial/Ethnic Groups in Clinical Trials of Pharmacological Treatments for Alcohol Use Disorder. Alcohol Clin Exp Res 2020; 44:1933-1951. [DOI: 10.1111/acer.14440] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Melissa R. Schick
- From the PATHS Lab Department of Psychology University of Rhode Island Kingston Rhode Island
| | - Nichea S. Spillane
- From the PATHS Lab Department of Psychology University of Rhode Island Kingston Rhode Island
| | - Katherine L. Hostetler
- From the PATHS Lab Department of Psychology University of Rhode Island Kingston Rhode Island
| |
Collapse
|
6
|
Cavicchioli M, Vassena G, Movalli M, Maffei C. Is craving a risk factor for substance use among treatment-seeking individuals with alcohol and other drugs use disorders? A meta-analytic review. Drug Alcohol Depend 2020; 212:108002. [PMID: 32413635 DOI: 10.1016/j.drugalcdep.2020.108002] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND It is still unclear whether craving should be considered a key risk factor for substance-use behaviors (SUB) among treatment-seeking individuals with alcohol (AUD) and other drugs use disorders. Therefore, this study aims at clarifying this topic using a meta-analytic approach. METHODS Cohen's d was computed as effect size (ES) measure. Heterogeneity of ESs was computed using the Q statistic and I2 index. The analyses also evaluated the impact assessment length on ESs. Furthermore, proximal effects of craving on SUB were compared to distal ones. The diagnoses of specific substance use disorders (SUDs), together with assessment instruments and research design were considered as additional moderators. RESULTS Thirty-six independent studies were included for a total of 4868 treatment-seeking individuals with SUDs. Patients who used substances showed slightly higher levels of craving than abstinent ones. The heterogeneity of results was large and significant. The length of period of assessment was positively related to ESs. The analyses highlighted no differences between pooled ESs of proximal and distal impacts of craving on SUB. The diagnoses of SUDs were significant moderators. Considering AUD, assessment instruments and research design were additional moderators. CONCLUSIONS Craving is a modest time-dependent proximal and distal risk factor for SUB among individuals with SUDs. Both the frequency of craving episodes and a heightened reactivity to craving cues are largely associated to SUB among individuals with AUD. Future studies should evaluate the mediating and moderating roles of self-regulatory mechanisms on the relationship between craving and SUB.
Collapse
Affiliation(s)
- Marco Cavicchioli
- Department of Psychology, University "Vita-Salute San Raffaele", Milan, Italy; Unit of Clinical Psychology and Psychotherapy, San Raffaele-Turro Hospital, Milan, Italy.
| | - Giulia Vassena
- Department of Psychology, University "Vita-Salute San Raffaele", Milan, Italy; Unit of Clinical Psychology and Psychotherapy, San Raffaele-Turro Hospital, Milan, Italy.
| | - Mariagrazia Movalli
- Department of Psychology, University "Vita-Salute San Raffaele", Milan, Italy; Unit of Clinical Psychology and Psychotherapy, San Raffaele-Turro Hospital, Milan, Italy.
| | - Cesare Maffei
- Department of Psychology, University "Vita-Salute San Raffaele", Milan, Italy; Unit of Clinical Psychology and Psychotherapy, San Raffaele-Turro Hospital, Milan, Italy.
| |
Collapse
|
7
|
Lee MR, Tapocik JD, Ghareeb M, Schwandt ML, Dias AA, Le AN, Cobbina E, Farinelli LA, Bouhlal S, Farokhnia M, Heilig M, Akhlaghi F, Leggio L. The novel ghrelin receptor inverse agonist PF-5190457 administered with alcohol: preclinical safety experiments and a phase 1b human laboratory study. Mol Psychiatry 2020; 25:461-475. [PMID: 29728704 PMCID: PMC6215751 DOI: 10.1038/s41380-018-0064-y] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 12/15/2022]
Abstract
Rodent studies indicate that ghrelin receptor blockade reduces alcohol consumption. However, no ghrelin receptor blockers have been administered to heavy alcohol drinking individuals. Therefore, we evaluated the safety, tolerability, pharmacokinetic (PK), pharmacodynamic (PD) and behavioral effects of a novel ghrelin receptor inverse agonist, PF-5190457, when co-administered with alcohol. We tested the effects of PF-5190457 combined with alcohol on locomotor activity, loss-of-righting reflex (a measure of alcohol sedative actions), and on blood PF-5190457 concentrations in rats. Then, we performed a single-blind, placebo-controlled, within-subject human study with PF-5190457 (placebo/0 mg b.i.d., 50 mg b.i.d., 100 mg b.i.d.). Twelve heavy drinkers during three identical visits completed an alcohol administration session, subjective assessments, and an alcohol cue-reactivity procedure, and gave blood samples for PK/PD testing. In rats, PF-5190457 did not interact with the effects of alcohol on locomotor activity or loss-of-righting reflex. Alcohol did not affect blood PF-5190457 concentrations. In humans, all adverse events were mild or moderate and did not require discontinuation or dose reductions. Drug dose did not alter alcohol concentration or elimination, alcohol-induced stimulation or sedation, or mood during alcohol administration. Potential PD markers of PF-5190457 were acyl-to-total ghrelin ratio and insulin-like growth factor-1. PF-5190457 (100 mg b.i.d.) reduced alcohol craving during the cue-reactivity procedure. This study provides the first translational evidence of safety and tolerability of the ghrelin receptor inverse agonist PF-5190457 when co-administered with alcohol. PK/PD/behavioral findings support continued research of PF-5190457 as a potential pharmacological agent to treat alcohol use disorder.
Collapse
Affiliation(s)
- Mary R Lee
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Jenica D Tapocik
- Section on Molecular Pathophysiology, Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Mwlod Ghareeb
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Melanie L Schwandt
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Alexandra A Dias
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - April N Le
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Enoch Cobbina
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Lisa A Farinelli
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Sofia Bouhlal
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Mehdi Farokhnia
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Markus Heilig
- Section on Molecular Pathophysiology, Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA.
- Department of Behavioral and Social Sciences, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA.
| |
Collapse
|
8
|
Intravenous administration of ghrelin increases serum cortisol and aldosterone concentrations in heavy-drinking alcohol-dependent individuals: Results from a double-blind, placebo-controlled human laboratory study. Neuropharmacology 2019; 158:107711. [PMID: 31310775 DOI: 10.1016/j.neuropharm.2019.107711] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/25/2019] [Accepted: 07/12/2019] [Indexed: 12/16/2022]
Abstract
Increasing evidence supports the role of appetite-regulating hormones, including ghrelin, in alcohol use disorder (AUD). Effects of ghrelin administration on cortisol and aldosterone, two hormones known to influence the development and maintenance of AUD, have been observed in ghrelin-exposed tissues or cells, as well as rodents and healthy volunteers, however whether these effects replicate in individuals with AUD is unknown. Here, we tested the hypothesis that intravenous administration of ghrelin leads to increase in endogenous serum cortisol and aldosterone concentrations in alcohol-dependent, heavy drinking individuals, and that these changes may predict ghrelin-induced alcohol craving. This was a double-blind, placebo-controlled human laboratory study in non-treatment-seeking, heavy-drinking, alcohol-dependent individuals randomized to receive either placebo, 1 mcg/kg or 3 mcg/kg of intravenous ghrelin. Then, participants underwent a cue-reactivity procedure in a bar-like setting, which included exposure to both neutral (juice) and alcohol cues. Repeated blood samples were collected and used to measure endogenous cortisol and aldosterone serum concentrations, in response to exogenous ghrelin administration. Furthermore, cortisol and aldosterone serum concentrations were used to develop a model to predict the effect of exogenous ghrelin administration on alcohol craving. Intravenous ghrelin administration increased endogenous cortisol and aldosterone serum concentrations. While the effects on cortisol were greater than those on aldosterone, only the ghrelin-induced changes in aldosterone serum concentrations predicted craving. These findings provide initial evidence of ghrelin effects on glucocorticoids and mineralocorticoids in individuals with AUD, thereby providing additional information on the potential mechanisms by which the ghrelin system may play a role in alcohol craving and seeking in AUD.
Collapse
|
9
|
Cannella N, Ubaldi M, Masi A, Bramucci M, Roberto M, Bifone A, Ciccocioppo R. Building better strategies to develop new medications in Alcohol Use Disorder: Learning from past success and failure to shape a brighter future. Neurosci Biobehav Rev 2019; 103:384-398. [PMID: 31112713 DOI: 10.1016/j.neubiorev.2019.05.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022]
Abstract
Alcohol Use Disorder (AUD) is a chronic disease that develops over the years. The complexity of the neurobiological processes contributing to the emergence of AUD and the neuroadaptive changes occurring during disease progression make it difficult to improve treatments. On the other hand, this complexity offers researchers the possibility to explore new targets. Over years of intense research several molecules were tested in AUD; in most cases, despite promising preclinical data, the clinical efficacy appeared insufficient to justify futher development. A prototypical example is that of corticotropin releasing factor type 1 receptor (CRF1R) antagonists that showed significant effectiveness in animal models of AUD but were largely ineffective in humans. The present article attempts to analyze the most recent venues in the development of new medications in AUD with a focus on the most promising drug targets under current exploration. Moreover, we delineate the importance of using a more integrated translational framework approach to correlate preclinical findings and early clinical data to enhance the probability to validate biological targets of interest.
Collapse
Affiliation(s)
- Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Alessio Masi
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Massimo Bramucci
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Marisa Roberto
- The Scripps Research Institute, Department of Neuroscience, La Jolla, CA, USA
| | - Angelo Bifone
- Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy; Department of Molecular Biotechnology and Health Science, University of Torino, Italy
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy.
| |
Collapse
|
10
|
Pujol CN, Paasche C, Laprevote V, Trojak B, Vidailhet P, Bacon E, Lalanne L. Cognitive effects of labeled addictolytic medications. Prog Neuropsychopharmacol Biol Psychiatry 2018; 81:306-332. [PMID: 28919445 DOI: 10.1016/j.pnpbp.2017.09.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/11/2017] [Accepted: 09/11/2017] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Alcohol, tobacco, and illegal drug usage is pervasive throughout the world, and abuse of these substances is a major contributor to the global disease burden. Many pharmacotherapies have been developed over the last 50years to target addictive disorders. While the efficacy of these pharmacotherapies is largely recognized, their cognitive impact is less known. However, all substance abuse disorders are known to promote cognitive disorders like executive dysfunction and memory impairment. These impairments are critical for the maintenance of addictive behaviors and impede cognitive behavioral therapies that are regularly administered in association with pharmacotherapies. It is also unknown if addictolytic medications have an impact on preexisting cognitive disorders, and if this impact is modulated by the indication of prescription, i.e. abstinence, reduction or substitution, or by the specific action of the medication. METHOD We reviewed the cognitive effects of labeled medications for tobacco addiction (varenicline, bupropion, nicotine patch and nicotine gums), alcohol addiction (naltrexone, nalmefene, baclofen, disulfiram, sodium oxybate, acamprosate), and opioid addiction (methadone, buprenorphine) in human studies. Studies were selected following MOOSE guidelines for systematic reviews of observational studies, using the keywords [Cognition] and [Cognitive disorders] and [treatment] for each medication. RESULTS 971 articles were screened and 77 studies met the inclusion criteria and were reported in this review (for alcohol abuse, n=21, for tobacco n=22, for opioid n=34. However, very few comparative clinical trials have explored the chronic effects of addictolytic medications on cognition in addictive behaviors, and there are no clinical trials on the cognitive impact of nalmefene in patients suffering from alcohol use disorders. DISCUSSION Although some medications seem to enhance cognition in patients suffering from cognitive disorders, others could promote cognitive impairments, and our work highlights a lack of literature on this subject. In conclusion, more comparative clinical trials are needed to better understand the cognitive impact of addictolytic medications.
