Skip to main content
Erschienen in: Drugs 3/2022

Open Access 01.02.2022 | Review Article

Novel Agents for the Pharmacological Treatment of Alcohol Use Disorder

verfasst von: Elizabeth M. Burnette, Steven J. Nieto, Erica N. Grodin, Lindsay R. Meredith, Brian Hurley, Karen Miotto, Artha J. Gillis, Lara A. Ray

Erschienen in: Drugs | Ausgabe 3/2022

Abstract

Alcohol use disorder (AUD) is a highly prevalent but severely under-treated disorder, with only three widely-approved pharmacotherapies. Given that AUD is a very heterogeneous disorder, it is unlikely that one single medication will be effective for all individuals with an AUD. As such, there is a need to develop new, more effective, and diverse pharmacological treatment options for AUD with the hopes of increasing utilization and improving care. In this qualitative literature review, we discuss the efficacy, mechanism of action, and tolerability of approved, repurposed, and novel pharmacotherapies for the treatment of AUD with a clinical perspective. Pharmacotherapies discussed include: disulfiram, acamprosate, naltrexone, nalmefene, topiramate, gabapentin, varenicline, baclofen, sodium oxybate, aripiprazole, ondansetron, mifepristone, ibudilast, suvorexant, prazosin, doxazosin, N-acetylcysteine, GET73, ASP8062, ABT-436, PF-5190457, and cannabidiol. Overall, many repurposed and novel agents discussed in this review demonstrate clinical effectiveness and promise for the future of AUD treatment. Importantly, these medications also offer potential improvements towards the advancement of precision medicine and personalized treatment for the heterogeneous AUD population. However, there remains a great need to improve access to treatment, increase the menu of approved pharmacological treatments, and de-stigmatize and increase treatment-seeking for AUD.
Key Points
Alcohol use disorder is severely under-treated, and the development of new and more effective pharmacotherapies is necessary.
The repurposed and novel agents discussed in this qualitative literature review offer promise for the future of AUD treatment, including advancements toward precision medicine for the heterogeneous AUD population.

1 Introduction

Alcohol use disorder (AUD) is a highly prevalent, chronic, relapsing condition characterized by an impaired ability to stop or control alcohol use despite clinically significant impairment, distress, or other adverse consequences [1, 2]. It is the most common substance use disorder: in 2016, 100.4 million people globally were estimated to have an AUD [3]. This disorder represents a significant public health concern. The WHO estimates that alcohol consumption is responsible for 5.9% of all deaths (7.6% in men, 4.0% in women) and 5.1% of global disease burden [4]. Alcohol use and misuse is thought to contribute to over 200 related diseases and health conditions globally, including cardiovascular disease, cancer, liver cirrhosis, and injuries [4]. AUD is also often comorbid with other substance use disorders, major depressive disorder, bipolar disorder, and other psychiatric disorders [2].
In the USA alone, AUD is estimated to contribute to about 88,000 deaths each year [5]. An estimated 44.6 million adults per year in the USA suffer from AUD, and 93.4 million (approximately 33%) adults in the USA will meet or have met AUD criteria at some point in their life [2]. Furthermore, the economic burden of AUD is estimated to be approximately $250 billion across the USA [6].
Although AUD is an important public health concern, the disorder remains severely under treated with only 7% of adults with AUD in the USA [7] and less than 10% in Europe [8] receiving pharmacotherapy and/or psychotherapy treatment. Furthermore, the lag between the age at which AUD onsets and the age of first AUD treatment has been estimated to be eight years on average [7]. Pharmacotherapies have seen limited use in the treatment of AUD, partially due to lack of addiction treatment training for medical professionals, lack of awareness regarding medication options (e.g., limited marketing), reluctance to prescribe and take medications, perceived low efficacy of medication, and stigma surrounding treatment. Indeed, only three pharmacotherapies are approved by the US Food and Drug Administration (FDA) for use in AUD treatment. These medications are disulfiram (Antabuse), acamprosate, and naltrexone (formulated for oral administration or extended-release injection) [911]. The European Medicines Agency (EMA) similarly recognizes only these same three medications, as well as nalmefene, as established pharmacotherapies for AUD [12]. These medications are only modestly effective [13] and are under-utilized in treatment. A study conducted in 2019 found that only 1.6% of adults in the USA with past-year AUD received evidence-based AUD pharmacotherapies [14].
Treatment outcomes for AUD differ widely across patients and medications. While abstinence may be desirable, it is infrequently obtained, such that only 16% of individuals in treatment for AUD achieve abstinence [15]. Furthermore, evidence does not support abstinence as the only approach in the treatment of AUD [16]. Not all individuals with AUD consider abstinence to be a goal of their recovery; only 2–6% of goals set in patient-driven treatment center on attaining alcohol abstinence [17]. Non-abstinent recovery, including reductions in drinking and heavy drinking in particular (e.g., controlled drinking / harm reduction), has been recognized to have health and societal value and has gained traction as a treatment target [1820]. Indeed, non-abstinent AUD recovery has been shown to be sustainable for up to 10 years following treatment [21]. Despite growing recognition of the benefits of harm reduction, however, the most commonly prescribed pharmacotherapy to treat AUD remains disulfiram, a medication advised strictly for abstinence [22]. Furthermore, the heterogeneity of AUD suggests that it will be unlikely that one single medication will be effective for all individuals with an AUD. Therefore, there is a pressing need for the development of novel, diverse, and effective pharmacological treatment options for AUD with the hopes of increasing utilization and improving care. The focus of the current review is to summarize pharmacotherapies for AUD with a clinical perspective. Specifically, this review provides a brief overview of currently approved medications and identifies new and repurposed agents “on the horizon” for which evidence indicates a potentially effective application toward AUD treatment.

2 Pharmacological Treatments for AUD

The following sections examine pharmacotherapies approved or in-development for AUD. Medications were selected for this qualitative review by considering gaps in existing review articles and the expertise of all authors; information was gathered via qualitative PubMed literature searches. This review is organized into sections based on drug approval status, namely (a) approved medications, (b) repurposed (i.e., off-label) medications, and (c) novel agents. Within each medication section, we examine the basic mechanism of action, evaluate preclinical research testing the efficacy of the medication in mitigating alcohol-related behaviors in animal models, and review clinical findings from human laboratory studies and randomized controlled trials (RCTs) where available. We also examine the tolerability and potential personalized applications of each drug by identifying populations in which the drug may be particularly effective, and indicate treatment targets (drinking reduction, achievement, or maintenance of abstinence). This information is summarized in Table 1. We conclude by discussing future directions for the development of pharmacological treatments and precision medicine for AUD.
Table 1
Pharmacotherapies approved and under development for the treatment of AUD
Agent
Mechanism of action
Approval status
Preclinical results
Clinical results
Target outcome
Disulfiram
Aldehyde dehydrogenase inhibitor; dopamine beta-hydroxylase inhibitor
FDA- and EMA-approved for AUD
Mixed results: prevents heavy but not moderate drinking [297]; chronic alcohol induces disulfiram tolerance [298]
Mixed results; drinking outcomes improved with supervised administration [2933]
Achieve and maintain abstinence
Acamprosate
Modulates glutamatergic activity
FDA- and EMA-approved for AUD
Reduced ethanol intake, withdrawal symptoms, place preference [299302]
Mixed results; Reduced risk of relapse, increased cumulative abstinence duration [36, 44, 49]. Negative studies show no benefit over placebo [3742]
Achieve and maintain abstinence
Naltrexone
Opioid receptor antagonist
FDA- and EMA-approved for AUD
Reduced binge drinking, ethanol preference, motor impairment and sedation [303305]
Reduced risk of relapse, binge drinking, and craving [5259]; modest effect sizes [6264]
Achieve and maintain abstinence and reduce drinking
Nalmefene
Opioid receptor antagonist and partial agonist
EMA-approved for AUD; FDA-approved for opioid overdose
Reduced alcohol seeking, relapse and binge-like drinking, neuroinflammation [306308]
Reduced binge drinking and total alcohol consumption [7173, 75]
Reduce drinking
Baclofen
GABAB agonist
Approved for AUD in France
Reduced ethanol self-administration and motivational properties [309312]
Mixed results; increased rates of abstinence, time to first relapse, possibly reduced heavy-drinking days [8486, 91, 92]
Achieve and maintain abstinence
Sodium oxybate
Modulates GABA activity
Approved for AUD in Italy and Austria
Reduced ethanol self-administration, withdrawal symptoms, and relapse-like drinking [247, 313]
Effective treatment of alcohol withdrawal syndrome, increased abstinence [99, 100]
Reduce withdrawal symptoms and achieve abstinence
Topiramate
Inhibits glutamatergic activity and increases GABA activity
Repurposed
Reduced ethanol intake in rodent models [113115]
Reduced drinks per day and percent heavy-drinking days and increased percent days abstinent [112, 116119]
Achieve abstinence and reduce drinking
Gabapentin
Modulates GABA activity
Repurposed
Mixed results: reduced or increased ethanol intake [123125]
Reduced percent heavy-drinking days, alcohol consumption, and abstinence rates and increased time to relapse, with outcomes improved among those with alcohol withdrawal symptoms [126135]
Achieve abstinence and reduce drinking
Varenicline
Nicotinic acetylcholine receptor agonist
Repurposed
Reduced ethanol seeking, intake, and binge-like consumption [138140]
Reduced percent heavy-drinking days and drinks per day with outcomes improved for those who smoke cigarettes [142144, 146149]
Reduce drinking
Aripiprazole
Dopamine receptor partial agonist and 5-HT2A receptor antagonist
Repurposed
Reduce ethanol-induced place preference and ethanol consumption [151153]
Mixed results with outcomes improved for more impulsive individuals; reduced heavy-drinking days and increased days abstinent at rates comparable to naltrexone [154160]
Reduce drinking
Ondansetron
5-HT3 receptor antagonist
Repurposed
Blocked sensitization to locomotor stimulant effects, reduced voluntary ethanol intake [167169]
Reduced drinks per day and increased days abstinent in patients with early-onset AUD [170174]
Achieve abstinence and reduce drinking
Mifepristone
Glucocorticoid receptor antagonist
Repurposed
Reduced ethanol-seeking in rodents [177, 178]
Mixed results in primates; reduced chronic voluntary alcohol use but did not prevent relapse [179, 180]
Reduced alcohol craving and consumption [178, 181, 182]
Reduce drinking
Ibudilast
Selective PDE inhibitor and MIF inhibitor
Repurposed
Reduced ethanol intake [188191]
Reduced craving, odds of heavy drinking, and neural cue-reactivity [192, 193]
Reduce drinking
Prazosin and Doxazosin
Adrenergic alpha-1 antagonists
Repurposed
Reduced relapse, ethanol intake, and stress-induced ethanol seeking [201205]
Mixed results; reduced craving and alcohol consumption. Outcomes improved among those with alcohol withdrawal symptoms [206211]
Reduce drinking
N-Acetylcysteine
Modulates synaptic glutamatergic activity
Repurposed
Reduced ethanol intake and relapse [215220]
Increased abstinence rates and reduced drinks per week among those with cannabis use disorder [222, 223]
Reduce drinking
Suvorexant
Dual orexin antagonist
Repurposed
Reduced ethanol consumption and relapse [229238]
Unknown
ABT-436
Highly selective vasopressin type 1B receptor antagonist
Novel agent
Attenuated ethanol reinstatement and reduced ethanol intake [241, 242]
Increased days abstinent but did not reduce heavy drinking or craving [244]
Achieve and maintain abstinence
GET73
Gamma-hydroxy-butyrate analogue; negative allosteric modulator at mGluR5
Novel agent
Reduced ethanol intake and suppressed relapse [246]
Unknown
ASP8062
Positive allosteric modulator of GABAB receptor
Novel agent
Reduced ethanol self-administration [259]
Unknown
PF-5190457
Ghrelin receptor inverse agonist
Novel agent
Reduced ethanol intake and preference [266270]
Reduced alcohol craving and cue-reactivity [272]
Unknown
Cannabidiol
Diverse pharmacological effects
Novel agent
Reduced ethanol administration and relapse-like behavior [279]
Unknown
5-HT3 serotonin receptor 3, 5-HT2A serotonin receptor 2A, AUD alcohol use disorder, EMA European Medicines Agency, FDA United States Food and Drug Administration, GABA gamma-aminobutyric acid, mGluR5 metabotropic glutamate receptor 5, MIF macrophage migration inhibitory factor, PDE phosphodiesterase

2.1 Approved Agents

This section briefly describes medications currently approved by agencies in many countries including the US FDA and EMA for the treatment of AUD: disulfiram, acamprosate, and naltrexone (oral and extended-release), as well as nalmefene, which is EMA-approved. This section also reviews baclofen and sodium oxybate, which are medications approved for AUD treatment by agencies in European countries but not by the FDA or EMA. As many of these pharmacotherapies have been extensively discussed in the literature, our review of these medications primarily focuses on clinical trials and recent meta-analyses.

2.1.1 Disulfiram

Disulfiram, the first medication FDA-approved for AUD treatment (in 1951), is an aldehyde dehydrogenase inhibitor that acts by blocking the metabolism of alcohol, increasing acetaldehyde concentration. Acetaldehyde, a toxic metabolite of ethanol, produces an alcohol-induced aversive response, characterized by nausea, vomiting, sweating, flushing, and heart palpitations [23]. Unsurprisingly, these unpleasant effects give disulfiram a relatively poor adherence rate [24, 25]. Disulfiram may also act on dopamine systems, as its major metabolite inhibits the enzyme dopamine beta-hydroxylase (DBH), which aids in metabolizing dopamine into noradrenaline [22, 26, 27]. Serum DBH activity is associated with withdrawal symptoms, and disulfiram has been shown to reduce serum DBH levels [28].
A meta-analysis of 22 RCTs found that disulfiram saw increased success rates compared to placebo in open-label studies only—blinded trials yielded no statistical significance between disulfiram and placebo [29]. However, while these findings do not appear to support the use of disulfiram for treating AUD, this outcome may be due to placebo effects. Research showed that placebo-treated individuals showed decreases in cue-reactivity to alcohol stimuli in a sample of 38 participants [30], and experiencing an acetaldehyde reaction did not necessarily improve treatment response in a sample of 46 participants [31]. Instead, a patient simply being aware of a potential adverse reaction appears to be enough to influence drinking behavior [32].
Disulfiram ingestion under supervision (to ensure adherence) saw significantly better success rates compared to non-supervised treatment, suggesting that supervised administration of disulfiram may still have a place in treating individuals struggling to attain sobriety [33], and unsupervised disulfiram may also be helpful for individuals highly motivated for abstinence. However, adherence management issues limit the utility of disulfiram in the treatment of AUD, and the disulfiram-ethanol interaction can sometimes present as a medical emergency. Therefore, disulfiram is only recommended in the maintenance of abstinence; using this medication to reduce drinking is not advised [34].

2.1.2 Acamprosate

While the specific mechanisms through which acamprosate works to treat AUD remain under investigation, it is thought to act on the glutamatergic system as an N-methyl-d-aspartic acid (NMDA) receptor partial co-agonist [35]. This may reduce neuronal hyperexcitability, a phenomenon that occurs in acute withdrawal and protracted abstinence from alcohol.
A meta-analysis of 27 RCTs with a total of 7519 participants found that acamprosate treatment reduced risk of abstinent patients returning to any drinking, but did not reduce rates of binge drinking [36]. A number of trials have also found that acamprosate did not show a significant benefit over placebo [3741]. In particular, a large scale trial (COMBINE), which compared acamprosate, naltrexone, and behavioral therapies, both individually and combined with each other, against placebo (N = 1383), found that acamprosate had no significant effect on drinking in comparison to placebo, either alone or in combination with naltrexone and/or behavioral intervention [42]. Placebo effects in this trial may explain some of these negative outcomes, as might differences in trial design and patient characteristics: COMBINE required 4 days of pre-trial abstinence while European trials with positive outcomes were typically conducted in inpatient populations requiring complete detoxification [43]. Overall, however, a Cochrane meta-analysis of 24 RCTs with 6915 participants found that acamprosate significantly reduced the risk of any drinking and increased cumulative duration of abstinence [44].
Acamprosate is generally well tolerated [34] and may also have neuroprotective effects [45, 46]. As chronic alcohol abuse is associated with neuronal changes related to NMDA receptors, this neuroprotection may be particularly important in AUD treatment [47, 48]. Acamprosate is recommended for the achievement and maintenance of complete abstinence, rather than for the reduction of drinking or prevention of relapse in the event of drinking [49]. It is FDA-approved for abstinence maintenance in AUD patients who are abstinent when beginning treatment.

2.1.3 Naltrexone (Oral and Extended-Release)

Naltrexone, the best-studied of these three commonly approved medications, was originally approved to treat opioid use disorder. Naltrexone is an antagonist of the mu opioid receptor (with additional affinity for kappa and delta opioid receptors). By attenuating alcohol-induced opioidergic activity in the mesolimbic dopamine system, opioid antagonists like naltrexone, modulate the rewarding effects of alcohol (mediated by dopamine), thereby reducing alcohol consumption [50, 51].
In 1994, the FDA approved oral naltrexone to treat alcohol dependence after two independent 12-week trials, which included 97 and 70 participants, respectively, found that naltrexone significantly decreased drinking days and relapse rates [52, 53]. Additional recent trials demonstrate that naltrexone reduces the rewarding effects of alcohol [54, 55], alcohol craving [40, 56], drinks per drinking day [57], and relapse rates [58, 59], strengthening the initial findings.
Extended-release injectable naltrexone (i.e., Vivitrol), administered once monthly by a medical professional, may be beneficial for individuals who are more sensitive to naltrexone’s adverse side effects or have difficulty adhering to oral medication [60]. A six-month multisite trial of 380 mg injectable naltrexone in 624 patients with alcohol dependence found significant reductions in heavy-drinking days versus placebo [61]. However, evidence for naltrexone remains mixed. Another RCT reported no significant differences between naltrexone and placebo (N = 183), and that the clinical utility of naltrexone was limited by its adverse effects [62]. Additionally, clinical trials of naltrexone often yield modest effect sizes [63]. A meta-analysis of 53 naltrexone RCTs with a total of 9140 participants found naltrexone to significantly reduce both the risk of return to any drinking and return to binge drinking [36]; however, both of these associations were modest in magnitude [64].
Oral and injectable naltrexone show similar decreases in likelihood of binge drinking [65] and both are generally well tolerated, with fairly mild side effects [66]. Importantly, however, naltrexone does block the therapeutic effects of opioid analgesics and can precipitate opioid withdrawal in patients who have developed physical dependence to opioids [34]; therefore, individuals who are prescribed naltrexone for AUD must be monitored for opioid use and withdrawal. Of note, naltrexone is contraindicated for patients with acute hepatitis or liver failure, and should be “carefully considered” in patients with acute liver disease [67], potentially limiting its use in the alcohol-associated liver disease (AALD) population. In summary, naltrexone appears to have a moderate effect on the reduction of alcohol use.

2.1.4 Nalmefene

Nalmefene works in a similar manner to naltrexone as an antagonist at the mu and delta opioid receptor, but is also a kappa opioid receptor partial agonist [68]. Via its kappa agonist activity, particularly centered in dopaminergic nucleus accumbens circuitry, nalmefene may reduce motivation for self-administration and withdrawal-induced alcohol consumption [69, 70].
Nalmefene was approved by the EMA in 2013 for the reduction of alcohol consumption among patients with AUD. Approval followed findings from three multicenter 6-month clinical trials enrolling 604, 667, and 718 individuals, respectively, in which participants took the medication or placebo on an as-needed basis [7173]. In these trials, nalmefene decreased total alcohol consumption and heavy-drinking days. However, the drug’s approval based on these studies has received criticism due to limited evidence of efficacy, especially as these trials were conducted only against placebo rather than an active medication comparison (e.g., naltrexone) [34, 74]. A more recent meta-analysis of the efficacy of nalmefene, which included five RCTs (N = 2567), found that participants treated with nalmefene had 1.65 fewer heavy-drinking days per month than participants treated with placebo after 6 months [75].
Studies indicate that nalmefene is associated with more adverse events and study dropouts compared to placebo, and the most frequently reported of these are dizziness, nausea, vomiting, insomnia, and headache [36, 76]. Overall, while nalmefene may reduce heavy-drinking rates, its effects on other outcomes [77] remain small-to-moderate, and study withdrawals related to adverse events are common in nalmefene trials. However, similar to naltrexone, this medication may appeal to patients with goals of reducing alcohol consumption and those reluctant to engage in abstinence-based treatment.

2.1.5 Baclofen

Baclofen acts on the γ-amino butyric acid (GABA) system as a GABAB agonist. It was approved for treatment of AUD in France in 2018 [78] and has been used off-label for AUD for over a decade in other countries, especially other European countries and Australia [79, 80].
Clinical trials of baclofen have produced mixed results [6567]. A recent meta-analysis of 12 RCTs (N = 703) showed that baclofen in comparison to placebo was associated with higher abstinence rates [8183]; however, it did not increase abstinent days or decrease craving, heavy drinking, depression, or anxiety [84]. Another meta-analysis of 13 RCTs (N = 1492) indicated that baclofen (again vs placebo) was associated with longer time to relapse and a larger percentage of abstinent patients [85]. Furthermore, greater alcohol use at baseline was correlated with a greater treatment effect [85]. In contrast, however, another recent meta-analysis of 12 RCTs with 1128 total participants found no significant differences between baclofen and placebo in primary (i.e., percent abstinent days, percent heavy-drinking days, return to any drinking, and study dropout) or secondary outcomes of interest (i.e., anxiety and craving); however, baclofen did increase depression and adverse effects including sedation and vertigo [86].
Baclofen’s significant adverse side effects include drowsiness, sedation, headache, vertigo, confusion, perspiration, muscle stiffness and/or abnormal movements, slurred speech, and numbness [8688]. Tolerance to baclofen may also develop with chronic administration [89, 90]. Additionally, dose and sex may moderate baclofen tolerability and response; escalation of dosage in response to developing tolerance can increase sedative side effects, which affect women significantly more than men at the same dose [91]. Importantly, cessation or reduction in dose can precipitate potentially life-threatening withdrawal syndrome [90].
The variability in baclofen’s effectiveness seen across studies may be partially explained by high baclofen pharmacokinetic variability seen among individuals with AUD. This heterogeneity is an important factor to take into account when considering baclofen as an AUD treatment [92]. Of note, there is also some evidence that baclofen might be particularly useful in treatment of AUD among individuals with liver disease [93, 94].
In summary, baclofen seems to effectively promote abstinence; however, it shows mixed results regarding its clinical effects on non-abstinence outcomes (e.g., reduction in heavy drinking), significant adverse side effects, and inter-individual variability in response. As such, baclofen as a treatment of AUD—as well as its optimal dosage—continues to be debated [95].

2.1.6 Sodium Oxybate

Sodium oxybate (SMO), the sodium salt of gamma-hydroxybutyrate (GHB), has been utilized as a medication for a number of disorders. SMO is approved in Italy and Austria for the treatment of AUD [96]. SMO acts on the GABA system both directly as a GABAB partial agonist and indirectly through GHB-derived GABA [97, 98].
A Cochrane meta-analysis of 13 RCTs with a total of 649 participants found that SMO (50 mg) was effective compared to placebo in the treatment of alcohol withdrawal syndrome (AWS) and in preventing relapses in previously detoxified participants, and that SMO was more effective than naltrexone and disulfiram in maintaining abstinence [99]. Another recent meta-analysis found that SMO, compared to placebo, increased abstinence rates by up to 34% in a sample of 711 participants with very high drinking risk levels [100]. However, use of GHB as a recreational drug raises concern regarding the abuse potential of SMO. Indeed, in another clinical study with a sample of 48 participants, craving for SMO was observed in 10% of participants with psychiatric comorbidities [101], indicating that patients may be at risk of developing craving for and abusing SMO.
SMO may be particularly effective in combination with other pharmacotherapies. It was shown to reduce relapses when administered with naltrexone and escitalopram [102]. A study of 52 subjects with chronic, treatment-resistant alcohol dependence found that participants stayed in treatment significantly longer when SMO was co-administered with disulfiram than with SMO alone [103]. Additionally, in a case study of seven partial- and non-responders to SMO treatment, co-administration with nalmefene was effective in promoting abstinence, reducing heavy-drinking episodes, and importantly, suppressing craving for SMO [104].
SMO craving and abuse potential may limit its use; however, at therapeutic doses, SMO abuse seems to be a relatively infrequent phenomenon among patients without psychiatric comorbidities or poly-drug use [96, 100, 105]. Safety data show that serious adverse events with SMO treatment are rare, with the most common adverse effects being transitory dizziness and vertigo [96]. Overall, the effectiveness of SMO lies largely in reducing withdrawal symptoms and increasing abstinence rates. Its therapeutic use remains controversial due to its potential for craving and abuse, and further study on co-administration with other pharmacotherapies appears promising.

2.2 Repurposed Agents

The following sections address medications that have been approved by the FDA for applications other than AUD but are in development to be repurposed (i.e., used off-label) to treat AUD.

2.2.1 Topiramate

Despite that the anti-convulsant medication topiramate has not been approved by the FDA or EMA for AUD treatment, the US Department of Veterans Affairs recommends topiramate as a treatment for AUD, and it has been suggested by the American Psychiatric Association (APA) for the pharmacological treatment of patients with AUD [106, 107]. Currently, the specific mechanisms of action of topiramate remain under investigation. However, the drug is thought to inhibit glutamate α-amino 3-hydroxy-5-methyl-4-isoxazoleproprionic acid (AMPA) and kainate receptors [108110] and L-type calcium channels [111], as well as enhance the inhibitory activity of GABA [109]. These effects, taken together, work to attenuate dopaminergic activity mesolimbic reward circuits, thereby reducing both alcohol craving and withdrawal symptoms [112].
Preclinically, topiramate was seen to decrease ethanol consumption in rodent models [113115]. Clinical studies have found that topiramate reduced craving and alcohol drinking in a sample of 150 participants over 12 weeks [112], and decreased drinking days, heavy-drinking days, and number of drinks per day in a sample of 85 participants in a 14-week trial [116]. A large, multisite 14-week RCT that enrolled 371 participants with alcohol dependence found reductions in heavy-drinking days and improvements in various self-reported drinking-related outcomes [117]. A meta-analysis including seven RCTs of topiramate for AUD with a total of 1125 participants found that topiramate significantly increased number of days abstinence and decreased heavy-drinking days compared to placebo [118]. Of interest, this meta-analysis also directly compared effect sizes for topiramate and naltrexone, finding that topiramate was significantly more effective than naltrexone in reducing binge drinking, but not in improving abstinence [118]. Another recent month-long study of 94 patients that directly compared topiramate and baclofen demonstrated that baclofen was overall more effective (i.e., 61% of participants receiving baclofen achieved abstinence vs 38% with topiramate) and better tolerated (i.e., 33% of participants receiving topiramate dropped out of the study vs 24% with baclofen) than topiramate [119].
The potential of reduced tolerability and significant side effects such as pruritis, paresthesia, anorexia, dysgeusia, dizziness, nervousness, and cognitive impairment, including difficulty with concentration and attention, are concerns for the use of topiramate in the treatment of AUD [50]. These cognitive impairments were found to be dose-associated and include impairments in working memory and verbal fluency, as well as mental slowing [116]. Topiramate’s side effects, especially those affecting memory and cognition, present reasons for concern and warrant long-term trials to further investigate these effects. Slow dose titration and ongoing patient monitoring is also recommended.
It is worth noting a potential strength of topiramate: the possibility of starting treatment while patients are still actively drinking, allowing topiramate to serve as a harm-reduction or abstinence-initiation treatment, rather than only being used to prevent relapse in already detoxified patients [120]. Topiramate has already been demonstrably effective in clinical applications across a number of drinking outcomes [121], but longer-term trials of topiramate are recommended to optimize dose and duration, as well as further exploring side effects.

2.2.2 Gabapentin

Another anti-convulsant medication, gabapentin, is also suggested by the APA for use in AUD treatment with the goal of reducing alcohol consumption or achieving abstinence [107]. It is thought to modulate GABA activity by indirectly interacting with voltage-gated calcium channels [122]. Preclinical studies of gabapentin’s effects on ethanol consumption have shown mixed results, with some studies finding that gabapentin reduced ethanol intake in alcohol-dependent rodents [123] and others finding that it increased self-administration and binge-like drinking [124, 125].
Gabapentin has shown promising results in human laboratory studies and clinical trials. Gabapentin reduced percent heavy-drinking days and drinks per day, as well as increased percentage abstinent days in a 28-day trial with 60 participants [126]. In another 10-day study with 21 participants, gabapentin significantly delayed return to heavy drinking [127]. In a proof-of-concept one-week human laboratory trial (N = 33), participants treated with gabapentin showed significant decreases in alcohol craving in comparison to placebo [128], and a follow-up 12-week RCT with 150 participants found that gabapentin significantly increased abstinence rates and percentage of no heavy-drinking days [129]. Gabapentin may also be particularly effective in combination with other medications. For instance, a combination of gabapentin and flumazenil over 39 days in 60 participants increased percentage of abstinent days and delayed time to first heavy-drinking day compared to placebo. This trial also showed an interaction effect with pre-study alcohol withdrawal, such that those with more severe withdrawal symptoms at baseline benefitted more from the treatment during the trial [130]. In combination with naltrexone, gabapentin was also shown to be more effective (i.e., decreased heavy-drinking days, fewer drinks per drinking day, and delayed time to heavy drinking) compared with naltrexone alone in a 6-week trial with 150 participants [131].
A Cochrane review of 25 studies (N = 2641) found anticonvulsants, including gabapentin, to significantly reduce both heavy-drinking days and drinks per drinking day in comparison to placebo. Anticonvulsants were also associated with longer time-to-relapse and fewer heavy-drinking days compared to naltrexone [132]. A recent meta-analysis of 7 RCTs with a total of 751 participants found that gabapentin compared to placebo only significantly decreased percent heavy-drinking days, although trend-level effect estimates were shown for other alcohol-related outcomes (i.e., relapse, drinks per day, complete abstinence, percent days abstinent, and concentration of gamma-glutamyl transferase (GGT)) [133]. In a 16-week RCT with 96 participants, gabapentin improved the percentage of individuals with no heavy-drinking days and increased total abstinence rates over placebo, especially among participants with more severe pre-study withdrawal [134]. However, another recent 6-month multisite RCT of extended-release gabapentin conducted in 346 participants with at least moderate AUD found no effects over placebo for any clinical outcomes including abstinence rate, percent heavy-drinking days, drinks per drinking day, or alcohol craving [135]. While these findings may be related to potential pharmacokinetic issues relating to the specific formulation of extended-release gabapentin used in the trial, it is also possible that gabapentin may simply be more effective in patients with more clinically relevant and severe symptoms of alcohol withdrawal [130, 134].
The most common adverse events associated with gabapentin treatment are somnolence, dizziness, peripheral edema, and ataxia or gait disorder [136]. Notably, gabapentin does carry the potential for misuse and abuse, particularly in individuals with opioid use disorders [137]; therefore, recommendation of gabapentin to individuals with comorbid substance use disorder should be carefully considered. In summary, gabapentin shows some clinical efficacy, especially in populations with more severe withdrawal symptomology, but more extensive investigation is recommended.

2.2.3 Varenicline

Varenicline is a nicotinic acetylcholine receptor agonist (partial at α4β2; full at α7) that is used for the treatment of nicotine dependence. In rodent models, varenicline was shown to reduce ethanol seeking, intake, and binge-like consumption [138140]. Additionally, the combination of varenicline and naltrexone decreased alcohol drinking more effectively than either drug on its own [141].
Clinically, varenicline was found to significantly reduce weekly percent heavy-drinking days, drinks per day, drinks per drinking day, and alcohol craving compared to placebo in a 13-week multisite RCT with 200 participants [142]. However, a 12-week RCT (N = 160) found no effect of varenicline over placebo on heavy-drinking days [143], and a recent 6-week human laboratory study in 47 participants found that varenicline did not significantly attenuate cue-induced alcohol craving relative to placebo [144].
Varenicline appears to be especially relevant to heavy-drinking smokers. Approximately 20%–25% of current smokers are estimated to also be heavy drinkers [145]. A human laboratory study with 20 heavy-drinking smokers on varenicline for 7 days found that varenicline significantly reduced the number of drinks consumed and increased the likelihood of complete abstinence in the human laboratory paradigm [146]. Moderator analyses of data collected in the above multisite trial [142] indicated that varenicline was more efficacious in reducing drinking among smokers who also reduced their cigarette smoking [147] and among individuals with lower severity of AUD, such that that varenicline significantly reduced percent heavy-drinking days and drinks per drinking day in low-severity individuals, while the most severe group showed no differences between varenicline and placebo on drinking outcomes [148].
Findings also indicate that varenicline may be more effective as an AUD treatment in men, as a 16-week RCT with 131 participants found that varenicline combined with medication management decreased percent heavy-drinking days only in men, but improved smoking abstinence in both men and women [149]. Varenicline is well tolerated, suggesting that it may serve as a promising option for AUD treatment, especially in the case of individuals co-using alcohol and nicotine.

2.2.4 Aripiprazole

Aripiprazole is an antipsychotic drug that acts as a partial agonist at the dopamine D2 and 5-HT1A receptors and as an antagonist at the 5-HT2A receptor [150]. Preclinically, aripiprazole has been shown to reduce ethanol-induced place preference and decrease drinking behaviors in mice [151], as well as reducing alcohol consumption in alcohol-preferring rats [152, 153].
A human laboratory study in 18 healthy participants indicated that aripiprazole affected outcomes related to alcohol consumption (i.e., subjective response to alcohol), including reducing the euphoric effects of alcohol and increasing sedative effects [154]. In a sample of 30 participants with AUD, aripiprazole compared to placebo, was found to attenuate cue-induced neural activation in the ventral striatum, a region associated with reward [155]. Another recent clinical laboratory study in which 99 participants with AUD took aripiprazole over eight days found that aripiprazole reduced the number of drinks consumed in a bar lab setting, especially among individuals with low self-control, and prolonged the latency to drink in individuals with high impulsivity [156].
Results from clinical trials of aripiprazole are mixed. A 16-week RCT directly comparing aripiprazole against naltrexone (N = 75) found that the two medications were overall comparable in reducing heavy-drinking days and increasing days of abstinence, although patients treated with naltrexone had reduced craving compared to aripiprazole [157]. A 35-day open-label trial assessing the combination of aripiprazole and topiramate in 13 heavy-drinking participants found significant reductions in alcohol use. Additionally, this trial provided no evidence that the side effects of the two medications are additive, indicating that the combination is generally safe and well tolerated [158]. However, another recent five-week study assessing topiramate, aripiprazole, and their combination with 90 participants found that effects on drinking reduction were due to topiramate, while no significant findings were seen for aripiprazole for any outcomes [159]. Another multicenter RCT enrolling 295 participants over 12 weeks found no significant difference between the aripiprazole and placebo groups in percentage of participants completely abstinent from alcohol throughout the study or time to first drinking day; however, the average number of drinks per drinking day was significantly lower for aripiprazole [160]. The aripiprazole group yielded significantly higher discontinuation rates and earlier discontinuation, largely due to adverse events associated with side effects, especially when dosage exceeded 15 mg/day. The most common side effects cited in cases of discontinuation were fatigue, insomnia, restlessness, anxiety, and deficits in attention. Of note, long-term use of antipsychotics like aripiprazole may be associated with more severe adverse effects such as tardive dyskinesia, with risk factors including older age and female sex [161, 162].
In summary, these findings suggest that aripiprazole may be more effective at lower doses and in more impulsive individuals, although larger confirmatory studies are needed to pursue these personalized medicine approaches.

2.2.5 Ondansetron

Ondansetron is a 5-HT3 antagonist that is used to treat nausea and vomiting. Although the specific mechanism of action remains under investigation, ondansetron may address serotonergic dysfunction common in early-onset AUD [163, 164], and may reduce alcohol craving via 5-HT3 projections to dopaminergic connections in the midbrain. Preclinically, 5-HT3 antagonism has been shown to block acquisition and maintenance of ethanol self-administration [165] and reduce ethanol-associated dopamine concentration in the nucleus accumbens [166]. Ondansetron was also found to block the development and expression of sensitization to the locomotor stimulant effects of ethanol [167] and reduce voluntary ethanol intake, preference, and withdrawal seizures in rodent models [168, 169].
Clinically, ondansetron may be particularly effective in combination with naltrexone. In an 8-week RCT in 20 participants with early-onset AUD, ondansetron and naltrexone (in comparison with placebo) significantly reduced drinks per drinking day and trended towards an increase in percentage of days abstinent [170]. Another combination study in 90 participants after 7 days on ondansetron and naltrexone found that the combination decreased craving for alcohol and ventral striatal activation to alcohol cues [171].
Ondansetron may also be suitable for individuals with biological predisposition to early-onset AUD. In an 11-week RCT of 271 participants, ondansetron was shown to reduce self-reported drinking such that patients with early-onset AUD who received ondansetron reported fewer drinks per day and more days of abstinence compared to placebo [172]. Ondansetron, combined with cognitive behavioral therapy in an 8-week open-label trial (N = 40) comparing effects in early-onset versus late-onset AUD, found that drinks per drinking day and alcohol-related problems (i.e., Drinker Inventory of Consequences; DrInC) were significantly decreased among those with early-onset AUD compared to those with late-onset AUD [173]. Furthermore, in an 11-week study with 253 participants, ondansetron at 4 μg/kg reduced overall craving significantly in participants with early-onset AUD, while a lower dose (1 μg/kg) reduced craving in participants with late-onset AUD. These reductions in craving were also associated with reduced drinking (i.e., drinks per drinking day) and an increased percentage of abstinent days [174].
Ondansetron is well tolerated with relatively mild side effects including diarrhea, constipation, and headache [175]. Overall, these studies suggest a potential role for ondansetron as an AUD treatment, especially in participants with early-onset AUD and possibly in combination with naltrexone.

2.2.6 Mifepristone

Mifepristone (RU-486), a glucocorticoid receptor antagonist, works on the stress system by regulating the amygdala [176]. Preclinically, both systemic and central amygdala injections of mifepristone were shown to suppress yohimbine stress-induced reinstatement of alcohol seeking, indicating that the central amygdala plays an important role in mifepristone’s effects on ethanol-seeking [177, 178]. Recent preclinical research in primates has shown mixed results, such that mifepristone was shown to decrease chronic voluntary alcohol consumption in rhesus macaques at doses of 30 and 56 mg/kg per day [179], but had no effect on alcohol-seeking or self-administration in baboons at slightly lower doses of 10–30 mg/kg per day [180]. Of note, in the former study, cessation of mifepristone treatment resulted in a rapid return to baseline intake levels, and mifepristone was not effective in preventing a relapse during early abstinence.
In a human laboratory study with 56 alcohol-dependent participants, mifepristone (taken for one week) was effective in reducing alcohol craving and consumption relative to placebo, improved liver-function markers, and was overall well tolerated [178]. A two-week Phase 4 RCT examining the effects of mifepristone on cognition in AUD was recently conducted, but recruitment challenges rendered the results inconclusive [181, 182]. Of note, in this trial, participants who received mifepristone had higher Beck Depression Inventory scores compared to placebo at 4 weeks post-randomization despite similar scores at baseline between the two groups, indicating a greater severity of depression symptoms caused by the medication. Additional clinical research on mifepristone as a treatment for AUD and its potential side effects is warranted.

2.2.7 Ibudilast

Ibudilast is an inhibitor of phosphodiesterases (PDE)-3, -4, -10, and -11 [183] and macrophage migration inhibitory factor (MIF) [184]. Ibudilast has been shown to dose-dependently suppress pro-inflammatory cytokines, such as interleukins IL-1β, IL-6, and tumor necrosis factor alpha (TNF-α), and to increase the anti-inflammatory cytokine IL-10 and neurotrophic factors [185]. As increases in inflammation are seen in AUD [186], the effects of ibudilast in treating AUD are thought to be driven by its anti-inflammatory and pro-neurotrophic qualities [187].
Preclinically, ibudilast was demonstrated to reduce alcohol intake in two rat models, and decreased drinking selectively in alcohol-dependent mice in comparison to non-dependent mice [188]. These preclinical findings align with prior rodent studies in which pharmacological inhibition of PDE also reduced alcohol intake [189191].
In a 7-day human laboratory crossover trial (N = 24), ibudilast was well tolerated and decreased tonic craving in comparison to placebo. Additionally, ibudilast improved mood during exposure to alcohol and stress cues, and reduced the mood-altering and stimulant effects of alcohol among participants with more severe depressive symptoms [192]. Another recent 2-week RCT enrolling 52 participants found that ibudilast also significantly decreased the odds of heavy drinking during the trial by 45% compared to placebo and attenuated neural response to alcohol cues in the ventral striatum [193, 194].
Ibudilast appears to be well tolerated [195], with common adverse side effects including gastrointestinal symptoms (nausea, vomiting, abdominal pain, and diarrhea), headaches, and depression [192, 196]. In the aforementioned 2-week RCT, no significant differences in side effects were seen between groups [193]. In summary, early findings from clinical studies of ibudilast for AUD treatment appear promising and warrant further clinical investigation.

2.2.8 Prazosin and Doxazosin

Prazosin and doxazosin are alpha-1 adrenergic receptor antagonists with similar chemical structures that can readily cross the blood-brain barrier and block noradrenergic excitation of the mesolimbic dopaminergic system [197, 198]. Both medications are used for the treatment of hypertension and benign prostatic hyperplasia. While these medications show good safety and tolerability, doxazosin appears to have a better clinical profile, such as improved absorption profile and a longer half-life, leading to less frequent dosing. Adrenergic receptors regulate sympathetic nervous system activity through activation of the neurotransmitter norepinephrine [199]. Stress physiology is disrupted with chronic alcohol use, particularly during early alcohol abstinence. In early abstinence, individuals with AUD experience more emotional dysregulation, stress, and alcohol cravings, all of which can increase the risk of relapse. Thus, alpha-1 blockers like prazosin and doxazosin may help to normalize these stress system changes seen in AUD [200].
Preclinical work found that prazosin reduced ethanol-related operant responding in acute withdrawal for dependent, but not non-dependent rodents [201]. In addition, prazosin treatment prevented yohimbine stress-induced reinstatement of ethanol seeking [202] and attenuated ethanol intake during relapse in alcohol-preferring rats [203]. Doxazosin decreased voluntary alcohol intake in alcohol-preferring rats in a dose-dependent manner, and this effect was likely not due to general motor impairment [204]. Further, doxazosin significantly reduced voluntary ethanol intake in a rodent model of AUD and stress exposure [205].
In humans, an early 6-week pilot study with 24 randomized participants with AUD revealed that prazosin treatment was associated with fewer drinking days per week and fewer drinks per week than placebo [206]. More recently, 92 participants with AUD completed a 12-week double-blind study with prazosin and medication management [207]. Results from intent-to-treat analyses showed that prazosin participants had greater reductions in heavy drinking and rates of drinking over time, although these effects were modest. A 10-week RCT comprising 41 individuals with AUD showed no significant effect of doxazosin over placebo on quantity of alcohol consumption (i.e., drinks per week, heavy-drinking days) [208]. However, further examination of clinical moderators revealed that doxazosin significantly reduced drinks per week and heavy-drinking days among individuals with higher family history of AUD and higher standing diastolic blood pressure [209]. Similarly, moderator analyses from a 12-week RCT of prazosin (N = 100) found that one’s degree of alcohol withdrawal symptoms predicted clinical response, such that participants with high levels of withdrawal symptoms benefited the most from treatment (e.g., reduced craving and heavy-drinking days; improved mood symptoms) [210]. A small meta-analysis of 6 studies with a total of 319 participants tested the effectiveness of drugs acting on adrenergic receptors for AUD and found a significant treatment effect of prazosin and doxazosin on alcohol consumption but not abstinence [211]. In summary, prazosin and doxazosin show some early promise as a treatment for AUD and may be particularly beneficial as a harm-reduction approach and for individuals with significant alcohol withdrawal symptoms or family history of AUD as well as comorbid post-traumatic stress disorder (PTSD).

2.2.9 N-Acetylcysteine

Alcohol use has been shown to alter the glutamate system. Chronic and binge drinking inhibit glutamate levels and transmission through blockade of NMDA receptors, subsequently leading to elevated glutamate levels during alcohol withdrawal [212214]. N-Acetylcysteine (NAC) is a cysteine prodrug that works to restore glutamatergic tone in reward circuitry by improving the expression and function of the cysteine-glutamate exchanger and normalizing glial glutamate transporters [215, 216].
Preclinically, NAC has been shown to reduce withdrawal effects [217], block the development of behavioral sensitization to alcohol [218], and attenuate biological adaptations induced by alcohol cessation (i.e., blocked the reduction of transcription factor ΔFosB in the nucleus accumbens; reduced corticosterone and leptin levels) [218220]. Additionally, NAC reduced ethanol-seeking and self-administration in rodents [215, 216]. NAC and aspirin co-administration also reduced ethanol intake and relapse binge drinking in ethanol-preferring rats [221].
In a recent meta-analysis of seven RCTs with a total of 245 participants, NAC compared to placebo was shown to reduce craving symptoms across a number of substance use disorders [222]. A secondary analysis of a 12-week, multisite RCT of NAC to treat cannabis use disorder in 277 participants found that NAC increased odds of between-visit abstinence, and reduced alcohol consumption (i.e., drinks per week and drinking days per week) by 30% [223]. However, a recent 5-day human laboratory study (N = 9) found that NAC did not attenuate alcohol self-administration [212]. Additional clinical trials will also examine the effectiveness of NAC in adolescent and adult samples of AUD ([224], NCT03707951). These trials include samples with comorbid psychopathology (i.e., PTSD) and will use neuroimaging methods to examine the neural circuitry underlying NAC’s modulation of relevant metabolites and neural reactivity to alcohol cues.
Orally-administered NAC appears to be well tolerated, with the most common adverse effects being nausea and diarrhea [212, 222]. Of note, NAC is currently being tested in adolescent AUD, a unique prospect as no other AUD pharmacotherapies are yet approved for adolescents [225]. In summary, NAC represents a promising potential treatment for AUD and merits further exploration.

2.2.10 Suvorexant

Suvorexant is a dual orexin antagonist that is used for the treatment of insomnia. The orexin/hypocretin system is well known for its role in sleep-wake regulation but has more recently been implicated in AUD [226]. Orexins are neuropeptides that are densely localized in the lateral hypothalamus. Orexins A and B bind to the G-protein coupled orexin-1 and orexin-2 receptors. Orexin A has equal affinity for both receptors, whereas orexin B has selectivity for orexin-2 receptors. The dense orexin projections from the lateral hypothalamus to the ventral tegmental area provide neurobiological support that orexins may influence responses to rewarding stimuli, including alcohol [227, 228].
Animal models demonstrate that orexin-1 receptor antagonists reduce alcohol drinking in alcohol-dependent mice and alcohol-preferring rats [229233]. Orexin-2 antagonists also reduce alcohol drinking and reinstatement/relapse in mice and rats [234236]. Similarly, dual orexin antagonists reduce alcohol consumption in alcohol-preferring rats [237, 238]. Given the effectiveness of orexin antagonists in reducing alcohol drinking at the preclinical level of analysis, suvorexant has garnered interest as a drug that can be repurposed to treat AUD [239]. While no animal or human laboratory study has directly examined the efficacy of suvorexant on alcohol-related behaviors, two ongoing Phase 2 RCTs will assess suvorexant’s potential as a treatment for AUD (NCT04229095) and comorbid AUD + insomnia (NCT03897062). The sedative effects of suvorexant are of notable concern, particularly if individuals engage in alcohol drinking during treatment [240]. Thus, the level of patient monitoring throughout the treatment phase and finding the optimal time of dosing to negate the additive sedative effects are important factors to consider to fully evaluate its therapeutic potential.

2.3 Novel Agents

The following sections discuss novel agents that have not yet been approved for any use. These medications are early in development and the majority do not yet have available clinical findings on their application toward the treatment of AUD.

2.3.1 ABT-436

ABT‐436 is an orally active, highly selective vasopressin type 1B (V1B) receptor antagonist. V1B antagonists attenuate basal hypothalamic-pituitary-adrenal (HPA) axis activity and have shown favorable effects in rat models of alcohol dependence, including attenuating reinstatement of alcohol self-administration [241], and diminishing alcohol intake by alcohol-preferring and alcohol-dependent rats [241, 242].
ABT-436 has been shown to attenuate basal HPA axis activity in humans [243]. A 12-week RCT enrolling 150 participants found ABT-436 to be associated with an increased percentage of days of abstinence compared to placebo, as well as significantly reduced cigarette use [244]. However, no differences were found on heavy-drinking days or alcohol craving. A subgroup analysis also showed that ABT-436 appeared to have greater efficacy among participants with high baseline stress levels. This study was the first Phase 2 clinical trial that tested a V1B receptor antagonist for AUD.
The finding that ABT-436 reduced both alcohol drinking and smoking indicate a potential use for V1B antagonists to treat co-use of alcohol and nicotine. ABT-436 is generally well tolerated in humans, with the most common side effect being diarrhea. Furthermore, results indicate that patients with high levels of stress may specifically benefit from medications targeting the vasopressin receptor.

2.3.2 GET73

N-[(4-Trifluoromethyl) benzyl] 4-methoxybutyramide (GET73) is a GHB analogue that has shown promising in vitro and in vivo preclinical results as a potential agent for the treatment of AUD. The addition of GET73 to cultures of rat hippocampal neurons rescued negative ethanol-induced effects, reductions in cell viability, and increases in reactive oxygen species production, providing evidence for a neuroprotective role of GET73 as an AUD treatment [245]. In vivo, GET73 treatment at low, non-sedative doses (5–50 mg/kg) reduced alcohol intake and suppressed relapse in alcohol-preferring rats, as well as exerting anxiolytic effects [246]. These effects are similar to those seen with GHB administration [247]; however, GET73 was shown not to bind to either high- or low-affinity GHB binding sites in rat cortical membranes [246]. Recent studies indicate that GET73 may act as a negative allosteric modulator at the metabotropic glutamate subtype 5 receptor (mGluR5) [248]; however, the complete mechanism of action of GET73 remains unclear.
A translational study examining GET73-alcohol interactions found that neither 30 nor 100 mg/kg GET73 administered in rats (equivalent to doses employed in humans) potentiated alcohol-induced intoxication. Additionally, GET73 administered both in the presence and absence of alcohol was well tolerated in two samples of 14 and 11 participants, with no severe adverse events and no difference in adverse events [249]. A Phase I clinical trial in two samples of 48 and 32 participants found that both single doses (up to 600 mg) and repeated ascending doses (up to 450 mg twice /day) of GET-73 were safe and well tolerated [250]. Another inpatient human laboratory study conducted by the same group confirmed the safety and tolerability of GET73 such that no serious or severe adverse events occurred when GET73 was co-administered with alcohol. Co-administration also did not affect the pharmacokinetics of either GET73 or alcohol. However, GET73 also had no effect on alcohol cue-induced craving or self-administration, warranting additional research [251]. A clinical trial of the effects of GET73 on magnetic resonance spectroscopy (MRS) measures of glutamate and GABA levels in individuals with AUD was recently completed (NCT03418623); however, the results have not yet been posted, and another trial, which includes a free-drinking bar lab component, is ongoing (NCT04831684).

2.3.3 ASP8062

In comparison to the drawbacks presented by GABAB agonists like baclofen and SMO, positive allosteric modulators (PAMs) at this receptor present a potential alternative [252]. By binding to a different, non-competitive allosteric site on the GABAB receptor, PAMs allow endogenous GABA, binding at its original orthosteric site, to retain its potency and efficacy, reducing the risk of tolerance development and side effects [253, 254]. A number of PAMs have been studied as potential pharmacotherapies for AUD, and have demonstrated reductions in alcohol-associated behaviors and ethanol self-administration in preclinical models [252, 255258]. When directly contrasted against baclofen, a GABAB PAM had a better profile, with dose-dependent reduction of relapse-like alcohol drinking and with no signs of sedation [257].
One such novel GABAB PAM, ASP8062, appears particularly promising as it has been shown to significantly increase the affinity and efficacy of endogenous GABA binding in human and rat GABAB receptors in vitro and with oral administration in an in vivo rodent model of fibromyalgia, demonstrating the ability of oral formulated ASP8062 to cross the blood-brain-barrier [259]. ASP8062 has recently progressed to clinical development. Two Phase I clinical trials with a combined total of 112 participants evaluated single ascending doses and multiple ascending doses of ASP8062, respectively [260]. These studies found that ASP8062 was well tolerated in humans, with no evidence of drug effects on safety, cognition, drug withdrawal, or suicidal ideation. One additional clinical trial, assessing the safety and efficacy of ASP8062 for alcohol use disorder (NCT05096117), is currently underway. Overall, ASP8062, and GABAB PAMs in general, appear to be well tolerated in humans and decrease alcohol self-administration in animals. These agents present a potential pathway to better utilize the GABAergic system and reduce the side effects seen with GABAB agonists.

2.3.4 PF-5190457

Ghrelin, a peptide produced by endocrine cells primarily in the stomach [261], is thought to regulate growth hormone secretion, food intake, and glucose homeostasis [262]. Ghrelin is also thought to play a role in AUD. Ghrelin signaling is required for stimulation of the reward system by alcohol, and higher ghrelin levels are associated with higher self-reported measures of alcohol craving [263]. In human laboratory studies, intravenous ghrelin administration has been shown to increase the urge to drink, increase alcohol self-administration, and modulate brain activity in regions involved in reward processing and stress regulation [264, 265].
Preclinical studies with ghrelin receptor (GHS-R1a) antagonists have shown reductions in alcohol conditioned place preference, alcohol intake and preference, and alcohol-elicited nucleus accumbens dopamine release in rodents [266270].
PF-5190457 is a ghrelin receptor inverse agonist that inhibits GHS-R1a constitutive activity as well as blocking its activation by ghrelin [271]. In a preliminary clinical study in 12 heavy-drinking individuals, PF-5190457, compared to placebo, reduced alcohol craving and cue-reactivity to alcohol. Additionally, when administered in combination with alcohol, PF-5190457 was safe and well-tolerated with no drug-alcohol interactions [272]. This was the first clinical study of a GHS-R1a inverse agonist in a sample of heavy alcohol drinkers.
PF-5190457 may increase somnolence, heart rate, and lower blood glucose concentrations [273], although clinical results indicate that these side effects were not exacerbated by alcohol co-administration and in general PF-5190457 is well tolerated. In summary, preclinical and early clinical evidence support additional research toward investigating PF-5190457 as a pharmacological approach to treat AUD.

2.3.5 Cannabidiol

Cannabidiol (CBD), one of the main compounds found in Cannabis sativa, has shown promise as a novel therapeutic to treat AUD. CBD is nonintoxicating and has diverse pharmacological effects throughout the central nervous system, including functioning as a negative allosteric modulator of CB1 and CB2 receptors [274, 275], and blocking anandamide update and inhibiting enzymatic hydrolysis [276]. CBD may also interact with non-endocannabinoid signaling systems, including the serotonergic system [277] and the opioidergic system [278], among others.
Preclinical studies have shown that CBD reduces alcohol administration, decreases motivation for alcohol, reduces relapse-like behavior, and improves withdrawal symptoms in animals exposed to chronic alcohol [279]. Evidence in healthy individuals demonstrates that CBD is well tolerated, does not interact with the subjective effects of alcohol, and has no abuse liability [280]. Two recent studies investigated signals for potential efficacy of CBD as a treatment for heroin use disorder [281] and cannabis use disorder [282]. Regarding heroin use disorder, acute CBD reduced cue-induced craving for heroin and reduced anxiety in a sample of 42 abstinent individuals, which was maintained one-week following the last CBD exposure. The cannabis use disorder study found that CBD was more efficacious at reducing cannabis use than placebo in a sample of 48 subjects. In both clinical samples, CBD was well tolerated and not associated with serious adverse events. CBD is currently being evaluated as a potential treatment for AUD, AUD with comorbid PTSD, and alcohol withdrawal in AUD in three clinical trials (NCT03252756, NCT03248167, NCT04205682). In brief, preclinical evidence and clinical evidence in other substance use disorders indicate the promise of CBD as a novel therapeutic for AUD.

3 Discussion

This qualitative literature review discusses the efficacy, mechanism of action, and tolerability of approved, repurposed, and novel pharmacotherapies for the treatment of AUD. This information is summarized in Table 1. As of 2018, the APA recommends acamprosate and naltrexone for the treatment of AUD and suggests gabapentin and topiramate for patients with the goal of reducing alcohol consumption or achieving abstinence, while disulfiram is suggested for achieving and maintaining abstinence only [107]. Similarly, while not included in the APA’s recommendations, aripiprazole and mifepristone are associated with drinking reduction, while baclofen shows association with abstinence and mixed results with drinking reduction. Additional repurposed medications show clinical effectiveness for the treatment of AUD. Some of these appear to have particular promise in specific cases, such as varenicline’s use for nicotine and alcohol co-users [147], baclofen for individuals with liver disease [93], and aripiprazole for more impulsive individuals [156]. Novel agents such as GET73 [245] and ASP8062 [260] have also reduced alcohol intake in preclinical studies. In summary, while currently approved medications are somewhat effective, there remains a crucial need to develop new and improved pharmacotherapies for AUD. Novel and repurposed agents show significant promise as treatments that may improve upon currently approved pharmacotherapies.
Medication development has been identified as a critical priority for AUD research [283]. While considerable progress has been made in this field, there are a number of areas which require our attention to realize the benefit of AUD pharmacotherapy. First, despite the prevalence of AUD, the rate of seeking treatment for AUD remains very low [284]. In order for anyone to benefit from the advances in medication development reviewed herein, the treatment gap must be closed. This will require engagement at multiple levels, from prevention to public education about AUD and the available treatments. Researchers and clinicians can help in these efforts by reducing stigma surrounding AUD and other substance use disorders by choosing appropriate language to describe these disorders and the people who are affected by them [285].
A related issue is the need to improve access to FDA-approved pharmacotherapies for AUD. A recent analysis of the 2019 National Survey on Drug Use and Health found that only 1.6% of people with a past-year AUD received an evidence-based medication to treat their AUD [14]. Medication use was associated with living in a large metropolitan area, use of the emergency department, and receiving mental health care, indicating that these services and residing in an urban environment appear to increase access to evidence-based medications. There is also a great need to improve the education of physicians and clinicians on the availability of evidence-based treatments for AUD. Ongoing efforts in this area are underway by the American Society of Addiction Medicine and the American Academy of Addiction Psychiatry, as well as by the National Institute on Alcohol Abuse and Alcoholism.
To further improve access to treatments and increase treatment-seeking, there is a need to increase the menu of approved pharmacological treatments towards AUD, especially those that have shown promise internationally. Currently, the FDA only accepts two primary outcomes for Phase 3 trials: abstinence and no heavy-drinking days. These outcomes do not always mirror the goals of patients with AUD for their recovery, which may be better reflected by a harm reduction endpoint. Recent work has found that harm reduction endpoints, including reductions in WHO-based drinking levels [286], are associated with improvements in physical health and quality of life [287] and can be used as efficacy outcomes in clinical trials [288]. The acceptance of these outcomes as clinical trial endpoints could have a substantial impact on the medication development field and ultimately result in a larger pharmacotherapy toolbox for clinicians.
Another area of development is the move towards personalized treatment, also referred to as precision medication. AUD is a highly heterogenous disorder, and it unlikely that any medication will work for all individuals with an AUD. As such, there have been efforts to use precision medicine approaches to tailor pharmacotherapies to individuals with different presentations of AUD. Studies have taken several approaches towards personalized treatments, such as pharmacogenetics, sex differences, family history, severity of alcohol withdrawal, drinking phenotypes, and biobehavioral markers [283, 289293]. However, even these efforts may be overly simplistic given the complex nature of AUD. It is likely that personalized treatment approaches will need to account for multiple factors to truly tailor treatments to individual patients [294]. Conversely, the public health significance of the improved efficacy of AUD pharmacotherapy with clinically accessible phenotypes argue for wider dissemination and implementation of precision treatment recommendations identified to date.
A final issue to consider is the need to develop treatments for individuals with AUD and comorbid psychiatric disorders and for individuals with AUD and AALD. AUD often co-occurs with other psychiatric disorders, including other substance use disorders, personality disorders, major depressive disorder, anxiety disorders, and PTSD [295]. The development of integrated treatments, including combined behavioral and pharmacological interventions, which simultaneously address AUD and other co-occurring disorders, is difficult due to the complexity of treating multiple disorders and the limited understanding of the underlying mechanisms. However, this is a necessary area of research given the high rates of comorbidity in the AUD population. Treatment options for individuals with AALD are limited; of the FDA-approved medications, only acamprosate can be used without concerns of hepatotoxicity [296]. Of the non-FDA medications that may prove useful in this population, only baclofen has been evaluated in an RCT [94]. There is a clear need to develop additional treatments for this population.

4 Conclusions

In conclusion, the three widely approved medications for AUD (i.e., disulfiram, acamprosate, and naltrexone) are both modestly effective and underutilized. The heterogeneity inherent in AUD also presents the issue that no one medication is likely to be effective for all individuals with AUD. Therefore, it is necessary to develop more effective, novel, and diverse pharmacotherapy options and to improve access to treatment. The repurposed and novel medications reviewed in this article demonstrate promise for the future of AUD treatment. Additionally, the future directions indicated above offer potential improvements toward the advancement of precision medicine and personalized treatment for the heterogeneous AUD population.

Declarations

Funding

This work was supported in part by the National Institute on Alcohol Abuse and Alcoholism (K24AA025704 to LAR; F31AA028976 to EMB; F32AA029288 to SJN).

Conflict of interest

The authors declare they have no conflict of interest.

Author contributions

This article was conceptualized by LAR, ENG, and EMB. Literature review and original draft preparation was conducted by EMB, ENG, SJN, and LRM. All authors reviewed and commented on previous versions of the manuscript, and all authors read and approved the final manuscript. All authors agree to be accountable for the work presented in the manuscript.

Ethics approval

Not applicable.
Not applicable.
Not applicable.

Code availability

Not applicable.

Availability of data and material

Not applicable.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

e.Dent – Das Online-Abo der Zahnmedizin

Online-Abonnement

Mit e.Dent erhalten Sie Zugang zu allen zahnmedizinischen Fortbildungen und unseren zahnmedizinischen und ausgesuchten medizinischen Zeitschriften.

Literatur
2.
Zurück zum Zitat Grant BF, Goldstein RB, Saha TD, Chou SP, Jung J, Zhang H, et al. Epidemiology of DSM-5 alcohol use disorder: results from the national epidemiologic survey on alcohol and related conditions III. JAMA Psychiat. 2015;72(8):757–66. Grant BF, Goldstein RB, Saha TD, Chou SP, Jung J, Zhang H, et al. Epidemiology of DSM-5 alcohol use disorder: results from the national epidemiologic survey on alcohol and related conditions III. JAMA Psychiat. 2015;72(8):757–66.
3.
Zurück zum Zitat Degenhardt L, Charlson F, Ferrari A, Santomauro D, Erskine H, Mantilla-Herrara A, et al. The global burden of disease attributable to alcohol and drug use in 195 countries and territories, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Psychiatry. 2018;5(12):987–1012. Degenhardt L, Charlson F, Ferrari A, Santomauro D, Erskine H, Mantilla-Herrara A, et al. The global burden of disease attributable to alcohol and drug use in 195 countries and territories, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Psychiatry. 2018;5(12):987–1012.
4.
Zurück zum Zitat World Health Organization. Global status report on alcohol and health 2014. World Health Organization; 2014. World Health Organization. Global status report on alcohol and health 2014. World Health Organization; 2014.
5.
Zurück zum Zitat Stahre M, Roeber J, Kanny D, Brewer RD, Zhang X. Contribution of excessive alcohol consumption to deaths and years of potential life lost in the United States. Prev Chronic Dis. 2014;26(11):E109. Stahre M, Roeber J, Kanny D, Brewer RD, Zhang X. Contribution of excessive alcohol consumption to deaths and years of potential life lost in the United States. Prev Chronic Dis. 2014;26(11):E109.
6.
Zurück zum Zitat Sacks JJ, Gonzales KR, Bouchery EE, Tomedi LE, Brewer RD. 2010 national and state costs of excessive alcohol consumption. Am J Prev Med. 2015;49(5):e73–9.PubMed Sacks JJ, Gonzales KR, Bouchery EE, Tomedi LE, Brewer RD. 2010 national and state costs of excessive alcohol consumption. Am J Prev Med. 2015;49(5):e73–9.PubMed
7.
Zurück zum Zitat Hasin DS, Stinson FS, Ogburn E, Grant BF. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States: results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2007;64(7):830–42.PubMed Hasin DS, Stinson FS, Ogburn E, Grant BF. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States: results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch Gen Psychiatry. 2007;64(7):830–42.PubMed
8.
Zurück zum Zitat Rehm J, Shield KD, Gmel G, Rehm MX, Frick U. Modeling the impact of alcohol dependence on mortality burden and the effect of available treatment interventions in the European Union. Eur Neuropsychopharmacol. 2013;23(2):89–97.PubMed Rehm J, Shield KD, Gmel G, Rehm MX, Frick U. Modeling the impact of alcohol dependence on mortality burden and the effect of available treatment interventions in the European Union. Eur Neuropsychopharmacol. 2013;23(2):89–97.PubMed
9.
Zurück zum Zitat Petrakis IL. A rational approach to the pharmacotherapy of alcohol dependence. J Clin Psychopharmacol. 2006;26(Suppl 1):S3-12.PubMed Petrakis IL. A rational approach to the pharmacotherapy of alcohol dependence. J Clin Psychopharmacol. 2006;26(Suppl 1):S3-12.PubMed
10.
Zurück zum Zitat Pettinati HM, Rabinowitz AR. Choosing the right medication for the treatment of alcoholism. Curr Psychiatry Rep. 2006;8(5):383–8.PubMed Pettinati HM, Rabinowitz AR. Choosing the right medication for the treatment of alcoholism. Curr Psychiatry Rep. 2006;8(5):383–8.PubMed
11.
Zurück zum Zitat Zindel LR, Kranzler HR. Pharmacotherapy of alcohol use disorders: seventy-five years of progress. J Stud Alcohol Drugs Suppl. 2014;75(Suppl 17):79–88.PubMedPubMedCentral Zindel LR, Kranzler HR. Pharmacotherapy of alcohol use disorders: seventy-five years of progress. J Stud Alcohol Drugs Suppl. 2014;75(Suppl 17):79–88.PubMedPubMedCentral
12.
Zurück zum Zitat Wallhed Finn S, Lundin A, Sjöqvist H, Danielsson A-K. Pharmacotherapy for alcohol use disorders—unequal provision across sociodemographic factors and co-morbid conditions. A cohort study of the total population in Sweden. Drug Alcohol Depend. 2021;227:108964.PubMed Wallhed Finn S, Lundin A, Sjöqvist H, Danielsson A-K. Pharmacotherapy for alcohol use disorders—unequal provision across sociodemographic factors and co-morbid conditions. A cohort study of the total population in Sweden. Drug Alcohol Depend. 2021;227:108964.PubMed
13.
Zurück zum Zitat Ray LA, Bujarski S, Grodin E, Hartwell E, Green R, Venegas A, et al. State-of-the-art behavioral and pharmacological treatments for alcohol use disorder. Am J Drug Alcohol Abuse. 2019;45(2):124–40. Ray LA, Bujarski S, Grodin E, Hartwell E, Green R, Venegas A, et al. State-of-the-art behavioral and pharmacological treatments for alcohol use disorder. Am J Drug Alcohol Abuse. 2019;45(2):124–40.
15.
Zurück zum Zitat Fan AZ, Chou SP, Zhang H, Jung J, Grant BF. Prevalence and correlates of past-year recovery from DSM-5 alcohol use disorder: results from national epidemiologic survey on alcohol and related conditions-III. Alcohol Clin Exp Res. 2019;43(11):2406–20.PubMed Fan AZ, Chou SP, Zhang H, Jung J, Grant BF. Prevalence and correlates of past-year recovery from DSM-5 alcohol use disorder: results from national epidemiologic survey on alcohol and related conditions-III. Alcohol Clin Exp Res. 2019;43(11):2406–20.PubMed
16.
Zurück zum Zitat Henssler J, Müller M, Carreira H, Bschor T, Heinz A, Baethge C. Controlled drinking—non-abstinent versus abstinent treatment goals in alcohol use disorder: a systematic review, meta-analysis and meta-regression. Addiction. 2021;116(8):1973–87.PubMed Henssler J, Müller M, Carreira H, Bschor T, Heinz A, Baethge C. Controlled drinking—non-abstinent versus abstinent treatment goals in alcohol use disorder: a systematic review, meta-analysis and meta-regression. Addiction. 2021;116(8):1973–87.PubMed
17.
Zurück zum Zitat Fentress TSP, Wald S, Brah A, Leemon G, Reyes R, Alkhamees F, et al. Dual study describing patient-driven harm reduction goal-setting among people experiencing homelessness and alcohol use disorder. Exp Clin Psychopharmacol. 2021;29(3):261–71.PubMed Fentress TSP, Wald S, Brah A, Leemon G, Reyes R, Alkhamees F, et al. Dual study describing patient-driven harm reduction goal-setting among people experiencing homelessness and alcohol use disorder. Exp Clin Psychopharmacol. 2021;29(3):261–71.PubMed
18.
Zurück zum Zitat Charlet K, Heinz A. Harm reduction-a systematic review on effects of alcohol reduction on physical and mental symptoms. Addict Biol. 2017;22(5):1119–59.PubMed Charlet K, Heinz A. Harm reduction-a systematic review on effects of alcohol reduction on physical and mental symptoms. Addict Biol. 2017;22(5):1119–59.PubMed
19.
Zurück zum Zitat Maremmani I, Cibin M, Pani PP, Rossi A, Turchetti G. Harm Reduction as “Continuum Care” in alcohol abuse disorder. Int J Environ Res Public Health. 2015;12(11):14828–41.PubMedPubMedCentral Maremmani I, Cibin M, Pani PP, Rossi A, Turchetti G. Harm Reduction as “Continuum Care” in alcohol abuse disorder. Int J Environ Res Public Health. 2015;12(11):14828–41.PubMedPubMedCentral
20.
Zurück zum Zitat Witkiewitz K, Morris J, Tucker JA. Commentary on Henssler et al.: the public health case for promoting and valuing drinking reductions in the treatment of alcohol use disorder. Addiction. 2021;116(8):1988–9.PubMed Witkiewitz K, Morris J, Tucker JA. Commentary on Henssler et al.: the public health case for promoting and valuing drinking reductions in the treatment of alcohol use disorder. Addiction. 2021;116(8):1988–9.PubMed
21.
Zurück zum Zitat Witkiewitz K, Wilson AD, Roos CR, Swan JE, Votaw VR, Stein ER, et al. Can individuals with alcohol use disorder sustain non-abstinent recovery? Non-abstinent outcomes 10 years after alcohol use disorder treatment. J Addict Med. 2021;15(4):303–10.PubMedPubMedCentral Witkiewitz K, Wilson AD, Roos CR, Swan JE, Votaw VR, Stein ER, et al. Can individuals with alcohol use disorder sustain non-abstinent recovery? Non-abstinent outcomes 10 years after alcohol use disorder treatment. J Addict Med. 2021;15(4):303–10.PubMedPubMedCentral
22.
23.
Zurück zum Zitat Vallari RC, Pietruszko R. Human aldehyde dehydrogenase: mechanism of inhibition of disulfiram. Science. 1982;216(4546):637–9.PubMed Vallari RC, Pietruszko R. Human aldehyde dehydrogenase: mechanism of inhibition of disulfiram. Science. 1982;216(4546):637–9.PubMed
24.
Zurück zum Zitat Fuller RK, Branchey L, Brightwell DR, Derman RM, Emrick CD, Iber FL, et al. Disulfiram treatment of alcoholism. A veterans administration cooperative study. JAMA. 1986;256(11):1449–55.PubMed Fuller RK, Branchey L, Brightwell DR, Derman RM, Emrick CD, Iber FL, et al. Disulfiram treatment of alcoholism. A veterans administration cooperative study. JAMA. 1986;256(11):1449–55.PubMed
25.
Zurück zum Zitat Garbutt JC, West SL, Carey TS, Lohr KN, Crews FT. Pharmacological treatment of alcohol dependence: a review of the evidence. JAMA. 1999;281(14):1318–25.PubMed Garbutt JC, West SL, Carey TS, Lohr KN, Crews FT. Pharmacological treatment of alcohol dependence: a review of the evidence. JAMA. 1999;281(14):1318–25.PubMed
26.
Zurück zum Zitat Kosten TR, Wu G, Huang W, Harding MJ, Hamon SC, Lappalainen J, et al. Pharmacogenetic randomized trial for cocaine abuse: disulfiram and dopamine β-hydroxylase. Biol Psychiatry. 2013;73(3):219–24.PubMed Kosten TR, Wu G, Huang W, Harding MJ, Hamon SC, Lappalainen J, et al. Pharmacogenetic randomized trial for cocaine abuse: disulfiram and dopamine β-hydroxylase. Biol Psychiatry. 2013;73(3):219–24.PubMed
27.
Zurück zum Zitat Devoto P, Flore G, Saba P, Cadeddu R, Gessa GL. Disulfiram stimulates dopamine release from noradrenergic terminals and potentiates cocaine-induced dopamine release in the prefrontal cortex. Psychopharmacology. 2012;219(4):1153–64.PubMed Devoto P, Flore G, Saba P, Cadeddu R, Gessa GL. Disulfiram stimulates dopamine release from noradrenergic terminals and potentiates cocaine-induced dopamine release in the prefrontal cortex. Psychopharmacology. 2012;219(4):1153–64.PubMed
28.
Zurück zum Zitat Bagdy G, Arató M. Serum dopamine-beta-hydroxylase activity and alcohol withdrawal symptoms. Drug Alcohol Depend. 1987;19(1):45–50.PubMed Bagdy G, Arató M. Serum dopamine-beta-hydroxylase activity and alcohol withdrawal symptoms. Drug Alcohol Depend. 1987;19(1):45–50.PubMed
29.
Zurück zum Zitat Skinner MD, Lahmek P, Pham H, Aubin H-J. Disulfiram efficacy in the treatment of alcohol dependence: a meta-analysis. PLoS ONE. 2014;9(2):e87366.PubMedPubMedCentral Skinner MD, Lahmek P, Pham H, Aubin H-J. Disulfiram efficacy in the treatment of alcohol dependence: a meta-analysis. PLoS ONE. 2014;9(2):e87366.PubMedPubMedCentral
30.
Zurück zum Zitat Skinner MD, Coudert M, Berlin I, Passeri E, Michel L, Aubin H-J. Effect of the threat of a disulfiram-ethanol reaction on cue reactivity in alcoholics. Drug Alcohol Depend. 2010;112(3):239–46.PubMed Skinner MD, Coudert M, Berlin I, Passeri E, Michel L, Aubin H-J. Effect of the threat of a disulfiram-ethanol reaction on cue reactivity in alcoholics. Drug Alcohol Depend. 2010;112(3):239–46.PubMed
31.
Zurück zum Zitat Mutschler J, Dirican G, Funke S, Obermann C, Grosshans M, Mann K, et al. Experienced acetaldehyde reaction does not improve treatment response in outpatients treated with supervised disulfiram. Clin Neuropharmacol. 2011;34(4):161–5.PubMed Mutschler J, Dirican G, Funke S, Obermann C, Grosshans M, Mann K, et al. Experienced acetaldehyde reaction does not improve treatment response in outpatients treated with supervised disulfiram. Clin Neuropharmacol. 2011;34(4):161–5.PubMed
32.
Zurück zum Zitat Mutschler J, Grosshans M, Soyka M, Rösner S. Current findings and mechanisms of action of disulfiram in the treatment of alcohol dependence. Pharmacopsychiatry. 2016;49(4):137–41.PubMed Mutschler J, Grosshans M, Soyka M, Rösner S. Current findings and mechanisms of action of disulfiram in the treatment of alcohol dependence. Pharmacopsychiatry. 2016;49(4):137–41.PubMed
33.
Zurück zum Zitat Fuller RK, Gordis E. Does disulfiram have a role in alcoholism treatment today? Addiction. 2004;99(1):21–4.PubMed Fuller RK, Gordis E. Does disulfiram have a role in alcoholism treatment today? Addiction. 2004;99(1):21–4.PubMed
34.
Zurück zum Zitat Kranzler HR, Soyka M. Diagnosis and pharmacotherapy of alcohol use disorder: a review. JAMA. 2018;320(8):815–24.PubMedPubMedCentral Kranzler HR, Soyka M. Diagnosis and pharmacotherapy of alcohol use disorder: a review. JAMA. 2018;320(8):815–24.PubMedPubMedCentral
35.
Zurück zum Zitat Mason BJ, Heyser CJ. Acamprosate: a prototypic neuromodulator in the treatment of alcohol dependence. CNS Neurol Disord Drug Targets. 2010;9(1):23–32.PubMedPubMedCentral Mason BJ, Heyser CJ. Acamprosate: a prototypic neuromodulator in the treatment of alcohol dependence. CNS Neurol Disord Drug Targets. 2010;9(1):23–32.PubMedPubMedCentral
36.
Zurück zum Zitat Jonas DE, Amick HR, Feltner C, Bobashev G, Thomas K, Wines R, et al. Pharmacotherapy for adults with alcohol use disorders in outpatient settings: a systematic review and meta-analysis. JAMA. 2014;311(18):1889–900.PubMed Jonas DE, Amick HR, Feltner C, Bobashev G, Thomas K, Wines R, et al. Pharmacotherapy for adults with alcohol use disorders in outpatient settings: a systematic review and meta-analysis. JAMA. 2014;311(18):1889–900.PubMed
37.
Zurück zum Zitat Paille FM, Guelfi JD, Perkins AC, Royer RJ, Steru L, Parot P. Double-blind randomized multicentre trial of acamprosate in maintaining abstinence from alcohol. Alcohol Alcohol. 1995;30(2):239–47.PubMed Paille FM, Guelfi JD, Perkins AC, Royer RJ, Steru L, Parot P. Double-blind randomized multicentre trial of acamprosate in maintaining abstinence from alcohol. Alcohol Alcohol. 1995;30(2):239–47.PubMed
38.
Zurück zum Zitat Chick J, Howlett H, Morgan MY, Ritson B. United Kingdom Multicentre Acamprosate Study (UKMAS): a 6-month prospective study of acamprosate versus placebo in preventing relapse after withdrawal from alcohol. Alcohol Alcohol. 2000;35(2):176–87.PubMed Chick J, Howlett H, Morgan MY, Ritson B. United Kingdom Multicentre Acamprosate Study (UKMAS): a 6-month prospective study of acamprosate versus placebo in preventing relapse after withdrawal from alcohol. Alcohol Alcohol. 2000;35(2):176–87.PubMed
39.
Zurück zum Zitat Kampman KM, Pettinati HM, Lynch KG, Xie H, Dackis C, Oslin DW, et al. Initiating acamprosate within-detoxification versus post-detoxification in the treatment of alcohol dependence. Addict Behav. 2009;34(6–7):581–6.PubMedPubMedCentral Kampman KM, Pettinati HM, Lynch KG, Xie H, Dackis C, Oslin DW, et al. Initiating acamprosate within-detoxification versus post-detoxification in the treatment of alcohol dependence. Addict Behav. 2009;34(6–7):581–6.PubMedPubMedCentral
40.
Zurück zum Zitat Richardson K, Baillie A, Reid S, Morley K, Teesson M, Sannibale C, et al. Do acamprosate or naltrexone have an effect on daily drinking by reducing craving for alcohol? Addiction. 2008;103(6):953–9.PubMed Richardson K, Baillie A, Reid S, Morley K, Teesson M, Sannibale C, et al. Do acamprosate or naltrexone have an effect on daily drinking by reducing craving for alcohol? Addiction. 2008;103(6):953–9.PubMed
41.
Zurück zum Zitat Mason BJ, Goodman AM, Chabac S, Lehert P. Effect of oral acamprosate on abstinence in patients with alcohol dependence in a double-blind, placebo-controlled trial: the role of patient motivation. J Psychiatr Res. 2006;40(5):383–93.PubMed Mason BJ, Goodman AM, Chabac S, Lehert P. Effect of oral acamprosate on abstinence in patients with alcohol dependence in a double-blind, placebo-controlled trial: the role of patient motivation. J Psychiatr Res. 2006;40(5):383–93.PubMed
42.
Zurück zum Zitat Anton RF, O’Malley SS, Ciraulo DA, Cisler RA, Couper D, Donovan DM, et al. Combined pharmacotherapies and behavioral interventions for alcohol dependence: the COMBINE study: a randomized controlled trial. JAMA. 2006;295(17):2003–17.PubMed Anton RF, O’Malley SS, Ciraulo DA, Cisler RA, Couper D, Donovan DM, et al. Combined pharmacotherapies and behavioral interventions for alcohol dependence: the COMBINE study: a randomized controlled trial. JAMA. 2006;295(17):2003–17.PubMed
43.
Zurück zum Zitat Swift RM, Aston ER. Pharmacotherapy for alcohol use disorder: current and emerging therapies. Harv Rev Psychiatry. 2015;23(2):122–33.PubMedPubMedCentral Swift RM, Aston ER. Pharmacotherapy for alcohol use disorder: current and emerging therapies. Harv Rev Psychiatry. 2015;23(2):122–33.PubMedPubMedCentral
44.
Zurück zum Zitat Rösner S, Hackl-Herrwerth A, Leucht S, Lehert P, Vecchi S, Soyka M. Acamprosate for alcohol dependence. Cochrane Database Syst Rev. 2010;(9):CD004332. Rösner S, Hackl-Herrwerth A, Leucht S, Lehert P, Vecchi S, Soyka M. Acamprosate for alcohol dependence. Cochrane Database Syst Rev. 2010;(9):CD004332.
45.
Zurück zum Zitat Littleton JM. Acamprosate in alcohol dependence: implications of a unique mechanism of action. J Addict Med. 2007;1(3):115–25.PubMed Littleton JM. Acamprosate in alcohol dependence: implications of a unique mechanism of action. J Addict Med. 2007;1(3):115–25.PubMed
46.
Zurück zum Zitat Koob GF, Mason BJ, De Witte P, Littleton J, Siggins GR. Potential neuroprotective effects of acamprosate. Alcohol Clin Exp Res. 2002;26(4):586–92.PubMed Koob GF, Mason BJ, De Witte P, Littleton J, Siggins GR. Potential neuroprotective effects of acamprosate. Alcohol Clin Exp Res. 2002;26(4):586–92.PubMed
47.
Zurück zum Zitat Davidson M, Shanley B, Wilce P. Increased NMDA-induced excitability during ethanol withdrawal: a behavioural and histological study. Brain Res. 1995;674(1):91–6.PubMed Davidson M, Shanley B, Wilce P. Increased NMDA-induced excitability during ethanol withdrawal: a behavioural and histological study. Brain Res. 1995;674(1):91–6.PubMed
48.
Zurück zum Zitat Grant KA, Valverius P, Hudspith M, Tabakoff B. Ethanol withdrawal seizures and the NMDA receptor complex. Eur J Pharmacol. 1990;176(3):289–96.PubMed Grant KA, Valverius P, Hudspith M, Tabakoff B. Ethanol withdrawal seizures and the NMDA receptor complex. Eur J Pharmacol. 1990;176(3):289–96.PubMed
49.
Zurück zum Zitat Rösner S, Leucht S, Lehert P, Soyka M. Acamprosate supports abstinence, naltrexone prevents excessive drinking: evidence from a meta-analysis with unreported outcomes. J Psychopharmacol. 2008;22(1):11–23.PubMed Rösner S, Leucht S, Lehert P, Soyka M. Acamprosate supports abstinence, naltrexone prevents excessive drinking: evidence from a meta-analysis with unreported outcomes. J Psychopharmacol. 2008;22(1):11–23.PubMed
50.
Zurück zum Zitat Mitchell JM, O’Neil JP, Janabi M, Marks SM, Jagust WJ, Fields HL. Alcohol consumption induces endogenous opioid release in the human orbitofrontal cortex and nucleus accumbens. Sci Transl Med. 2012;4(116):116ra6.PubMed Mitchell JM, O’Neil JP, Janabi M, Marks SM, Jagust WJ, Fields HL. Alcohol consumption induces endogenous opioid release in the human orbitofrontal cortex and nucleus accumbens. Sci Transl Med. 2012;4(116):116ra6.PubMed
51.
Zurück zum Zitat Nestler EJ. Is there a common molecular pathway for addiction? Nat Neurosci. 2005;8(11):1445–9.PubMed Nestler EJ. Is there a common molecular pathway for addiction? Nat Neurosci. 2005;8(11):1445–9.PubMed
52.
Zurück zum Zitat O’Malley SS, Jaffe AJ, Chang G, Schottenfeld RS, Meyer RE, Rounsaville B. Naltrexone and coping skills therapy for alcohol dependence. A controlled study. Arch Gen Psychiatry. 1992;49(11):881–7.PubMed O’Malley SS, Jaffe AJ, Chang G, Schottenfeld RS, Meyer RE, Rounsaville B. Naltrexone and coping skills therapy for alcohol dependence. A controlled study. Arch Gen Psychiatry. 1992;49(11):881–7.PubMed
53.
Zurück zum Zitat Volpicelli JR, Alterman AI, Hayashida M, O’Brien CP. Naltrexone in the treatment of alcohol dependence. Arch Gen Psychiatry. 1992;49(11):876–80.PubMed Volpicelli JR, Alterman AI, Hayashida M, O’Brien CP. Naltrexone in the treatment of alcohol dependence. Arch Gen Psychiatry. 1992;49(11):876–80.PubMed
54.
Zurück zum Zitat Drobes DJ, Anton RF, Thomas SE, Voronin K. Effects of naltrexone and nalmefene on subjective response to alcohol among non-treatment-seeking alcoholics and social drinkers. Alcohol Clin Exp Res. 2004;28(9):1362–70.PubMed Drobes DJ, Anton RF, Thomas SE, Voronin K. Effects of naltrexone and nalmefene on subjective response to alcohol among non-treatment-seeking alcoholics and social drinkers. Alcohol Clin Exp Res. 2004;28(9):1362–70.PubMed
55.
Zurück zum Zitat Ray LA, Hutchison KE. Effects of naltrexone on alcohol sensitivity and genetic moderators of medication response: a double-blind placebo-controlled study. Arch Gen Psychiatry. 2007;64(9):1069–77.PubMed Ray LA, Hutchison KE. Effects of naltrexone on alcohol sensitivity and genetic moderators of medication response: a double-blind placebo-controlled study. Arch Gen Psychiatry. 2007;64(9):1069–77.PubMed
56.
Zurück zum Zitat O’Malley SS, Krishnan-Sarin S, Farren C, Sinha R, Kreek MJ. Naltrexone decreases craving and alcohol self-administration in alcohol-dependent subjects and activates the hypothalamo-pituitary-adrenocortical axis. Psychopharmacology. 2002;160(1):19–29.PubMed O’Malley SS, Krishnan-Sarin S, Farren C, Sinha R, Kreek MJ. Naltrexone decreases craving and alcohol self-administration in alcohol-dependent subjects and activates the hypothalamo-pituitary-adrenocortical axis. Psychopharmacology. 2002;160(1):19–29.PubMed
57.
Zurück zum Zitat O’Malley SS, Corbin WR, Leeman RF, DeMartini KS, Fucito LM, Ikomi J, et al. Reduction of alcohol drinking in young adults by naltrexone: a double-blind, placebo-controlled, randomized clinical trial of efficacy and safety. J Clin Psychiatry. 2015;76(2):e207-213.PubMedPubMedCentral O’Malley SS, Corbin WR, Leeman RF, DeMartini KS, Fucito LM, Ikomi J, et al. Reduction of alcohol drinking in young adults by naltrexone: a double-blind, placebo-controlled, randomized clinical trial of efficacy and safety. J Clin Psychiatry. 2015;76(2):e207-213.PubMedPubMedCentral
58.
Zurück zum Zitat Morris PL, Hopwood M, Whelan G, Gardiner J, Drummond E. Naltrexone for alcohol dependence: a randomized controlled trial. Addiction. 2001;96(11):1565–73.PubMed Morris PL, Hopwood M, Whelan G, Gardiner J, Drummond E. Naltrexone for alcohol dependence: a randomized controlled trial. Addiction. 2001;96(11):1565–73.PubMed
59.
Zurück zum Zitat Bouza C, Angeles M, Magro A, Muñoz A, Amate JM. Efficacy and safety of naltrexone and acamprosate in the treatment of alcohol dependence: a systematic review. Addiction. 2004;99(7):811–28.PubMed Bouza C, Angeles M, Magro A, Muñoz A, Amate JM. Efficacy and safety of naltrexone and acamprosate in the treatment of alcohol dependence: a systematic review. Addiction. 2004;99(7):811–28.PubMed
60.
Zurück zum Zitat Johnson BA. Naltrexone long-acting formulation in the treatment of alcohol dependence. Ther Clin Risk Manag. 2007;3(5):741–9.PubMedPubMedCentral Johnson BA. Naltrexone long-acting formulation in the treatment of alcohol dependence. Ther Clin Risk Manag. 2007;3(5):741–9.PubMedPubMedCentral
61.
Zurück zum Zitat Garbutt JC, Kranzler HR, O’Malley SS, Gastfriend DR, Pettinati HM, Silverman BL, et al. Efficacy and tolerability of long-acting injectable naltrexone for alcohol dependence: a randomized controlled trial. JAMA. 2005;293(13):1617–25.PubMed Garbutt JC, Kranzler HR, O’Malley SS, Gastfriend DR, Pettinati HM, Silverman BL, et al. Efficacy and tolerability of long-acting injectable naltrexone for alcohol dependence: a randomized controlled trial. JAMA. 2005;293(13):1617–25.PubMed
62.
Zurück zum Zitat Kranzler HR, Modesto-Lowe V, Van Kirk J. Naltrexone vs. nefazodone for treatment of alcohol dependence. A placebo-controlled trial. Neuropsychopharmacology. 2000;22(5):493–503.PubMed Kranzler HR, Modesto-Lowe V, Van Kirk J. Naltrexone vs. nefazodone for treatment of alcohol dependence. A placebo-controlled trial. Neuropsychopharmacology. 2000;22(5):493–503.PubMed
63.
Zurück zum Zitat Streeton C, Whelan G. Naltrexone, a relapse prevention maintenance treatment of alcohol dependence: a meta-analysis of randomized controlled trials. Alcohol Alcohol. 2001;36(6):544–52.PubMed Streeton C, Whelan G. Naltrexone, a relapse prevention maintenance treatment of alcohol dependence: a meta-analysis of randomized controlled trials. Alcohol Alcohol. 2001;36(6):544–52.PubMed
64.
Zurück zum Zitat Mark TL, Kassed CA, Vandivort-Warren R, Levit KR, Kranzler HR. Alcohol and opioid dependence medications: prescription trends, overall and by physician specialty. Drug Alcohol Depend. 2009;99(1–3):345–9.PubMed Mark TL, Kassed CA, Vandivort-Warren R, Levit KR, Kranzler HR. Alcohol and opioid dependence medications: prescription trends, overall and by physician specialty. Drug Alcohol Depend. 2009;99(1–3):345–9.PubMed
65.
Zurück zum Zitat Busch AC, Denduluri M, Glass J, Hetzel S, Gugnani SP, Gassman M, et al. Predischarge injectable versus oral naltrexone to improve postdischarge treatment engagement among hospitalized veterans with alcohol use disorder: a randomized pilot proof-of-concept study. Alcohol Clin Exp Res. 2017;41(7):1352–60.PubMedPubMedCentral Busch AC, Denduluri M, Glass J, Hetzel S, Gugnani SP, Gassman M, et al. Predischarge injectable versus oral naltrexone to improve postdischarge treatment engagement among hospitalized veterans with alcohol use disorder: a randomized pilot proof-of-concept study. Alcohol Clin Exp Res. 2017;41(7):1352–60.PubMedPubMedCentral
66.
Zurück zum Zitat Rösner S, Hackl-Herrwerth A, Leucht S, Vecchi S, Srisurapanont M, Soyka M. Opioid antagonists for alcohol dependence. Cochrane Database Syst Rev. 2010;(12):CD001867. Rösner S, Hackl-Herrwerth A, Leucht S, Vecchi S, Srisurapanont M, Soyka M. Opioid antagonists for alcohol dependence. Cochrane Database Syst Rev. 2010;(12):CD001867.
68.
Zurück zum Zitat Bart G, Schluger JH, Borg L, Ho A, Bidlack JM, Kreek MJ. Nalmefene induced elevation in serum prolactin in normal human volunteers: partial kappa opioid agonist activity? Neuropsychopharmacology. 2005;30(12):2254–62.PubMed Bart G, Schluger JH, Borg L, Ho A, Bidlack JM, Kreek MJ. Nalmefene induced elevation in serum prolactin in normal human volunteers: partial kappa opioid agonist activity? Neuropsychopharmacology. 2005;30(12):2254–62.PubMed
69.
Zurück zum Zitat Rose JH, Karkhanis AN, Steiniger-Brach B, Jones SR. Distinct effects of nalmefene on dopamine uptake rates and kappa opioid receptor activity in the nucleus accumbens following chronic intermittent ethanol exposure. Int J Mol Sci. 2016;17(8):1216.PubMedCentral Rose JH, Karkhanis AN, Steiniger-Brach B, Jones SR. Distinct effects of nalmefene on dopamine uptake rates and kappa opioid receptor activity in the nucleus accumbens following chronic intermittent ethanol exposure. Int J Mol Sci. 2016;17(8):1216.PubMedCentral
70.
Zurück zum Zitat Nealey KA, Smith AW, Davis SM, Smith DG, Walker BM. κ-opioid receptors are implicated in the increased potency of intra-accumbens nalmefene in ethanol-dependent rats. Neuropharmacology. 2011;61:35–42.PubMedPubMedCentral Nealey KA, Smith AW, Davis SM, Smith DG, Walker BM. κ-opioid receptors are implicated in the increased potency of intra-accumbens nalmefene in ethanol-dependent rats. Neuropharmacology. 2011;61:35–42.PubMedPubMedCentral
71.
Zurück zum Zitat Mann K, Bladström A, Torup L, Gual A, van den Brink W. Extending the treatment options in alcohol dependence: a randomized controlled study of as-needed nalmefene. Biol Psychiatry. 2013;73(8):706–13.PubMed Mann K, Bladström A, Torup L, Gual A, van den Brink W. Extending the treatment options in alcohol dependence: a randomized controlled study of as-needed nalmefene. Biol Psychiatry. 2013;73(8):706–13.PubMed
72.
Zurück zum Zitat van den Brink W, Aubin H-J, Bladström A, Torup L, Gual A, Mann K. Efficacy of as-needed nalmefene in alcohol-dependent patients with at least a high drinking risk level: results from a subgroup analysis of two randomized controlled 6-month studies. Alcohol Alcohol. 2013;48(5):570–8.PubMedPubMedCentral van den Brink W, Aubin H-J, Bladström A, Torup L, Gual A, Mann K. Efficacy of as-needed nalmefene in alcohol-dependent patients with at least a high drinking risk level: results from a subgroup analysis of two randomized controlled 6-month studies. Alcohol Alcohol. 2013;48(5):570–8.PubMedPubMedCentral
73.
Zurück zum Zitat Gual A, He Y, Torup L, van den Brink W, Mann K, ESENSE 2 Study Group. A randomised, double-blind, placebo-controlled, efficacy study of nalmefene, as-needed use, in patients with alcohol dependence. Eur Neuropsychopharmacol. 2013;23(11):1432–42.PubMed Gual A, He Y, Torup L, van den Brink W, Mann K, ESENSE 2 Study Group. A randomised, double-blind, placebo-controlled, efficacy study of nalmefene, as-needed use, in patients with alcohol dependence. Eur Neuropsychopharmacol. 2013;23(11):1432–42.PubMed
74.
Zurück zum Zitat Naudet F, Fitzgerald N, Braillon A. Nalmefene for alcohol dependence: a NICE decision? Lancet Psychiatry. 2016;3(12):1104–5.PubMed Naudet F, Fitzgerald N, Braillon A. Nalmefene for alcohol dependence: a NICE decision? Lancet Psychiatry. 2016;3(12):1104–5.PubMed
75.
Zurück zum Zitat Palpacuer C, Laviolle B, Boussageon R, Reymann JM, Bellissant E, Naudet F. Risks and benefits of nalmefene in the treatment of adult alcohol dependence: a systematic literature review and meta-analysis of published and unpublished double-blind randomized controlled trials. PLoS Med. 2015;12(12):e1001924.PubMedPubMedCentral Palpacuer C, Laviolle B, Boussageon R, Reymann JM, Bellissant E, Naudet F. Risks and benefits of nalmefene in the treatment of adult alcohol dependence: a systematic literature review and meta-analysis of published and unpublished double-blind randomized controlled trials. PLoS Med. 2015;12(12):e1001924.PubMedPubMedCentral
76.
Zurück zum Zitat Johansen KGV, Tarp S, Astrup A, Lund H, Pagsberg AK, Christensen R. Harms associated with taking nalmefene for substance use and impulse control disorders: a systematic review and meta-analysis of randomised controlled trials. PLoS ONE. 2017;12(8):e0183821.PubMedPubMedCentral Johansen KGV, Tarp S, Astrup A, Lund H, Pagsberg AK, Christensen R. Harms associated with taking nalmefene for substance use and impulse control disorders: a systematic review and meta-analysis of randomised controlled trials. PLoS ONE. 2017;12(8):e0183821.PubMedPubMedCentral
77.
Zurück zum Zitat Palpacuer C, Duprez R, Huneau A, Locher C, Boussageon R, Laviolle B, et al. Pharmacologically controlled drinking in the treatment of alcohol dependence or alcohol use disorders: a systematic review with direct and network meta-analyses on nalmefene, naltrexone, acamprosate, baclofen and topiramate. Addiction. 2018;113(2):220–37.PubMed Palpacuer C, Duprez R, Huneau A, Locher C, Boussageon R, Laviolle B, et al. Pharmacologically controlled drinking in the treatment of alcohol dependence or alcohol use disorders: a systematic review with direct and network meta-analyses on nalmefene, naltrexone, acamprosate, baclofen and topiramate. Addiction. 2018;113(2):220–37.PubMed
78.
Zurück zum Zitat Naudet F, Braillon A. Baclofen and alcohol in France. Lancet Psychiatry. 2018;5(12):961–2.PubMed Naudet F, Braillon A. Baclofen and alcohol in France. Lancet Psychiatry. 2018;5(12):961–2.PubMed
79.
Zurück zum Zitat Agabio R, Sinclair JM, Addolorato G, Aubin H-J, Beraha EM, Caputo F, et al. Baclofen for the treatment of alcohol use disorder: the Cagliari statement. Lancet Psychiatry. 2018;5(12):957–60.PubMed Agabio R, Sinclair JM, Addolorato G, Aubin H-J, Beraha EM, Caputo F, et al. Baclofen for the treatment of alcohol use disorder: the Cagliari statement. Lancet Psychiatry. 2018;5(12):957–60.PubMed
80.
Zurück zum Zitat Leggio L, Garbutt JC, Addolorato G. Effectiveness and safety of baclofen in the treatment of alcohol dependent patients. CNS Neurol Disord Drug Targets. 2010;9(1):33–44.PubMed Leggio L, Garbutt JC, Addolorato G. Effectiveness and safety of baclofen in the treatment of alcohol dependent patients. CNS Neurol Disord Drug Targets. 2010;9(1):33–44.PubMed
81.
Zurück zum Zitat Garbutt JC, Kampov-Polevoy AB, Gallop R, Kalka-Juhl L, Flannery BA. Efficacy and safety of baclofen for alcohol dependence: a randomized, double-blind, placebo-controlled trial. Alcohol Clin Exp Res. 2010;34(11):1849–57.PubMedPubMedCentral Garbutt JC, Kampov-Polevoy AB, Gallop R, Kalka-Juhl L, Flannery BA. Efficacy and safety of baclofen for alcohol dependence: a randomized, double-blind, placebo-controlled trial. Alcohol Clin Exp Res. 2010;34(11):1849–57.PubMedPubMedCentral
82.
Zurück zum Zitat Addolorato G, Caputo F, Capristo E, Domenicali M, Bernardi M, Janiri L, et al. Baclofen efficacy in reducing alcohol craving and intake: a preliminary double-blind randomized controlled study. Alcohol Alcohol. 2002;37(5):504–8.PubMed Addolorato G, Caputo F, Capristo E, Domenicali M, Bernardi M, Janiri L, et al. Baclofen efficacy in reducing alcohol craving and intake: a preliminary double-blind randomized controlled study. Alcohol Alcohol. 2002;37(5):504–8.PubMed
83.
Zurück zum Zitat Morley KC, Baillie A, Leung S, Addolorato G, Leggio L, Haber PS. Baclofen for the treatment of alcohol dependence and possible role of comorbid anxiety. Alcohol Alcohol. 2014;49(6):654–60.PubMedPubMedCentral Morley KC, Baillie A, Leung S, Addolorato G, Leggio L, Haber PS. Baclofen for the treatment of alcohol dependence and possible role of comorbid anxiety. Alcohol Alcohol. 2014;49(6):654–60.PubMedPubMedCentral
84.
Zurück zum Zitat Rose AK, Jones A. Baclofen: its effectiveness in reducing harmful drinking, craving, and negative mood. A meta-analysis. Addiction. 2018;113(8):1396–406.PubMed Rose AK, Jones A. Baclofen: its effectiveness in reducing harmful drinking, craving, and negative mood. A meta-analysis. Addiction. 2018;113(8):1396–406.PubMed
85.
Zurück zum Zitat Pierce M, Sutterland A, Beraha EM, Morley K, van den Brink W. Efficacy, tolerability, and safety of low-dose and high-dose baclofen in the treatment of alcohol dependence: a systematic review and meta-analysis. Eur Neuropsychopharmacol. 2018;28(7):795–806.PubMed Pierce M, Sutterland A, Beraha EM, Morley K, van den Brink W. Efficacy, tolerability, and safety of low-dose and high-dose baclofen in the treatment of alcohol dependence: a systematic review and meta-analysis. Eur Neuropsychopharmacol. 2018;28(7):795–806.PubMed
86.
Zurück zum Zitat Minozzi S, Saulle R, Rösner S. Baclofen for alcohol use disorder. Cochrane Database Syst Rev. 2018;11:CD012557. Minozzi S, Saulle R, Rösner S. Baclofen for alcohol use disorder. Cochrane Database Syst Rev. 2018;11:CD012557.
87.
Zurück zum Zitat Hauser P, Fuller B, Ho SB, Thuras P, Kern S, Dieperink E. The safety and efficacy of baclofen to reduce alcohol use in veterans with chronic hepatitis C: a randomized controlled trial. Addiction. 2017;112(7):1173–83.PubMed Hauser P, Fuller B, Ho SB, Thuras P, Kern S, Dieperink E. The safety and efficacy of baclofen to reduce alcohol use in veterans with chronic hepatitis C: a randomized controlled trial. Addiction. 2017;112(7):1173–83.PubMed
88.
Zurück zum Zitat Reynaud M, Aubin H-J, Trinquet F, Zakine B, Dano C, Dematteis M, et al. A randomized, placebo-controlled study of high-dose baclofen in alcohol-dependent patients—the ALPADIR study. Alcohol Alcohol. 2017;52(4):439–46.PubMed Reynaud M, Aubin H-J, Trinquet F, Zakine B, Dano C, Dematteis M, et al. A randomized, placebo-controlled study of high-dose baclofen in alcohol-dependent patients—the ALPADIR study. Alcohol Alcohol. 2017;52(4):439–46.PubMed
89.
Zurück zum Zitat Chartier M, Tannous S, Benturquia N, Labat L, Reis R, Risède P, et al. Baclofen-induced neuro-respiratory toxicity in the rat: contribution of tolerance and characterization of withdrawal syndrome. Toxicol Sci. 2018;164(1):153–65.PubMed Chartier M, Tannous S, Benturquia N, Labat L, Reis R, Risède P, et al. Baclofen-induced neuro-respiratory toxicity in the rat: contribution of tolerance and characterization of withdrawal syndrome. Toxicol Sci. 2018;164(1):153–65.PubMed
90.
Zurück zum Zitat Santos C, Olmedo RE. Sedative-hypnotic drug withdrawal syndrome: recognition and treatment. Emerg Med Pract. 2017;19(3):1–20.PubMed Santos C, Olmedo RE. Sedative-hypnotic drug withdrawal syndrome: recognition and treatment. Emerg Med Pract. 2017;19(3):1–20.PubMed
91.
Zurück zum Zitat Garbutt JC, Kampov-Polevoy AB, Pedersen C, et al. Efficacy and tolerability of baclofen in a U.S. community population with alcohol use disorder: a dose-response, randomized, controlled trial. Neuropsychopharmacol. 2021;46:2250–6. Garbutt JC, Kampov-Polevoy AB, Pedersen C, et al. Efficacy and tolerability of baclofen in a U.S. community population with alcohol use disorder: a dose-response, randomized, controlled trial. Neuropsychopharmacol. 2021;46:2250–6.
92.
Zurück zum Zitat Farokhnia M, Deschaine SL, Sadighi A, Farinelli LA, Lee MR, Akhlaghi F, et al. A deeper insight into how GABA-B receptor agonism via baclofen may affect alcohol seeking and consumption: lessons learned from a human laboratory investigation. Mol Psychiatry. 2021;26(2):545–55.PubMed Farokhnia M, Deschaine SL, Sadighi A, Farinelli LA, Lee MR, Akhlaghi F, et al. A deeper insight into how GABA-B receptor agonism via baclofen may affect alcohol seeking and consumption: lessons learned from a human laboratory investigation. Mol Psychiatry. 2021;26(2):545–55.PubMed
93.
Zurück zum Zitat Morley KC, Baillie A, Fraser I, Furneaux-Bate A, Dore G, Roberts M, et al. Baclofen in the treatment of alcohol dependence with or without liver disease: multisite, randomised, double-blind, placebo-controlled trial. Br J Psychiatry. 2018;212(6):362–9.PubMed Morley KC, Baillie A, Fraser I, Furneaux-Bate A, Dore G, Roberts M, et al. Baclofen in the treatment of alcohol dependence with or without liver disease: multisite, randomised, double-blind, placebo-controlled trial. Br J Psychiatry. 2018;212(6):362–9.PubMed
94.
Zurück zum Zitat Addolorato G, Leggio L, Ferrulli A, Cardone S, Vonghia L, Mirijello A, et al. Effectiveness and safety of baclofen for maintenance of alcohol abstinence in alcohol-dependent patients with liver cirrhosis: randomised, double-blind controlled study. Lancet. 2007;370(9603):1915–22.PubMed Addolorato G, Leggio L, Ferrulli A, Cardone S, Vonghia L, Mirijello A, et al. Effectiveness and safety of baclofen for maintenance of alcohol abstinence in alcohol-dependent patients with liver cirrhosis: randomised, double-blind controlled study. Lancet. 2007;370(9603):1915–22.PubMed
95.
Zurück zum Zitat Leggio L, Litten RZ. The GABA-B receptor agonist baclofen helps patients with alcohol use disorder: why these findings matter. Neuropsychopharmacol. 2021;46:2228–9. Leggio L, Litten RZ. The GABA-B receptor agonist baclofen helps patients with alcohol use disorder: why these findings matter. Neuropsychopharmacol. 2021;46:2228–9.
96.
Zurück zum Zitat Addolorato G, Lesch O-M, Maremmani I, Walter H, Nava F, Raffaillac Q, et al. Post-marketing and clinical safety experience with sodium oxybate for the treatment of alcohol withdrawal syndrome and maintenance of abstinence in alcohol-dependent subjects. Expert Opin Drug Saf. 2020;19(2):159–66.PubMed Addolorato G, Lesch O-M, Maremmani I, Walter H, Nava F, Raffaillac Q, et al. Post-marketing and clinical safety experience with sodium oxybate for the treatment of alcohol withdrawal syndrome and maintenance of abstinence in alcohol-dependent subjects. Expert Opin Drug Saf. 2020;19(2):159–66.PubMed
97.
Zurück zum Zitat Skala K, Caputo F, Mirijello A, Vassallo G, Antonelli M, Ferrulli A, et al. Sodium oxybate in the treatment of alcohol dependence: from the alcohol withdrawal syndrome to the alcohol relapse prevention. Expert Opin Pharmacother. 2014;15(2):245–57.PubMed Skala K, Caputo F, Mirijello A, Vassallo G, Antonelli M, Ferrulli A, et al. Sodium oxybate in the treatment of alcohol dependence: from the alcohol withdrawal syndrome to the alcohol relapse prevention. Expert Opin Pharmacother. 2014;15(2):245–57.PubMed
98.
Zurück zum Zitat Busardò FP, Kyriakou C, Napoletano S, Marinelli E, Zaami S. Clinical applications of sodium oxybate (GHB): from narcolepsy to alcohol withdrawal syndrome. Eur Rev Med Pharmacol Sci. 2015;19(23):4654–63.PubMed Busardò FP, Kyriakou C, Napoletano S, Marinelli E, Zaami S. Clinical applications of sodium oxybate (GHB): from narcolepsy to alcohol withdrawal syndrome. Eur Rev Med Pharmacol Sci. 2015;19(23):4654–63.PubMed
99.
Zurück zum Zitat Leone MA, Vigna-Taglianti F, Avanzi G, Brambilla R, Faggiano F. Gamma-hydroxybutyrate (GHB) for treatment of alcohol withdrawal and prevention of relapses. Cochrane Database Syst Rev. 2010;(2):CD006266. Leone MA, Vigna-Taglianti F, Avanzi G, Brambilla R, Faggiano F. Gamma-hydroxybutyrate (GHB) for treatment of alcohol withdrawal and prevention of relapses. Cochrane Database Syst Rev. 2010;(2):CD006266.
100.
Zurück zum Zitat van den Brink W, Addolorato G, Aubin H-J, Benyamina A, Caputo F, Dematteis M, et al. Efficacy and safety of sodium oxybate in alcohol-dependent patients with a very high drinking risk level. Addict Biol. 2018;23(4):969–86.PubMed van den Brink W, Addolorato G, Aubin H-J, Benyamina A, Caputo F, Dematteis M, et al. Efficacy and safety of sodium oxybate in alcohol-dependent patients with a very high drinking risk level. Addict Biol. 2018;23(4):969–86.PubMed
101.
Zurück zum Zitat Caputo F, Francini S, Brambilla R, Vigna-Taglianti F, Stoppo M, Del Re A, et al. Sodium oxybate in maintaining alcohol abstinence in alcoholic patients with and without psychiatric comorbidity. Eur Neuropsychopharmacol. 2011;21(6):450–6.PubMed Caputo F, Francini S, Brambilla R, Vigna-Taglianti F, Stoppo M, Del Re A, et al. Sodium oxybate in maintaining alcohol abstinence in alcoholic patients with and without psychiatric comorbidity. Eur Neuropsychopharmacol. 2011;21(6):450–6.PubMed
102.
Zurück zum Zitat Mannucci C, Pichini S, Spagnolo EV, Calapai F, Gangemi S, Navarra M, et al. Sodium oxybate therapy for alcohol withdrawal syndrome and keeping of alcohol abstinence. Curr Drug Metab. 2018;19(13):1056–64.PubMed Mannucci C, Pichini S, Spagnolo EV, Calapai F, Gangemi S, Navarra M, et al. Sodium oxybate therapy for alcohol withdrawal syndrome and keeping of alcohol abstinence. Curr Drug Metab. 2018;19(13):1056–64.PubMed
103.
Zurück zum Zitat Maremmani AGI, Pani PP, Rovai L, Pacini M, Dell’Osso L, Maremmani I. Long-term γ-hydroxybutyric acid (GHB) and disulfiram combination therapy in GHB treatment-resistant chronic alcoholics. Int J Environ Res Public Health. 2011;8(7):2816–27.PubMedPubMedCentral Maremmani AGI, Pani PP, Rovai L, Pacini M, Dell’Osso L, Maremmani I. Long-term γ-hydroxybutyric acid (GHB) and disulfiram combination therapy in GHB treatment-resistant chronic alcoholics. Int J Environ Res Public Health. 2011;8(7):2816–27.PubMedPubMedCentral
104.
Zurück zum Zitat Caputo F, Maremmani AGI, Addolorato G, Domenicali M, Zoli G, D’Amore A, et al. Sodium oxybate plus nalmefene for the treatment of alcohol use disorder: a case series. J Psychopharmacol. 2016;30(4):402–9.PubMed Caputo F, Maremmani AGI, Addolorato G, Domenicali M, Zoli G, D’Amore A, et al. Sodium oxybate plus nalmefene for the treatment of alcohol use disorder: a case series. J Psychopharmacol. 2016;30(4):402–9.PubMed
105.
Zurück zum Zitat Caputo F, Francini S, Stoppo M, Lorenzini F, Vignoli T, Del Re A, et al. Incidence of craving for and abuse of gamma-hydroxybutyric acid (GHB) in different populations of treated alcoholics: an open comparative study. J Psychopharmacol. 2009;23(8):883–90.PubMed Caputo F, Francini S, Stoppo M, Lorenzini F, Vignoli T, Del Re A, et al. Incidence of craving for and abuse of gamma-hydroxybutyric acid (GHB) in different populations of treated alcoholics: an open comparative study. J Psychopharmacol. 2009;23(8):883–90.PubMed
106.
Zurück zum Zitat Department of Defense. VA/DoD clinical practice guideline for the management of substance use disorders. 2015. Department of Defense. VA/DoD clinical practice guideline for the management of substance use disorders. 2015.
107.
Zurück zum Zitat Reus VI, Fochtmann LJ, Bukstein O, Eyler AE, Hilty DM, Horvitz-Lennon M, et al. The American psychiatric association practice guideline for the pharmacological treatment of patients with alcohol use disorder. Am J Psychiatry. 2018;175(1):86–90.PubMed Reus VI, Fochtmann LJ, Bukstein O, Eyler AE, Hilty DM, Horvitz-Lennon M, et al. The American psychiatric association practice guideline for the pharmacological treatment of patients with alcohol use disorder. Am J Psychiatry. 2018;175(1):86–90.PubMed
108.
Zurück zum Zitat Shalaby HN, El-Tanbouly DM, Zaki HF. Topiramate mitigates 3-nitropropionic acid-induced striatal neurotoxicity via modulation of AMPA receptors. Food Chem Toxicol. 2018;118:227–34.PubMed Shalaby HN, El-Tanbouly DM, Zaki HF. Topiramate mitigates 3-nitropropionic acid-induced striatal neurotoxicity via modulation of AMPA receptors. Food Chem Toxicol. 2018;118:227–34.PubMed
109.
Zurück zum Zitat Huang Y-L, Peng M, Zhu G. