Exenatide once weekly for alcohol use disorder investigated in a randomized, placebo-controlled clinical trial

Mette Kruse Klausen, Mathias Ebbesen Jensen, Marco Møller, Nina Le Dous, Anne-Marie Østergaard Jensen, Victoria Alberte Zeeman, Claas-Frederik Johannsen, Alycia Lee, Gerda Krog Thomsen, Julian Macoveanu, Patrick MacDonald Fisher, Matthew Paul Gillum, Niklas Rye Jørgensen, Marianne Lerbæk Bergmann, Henrik Enghusen Poulsen, Ulrik Becker, Jens Juul Holst, Helene Benveniste, Nora D Volkow, Sabine Vollstädt-Klein, Kamilla Woznica Miskowiak, Claus Thorn Ekstrøm, Gitte Moos Knudsen, Tina Vilsbøll, Anders Fink-Jensen, Mette Kruse Klausen, Mathias Ebbesen Jensen, Marco Møller, Nina Le Dous, Anne-Marie Østergaard Jensen, Victoria Alberte Zeeman, Claas-Frederik Johannsen, Alycia Lee, Gerda Krog Thomsen, Julian Macoveanu, Patrick MacDonald Fisher, Matthew Paul Gillum, Niklas Rye Jørgensen, Marianne Lerbæk Bergmann, Henrik Enghusen Poulsen, Ulrik Becker, Jens Juul Holst, Helene Benveniste, Nora D Volkow, Sabine Vollstädt-Klein, Kamilla Woznica Miskowiak, Claus Thorn Ekstrøm, Gitte Moos Knudsen, Tina Vilsbøll, Anders Fink-Jensen

Abstract

BackgroundAlcohol use disorder (AUD) is a chronic, relapsing brain disorder that accounts for 5% of deaths annually, and there is an urgent need to develop new targets for therapeutic intervention. The glucagon-like peptide-1 (GLP-1) receptor agonist exenatide reduces alcohol consumption in rodents and nonhuman primates, but its efficacy in patients with AUD is unknown.MethodsIn a randomized, double-blinded, placebo-controlled clinical trial, treatment-seeking AUD patients were assigned to receive exenatide (2 mg subcutaneously) or placebo once weekly for 26 weeks, in addition to standard cognitive-behavioral therapy. The primary outcome was reduction in number of heavy drinking days. A subgroup also completed functional MRI (fMRI) and single-photon emission CT (SPECT) brain scans.ResultsA total of 127 patients were enrolled. Our data revealed that although exenatide did not significantly reduce the number of heavy drinking days compared with placebo, it significantly attenuated fMRI alcohol cue reactivity in the ventral striatum and septal area, which are crucial brain areas for drug reward and addiction. In addition, dopamine transporter availability was lower in the exenatide group compared with the placebo group. Exploratory analyses revealed that exenatide significantly reduced heavy drinking days and total alcohol intake in a subgroup of obese patients (BMI > 30 kg/m2). Adverse events were mainly gastrointestinal.ConclusionThis randomized controlled trial on the effects of a GLP-1 receptor agonist in AUD patients provides new important knowledge on the effects of GLP-1 receptor agonists as a novel treatment target in addiction.Trial registrationEudraCT: 2016-003343-11. ClinicalTrials.gov (NCT03232112).FundingNovavi Foundation; Research Foundation, Mental Health Services, Capital Region of Denmark; Research Foundation, Capital Region of Denmark; Ivan Nielsen Foundation; A.P. Moeller Foundation; Augustinus Foundation; Woerzner Foundation; Grosserer L.F. Foghts Foundation; Hartmann Foundation; Aase and Ejnar Danielsen Foundation; P.A. Messerschmidt and Wife Foundation; and Lundbeck Foundation.

Keywords: Addiction; Clinical Trials; Neuroimaging; Neuroscience; Pharmacology.

Conflict of interest statement

Conflict of interest: AFJ has received an unrestricted research grant from Novo Nordisk A/S to investigate the effects of GLP-1 receptor stimulation on weight gain and metabolic disturbances in patients with schizophrenia treated with an antipsychotic. TV has served on scientific advisory panels for, been part of speaker’s bureaus for, served as a consultant to, and/or received research support from Amgen, AstraZeneca, Boehringer Ingelheim, Eli Lilly, Gilead, Mundipharma, MSD/Merck, Novo Nordisk, and Sun Pharmaceutical Industries. HB has received honoraria from Washington University Seminar. JJH has received consulting fees from Novo Nordisk A/S and grants from the Novo Nordisk Foundation. GMK has received personal honoraria from Sage Biogen, H. Lundbeck A/S, and Sanos and serves as president of the European College of Neuropsychopharmacology (unpaid) and chair of the Science and Infrastructure Advisory Board for the Human Brain Project (personal honorarium). SVK has received grants from the German Research Foundation. JM has received honoraria from H. Lundbeck A/S. KWM has received honoraria from H. Lundbeck A/S and Janssen. The funding sources and the manufacturer of exenatide once weekly (Bydureon, AstraZeneca) had no influence on the trial design or data analysis.

Figures

Figure 1. CONSORT flow diagram.
Figure 1. CONSORT flow diagram.
Study diagram of patient flow according to CONSORT 2010 statement. Details regarding initial meetings and ineligibility for screening can be found in Supplemental Figure 3, and a flowchart for the 6-month follow-up can be found in Supplemental Figure 1. Of the 127 patients included in the study, 65 patients were randomized to 2 mg exenatide once weekly, and 62 patients were randomized to placebo. Thirty-two patients from the exenatide group and 26 patients from the placebo group completed the study after 26 weeks of trial participation. AUDIT, Alcohol Use Disorders Identification Test; CIWA-Ar, Clinical Institute Withdrawal Assessment for Alcohol, Revised.
Figure 2. Kaplan-Meier survival curve of patients…
Figure 2. Kaplan-Meier survival curve of patients who withdrew from the trial or were lost to follow-up.
The time to discontinuation was not significantly different in the 2 groups (P = 0.46). Input data are the number of injections + 1 because patients were registered as discontinued in the week after the last injection was received. All patients are included (n = 127). Censoring is indicated by the + mark.
Figure 3. Reduction in heavy drinking days.
Figure 3. Reduction in heavy drinking days.
Mean percentage heavy drinking days in the last 30 days, measured with the Time-Line Follow Back (TLFB) method, at all assessments (week 0, week 4, week 12, week 20, week 26). Data were analyzed with an ANOVA adjusted for baseline, and missing data were imputed with the use of multiple imputations as described in the text (n = 127). Data represent mean ± SEM.
Figure 4. Reduction in heavy drinking days…
Figure 4. Reduction in heavy drinking days in BMI subgroups.
Exploratory analysis of mean percentage heavy drinking days in the last 30 days, measured with the TLFB method, at all assessments (week 0, week 4, week 12, week 20, week 26) within the BMI subgroups. Normal weight, n = 52; BMI > 30, n = 30. Only significant findings from Supplemental Table 7 are included. Data were analyzed with an ANOVA adjusted for baseline, and missing data were imputed with the use of multiple imputations as described in the text. Data represent mean ± SEM.
Figure 5. Reduction in total alcohol intake…
Figure 5. Reduction in total alcohol intake in BMI subgroups.
Exploratory analysis of mean total alcohol intake in the last 30 days, measured with the TLFB method, at all assessments (week 0, week 4, week 12, week 20, week 26) within the BMI subgroups. BMI > 25, n = 75; BMI > 30, n = 30. Only significant findings from Supplemental Table 7 are included. Data were analyzed with an ANOVA adjusted for baseline, and missing data were imputed with the use of multiple imputations as described in the text. Data represent mean ± SEM.
Figure 6. fMRI ALCUE ROI results.
Figure 6. fMRI ALCUE ROI results.
(A) Key fMRI findings showed reduced cue reactivity after 26 weeks of treatment with exenatide compared with placebo. Analysis revealed statistically significant interaction between the treatment and time on fMRI response in all 3 ROIs: ventral striatum [F(1,31) = 4.744, P = 0.037, partial η2 =0.133], dorsal striatum [F(1,31) = 6.124, P = 0.019, partial η2 = 0.165], putamen [F(1,31) = 4.730, P = 0.037, partial η2 = 0.132]. *P < 0.05. (B) In more detail, we found that at week 26, cue-induced activity was significantly lower in ventral striatum after treatment with exenatide compared with placebo (M = –0.176, SE = 0.075, P = 0.025), but not in dorsal striatum (M = –0.142, SE = 0.076, P = 0.073) nor in putamen (M = –0.123, SE = 0.084, P = 0.153). At baseline, cue-induced activity did not differ significantly between groups. Within the exenatide group, cue-induced activity was significantly reduced from baseline to week 26 in ventral striatum (M = –0.254, SE = 0.116, P = 0.044) and in dorsal striatum (M = –0.351, SE = 0.156, P = 0.039), but not in putamen (M = –0.405, SE = 0.202, P = 0.063). Within the placebo group, no statistically significant differences were found. (A and B) ROI data were analyzed using a repeated-measures ANOVA including factors group and time and an independent sample 2-tailed t test comparing groups (placebo and exenatide). Placebo, n = 16; exenatide, n = 17. Boxes represent upper and lower quartiles, the line represents the median, and the X represents the mean. Horizontal lines indicate significant interactions between treatment and time (*P < 0.05), and brackets indicate significant simple effects (*P < 0.05).
Figure 7. fMRI ALCUE whole-brain results.
Figure 7. fMRI ALCUE whole-brain results.
Reduced cue-induced activation in the exenatide group compared with the placebo group after 26 weeks of treatment in the left caudate and septal area (x, y, z coordinates = 0, 0, 4) (A) and right middle frontal gyrus (x, y, z coordinates = 36, 20, 48) (B). A 2-sample 2-tailed t test was performed for the post hoc analyses to compare groups (placebo, exenatide) and within a group across time (placebo/exenatide: T1, T2). For the group comparisons, the contrast of interest used was ‘alcohol > neutral stimuli’, where the probability of a family wise error (FWE) was set to 0.05 to control for multiple statistical testing. Using the AlphaSim (3dClustSim) method, a combined voxel wise threshold of P < 0.001 and a cluster extent threshold of 101 voxels were calculated (n = 22).
Figure 8. fMRI spatial working memory task…
Figure 8. fMRI spatial working memory task (N-back task).
The exenatide group showed a reduction at follow-up in the response to the 2-back > 1-back task compared with the placebo group (2-way mixed-effect ANOVA; placebo, n = 16; exenatide, n = 17; control, n = 25) in 2 prefrontal clusters (frontal pole x, y, z = 34, 54, 20, corrected P < 0.002; superior frontal gyrus x, y, z = 4, 46, 46, corrected P < 0.001). Boxes represent upper and lower quartiles, the line represents the median, and the X represents the mean. dlPFC, dorsolateral prefrontal cortex; dmPFC, dorsomedial prefrontal cortex.
Figure 9. SPECT DAT results combined.
Figure 9. SPECT DAT results combined.
(A) Baseline DAT availability in striatum, caudate, and putamen in AUD patients did not differ from that in healthy controls. Data were analyzed with a 1-way ANCOVA, adjusted for baseline DAT availability. Healthy controls, n = 21; patients at baseline, n = 45. (B) At the week 26 rescan, DAT availability in striatum, caudate, and putamen was significantly lower in the exenatide group compared with the placebo group [striatum, F(1,13) = 4.978, P = 0.044; caudate, F(1,13) = 8.066, P = 0.014; putamen, F(1,13) = 6.571, P = 0.024]. Data were analyzed with an ANCOVA adjusted for age. Placebo, n = 9; exenatide, n = 7. *P < 0.05. (A and B) Boxes represent upper and lower quartiles, the line represents the median, and the X represents the mean.

References

    1. Carvalho AF, et al. Alcohol use disorders. Lancet. 2019;394(10200):781–792. doi: 10.1016/S0140-6736(19)31775-1.
    1. Askgaard G, et al. Hospital admissions and mortality in the 15 years after a first-time hospital contact with an alcohol problem: a prospective cohort study using the entire Danish population. Int J Epidemiol. 2020;49(1):94–102. doi: 10.1093/ije/dyz159.
    1. Kranzler HR, Soyka M. Diagnosis and pharmacotherapy of alcohol use disorder: a review. JAMA. 2018;320(8):815–824. doi: 10.1001/jama.2018.11406.
    1. Bottlender M, Soyka M. Outpatient alcoholism treatment: predictors of outcome after 3 years. Drug Alcohol Depend. 2005;80(1):83–89. doi: 10.1016/j.drugalcdep.2005.03.011.
    1. Holst JJ. The physiology of glucagon-like peptide 1. Physiol Rev. 2007;87(4):1409–1439. doi: 10.1152/physrev.00034.2006.
    1. Reddy IA, et al. Glucagon-like peptide 1 receptor activation regulates cocaine actions and dopamine homeostasis in the lateral septum by decreasing arachidonic acid levels. Transl Psychiatry. 2016;6(5):e809. doi: 10.1038/tp.2016.86.
    1. Nuffer WA, Trujillo JM. Liraglutide: a new option for the treatment of obesity. Pharmacotherapy. 2015;35(10):926–934. doi: 10.1002/phar.1639.
    1. Volkow ND, et al. Obesity and addiction: neurobiological overlaps. Obes Rev. 2013;14(1):2–18. doi: 10.1111/j.1467-789X.2012.01031.x.
    1. Cork SC, et al. Distribution and characterisation of glucagon-like peptide-1 receptor expressing cells in the mouse brain. Mol Metab. 2015;4(10):718–731. doi: 10.1016/j.molmet.2015.07.008.
    1. Han VKM, et al. Cellular localization of proglucagon/glucagon-like peptide I messenger RNAs in rat brain. J Neurosci Res. 1986;16(1):97–107. doi: 10.1002/jnr.490160110.
    1. Jensen CB, et al. Characterization of the glucagonlike peptide-1 receptor in male mouse brain using a novel antibody and in situ hybridization. Endocrinology. 2018;159(2):665–675. doi: 10.1210/en.2017-00812.
    1. Merchenthaler I, et al. Distribution of pre-pro-glucagon and glucagon-like peptide-1 receptor messenger RNAs in the rat central nervous system. J Comp Neurol. 1999;403(2):261–280. doi: 10.1002/(SICI)1096-9861(19990111)403:2<261::AID-CNE8>;2-5.
    1. Rinaman L. Ascending projections from the caudal visceral nucleus of the solitary tract to brain regions involved in food intake and energy expenditure. Brain Res. 2010;1350(412):18–34.
    1. Vrang N, Grove K. The brainstem preproglucagon system in a non-human primate (Macaca mulatta) Brain Res. 2011;1397:28–37. doi: 10.1016/j.brainres.2011.05.002.
    1. Olds J, Milner P. Positive reinforcement produced by electrical stimulation of septal area and other regions of rat brain. J Comp Physiol Psychol. 1954;47(6):419–427. doi: 10.1037/h0058775.
    1. Heppner KM, et al. Expression and distribution of glucagon-like peptide-1 receptor mRNA, protein and binding in the male nonhuman primate (Macaca mulatta) brain. Endocrinology. 2015;156(1):255–267. doi: 10.1210/en.2014-1675.
    1. Thomsen M, et al. Effects of glucagon-like peptide 1 analogs on alcohol intake in alcohol-preferring vervet monkeys. Psychopharmacology (Berl) 2018;236(2):603–611.
    1. Brunchmann A, et al. The effect of glucagon-like peptide-1 (GLP-1) receptor agonists on substance use disorder (SUD)-related behavioural effects of drugs and alcohol: a systematic review. Physiol Behav. 2019;206:232–242. doi: 10.1016/j.physbeh.2019.03.029.
    1. Suchankova P, et al. The glucagon-like peptide-1 receptor as a potential treatment target in alcohol use disorder: evidence from human genetic association studies and a mouse model of alcohol dependence. Transl Psychiatry. 2015;5(6):e583–e583. doi: 10.1038/tp.2015.68.
    1. Athauda D, et al. Exenatide once weekly versus placebo in Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet. 2017;390(10103):1664–1675. doi: 10.1016/S0140-6736(17)31585-4.
    1. Yammine L, et al. Exenatide adjunct to nicotine patch facilitates smoking cessation and may reduce post-cessation weight gain: a pilot randomized controlled trial. Nicotine Tob Res. 2021;23(10):1682–1690. doi: 10.1093/ntr/ntab066.
    1. Klausen MK, et al. The role of glucagon-like peptide 1 (GLP-1) in addictive disorders. Br J Pharmacol. 2021;197(4):625–641.
    1. Sobell MB, et al. The reliability of a timeline method for assessing normal drinker college students’ recent drinking history: utility for alcohol research. Addict Behav. 1986;11(2):149–161. doi: 10.1016/0306-4603(86)90040-7.
    1. Antonsen KK, et al. Does glucagon-like peptide-1 (GLP-1) receptor agonist stimulation reduce alcohol intake in patients with alcohol dependence: study protocol of a randomised, double-blinded, placebo-controlled clinical trial. BMJ Open. 2018;8(7):e019562. doi: 10.1136/bmjopen-2017-019562.
    1. Yaribeygi H, et al. GLP-1 mimetics and cognition. Life Sci. 2021;264:118645. doi: 10.1016/j.lfs.2020.118645.
    1. Nutt DJ, et al. The dopamine theory of addiction: 40 years of highs and lows. Nat Rev Neurosci. 2015;16(5):305–312. doi: 10.1038/nrn3939.
    1. Volkow ND, et al. Role of dopamine, the frontal cortex and memory circuits in drug addiction: insight from imaging studies. Neurobiol Learn Mem. 2002;78(3):610–624. doi: 10.1006/nlme.2002.4099.
    1. Witkiewitz K, et al. Clinical validation of reduced alcohol consumption after treatment for alcohol dependence using the World Health Organization risk drinking levels. Alcohol Clin Exp Res. 2017;41(1):179–186. doi: 10.1111/acer.13272.
    1. Maldjian JA, et al. An automated method for neuroanatomic and cytoarchitectonic atlas-based interrogation of fMRI data sets. Neuroimage. 2003;19(3):1233–1239. doi: 10.1016/S1053-8119(03)00169-1.
    1. Vallöf D, et al. The glucagon-like peptide 1 receptor agonist liraglutide attenuates the reinforcing properties of alcohol in rodents. Addict Biol. 2016;21(2):422–437. doi: 10.1111/adb.12295.
    1. Shirazi RH, et al. Gut peptide GLP-1 and its analogue, Exendin-4, decrease alcohol intake and reward. PLoS One. 2013;8(4):1–7.
    1. Anton RF, et al. Combined pharmacotherapies and behavioral interventions for alcohol dependence: the COMBINE study: a randomized controlled trial. JAMA. 2006;295(17):2003–2017. doi: 10.1001/jama.295.17.2003.
    1. Johnson BA, et al. Oral topiramate for treatment of alcohol dependence: a randomised controlled trial. Lancet. 2003;361(9370):1677–1685. doi: 10.1016/S0140-6736(03)13370-3.
    1. Carroll KM, et al. Cognitive Behavioral Interventions for Alcohol and Drug Use Disorders: Through the Stage Model and Back Again. Psychol Addict Behav. 2017;31(8):847–861. doi: 10.1037/adb0000311.
    1. Scherrer B, et al. Baseline severity and the prediction of placebo response in clinical trials for alcohol dependence: a meta-regression analysis to develop an enrichment strategy. Alcohol Clin Exp Res. 2021;45(9):1722–1734. doi: 10.1111/acer.14670.
    1. Litten RZ, et al. The placebo effect in clinical trials for alcohol dependence: an exploratory analysis of 51 naltrexone and acamprosate studies. Alcohol Clin Exp Res. 2013;37(12):2128–2137. doi: 10.1111/acer.12197.
    1. Butler T, et al. Comparison of human septal nuclei MRI measurements using automated segmentation and a new manual protocol based on histology. Neuroimage. 2014;97(1):245–251.
    1. Bruijnen CJWH, et al. Prevalence of cognitive impairment in patients with substance use disorder. Drug Alcohol Rev. 2019;38(4):435–442. doi: 10.1111/dar.12922.
    1. Wilcox CE, et al. Cognitive control in alcohol use disorder: deficits and clinical relevance. Rev Neurosci. 2014;25(1):1–24. doi: 10.1515/revneuro-2013-0054.
    1. Petersen CS, Miskowiak KW. Toward a transdiagnostic neurocircuitry-based biomarker model for pro-cognitive effects: challenges, opportunities, and next steps. CNS Spectr. 2020;26(4):333–337.
    1. Volkow ND, et al. Decreases in dopamine receptors but not in dopamine transporters in alcoholics. Alcohol Clin Exp Res. 1996;20(9):1594–1598. doi: 10.1111/j.1530-0277.1996.tb05936.x.
    1. Yen CH, et al. Reduced dopamine transporter availability and neurocognitive deficits in male patients with alcohol dependence. PLoS One. 2015;10(6):1–14.
    1. Volkow ND, et al. Profound decreases in dopamine release in striatum in detoxified alcoholics: possible orbitofrontal involvement. J Neurosci. 2007;27(46):12700–12706. doi: 10.1523/JNEUROSCI.3371-07.2007.
    1. Jensen ME, et al. Glucagon-like peptide-1 receptor regulation of basal dopamine transporter activity is species-dependent. Neurochem Int. 2020;138:104772. doi: 10.1016/j.neuint.2020.104772.
    1. Anandhakrishnan A, Korbonits M. Glucagon-like peptide 1 in the pathophysiology and pharmacotherapy of clinical obesity. World J Diabetes. 2016;7(20):572–598. doi: 10.4239/wjd.v7.i20.572.
    1. Eldor R, et al. Discordance between central (brain) and pancreatic action of exenatide in lean and obese subjects. Diabetes Care. 2016;39(10):1804–1810. doi: 10.2337/dc15-2706.
    1. Edwards CMB, et al. Exendin-4 reduces fasting and postprandial glucose and decreases energy intake in healthy volunteers. Am J Physiol Endocrinol Metab. 2001;281(1 44-1):155–E161.
    1. Biery JR, et al. Alcohol craving in rehabilitation: assessment of nutrition therapy. J Am Diet Assoc. 1991;91(4):463–466. doi: 10.1016/S0002-8223(21)01147-0.
    1. Johansen NJ, et al. Effect of short-acting exenatide administered three times daily on markers of cardiovascular disease in type 1 diabetes: a randomized double-blind placebo-controlled trial. Diabetes Obes Metab. 2020;22(9):1639–1647. doi: 10.1111/dom.14078.
    1. Skytte MJ, et al. Effects of a highly controlled carbohydrate-reduced high-protein diet on markers of oxidatively generated nucleic acid modifications and inflammation in weight stable participants with type 2 diabetes; a randomized controlled trial. Scand J Clin Lab Invest. 2020;80(5):401–407. doi: 10.1080/00365513.2020.1759137.
    1. Kjær LK, et al. Cardiovascular and all-cause mortality risk associated with urinary excretion of 8-oxoguo, a biomarker for RNA oxidation, in patients with type 2 diabetes: a prospective cohort study. Diabetes Care. 2017;40(12):1771–1778. doi: 10.2337/dc17-1150.
    1. Mabilleau G, et al. Novel skeletal effects of glucagon-like peptide-1 (GLP-1) receptor agonists. J Endocrinol. 2018;236(1):R29–R42. doi: 10.1530/JOE-17-0278.
    1. Storgaard H, et al. Glucagon-like peptide-1 receptor agonists and risk of acute pancreatitis in patients with type 2 diabetes. Diabetes Obes Metab. 2017;19(6):906–908. doi: 10.1111/dom.12885.
    1. Sattar N, et al. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of randomised trials. Lancet Diabetes Endocrinol. 2021;9(10):653–662. doi: 10.1016/S2213-8587(21)00203-5.
    1. Jones SC, et al. Injection-site nodules associated with the use of exenatide extended-release reported to the U.S. Food and Drug Administration Adverse Event Reporting System. Diabetes Spectr. 2015;28(4):283–288.
    1. Trujillo J. Safety and tolerability of once-weekly GLP-1 receptor agonists in type 2 diabetes. J Clin Pharm Ther. 2020;45(s1):43–60. doi: 10.1111/jcpt.13225.
    1. Drucker DJ, et al. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority study. Lancet. 2008;372(9645):1240–1250. doi: 10.1016/S0140-6736(08)61206-4.
    1. MacConell L, et al. Safety and tolerability of exenatide once weekly in patients with type 2 diabetes: an integrated analysis of 4,328 patients. Diabetes Metab Syndr Obes. 2015;8:241–253.
    1. Haber PS, Kortt NC. Alcohol use disorder and the gut. Addiction. 2021;116(3):658–667. doi: 10.1111/add.15147.
    1. Ellero C, et al. Prophylactic use of anti-emetic medications reduced nausea and vomiting associated with exenatide treatment: a retrospective analysis of an open-label, parallel-group, single-dose study in healthy subjects. Diabet Med. 2010;27(10):1168–1173. doi: 10.1111/j.1464-5491.2010.03085.x.
    1. Hallgren KA, Witkiewitz K. Missing data in alcohol clinical trials: a comparison of methods. Alcohol Clin Exp Res. 2013;37(12):2152–2160. doi: 10.1111/acer.12205.
    1. European Medicines Agency. Guideline on the Development of Medicinal Products for the Treatment of Alcohol Dependence. Updated February 18, 2010. Accessed September 1, 2022. .
    1. Wallach JD, et al. Characteristics of ongoing clinical trials for alcohol use disorder registered on . JAMA Psychiatry. 2020;77(10):1081–1084. doi: 10.1001/jamapsychiatry.2020.1167.
    1. Fineman M, et al. Pharmacokinetics and pharmacodynamics of exenatide extended-release after single and multiple dosing. Clin Pharmacokinet. 2011;50(1):65–74. doi: 10.2165/11585880-000000000-00000.
    1. Dickson SL, et al. The glucagon-like peptide 1 (GLP-1) analogue, exendin-4, decreases the rewarding value of food: a new role for mesolimbic GLP-1 receptors. J Neurosci. 2012;32(14):4812–4820. doi: 10.1523/JNEUROSCI.6326-11.2012.
    1. Thomsen M, et al. The glucagon-like peptide 1 receptor agonist Exendin-4 decreases relapse-like drinking in socially housed mice. Pharmacol Biochem Behav. 2017;160:14–20. doi: 10.1016/j.pbb.2017.07.014.
    1. Egecioglu E, et al. The glucagon-like peptide 1 analogue Exendin-4 attenuates alcohol mediated behaviors in rodents. Psychoneuroendocrinology. 2013;38(8):1259–1270. doi: 10.1016/j.psyneuen.2012.11.009.
    1. Taylor K, et al. Day-long subcutaneous infusion of exenatide lowers glycemia in patients with type 2 diabetes. Horm Metab Res. 2005;37(10):627–632. doi: 10.1055/s-2005-870529.
    1. Fineman MS, et al. Clinical relevance of anti-exenatide antibodies: safety, efficacy and cross-reactivity with long-term treatment. Diabetes Obes Metab. 2012;14(6):546–554. doi: 10.1111/j.1463-1326.2012.01561.x.
    1. Schacht J, et al. Functional neuroimaging studies of alcohol cue reactivity: a quantitative meta-analysis and systematic review. Addict Biol. 2013;18(1):121–133. doi: 10.1111/j.1369-1600.2012.00464.x.
    1. Courtney KE, et al. Neural substrates of cue reactivity: association with treatment outcomes and relapse. Addict Biol. 2016;21(1):3–22. doi: 10.1111/adb.12314.
    1. Bernhoft IM, Behrensdorff I. Effect of lowering the alcohol limit in Denmark. Accid Anal Prev. 2003;35(4):515–525. doi: 10.1016/S0001-4575(02)00029-5.
    1. Sobell L, Sobell M. Alcohol consumption measures. In: Allen JP, ed. Assessing Alcohol Problems: A Guide for Clinicians and Researchers. US Department of Health and Human Services; 1995:55–73.
    1. Harris PA, et al. The REDCap Consortium: building an international community of software platform partners. J Biomed Inform. 2019;95:103208. doi: 10.1016/j.jbi.2019.103208.
    1. Elashoff M, et al. Pancreatitis, pancreatic, and thyroid cancer with glucagon-like peptide-1-based therapies. Gastroenterology. 2011;141(1):150–156. doi: 10.1053/j.gastro.2011.02.018.
    1. Klausen MK, et al. Exenatide once weekly for alcohol use disorder investigated in a randomized, placebo-controlled clinical trial. Mendeley Data. Published September 16, 2022. Accessed September 22, 2022.
    1. van Buuren S, Groothuis-Oudshoorn K. mice: multivariate imputation by chained equations in R. J Stat Softw. 2011;45(3):1–67.
    1. R: A Language and Environment for Statistical Computing. Version 4.2.0. R Foundation for Statistical Computing; 2022.
    1. World Health Organization. Obesity: preventing and managing the global epidemic. Report of a WHO consultation. World Health Organ Tech Rep Ser. 2000;894:1–253.
    1. Fink-Jensen A. Does Treatment With GLP-1 Reduce Alcohol Intake in Patients With Alcohol Dependence? (EXALT). US National Library of Medicine. . Published July 27, 2017. Updated June 4, 2021. Accessed September 22, 2022.

Source: PubMed

3
Sottoscrivi