Collapse
Affiliation(s)
- Camille Noélie Pujol
- Department of Neurosciences, Institute for Functional Genomics, INSERM U-661, CNRS UMR-5203, 34094 Montpellier, France
| | - Cecilia Paasche
- INSERM 1114, Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Vincent Laprevote
- Centre Psychothérapique de Nancy, Laxou, F-54520, France.; EA 7298, INGRES, Université de Lorraine, Vandoeuvre-lès-, Nancy F-54000, France; CHU Nancy, Maison des Addictions, Nancy, F-54000, France.
| | - Benoit Trojak
- Department of Psychiatry and Addictology, University Hospital of Dijon, France; EA 4452, LPPM, University of Burgundy, France.
| | - Pierre Vidailhet
- INSERM 1114, Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France; Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France..
| | - Elisabeth Bacon
- INSERM 1114, Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France.
| | - Laurence Lalanne
- INSERM 1114, Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France; Department of Psychiatry and Addictology, University Hospital of Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France..
| |
Collapse
|
11
|
Hendershot CS, Wardell JD, Samokhvalov AV, Rehm J. Effects of naltrexone on alcohol self-administration and craving: meta-analysis of human laboratory studies. Addict Biol 2017; 22:1515-1527. [PMID: 27411969 PMCID: PMC6139429 DOI: 10.1111/adb.12425] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 05/19/2016] [Accepted: 06/21/2016] [Indexed: 12/21/2022]
Abstract
Randomized clinical trials have established the efficacy of naltrexone for reducing quantity of alcohol consumption and incidence of relapse to heavy drinking. To evaluate putative treatment mechanisms, human laboratory studies have examined naltrexone's effects on alcohol responses and self-administration during short-term medication protocols. Results from these studies are inconsistent and have yet to be examined in aggregate. This meta-analysis aimed to quantify naltrexone's effects on alcohol self-administration and craving in the context of placebo-controlled human laboratory trials. Potential moderators of medication effects were also examined. Meta-analyses of alcohol self-administration (k = 9, N = 490) and craving (k = 16, N = 748) confirmed that, under controlled experimental conditions, naltrexone reduces the quantity of consumption (Hedges' g = -.277, SE = .074, 95 percent CI = -.421, -.133, p < .001) and magnitude of self-reported craving (g = -.286, SE = .066, 95 percent CI = -.416, -.156, p < .001) relative to placebo. Subgroup and moderation analyses found no evidence that effect sizes differed by study population (dependent versus non-dependent drinkers), laboratory paradigm or duration of medication exposure. These results substantiate prior evidence for reductions in event-level craving and consumption as potential treatment mediators, also establishing effect sizes to inform future human laboratory trials. From a clinical perspective, these results may provide additional evidence regarding naltrexone's efficacy in the context of acute or subacute dosing regimens.
Collapse
Affiliation(s)
- Christian S. Hendershot
- Campbell Family Mental Health Research InstituteCentre for Addiction and Mental HealthCanada
- Department of Psychiatry, University of TorontoCanada
- Department of PsychologyUniversity of TorontoCanada
- Institute for Mental Health Policy ResearchCentre for Addiction and Mental HealthCanada
| | - Jeffrey D. Wardell
- Campbell Family Mental Health Research InstituteCentre for Addiction and Mental HealthCanada
| | - Andriy V. Samokhvalov
- Department of Psychiatry, University of TorontoCanada
- Institute for Mental Health Policy ResearchCentre for Addiction and Mental HealthCanada
- Institute for Medical SciencesUniversity of TorontoCanada
| | - Jürgen Rehm
- Campbell Family Mental Health Research InstituteCentre for Addiction and Mental HealthCanada
- Department of Psychiatry, University of TorontoCanada
- Institute for Mental Health Policy ResearchCentre for Addiction and Mental HealthCanada
- Dalla Lana School of Public HealthUniversity of TorontoCanada
- Technische UniversitätGermany
| |
Collapse
|
12
|
Naloxone effects on extinction of ethanol- and cocaine-induced conditioned place preference in mice. Psychopharmacology (Berl) 2017; 234:2747-2759. [PMID: 28653079 PMCID: PMC5709191 DOI: 10.1007/s00213-017-4672-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/06/2017] [Indexed: 01/07/2023]
Abstract
RATIONALE Previous studies found that naloxone (NLX) facilitated choice extinction of ethanol conditioned place preference (CPP) using long (60 min) test sessions, but there is little information on the variables determining this effect. OBJECTIVES These studies examined repeated exposure to NLX during extinction of ethanol- or cocaine-induced CPP using both short and long tests. METHODS DBA/2J mice were injected with NLX (0 or 10 mg/kg) before three 10- or 60-min choice extinction tests (experiment 1). All mice received a final 60-min test without NLX. Post-test NLX was given in experiment 2. Experiment 3 tested whether NLX would affect a forced extinction procedure. Experiment 4 tested its effect on extinction of cocaine-induced CPP. RESULTS Pre-test (but not post-test) injections of NLX-facilitated choice extinction of ethanol CPP at both test durations. Pre-test NLX also facilitated forced extinction. However, pre-test NLX had no effect on choice extinction of cocaine CPP. CONCLUSIONS Extinction test duration is not critical for engaging the opioid system during ethanol CPP extinction (experiment 1). Moreover, NLX's effect does not depend on CPP expression during extinction, just exposure to previously conditioned cues (experiment 3). The null effect of post-test NLX eliminates a memory consolidation interpretation (experiment 2) and the failure to alter cocaine CPP extinction argues against alteration of general learning or memory processes (experiment 4). Overall, these data suggest that the endogenous opioid system mediates a conditioned motivational effect that normally maintains alcohol-induced seeking behavior, which may underlie the efficacy of opiate antagonists in the treatment of alcoholism.
Collapse
|
13
|
Lichtenberg NT, Wassum KM. Amygdala mu-opioid receptors mediate the motivating influence of cue-triggered reward expectations. Eur J Neurosci 2017; 45:381-387. [PMID: 27862489 PMCID: PMC5293612 DOI: 10.1111/ejn.13477] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 01/16/2023]
Abstract
Environmental reward-predictive stimuli can retrieve from memory a specific reward expectation that allows them to motivate action and guide choice. This process requires the basolateral amygdala (BLA), but little is known about the signaling systems necessary within this structure. Here we examined the role of the neuromodulatory opioid receptor system in the BLA in such cue-directed action using the outcome-specific Pavlovian-to-instrumental transfer (PIT) test in rats. Inactivation of BLA mu-, but not delta-opioid receptors was found to dose-dependently attenuate the ability of a reward-predictive cue to selectively invigorate the performance of actions directed at the same unique predicted reward (i.e. to express outcome-specific PIT). BLA mu-opioid receptor inactivation did not affect the ability of a reward itself to similarly motivate action (outcome-specific reinstatement), suggesting a more selective role for the BLA mu-opioid receptor in the motivating influence of currently unobservable rewarding events. These data reveal a new role for BLA mu-opioid receptor activation in the cued recall of precise reward memories and the use of this information to motivate specific action plans.
Collapse
Affiliation(s)
- Nina T Lichtenberg
- Department of Psychology, UCLA, 1285 Franz Hall, Box 951563, Los Angeles, CA, 90095, USA
| | - Kate M Wassum
- Department of Psychology, UCLA, 1285 Franz Hall, Box 951563, Los Angeles, CA, 90095, USA
- Brain Research Institute, UCLA, Los Angeles, CA, USA
| |
Collapse
|
14
|
Potthast N, Neuner F, Catani C. Automatic activation of alcohol cues by child maltreatment related words: a replication attempt in a different treatment setting. BMC Res Notes 2017; 10:17. [PMID: 28057039 PMCID: PMC5217616 DOI: 10.1186/s13104-016-2324-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 12/07/2016] [Indexed: 11/10/2022] Open
|
15
|
Metrik J, Aston ER, Kahler CW, Rohsenow DJ, McGeary JE, Knopik VS, MacKillop J. Cue-elicited increases in incentive salience for marijuana: Craving, demand, and attentional bias. Drug Alcohol Depend 2016; 167:82-8. [PMID: 27515723 PMCID: PMC5037029 DOI: 10.1016/j.drugalcdep.2016.07.027] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/26/2016] [Accepted: 07/26/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND Incentive salience is a multidimensional construct that includes craving, drug value relative to other reinforcers, and implicit motivation such as attentional bias to drug cues. Laboratory cue reactivity (CR) paradigms have been used to evaluate marijuana incentive salience with measures of craving, but not with behavioral economic measures of marijuana demand or implicit attentional processing tasks. METHODS This within-subjects study used a new CR paradigm to examine multiple dimensions of marijuana's incentive salience and to compare CR-induced increases in craving and demand. Frequent marijuana users (N=93, 34% female) underwent exposure to neutral cues then to lit marijuana cigarettes. Craving, marijuana demand via a marijuana purchase task, and heart rate were assessed after each cue set. A modified Stroop task with cannabis and control words was completed after the marijuana cues as a measure of attentional bias. RESULTS Relative to neutral cues, marijuana cues significantly increased subjective craving and demand indices of intensity (i.e., drug consumed at $0) and Omax (i.e., peak drug expenditure). Elasticity significantly decreased following marijuana cues, reflecting sustained purchase despite price increases. Craving was correlated with demand indices (r's: 0.23-0.30). Marijuana users displayed significant attentional bias for cannabis-related words after marijuana cues. Cue-elicited increases in intensity were associated with greater attentional bias for marijuana words. CONCLUSIONS Greater incentive salience indexed by subjective, behavioral economic, and implicit measures was observed after marijuana versus neutral cues, supporting multidimensional assessment. The study highlights the utility of a behavioral economic approach in detecting cue-elicited changes in marijuana incentive salience.
Collapse
Affiliation(s)
- Jane Metrik
- Center for Alcohol and Addiction Studies, Brown University School of Public Health, Providence, RI, 02903, USA; Providence Veterans Affairs Medical Center, Providence, RI, 02908, USA.
| | - Elizabeth R. Aston
- Center for Alcohol and Addiction Studies, Brown University School of Public Health, Providence, RI, 02903
| | - Christopher W. Kahler
- Center for Alcohol and Addiction Studies, Brown University School of Public Health, Providence, RI, 02903
| | - Damaris J. Rohsenow
- Center for Alcohol and Addiction Studies, Brown University School of Public Health, Providence, RI, 02903,Providence Veterans Affairs Medical Center, Providence, RI, 02908
| | - John E. McGeary
- Providence Veterans Affairs Medical Center, Providence, RI, 02908,Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903
| | - Valerie S. Knopik
- Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903
| | - James MacKillop
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, L8N 3K7 Canada
| |
Collapse
|
16
|
Magnitude and duration of cue-induced craving for marijuana in volunteers with cannabis use disorder. Drug Alcohol Depend 2016; 166:143-9. [PMID: 27436749 PMCID: PMC5113710 DOI: 10.1016/j.drugalcdep.2016.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 06/30/2016] [Accepted: 07/04/2016] [Indexed: 11/22/2022]
Abstract
AIMS Evaluate magnitude and duration of subjective and physiologic responses to neutral and marijuana (MJ)-related cues in cannabis dependent volunteers. METHODS 33 volunteers (17 male) who met DSM-IV criteria for Cannabis Abuse or Dependence were exposed to neutral (first) then MJ-related visual, auditory, olfactory and tactile cues. Mood, drug craving and physiology were assessed at baseline, post-neutral, post-MJ and 15-min post MJ cue exposure to determine magnitude of cue- responses. For a subset of participants (n=15; 9 male), measures of craving and physiology were collected also at 30-, 90-, and 150-min post-MJ cue to examine duration of cue-effects. RESULTS In cue-response magnitude analyses, visual analog scale (VAS) items craving for, urge to use, and desire to smoke MJ, Total and Compulsivity subscale scores of the Marijuana Craving Questionnaire, anxiety ratings, and diastolic blood pressure (BP) were significantly elevated following MJ vs. neutral cue exposure. In cue-response duration analyses, desire and urge to use MJ remained significantly elevated at 30-, 90- and 150-min post MJ-cue exposure, relative to baseline and neutral cues. CONCLUSIONS Presentation of polysensory MJ cues increased MJ craving, anxiety and diastolic BP relative to baseline and neutral cues. MJ craving remained elevated up to 150-min after MJ cue presentation. This finding confirms that carry-over effects from drug cue presentation must be considered in cue reactivity studies.
Collapse
|
17
|
Haass-Koffler CL, Giovenco DE, Lee MR, Zywiak WH, de la Monte SM, Kenna GA, Swift RM, Leggio L. Serum Insulin Levels Are Reduced by Intravenous Ghrelin Administration but Do Not Correlate with Alcohol Craving in Alcohol-Dependent Individuals. Int J Neuropsychopharmacol 2016; 19:pyw048. [PMID: 27207912 PMCID: PMC5091823 DOI: 10.1093/ijnp/pyw048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/25/2016] [Accepted: 05/03/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Increasing evidence supports a role for appetite-regulating pathways like ghrelin, insulin, and leptin in alcoholism. We previously reported that intravenous (i.v.) exogenous ghrelin increases alcohol craving. We also reported i.v. ghrelin reduces endogenous serum leptin, whose levels, in turn, negatively correlated with alcohol craving. Exogenous ghrelin administration decreases insulin secretion both in vitro and in vivo experiments. This study tested the hypothesis that i.v. ghrelin may also decrease endogenous serum insulin levels in alcoholic individuals. Additionally, we explored possible correlations between serum insulin and alcohol craving, since a correlation between insulin and alcohol craving was previously reported. METHODS This was a double-blind, placebo-controlled human laboratory study ( n =43). Non-treatment-seeking, alcohol-dependent, heavy drinkers were randomized to receive i.v. ghrelin or placebo, followed by an alcohol cue-reactivity procedure. RESULTS There was a main effect for i.v. ghrelin, compared to placebo in reducing serum insulin ( P <.05). There was also a time effect ( P <.001) but not ghrelin x time interaction ( P >.05). We did not find a correlation between the reduction of serum insulin and alcohol craving ( P >.05). The change in serum insulin was consistent with a parallel reduction in serum connective-peptide in the ghrelin group compared with placebo, although this difference did not reach statistical significance ( P =.076). No similar effects were found for other glucose-regulating hormones analyzed i.e. glucagon, glucagon-like peptide-1, and gastric inhibitory peptide ( P s>.05). CONCLUSIONS These findings indicate i.v. ghrelin administration has an effect on reducing serum insulin in alcohol-dependent individuals; however, the reduction of insulin did not correlate with changes in alcohol cue-elicited craving. We speculate that, unlike for leptin, the interactions between ghrelin and insulin relationship are limited at the peripheral level. However, mechanistic studies are needed to investigate this hypothesis.
Collapse
|
18
|
Giuliano C, Goodlett CR, Economidou D, García-Pardo MP, Belin D, Robbins TW, Bullmore ET, Everitt BJ. The Novel μ-Opioid Receptor Antagonist GSK1521498 Decreases Both Alcohol Seeking and Drinking: Evidence from a New Preclinical Model of Alcohol Seeking. Neuropsychopharmacology 2015; 40:2981-92. [PMID: 26044906 PMCID: PMC4864633 DOI: 10.1038/npp.2015.152] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 02/02/2023]
Abstract
Distinct environmental and conditioned stimuli influencing ethanol-associated appetitive and consummatory behaviors may jointly contribute to alcohol addiction. To develop an effective translational animal model that illuminates this interaction, daily seeking responses, maintained by alcohol-associated conditioned stimuli (CSs), need to be dissociated from alcohol drinking behavior. For this, we established a procedure whereby alcohol seeking maintained by alcohol-associated CSs is followed by a period during which rats have the opportunity to drink alcohol. This cue-controlled alcohol-seeking procedure was used to compare the effects of naltrexone and GSK1521498, a novel selective μ-opioid receptor antagonist, on both voluntary alcohol-intake and alcohol-seeking behaviors. Rederived alcohol-preferring, alcohol-nonpreferring, and high-alcohol-drinking replicate 1 line of rats (Indiana University) first received 18 sessions of 24 h home cage access to 10% alcohol and water under a 2-bottle choice procedure. They were trained subsequently to respond instrumentally for access to 15% alcohol under a second-order schedule of reinforcement, in which a prolonged period of alcohol-seeking behavior was maintained by contingent presentations of an alcohol-associated CS acting as a conditioned reinforcer. This seeking period was terminated by 20 min of free alcohol drinking access that achieved significant blood alcohol concentrations. The influence of pretreatment with either naltrexone (0.1-1-3 mg/kg) or GSK1521498 (0.1-1-3 mg/kg) before instrumental sessions was measured on both seeking and drinking behaviors, as well as on drinking in the 2-bottle choice procedure. Naltrexone and GSK1521498 dose-dependently reduced both cue-controlled alcohol seeking and alcohol intake in the instrumental context as well as alcohol intake in the choice procedure. However, GSK1521498 showed significantly greater effectiveness than naltrexone, supporting its potential use for promoting abstinence and preventing relapse in alcohol addiction.
Collapse
Affiliation(s)
- Chiara Giuliano
- Behavioral and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Cambridge, UK,Department of Psychology, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK, Tel: +44 (0)1223 65292, Fax: +44 (0)1223 333564, E-mail:
| | - Charles R Goodlett
- Department of Psychology, Indiana University–Purdue University Indianapolis, Indianapolis, IN, USA
| | - Daina Economidou
- Behavioral and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Cambridge, UK
| | - Maria P García-Pardo
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, School of Psychology, University of Valencia, Valencia, Spain
| | - David Belin
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Trevor W Robbins
- Behavioral and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Cambridge, UK
| | - Edward T Bullmore
- Behavioral and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Cambridge, UK,Department of Psychiatry, University of Cambridge, Cambridge, UK,Clinical Unit Cambridge and Academic DPU, GlaxoSmithKline R&D, Clinical Unit Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Barry J Everitt
- Behavioral and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Cambridge, UK
| |
Collapse
|
19
|
Haass-Koffler CL, Aoun EG, Swift RM, de la Monte SM, Kenna GA, Leggio L. Leptin levels are reduced by intravenous ghrelin administration and correlated with cue-induced alcohol craving. Transl Psychiatry 2015; 5:e646. [PMID: 26418274 PMCID: PMC5545639 DOI: 10.1038/tp.2015.140] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/08/2015] [Accepted: 06/01/2015] [Indexed: 01/24/2023] Open
Abstract
Increasing evidence supports the role of appetite-regulating pathways, including ghrelin and leptin, in alcoholism. This study tested the hypothesis that intravenous exogenous ghrelin administration acutely decreases endogenous serum leptin levels, and that changes in leptin levels negatively correlate with alcohol craving. This was a double-blind, placebo-controlled human laboratory study. Non-treatment-seeking, alcohol-dependent, heavy drinkers (n=45) were randomized to receive intravenous ghrelin or placebo, followed by a cue-reactivity procedure, during which participants were exposed to neutral (juice) and alcohol trial cues. There was a main effect for intravenous ghrelin administration, compared with placebo, in reducing serum leptin levels (P<0.01). Post hoc analysis showed significant differences in serum leptin levels at the alcohol trial (P<0.05) that persisted at the end of the experiment (P<0.05). By contrast, there were no significant differences in serum leptin levels at the juice trial (P=not significant (NS)). The change of serum leptin level at the alcohol trial correlated with the increase in alcohol urge (P<0.05), whereas urge to drink juice was not correlated with the leptin change at the juice trial (P=NS). These findings provide preliminary evidence of ghrelin-leptin cross-talk in alcoholic individuals and suggest that their relationship may have a role in alcohol craving.
Collapse
Affiliation(s)
- C L Haass-Koffler
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA,Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
| | - E G Aoun
- Department of Psychiatry and Human Behavior, Brown University, Providence, RI, USA
| | - R M Swift
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI, USA,Veterans Affairs Medical Center, Providence, RI, USA
| | - S M de la Monte
- Departments of Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - G A Kenna
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI, USA
| | - L Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA,Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA,Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, 10 Center Drive (10CRC/15330) MSC 1108, Room 1-5429, Bethesda, MD 20892-1108, USA. E-mail:
| |
Collapse
|
20
|
Lundahl LH, Greenwald MK. Effect of oral THC pretreatment on marijuana cue-induced responses in cannabis dependent volunteers. Drug Alcohol Depend 2015; 149:187-93. [PMID: 25725933 PMCID: PMC10369174 DOI: 10.1016/j.drugalcdep.2015.01.046] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/30/2015] [Accepted: 01/31/2015] [Indexed: 10/24/2022]
Abstract
BACKGROUND The current study tested whether oral Δ(9)-tetrahydrocannabinol (THC: 0-, 10-, and 20-mg) pretreatment would attenuate polysensory cue-induced craving for marijuana. METHODS Cannabis dependent participants (7 males and 7 females, who smoked on average 5.4 ± 1.1 blunts daily) completed 3 experimental sessions (oral THC pretreatment dose; counterbalanced order) using a placebo-controlled within-subject crossover design. During each session, participants completed a baseline evaluation and were first exposed to neutral cues then marijuana cues while physiological measures and subjective ratings of mood, craving, and drug effect were recorded. RESULTS Following placebo oral THC pretreatment, marijuana (vs. neutral) cues significantly increased ratings of marijuana craving (desire and urge to use, Marijuana Craving Questionnaire (MCQ)-Compulsivity scale), anxious mood and feeling hungry. Males also reported feeling more "Down" during marijuana cues relative to females. Pretreatment with oral THC (10-mg and/or 20-mg vs. placebo) significantly attenuated marijuana cue-induced increases in craving and anxiety but not hunger. Oral THC attenuation of the cue-induced increase in MCQ-Compulsivity ratings was observed in females only. Oral THC produced statistically (but not clinically) significant increases in heart rate and decreases in diastolic blood pressure, independent of cues. CONCLUSIONS These marijuana-cue findings replicate earlier results and further demonstrate that oral THC can attenuate selected effects during marijuana multi-cue exposure, and that some of these effects may be sex-related. Results of this study suggest oral THC may be effective for reducing marijuana cue-elicited (conditioned) effects. Further study is needed to determine whether females may selectively benefit from oral THC for this purpose.
Collapse
Affiliation(s)
- Leslie H Lundahl
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI, USA.
| | - Mark K Greenwald
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI, USA; Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| |
Collapse
|
21
|
Alleviating waiting impulsivity and perseverative responding by μ-opioid receptor antagonism in two inbred mouse strains. Psychopharmacology (Berl) 2015; 232:1483-92. [PMID: 25381183 DOI: 10.1007/s00213-014-3786-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/18/2014] [Indexed: 10/24/2022]
Abstract
RATIONALE Recent evidence has implicated the opioid system in exaggerated ethanol consumption and impulsivity deficits. The opioid receptor antagonist naltrexone (NTX) has proven efficient in reducing alcohol consumption; however, its role on impulsive behaviour is not fully characterised. OBJECTIVE The aim of this study was to investigate the effects of NTX on two measures of impulsive behaviour in two inbred mouse strains that differ in ethanol preference and impulsive phenotype. METHODS Two separate groups of C57BL/6J (B6, n = 24) and DBA2/J (D2, n = 24) male mice were exposed to intermittent ethanol (IEE; 2 g/kg) during early (PND 30-45, IEE_Early) or late (PND 45-60, IEE_Late) adolescence or the respective saline control. The ability of NTX (10 mg/kg) alone, or co-administered with ethanol (0.5 g/kg), to diminish waiting impulsivity in the five-choice serial reaction time task (5-CSRTT), or improve decision-making in a mouse version of the Iowa Gambling Task (mIGT), was examined in adulthood. RESULTS In the 5-CSRTT, NTX diminished impulsivity in both strains of mice, irrespective of previous ethanol experience. In the mIGT, NTX failed to alter risky decision-making but decreased perseverative responding. CONCLUSIONS Blocking the actions of endogenous opioids may attenuate waiting impulsivity, in addition to alleviating perseverative responding. In a broader context, μ-opiate antagonism may be of potential interest for impulse-control disorders.
Collapse
|
22
|
Leggio L, Zywiak WH, Edwards SM, Tidey JW, Swift RM, Kenna GA. A preliminary double-blind, placebo-controlled randomized study of baclofen effects in alcoholic smokers. Psychopharmacology (Berl) 2015; 232:233-43. [PMID: 24973894 PMCID: PMC4278944 DOI: 10.1007/s00213-014-3652-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 06/04/2014] [Indexed: 12/18/2022]
Abstract
RATIONALE There is presently no approved single treatment for dual alcohol and nicotine dependencies. OBJECTIVE This pilot study investigated baclofen effects in alcoholic smokers. METHODS This was a preliminary double-blind placebo-controlled randomized clinical study with 30 alcoholic smokers randomized to baclofen at 80 mg/day or placebo. A subgroup (n=18) participated in an alcohol cue-reactivity experiment. RESULTS Baclofen, compared with placebo, significantly decreased the percent days of abstinence from alcohol-tobacco co-use (p=0.004). Alcohol dependence severity moderated baclofen effects, with the higher severity group having the greater baclofen response (p<0.001). Although the percent days of alcohol-tobacco co-use declined in both groups, this decline was greater after placebo than baclofen (p<0.001). Secondary analyses on alcohol or tobacco use alone suggested that the increase in percent days of co-abstinence was driven by the medication differences on heavy drinking days and on percent days smoking. In the cue-reactivity substudy, baclofen slightly decreased alcohol urge (p=0.058) and significantly reduced salivation (p=0.001), but these effects were not related to cue type. CONCLUSIONS This study provides preliminary evidence suggesting a possible role of baclofen in the treatment of alcoholic smokers. However, the mixed results and the small sample require larger confirmatory studies.
Collapse
Affiliation(s)
- Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 10 Center Drive (10CRC/15330) MSC 1108; Room 1-5429, Bethesda, MD, 20892-1108, USA,
| | - William H. Zywiak
- Decision Sciences Institute, P.I.R.E, Pawtucket, RI, USA. Butler Hospital, Providence, RI, USA. Department of Psychiatry and Human Behavior, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
| | - Steven M. Edwards
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jennifer W. Tidey
- Department of Psychiatry and Human Behavior, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
| | - Robert M. Swift
- Department of Psychiatry and Human Behavior, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA. Veterans Affairs Medical Center, Providence, RI, USA. Roger Williams Medical Center, Providence, RI, USA
| | - George A. Kenna
- Department of Psychiatry and Human Behavior, Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
| |
Collapse
|
23
|
Leggio L, Zywiak WH, Fricchione SR, Edwards SM, de la Monte SM, Swift RM, Kenna GA. Intravenous ghrelin administration increases alcohol craving in alcohol-dependent heavy drinkers: a preliminary investigation. Biol Psychiatry 2014; 76:734-41. [PMID: 24775991 PMCID: PMC4176606 DOI: 10.1016/j.biopsych.2014.03.019] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/14/2014] [Accepted: 03/14/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND There is a need to identify novel pharmacologic targets to treat alcoholism. Animal and human studies suggest a role for ghrelin in the neurobiology of alcohol dependence and craving. Here, we were the first to test the hypothesis that intravenous administration of exogenous ghrelin acutely increases alcohol craving. METHODS This was a double-blind, placebo-controlled human laboratory proof-of-concept study. Nontreatment-seeking, alcohol-dependent, heavy-drinking individuals were randomized to receive intravenous ghrelin 1 mcg/kg, 3 mcg/kg or 0 mcg/kg (placebo), followed by a cue-reactivity procedure, during which participants were exposed to neutral (juice) and alcohol cues. The primary outcome variable was the increase in alcohol craving (also called urge) for alcohol, assessed by the Alcohol Visual Analogue Scale. RESULTS Out of 103 screenings, 45 individuals received the study drug. Repeated measures of analysis of covariance revealed a group effect across ghrelin doses in increasing alcohol craving (p < .05). A dose-specific examination revealed a significant effect of ghrelin 3 mcg/kg versus placebo in increasing alcohol craving (p < .05) with a large effect size (d = .94). By contrast, no significant ghrelin effect was found in increasing either urge to drink juice or food craving (p = ns). No significant differences in side effects were found (p = ns). CONCLUSIONS Intravenous administration of exogenous ghrelin increased alcohol craving in alcohol-dependent heavy-drinking individuals. Although the small sample requires confirmatory studies, these findings provide preliminary evidence that ghrelin may play a role in the neurobiology of alcohol craving, thus demonstrating a novel pharmacologic target for treatment.
Collapse
Affiliation(s)
- Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland; Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland; Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence.
| | - William H. Zywiak
- Decision Sciences Institute, P.I.R.E., Pawtucket, RI,Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI
| | - Samuel R. Fricchione
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI
| | - Steven M. Edwards
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE
| | - Suzanne M. de la Monte
- Departments of Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI
| | - Robert M. Swift
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI,Veterans Affairs Medical Center, Providence, RI
| | - George A. Kenna
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI
| |
Collapse
|
24
|
Ciccocioppo R, Stopponi S, Economidou D, Kuriyama M, Kinoshita H, Heilig M, Roberto M, Weiss F, Teshima K. Chronic treatment with novel brain-penetrating selective NOP receptor agonist MT-7716 reduces alcohol drinking and seeking in the rat. Neuropsychopharmacology 2014; 39:2601-10. [PMID: 24863033 PMCID: PMC4207340 DOI: 10.1038/npp.2014.113] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 03/31/2014] [Accepted: 04/22/2014] [Indexed: 11/09/2022]
Abstract
Since its discovery, the nociceptin/orphanin FQ (N/OFQ)-NOP receptor system has been extensively investigated as a promising target to treat alcoholism. Encouraging results obtained with the endogenous ligand N/OFQ stimulated research towards the development of novel brain-penetrating NOP receptor agonists with a pharmacological and toxicological profile compatible with clinical development. Here we describe the biochemical and alcohol-related behavioral effects of the novel NOP receptor agonist MT-7716. MT-7716 has high affinity for human NOP receptors expressed in HEK293 cells with a Ki value of 0.21 nM. MT-7716 concentration-dependently stimulated GTPγ(35)S binding with an EC50 value of 0.30 nM and its efficacy was similar to N/OFQ, suggesting that MT7716 is a full agonist at NOP receptors. In the two bottle choice test MT-7716 (0, 0.3, 1, and 3 mg/kg, bid) given orally for 14 days dose-dependently decreased voluntary alcohol intake in Marchigian Sardinian rats. The effect became gradually stronger following repeated administration, and was still significant 1 week after discontinuation of the drug. Oral naltrexone (30 mg/kg, bid) for 14 days also reduced ethanol intake; however, the effect decreased over the treatment period and rapidly disappeared when drug treatment was discontinued. MT-7716 is also effective for preventing reinstatement caused by both ethanol-associated environmental stimuli and stress. Finally, to investigate the effect of MT-7716 on alcohol withdrawal symptoms, Wistar rats were withdrawn from a 7-day alcohol liquid diet. MT-7716 significantly attenuated somatic alcohol withdrawal symptoms. Together these findings indicate that MT-7716 is a promising candidate for alcoholism treatment remaining effective with chronic administration.
Collapse
Affiliation(s)
- Roberto Ciccocioppo
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Serena Stopponi
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Daina Economidou
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Makoto Kuriyama
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Hiroshi Kinoshita
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Markus Heilig
- Laboratory of Clinical and Translational Studies, National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Koji Teshima
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| |
Collapse
|
25
|
Ramirez J, Miranda R. Alcohol craving in adolescents: bridging the laboratory and natural environment. Psychopharmacology (Berl) 2014; 231:1841-51. [PMID: 24363093 PMCID: PMC4127892 DOI: 10.1007/s00213-013-3372-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 11/20/2013] [Indexed: 12/11/2022]
Abstract
RATIONALE Initial lab studies suggest that adolescent drinkers crave alcohol when presented with alcohol cues. Whether this effect generalizes to the natural environment, however, remains unknown, and studies have not examined whether craving predicts drinking among youths. OBJECTIVES This study builds on existing research by pairing controlled lab-based cue reactivity assessments (CRAs) with data collected in the natural environment using ecological momentary assessment (EMA) methods. We examined whether alcohol cues evoke craving among adolescent drinkers in the lab and natural environment, and tested the clinical relevance of craving during adolescence by examining the prospective association between craving and alcohol use. METHODS Non-treatment-seeking adolescent drinkers (N = 42; ages 15 to 20 years) completed a lab-based CRA followed by a 1-week EMA monitoring period. During the EMA period, youth were prompted randomly throughout the day to record momentary data on craving and contextual factors (e.g., alcohol cues, peers present). RESULTS Alcohol cues elicited craving in the lab, and this effect generalized to the natural environment, especially among adolescents with more alcohol problems. In addition, craving predicted subsequent drinking levels in the natural environment. CONCLUSIONS This study demonstrates the utility of pairing lab paradigms with EMA methods to better characterize adolescents' reactivity to alcohol cues. Results implicate craving as a clinically meaningful motivator for drinking among adolescents and highlight a potentially important target of pharmacological or behavioral intervention.
Collapse
Affiliation(s)
- Jason Ramirez
- Center for Alcohol and Addiction Studies, Brown University, Box S121-5, Providence, RI, 02912, USA
| | | |
Collapse
|
26
|
Lieb M, Palm U, Chiang S, Laubender RP, Nothdurfter C, Sarubin N, Mokhtari-Nejad R, Koller G, Soyka M. Pharmacological challenge with naloxone and cue exposure in alcohol dependence: results of a randomized, double-blind placebo-controlled trial. World J Biol Psychiatry 2013; 14:539-46. [PMID: 24020866 DOI: 10.3109/15622975.2013.812239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Animal and clinical studies implicated opioid dysfunction in the pathogenesis of alcohol abuse and dependence. The π-opioid antagonist naltrexone reduces craving, eventually modulated by hypothalamic-pituitary-adrenal axis. Altered cortisol response to opioid receptor blockade not only in alcohol dependent persons, but also in persons with a family history of alcohol dependency was reported. METHODS Twenty patients with alcohol dependence who had undergone detoxification were recruited. Naloxone (3.2 mg/70 kg body weight) having a very similar receptor profile to naltrexone and placebo were administered in cross-over fashion on two separate days 48 h apart. Mood and craving was assessed with well-established instruments (Alcohol Craving Questionnaire (ACQ), Profile of Mood Scale (POMS)). Both patients and raters were blind to all treatments. Twelve patients were first treated with naloxone, eight were first treated with placebo. RESULTS No significant differences were found between the placebo and naloxone groups according to ACQ and POMS. Cortisol levels were significantly higher in naloxone group. CONCLUSIONS We could not replicate the result, that blocking of the endogenous opioid system leads to reduced craving in alcohol-dependent individuals, while increase of cortisol after naloxone challenge is the expected biological effect of opioid receptor blockade on the hypothalamic-pituitary-adrenal (HPA) axis.
Collapse
Affiliation(s)
- Martin Lieb
- Department of Psychiatry, Bezirksklinikum Regensburg , Regensburg , Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Stopponi S, de Guglielmo G, Somaini L, Cippitelli A, Cannella N, Kallupi M, Ubaldi M, Heilig M, Demopulos G, Gaitanaris G, Ciccocioppo R. Activation of PPARγ by pioglitazone potentiates the effects of naltrexone on alcohol drinking and relapse in msP rats. Alcohol Clin Exp Res 2013; 37:1351-60. [PMID: 23550625 DOI: 10.1111/acer.12091] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 01/08/2013] [Indexed: 12/15/2022]
Abstract
BACKGROUND Pioglitazone is a selective peroxisome proliferator-activated receptor γ (PPARγ) agonist used for the treatment of insulin resistance and type 2 diabetes. Previous studies conducted in our laboratory showed that activation of PPARγ by pioglitazone reduces alcohol drinking, stress-induced relapse, and alcohol withdrawal syndrome in rats. Pioglitazone was not able to prevent relapse elicited by alcohol cues. Conversely, the nonselective opioid antagonist naltrexone has been shown to reduce alcohol drinking and cue- but not stress-induced relapse in rodents. METHODS Based on these findings, this study was sought to determine the efficacy of pioglitazone and naltrexone combination on alcohol intake and relapse behavior. Genetically selected alcohol-preferring Marchigian Sardinian (msP) rats were used for the study. RESULTS Pioglitazone (10 and 30 mg/kg) and naltrexone (0.25 and 1.0 mg/kg) each individually reduced alcohol drinking in msP rats. The combination of the 2 drugs resulted in a more potent alcohol drinking reduction than single agents. Confirming previous studies, pioglitazone (10 and 30 mg/kg) significantly reduced relapse induced by the pharmacological stressor yohimbine (1.25 mg/kg) but not by cues predictive of alcohol availability. Conversely, naltrexone reduced reinstatement of drug seeking elicited by alcohol cues but not by yohimbine. CONCLUSIONS The drug combination was effective in reducing both relapse behaviors. These findings open new vistas in the use pioglitazone in combination with naltrexone for the treatment of alcoholism.
Collapse
Affiliation(s)
- Serena Stopponi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hofmann SG, Hüweler R, MacKillop J, Kantak KM. Effects of d-Cycloserine on Craving to Alcohol Cues in Problem Drinkers: Preliminary Findings. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2011; 38:101-7. [DOI: 10.3109/00952990.2011.600396] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
| | - Ruth Hüweler
- Department of Psychology, Boston University,
Boston, MA, USA
- Department of Psychology, University of Cologne,
Koeln, Germany
| | - James MacKillop
- Department of Psychology, University of Georgia,
Athens, GA, USA
| | | |
Collapse
|
29
|
Altamirano LJ, Fields HL, D'Esposito M, Boettiger CA. Interaction between family history of alcoholism and Locus of Control in the opioid regulation of impulsive responding under the influence of alcohol. Alcohol Clin Exp Res 2011; 35:1905-14. [PMID: 21569055 DOI: 10.1111/j.1530-0277.2011.01535.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Naltrexone (NTX) is an opioid antagonist indicated for the treatment of alcoholism, which is not universally effective. Thus, identifying individual predictors of NTX's behavioral effects is critical to optimizing its therapeutic use. Moreover, given the high rate of relapse during treatment for alcoholism, understanding NTX's behavioral effects when combined with moderate ethanol intake is important. Our previous study of abstinent alcoholics and control subjects showed that a more internal Locus of Control score predicted increased impulsive choice on NTX (Mitchell et al., 2007, Neuropsychopharmacology 32:439-449). Here, we tested whether this predictive relationship remains in the context of moderate alcohol intake. METHODS In this study, we tested the effect of acute NTX (50 mg) on impulsive choice, motor inhibition, and attentional bias after ingestion of moderate ethanol (∼0.3 g/kg, n = 30 subjects). Subjects included those recruited from a pool of ∼1,200 UC Berkeley undergraduates on the basis of scores on the Barratt Impulsiveness Scale (BIS). RESULTS Impulsive choice was positively correlated with breath alcohol concentration in placebo sessions. Locus of Control was again the sole predictor of NTX's effect on decision making among subjects with a family history of alcoholism. We also found a weak interaction between BIS scores and NTX's effect on impulsive choice. CONCLUSIONS Our results reinforce the predictive relationship between Locus of Control and NTX's effect on decision making in those with a family history of alcoholism, suggesting a possible biological basis to this relationship.
Collapse
Affiliation(s)
- Lee J Altamirano
- Department of Psychology and Neuroscience, University of Colorado, Boulder, Colorado, USA
| | | | | | | |
Collapse
|
30
|
Häggkvist J, Björkholm C, Steensland P, Lindholm S, Franck J, Schilström B. Naltrexone attenuates amphetamine-induced locomotor sensitization in the rat. Addict Biol 2011; 16:20-9. [PMID: 20192948 DOI: 10.1111/j.1369-1600.2009.00199.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Amphetamine, and other stimulants, readily induces behavioral sensitization, an effect hypothesized to reflect neurobiological changes that may underlie certain aspects of drug addiction. Apart from the effects on the dopamine system, previous studies have also shown that amphetamine interacts with other neurotransmitters, including the endogenous opioid system. The unselective opioid receptor antagonist naltrexone (NTX) modulates amphetamine-induced effects in both laboratory animals and humans. To further examine this interaction, the aim of the present study was to investigate the effect of NTX on the expression of locomotor sensitization and conditioned locomotor response in animals previously conditioned with amphetamine. Sensitization was induced by repeated administration of amphetamine (2 mg/kg) for 10 consecutive days. After a 10-day drug-free period, the rats were administered NTX (3 mg/kg) 30 minutes prior to the administration of a challenge dose of either amphetamine (0.5 mg/kg) (test for drug-induced sensitization) or saline (test for conditioned locomotor response). NTX had no effect on acute amphetamine-induced locomotor activity or on general locomotor activity in animals without a history of amphetamine conditioning. However, animals previously conditioned with amphetamine showed a sensitized locomotor response to the amphetamine challenge following the 10-day drug-free period. This sensitized response was significantly inhibited by NTX pre-treatment. In addition, NTX pre-treatment blocked the conditioned locomotor response when the amphetamine-conditioned animals were placed in the previously amphetamine-paired context. This study showed that NTX attenuates drug- and cue-induced locomotor behavior in amphetamine-conditioned animals, supporting recent clinical findings that indicated a potential role of NTX as a treatment for amphetamine dependence.
Collapse
Affiliation(s)
- Jenny Häggkvist
- Department of Clinical Neuroscience, Karolinska Institutet, Sweden.
| | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Skinner MD, Coudert M, Berlin I, Passeri E, Michel L, Aubin HJ. Effect of the threat of a disulfiram-ethanol reaction on cue reactivity in alcoholics. Drug Alcohol Depend 2010; 112:239-46. [PMID: 20708858 DOI: 10.1016/j.drugalcdep.2010.06.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2010] [Revised: 06/28/2010] [Accepted: 06/28/2010] [Indexed: 11/19/2022]
Abstract
RATIONALE Little is known about the effect of disulfiram on subjective and autonomic nervous system cue reactivity in the laboratory. The dissuasive psychological effect manifested as a threat would seem to prevail over the pharmacological effect. OBJECTIVES The primary objective was to determine whether there was a difference in cue reactivity responses during a threat condition compared to a neutral condition during alcohol cue exposure. METHODS In a crossover randomized study, participants received threat and neutral messages during two cue exposure sessions. The threat condition consisted of leading the patients to believe they had ingested 500 mg of disulfiram and the neutral condition of informing them that they had ingested a placebo, while in both condition they received the same placebo. RESULTS Physiological cue reactivity was demonstrated by a decrease in diastolic blood pressure during the threat compared to the neutral condition (p=0.04). Heart rate and subjective cue reactivity measures remained unchanged. There was a negative affect (assessed by the Positive and Negative Affect Scale) by condition by exposure interaction. CONCLUSIONS The threat of a disulfiram-ethanol reaction appears to affect cue reactivity physiologically rather than subjectively. While the data does not show changes in subjective ratings, it is possible that there are alternative beneficial effects arising from other cognitive processes that are not captivated by self-reported craving scales, reflected by decreases in negative affect and blood pressure. From this perspective, disulfiram might be recast to be more acceptable to patients.
Collapse
Affiliation(s)
- Marilyn D Skinner
- Centre Hospitalier Emile Roux, Centre de Traitement des Addictions, Limeil-Brévannes, France.
| | | | | | | | | | | |
Collapse
|
33
|
Weiss F. Advances in Animal Models of Relapse for Addiction Research. ADVANCES IN THE NEUROSCIENCE OF ADDICTION 2010. [DOI: 10.1201/9781420007350-c1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
34
|
Marinelli PW, Funk D, Harding S, Li Z, Juzytsch W, Lê AD. Roles of opioid receptor subtypes in mediating alcohol-seeking induced by discrete cues and context. Eur J Neurosci 2009; 30:671-8. [PMID: 19686472 DOI: 10.1111/j.1460-9568.2009.06851.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The aim of this study was to assess the effects of selective blockade of the delta (DOP) or mu (MOP) opioid receptors on alcohol-seeking induced by discrete cues and context. In Experiment 1, rats were trained to self-administer alcohol in an environment with distinct sensory properties. After extinction in a different context with separate sensory properties, rats were tested for context-induced renewal in the original context following treatment with the DOP receptor antagonist naltrindole (0-15 mg/kg, i.p.) or the MOP receptor antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH(2) (CTOP) (0-3 microg/4 microL, i.c.v.). In Experiment 2, reinstatement was tested with the presentation of a discrete light + tone cue previously associated with alcohol delivery, following extinction without the cue. The effects of naltrindole (0-5 mg/kg, i.p.) or CTOP (0-3 microg/4 microL, i.c.v.) were assessed. For context-induced renewal, 7.5 mg/kg naltrindole reduced responding without affecting locomotor activity. Both doses of CTOP attenuated responding in the first 15 min of the renewal test session; however, total responses did not differ at the end of the session. For discrete-cue-induced reinstatement, 1 and 5 mg/kg naltrindole attenuated responding but CTOP had no effect. We conclude that whereas DOP receptors mediate alcohol-seeking induced by discrete cues and context, MOP receptors may play a modest role only in context-induced renewal. These findings point to a differential involvement of opioid receptor subtypes in the effects of different kinds of conditioned stimuli on alcohol-seeking and support a more prominent role for DOP receptors.
Collapse
Affiliation(s)
- Peter W Marinelli
- Department of Neuroscience, Neurobiology of Alcohol Laboratory, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | | | | | | | | | | |
Collapse
|
35
|
Ooteman W, Verheul R, Naassila M, Daoust M, Schippers GM, Koeter MWJ, van den Brink W. Patient‐treatment matching with anti‐craving medications in alcohol‐dependent patients: A review on phenotypic, endophenotypic and genetic indicators. JOURNAL OF SUBSTANCE USE 2009. [DOI: 10.1080/14659890500038764] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Wendy Ooteman
- Amsterdam Institute for Addiction Research, Amsterdam, The Netherlands
| | - Roel Verheul
- Department of Psychiatry, Academic Medical Center University of Amsterdam, Amsterdam, The Netherlands
| | - Mickaël Naassila
- Viersprong Institute for Studies on Personality Disorders (VISPD), Halsteren, The Netherlands
| | - Martine Daoust
- Department of Clinical Psychology, University of Amsterdam, Amsterdam, The Netherlands
| | - Gerard M. Schippers
- Groupe de Recherche sur l'Alcool et les Pharmacodépendances (GRAP), Université de Picardie Jules Verne, Faculté de Pharmacie, Amiens, France
| | | | - Wim van den Brink
- Department of Psychiatry, Academic Medical Center University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
36
|
Liu X, Palmatier MI, Caggiula AR, Sved AF, Donny EC, Gharib M, Booth S. Naltrexone attenuation of conditioned but not primary reinforcement of nicotine in rats. Psychopharmacology (Berl) 2009; 202:589-98. [PMID: 18807246 PMCID: PMC2811405 DOI: 10.1007/s00213-008-1335-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Accepted: 09/04/2008] [Indexed: 12/01/2022]
Abstract
RATIONALE Opioid neurotransmission has been implicated in reinforcement-related processes for several drugs of abuse, including opiates, stimulants, and alcohol. However, less is known about its role in the motivational effects of nicotine and nicotine-associated environmental cues. OBJECTIVE This study investigated whether pretreatment with naltrexone, an opioid receptor antagonist, alters conditioned incentive salience of nicotine cues under two conditions: cue-induced reinstatement of nicotine-seeking after extinction and cue-maintained responding during extinction. The effect of naltrexone on nicotine self-administration during the maintenance phase was also examined. MATERIALS AND METHODS Male Sprague-Dawley rats were trained in daily 1-h sessions to self-administer nicotine (0.03 mg/kg/infusion, i.v.) on a fixed-ratio 5 schedule and associate a conditioned stimulus (CS) with each nicotine delivery. Once responding was extinguished by saline substitution for nicotine and omission of the CS, the reinstatement tests were conducted following subcutaneous administration of naltrexone (0, 0.25, 1, 2 mg/kg). In separate groups of rats, naltrexone (0, 2 mg/kg) was chronically given before each extinction sessions, where responses on the active lever resulted in presentations of the CS without nicotine infusion (saline substitution). Self-administration/naltrexone tests were conducted in different groups of rats receiving similar nicotine self-administration training. RESULTS Naltrexone significantly attenuated the CS-reinstated responding on the active, previously nicotine-reinforced lever in the reinstatement tests and the CS-maintained active lever responding during the extinction tests. In contrast, neither acute nor chronic naltrexone produced an effect on nicotine self-administration behavior. CONCLUSIONS These results indicate that activation of opioid receptors is implicated in mediation of the conditioned incentive properties of nicotine cues but not in the maintenance of nicotine self-administration. Therefore, these findings suggest that opioid receptor antagonists might have clinical potential for prevention of smoking relapse associated with exposure to environmental cues.
Collapse
Affiliation(s)
- Xiu Liu
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 N. State St., Jackson, MS 39216, USA.
| | | | - Anthony R. Caggiula
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Alan F. Sved
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Eric C. Donny
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Maysa Gharib
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Sheri Booth
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
37
|
Boettiger CA, Kelley EA, Mitchell JM, D'Esposito M, Fields HL. Now or Later? An fMRI study of the effects of endogenous opioid blockade on a decision-making network. Pharmacol Biochem Behav 2009; 93:291-9. [PMID: 19258022 DOI: 10.1016/j.pbb.2009.02.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Revised: 02/02/2009] [Accepted: 02/13/2009] [Indexed: 12/12/2022]
Abstract
Previously, we found that distinct brain areas predict individual selection bias in decisions between small immediate ("Now") and larger delayed rewards ("Later"). Furthermore, such selection bias can be manipulated by endogenous opioid blockade. To test whether blocking endogenous opioids with naltrexone (NTX) alters brain activity during decision-making in areas predicting individual bias, we compared fMRI BOLD signal correlated with Now versus Later decision-making after acute administration of NTX (50 mg) or placebo. We tested abstinent alcoholics and control subjects in a double-blind two-session design. We defined regions of interest (ROIs) centered on activation peaks predicting Now versus Later selection bias. NTX administration significantly increased BOLD signal during decision-making in the right lateral orbital gyrus ROI, an area where enhanced activity during decision-making predicts Later bias. Exploratory analyses identified additional loci where BOLD signal during decision-making was enhanced (left orbitofrontal cortex, left inferior temporal gyrus, and cerebellum) or reduced (right superior temporal pole) by NTX. Additional analyses identified sites, including the right lateral orbital gyrus, in which NTX effects on BOLD signal predicted NTX effects on selection bias. These data agree with opioid receptor expression in human frontal and temporal cortices, and suggest possible mechanisms of NTX's therapeutic effects.
Collapse
Affiliation(s)
- Charlotte A Boettiger
- Department of Psychology, Biomedical Research Imaging Center, University of North Carolina, Chapel Hill 27599-3270, United States.
| | | | | | | | | |
Collapse
|
38
|
Abstract
Use of human laboratory analogues of smoking behavior can provide an efficient, cost-effective mechanistic evaluation of a medication signal on smoking behavior, with the result of facilitating translational work in medications development. Although a number of human laboratory models exist to investigate various aspects of smoking behavior and nicotine dependence phenomena, none have yet modeled smoking lapse behavior. The first instance of smoking during a quit attempt (i.e. smoking lapse) is highly predictive of relapse and represents an important target for medications development. Focusing on an abstinence outcome is critical for medication screening as the US Food and Drug Administration approval for cessation medications is contingent on demonstrating effects on smoking abstinence. This paper outlines a three-stage process for the development of a smoking lapse model for the purpose of medication screening. The smoking lapse paradigm models two critical features of lapse behavior: the ability to resist the first cigarette and subsequent ad libitum smoking. Within the context of the model, smokers are first exposed to known precipitants of smoking relapse (e.g. nicotine deprivation, alcohol, stress), and then presented their preferred brand of cigarettes. Their ability to resist smoking is then modeled and once smokers 'give in' and decide to smoke, they participate in a tobacco self-administration session. Ongoing and completed work developing and validating these models for the purpose of medication screening is discussed.
Collapse
Affiliation(s)
- Sherry A McKee
- Yale University School of Medicine, New Haven, CT 06519, USA.
| |
Collapse
|
39
|
Williams KL, Schimmel JS. Effect of naltrexone during extinction of alcohol-reinforced responding and during repeated cue-conditioned reinstatement sessions in a cue exposure style treatment. Alcohol 2008; 42:553-63. [PMID: 18774673 DOI: 10.1016/j.alcohol.2008.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2008] [Revised: 06/04/2008] [Accepted: 06/09/2008] [Indexed: 10/21/2022]
Abstract
The ability of alcohol-related cues to promote craving can be attenuated independently by giving the opioid antagonist naltrexone (NTX) or by subjecting alcohol-dependent patients to a cue exposure treatment. The effects of cue exposure treatment may be enhanced if conducted in the presence of NTX. The purpose of these experiments was to determine if NTX given during extinction of responding for alcohol in rats would alter cue-conditioned reinstatement of responding and to determine if NTX, paired with repeated cue-conditioned reinstatement, would reduce subsequent cue-conditioned reinstatement or reacquisition of self-administration in the absence of NTX. Rats lever pressed for alcohol in the presence of an olfactory cue. Visual and auditory stimuli were presented during alcohol delivery. In the first experiment, rats were injected with saline or 3mg/kg NTX prior to extinction sessions followed by cue-conditioned reinstatement tests. In the second experiment, extinction was followed by cue-conditioned reinstatement sessions presented twice per week. The rats received saline or NTX (3 and 10mg/kg) prior to several sessions. All rats received reinstatement tests with and without a pretreatment of NTX followed by reacquisition of alcohol self-administration. NTX had no effect on responding during extinction or on subsequent cue-conditioned reinstatement. Only 10mg/kg NTX reduced responding during the twice weekly reinstatement sessions. The twice weekly NTX treatment had no effect on subsequent cue-conditioned reinstatement in the absence of NTX. Reacquisition of responding for alcohol was reduced in the group receiving saline during repeated reinstatement sessions, whereas this effect was blocked in the NTX group. These findings support the notion that NTX given during a brief abstinence period (i.e., extinction) has minimal effects on future sensitivity to alcohol cues and alcohol consumption. NTX given during the repeated alcohol cue exposure does not alter the subsequent incentive value of alcohol cues in the absence of NTX or enhance the beneficial effects of cue exposure treatment.
Collapse
|
40
|
Miranda R, MacKillop J, Monti PM, Rohsenow DJ, Tidey J, Gwaltney C, Swift R, Ray L, McGeary J. Effects of topiramate on urge to drink and the subjective effects of alcohol: a preliminary laboratory study. Alcohol Clin Exp Res 2008; 32:489-97. [PMID: 18215213 DOI: 10.1111/j.1530-0277.2007.00592.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Topiramate was recently reported to be efficacious in reducing drinking rates and craving among individuals with alcohol dependence in a randomized controlled trial, but dose effects could not be determined. This laboratory study systematically examined the dose-dependent effects of topiramate on cue-elicited craving and other putative mechanisms of its pharmacotherapeutic effects on drinking. METHODS Male and female heavy drinkers (n = 61) were randomized to 1 of 3 medication conditions (200 mg/d; 300 mg/d; placebo) in a double-blind study. Participants reached the target dose after a 32-day titration period, then were stabilized for approximately 1 week. All then participated in a laboratory assessment of alcohol cue reactivity and of the subjective effects of a moderate dose of alcohol. RESULTS Both doses of topiramate reduced the frequency of heavy drinking during the titration period as compared to placebo. However, topiramate did not affect self-reported craving for alcohol during the titration period, during the cue reactivity protocol, or in response to the alcohol challenge procedure. Topiramate reduced the stimulating effects of alcohol ingestion compared to placebo, but only in the 200 mg group. CONCLUSIONS The results of this study support previous findings that topiramate reduces drinking, but the behavioral mechanism underlying this effect does not appear to be attenuation of craving for alcohol as measured using the approaches employed in this study. Rather, the results tentatively suggest that topiramate may exert its beneficial effects by altering the subjective experiences of alcohol consumption. Limitations of the current study are discussed and complementary methods are recommended for future studies, such as the use of behavioral economic paradigms and ecological momentary assessment.
Collapse
Affiliation(s)
- Robert Miranda
- Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island 02912, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Tidey JW, Monti PM, Rohsenow DJ, Gwaltney CJ, Miranda R, McGeary JE, MacKillop J, Swift RM, Abrams DB, Shiffman S, Paty JA. Moderators of naltrexone's effects on drinking, urge, and alcohol effects in non-treatment-seeking heavy drinkers in the natural environment. Alcohol Clin Exp Res 2007; 32:58-66. [PMID: 18028530 DOI: 10.1111/j.1530-0277.2007.00545.x] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Naltrexone (NTX) has proven to be effective with alcoholics in treatment, with most controlled clinical trials showing beneficial effects on heavy drinking rates. However, little is known about the behavioral mechanisms underlying the effects of NTX on drinking, or about patient characteristics that may moderate NTX's effects on drinking. In this study, ecological momentary assessment (EMA) techniques were used to investigate some of the putative mechanisms of naltrexone's effects on drinking in heavy drinkers who were not seeking treatment for alcohol problems. Polymorphisms in the D4 dopamine receptor (DRD4) gene and the mu-opiate receptor (OPRM1) gene, family history of alcohol problems, age of onset of alcoholism and gender were explored as potential moderators of NTX's effects. METHODS After a 1-week placebo lead-in period, heavy drinkers (n = 180), 63% of whom were alcohol-dependent, were randomized to 3 weeks of daily naltrexone (50 mg) or placebo. Throughout the study, participants used EMA on palm-pilot computers to enter, in real time, drink data, urge levels, and subjective effects of alcohol consumption. RESULTS Naltrexone reduced percentage drinking days in all participants and reduced percent heavy drinking days in DRD4-L individuals; NTX decreased urge levels in participants with younger age of alcoholism onset; NTX increased time between drinks in participants who had more relatives with alcohol problems; and NTX reduced the stimulating effects of alcohol in women. OPRM1 status did not moderate any of NTX's effects. CONCLUSIONS These results confirm earlier findings of NTX's effects on drinking and related subjective effects, and extend them by describing individual difference variables that moderate these effects in the natural environment, using data collected in real time.
Collapse
Affiliation(s)
- Jennifer W Tidey
- Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island 02912, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ooteman W, Koeter MWJ, Verheul R, Schippers GM, van den Brink W. The effect of naltrexone and acamprosate on cue-induced craving, autonomic nervous system and neuroendocrine reactions to alcohol-related cues in alcoholics. Eur Neuropsychopharmacol 2007; 17:558-66. [PMID: 17379484 DOI: 10.1016/j.euroneuro.2007.02.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2006] [Revised: 02/01/2007] [Accepted: 02/13/2007] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Acamprosate and naltrexone have been shown to be effective in relapse prevention of alcoholism. It is hypothesized that naltrexone exerts its effects primarily on cue-induced craving and neuroendocrine cue reactivity, whereas acamprosate exerts its effect primarily on autonomic nervous system reactions to alcohol-related cues. EXPERIMENTAL PROCEDURES In a randomized double-blind experiment, 131 abstinent alcoholics received either acamprosate (n=56), naltrexone (n=52) or placebo (n=23) for three weeks and participated in two cue-exposure sessions: the first the day before and the second at the last day of medication. RESULTS Consistent with the hypotheses, naltrexone reduced craving more than acamprosate, and acamprosate reduced heart rate more than naltrexone. No medication effect was found on cue-induced cortisol. DISCUSSION The findings provide some evidence for differential effects of naltrexone and acamprosate: naltrexone may exert its effect, at least partly, by the reduction of cue-induced craving, whereas acamprosate may exert its effect, at least partly, by the reduction of autonomic nervous system reactions to alcohol-related cues.
Collapse
Affiliation(s)
- Wendy Ooteman
- Amsterdam Institute for Addiction Research, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
43
|
van den Wildenberg E, Janssen RGJH, Hutchison KE, van Breukelen GJP, Wiers RW. Polymorphisms of the dopamine D4 receptor gene (DRD4 VNTR) and cannabinoid CB1 receptor gene (CNR1) are not strongly related to cue-reactivity after alcohol exposure. Addict Biol 2007; 12:210-20. [PMID: 17508995 DOI: 10.1111/j.1369-1600.2007.00064.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Polymorphisms in the D4 dopamine receptor gene (DRD4) and the CB1 cannabinoid receptor gene (CNR1) have been associated with a differential response to alcohol after consumption. The goal of the present study was to investigate whether heavy drinkers with these polymorphisms would respond with enhanced cue-reactivity after alcohol exposure. Eighty-eight male heavy drinkers were genotyped for the DRD4 variable number of tandem repeats (VNTR) [either DRD4 long (L) or short (S)] and the CNR1 rs2023239 polymorphism (either CT/CC or TT). Participants were exposed to water and beer in 3-minute trials. Dependent variables of main interest were subjective craving for alcohol, subjective arousal and salivary reactivity. Overall, no strong evidence was found for stronger cue-reactivity (= outcome difference between beer and water trial) in the DRD4 L and CNR1 C allele groups. The DRD4 VNTR polymorphism tended to moderate salivary reactivity such that DRD4 L participants showed a larger beverage effect than the DRD4 S participants. Unexpectedly, the DRD4 L participants reported, on average, less craving for alcohol and more subjective arousal during cue exposure, compared with the DRD4 S participants. As weekly alcohol consumption increased, the CNR1 C allele group tended to report more craving for alcohol during the alcohol exposure than the T allele group. The DRD4 and CNR1 polymorphisms do not appear to strongly moderate cue-reactivity after alcohol cue exposure, in male heavy drinkers.
Collapse
Affiliation(s)
- Esther van den Wildenberg
- Faculty of Psychology, Experimental Psychology, University of Maastricht, Maastricht, the Netherlands.
| | | | | | | | | |
Collapse
|
44
|
Economidou D, Mattioli L, Ubaldi M, Lourdusamy A, Soverchia L, Hardiman G, Campolongo P, Cuomo V, Ciccocioppo R. Role of cannabinoidergic mechanisms in ethanol self-administration and ethanol seeking in rat adult offspring following perinatal exposure to Delta9-tetrahydrocannabinol. Toxicol Appl Pharmacol 2007; 223:73-85. [PMID: 17618662 DOI: 10.1016/j.taap.2007.05.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 05/04/2007] [Accepted: 05/16/2007] [Indexed: 10/23/2022]
Abstract
The present study evaluated the consequences of perinatal Delta(9)-tetrahydrocannabinol (Delta(9)-THC) treatment (5 mg/kg/day by gavage), either alone or combined with ethanol (3% v/v as the only fluid available), on ethanol self-administration and alcohol-seeking behavior in rat adult offspring. Furthermore, the effect of the selective cannabinoid CB(1) receptor antagonist, SR-141716A, on ethanol self-administration and on reinstatement of ethanol-seeking behavior induced either by stress or conditioned drug-paired cues was evaluated in adult offspring of rats exposed to the same perinatal treatment. Lastly, microarray experiments were conducted to evaluate if perinatal treatment with Delta(9)-tetrahydrocannabinol, ethanol or their combination causes long-term changes in brain gene expression profile in rats. The results of microarray data analysis showed that 139, 112 and 170 genes were differentially expressed in the EtOH, Delta(9)-THC, or EtOH+Delta(9)-THC group, respectively. No differences in alcohol self-administration and alcohol seeking were observed between rat groups. Intraperitoneal (IP) administration of SR-141716A (0.3-3.0 mg/kg) significantly reduced lever pressing for ethanol and blocked conditioned reinstatement of alcohol seeking. At the same doses SR-141716A failed to block foot-shock stress-induced reinstatement of alcohol seeking. The results reveal that perinatal exposure to Delta(9)-THC ethanol or their combination results in evident changes in gene expression patterns. However, these treatments do not significantly affect vulnerability to ethanol abuse in adult offspring. On the other hand, the results obtained with SR-141716A emphasize that endocannabinoid mechanisms play a major role in ethanol self-administration, as well as in the reinstatement of ethanol-seeking behavior induced by conditioned cues, supporting the idea that cannabinoid CB(1) receptor antagonists may represent interesting agents for the pharmacotherapy of alcoholism.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Animals, Newborn
- Behavior, Animal/drug effects
- Central Nervous System Depressants/administration & dosage
- Choice Behavior/drug effects
- Conditioning, Operant/drug effects
- Dronabinol/toxicity
- Drug Therapy, Combination
- Ethanol/administration & dosage
- Female
- Gene Expression Profiling
- Oligonucleotide Array Sequence Analysis/methods
- Piperidines/pharmacology
- Pregnancy
- Psychotropic Drugs/toxicity
- Pyrazoles/pharmacology
- Rats
- Rats, Wistar
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Rimonabant
- Self Administration
Collapse
Affiliation(s)
- Daina Economidou
- Department of Experimental Medicine and Public Health, University of Camerino, Via Scalzino 3, 62032 Camerino, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Dayas CV, Liu X, Simms JA, Weiss F. Distinct patterns of neural activation associated with ethanol seeking: effects of naltrexone. Biol Psychiatry 2007; 61:979-89. [PMID: 17098214 PMCID: PMC2831298 DOI: 10.1016/j.biopsych.2006.07.034] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2005] [Revised: 06/23/2006] [Accepted: 07/19/2006] [Indexed: 11/27/2022]
Abstract
BACKGROUND Alcoholism, like other substance abuse disorders, is a chronically relapsing condition. Compared with other abused drugs, however, little is known about the neural mechanisms mediating ethanol (EtOH)-craving and -seeking behavior leading to relapse. This study, therefore, was conducted to identify candidate brain regions that are recruited by an EtOH-associated contextual stimulus (S(+)). A secondary objective was to determine whether EtOH S(+)-elicited neural recruitment patterns are modified by the opiate antagonist naltrexone (NTX), a compound that reduces cue-induced craving in alcoholics and attenuates ethanol seeking in animal models of relapse. METHODS Rats were tested in a conditioned reinstatement model of relapse with subsequent examination of brain c-fos expression patterns elicited by an EtOH S(+) versus a cue associated with nonreward (S(-)). In addition, modification of these expression patterns by NTX was examined. RESULTS The EtOH S(+) reinstated extinguished responding and increased c-fos expression within the prefrontal cortex, hippocampus, nucleus accumbens, and hypothalamic paraventricular nucleus (PVN). Naltrexone suppressed the S(+)-induced reinstatement and attenuated hippocampal CA3 c-fos expression, while increasing neural activity in the extended amygdala and PVN. CONCLUSIONS Ethanol-associated contextual stimuli recruit key brain regions that regulate associative learning, goal-directed behavior, and Pavlovian conditioning of emotional significance to previously neutral stimuli. In addition, the data implicate the hippocampus, amygdala, and PVN as potential substrates for the inhibitory effects of NTX on conditioned reinstatement.
Collapse
Affiliation(s)
- Christopher V Dayas
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | | | |
Collapse
|
46
|
van den Wildenberg E, Wiers RW, Dessers J, Janssen RGJH, Lambrichs EH, Smeets HJM, van Breukelen GJP. A functional polymorphism of the mu-opioid receptor gene (OPRM1) influences cue-induced craving for alcohol in male heavy drinkers. Alcohol Clin Exp Res 2007; 31:1-10. [PMID: 17207095 DOI: 10.1111/j.1530-0277.2006.00258.x] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND The mu-opioid receptor gene (OPRM1) codes for the mu-opioid receptor, which binds beta-endorphin. The A118G polymorphism in this gene affects beta-endorphin binding such that the Asp40 variant (G allele) binds beta-endorphin 3 times more tightly than the more common Asn40 variant (A allele). This study investigated the influence of the A118G polymorphism on cue reactivity after exposure to an alcoholic beverage in male heavy drinkers. METHODS Participants were either homozygous for the A allele (n=84) or carrying at least 1 copy of the G allele (n=24). All participants took part in a cue-reactivity paradigm where they were exposed to water and beer in 3-minute trials. The dependent variables of main interest were subjective craving for alcohol, subjective arousal, and saliva production. RESULTS G allele carriers reported significantly more craving for alcohol than the A allele participants (as indicated by the within-subject difference in craving after beer vs after water exposure). No differences were found for subjective arousal and saliva. Both groups did not differ in family history of alcoholism. Participants with the G allele reported a significantly higher lifetime prevalence of drug use than participants homozygous for the A allele. CONCLUSIONS A stronger urge to drink alcohol after exposure to an alcoholic beverage might contribute to a heightened risk for developing alcohol-related problems in individuals with a copy of the G allele. The G allele might also predispose to drug use in general.
Collapse
|
47
|
Mitchell JM, Tavares VC, Fields HL, D'Esposito M, Boettiger CA. Endogenous opioid blockade and impulsive responding in alcoholics and healthy controls. Neuropsychopharmacology 2007; 32:439-49. [PMID: 17047667 DOI: 10.1038/sj.npp.1301226] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The opioid receptor antagonist naltrexone (NTX) is one of few approved treatments for alcoholism, yet the mechanism by which it reduces drinking remains unclear. In rats, NTX reduces morphine-induced impulsive choice bias; however, nothing is known about the drug's effect on discrete aspects of impulsive behavior in humans, such as decision-making and inhibitory control. Here, we used a modified delay discounting procedure to investigate whether NTX improves decision-making or inhibitory control in humans. We measured the effect of acute NTX (50 mg) on choice between smaller sooner (SS) and larger later monetary rewards and on response errors (motor mismatch) in a high conflict condition in a group of abstinent alcoholics (AA) and healthy control subjects (CS). We previously reported that AA selected the SS option significantly more often than did CS in this paradigm. If the choice bias of AA is due to enhanced endogenous opioid signaling in response to potential reward, NTX should reduce such bias in the AA group. We found that NTX did not reliably reduce impulsive choice in the AA group; however, NTX's effect on choice bias across individuals was robustly predictable. NTX's effect on choice bias was significantly correlated with scores on Rotter's Locus of Control (LOC) scale; increasingly internal LOC scores predicted increasing likelihood of impulsive choices on NTX. In addition, we found that NTX significantly enhanced control of motor responses, particularly within the CS group. These results suggest that endogenous opioids may impair response selection during decision-making under conflict, and that NTX's effects on explicit decision-making are personality-dependent. Determining the biological basis of this dependence could have important implications for effective alcoholism treatment.
Collapse
Affiliation(s)
- Jennifer M Mitchell
- Ernest Gallo Clinic and Research Center, University of California at San Francisco, Emeryville, CA 94608, USA
| | | | | | | | | |
Collapse
|
48
|
Psychosocial Predictors of Treatment Outcome, Dropout, and Change Processes in a Pharmacological Clinical Trial for Alcohol Dependence. ADDICTIVE DISORDERS & THEIR TREATMENT 2006. [DOI: 10.1097/01.adt.0000210701.63165.5a] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Higley AE, Kiefer SW. Delta receptor antagonism, ethanol taste reactivity, and ethanol consumption in outbred male rats. Alcohol 2006; 40:143-50. [PMID: 17418693 DOI: 10.1016/j.alcohol.2007.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Revised: 01/30/2007] [Accepted: 01/30/2007] [Indexed: 10/23/2022]
Abstract
Naltrexone, a nonspecific opioid antagonist, produces significant changes in ethanol responsivity in rats by rendering the taste of ethanol aversive as well as producing a decrease in voluntary ethanol consumption. The present study investigated the effect of naltrindole, a specific antagonist of delta opioid receptors, on ethanol taste reactivity and ethanol consumption in outbred rats. In the first experiment, rats received acute treatment of naltrexone, naltrindole, or saline followed by the measurement of ethanol consumption in a short-term access period. The second experiment involved the same treatments and investigated ethanol palatability (using the taste-reactivity test) as well as ethanol consumption. Results indicated that treatment with 3 mg/kg naltrexone significantly affected palatability (rendered ethanol more aversive, Experiment 2) and decreased voluntary ethanol consumption (Experiments 1 and 2). The effects of naltrindole were inconsistent. In Experiment 1, 8 mg/kg naltrindole significantly decreased voluntary ethanol consumption but this was not replicated in Experiment 2. The 8 mg/kg dose produced a significant increase in aversive responding (Experiment 2) but did not affect ingestive responding. Lower doses of naltrindole (2 and 4 mg/kg) were ineffective in altering rats' taste-reactivity response to and consumption of ethanol. While these data suggest that delta receptors are involved in rats' taste-reactivity response to ethanol and rats' ethanol consumption, it is likely that multiple opioid receptors mediate both behavioral responses.
Collapse
Affiliation(s)
- Amanda E Higley
- Department of Psychology, Kansas State University, 492 Bluemont Hall, Manhattan, KS 66506-5302, USA
| | | |
Collapse
|
50
|
McGeary JE, Monti PM, Rohsenow DJ, Tidey J, Swift R, Miranda R. Genetic moderators of naltrexone's effects on alcohol cue reactivity. Alcohol Clin Exp Res 2006; 30:1288-96. [PMID: 16899031 DOI: 10.1111/j.1530-0277.2006.00156.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Naltrexone (NTX) reduces drinking and craving in alcoholic individuals in treatment and also in heavy drinkers. Polymorphisms in the D4 dopamine receptor (DRD4) gene and mu-opiate receptor gene (OPRM1) may moderate NTX's effects on craving. This study examined these candidate genes as moderators of the effects of NTX on cue-elicited urge to drink in non-treatment-seeking heavy drinkers. METHOD Data from the subset of 93 participants who consented for genetic testing in a larger study of medication effects were used to examine pharmacogenetic hypotheses. The non-treatment-seeking male and female heavy drinkers (62% alcohol dependent) were genotyped for the variable number of tandem repeats polymorphism in the DRD4 gene [L=7 or more (n=34), S=less than 7 (n=56)] and Asn40Asp single-nucleotide polymorphism in the OPRM1 gene [29 aspartate (Asp) carriers and 59 asparagine (Asn) homozygotes]. Ten days after randomization to NTX (50 mg) or placebo, participants completed an alcohol cue reactivity assessment. RESULTS Any medication effects were all accounted for by interaction with genotype. Naltrexone increased urge for alcohol in Asp carriers across alcohol and neutral beverage cue trials and had no effect on homozygous Asn carriers. Asp40 carriers on either medication had greater decreases (from resting baseline) in mean arterial blood pressure across all beverage cue trials compared with Asn carriers. For DRD4, no differential medication effects by DRD4 polymorphism were found. Alcohol dependence diagnosis did not moderate the effects of gene and medication on cue-elicited measures. DISCUSSION The differential responses to NTX due to variation in the OPRM1 gene may help explain conflicting results in clinical trials and suggest directions for patient-treatment matching.
Collapse
Affiliation(s)
- John E McGeary
- Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island 02906, USA.
| | | | | | | | | | | |
Collapse
|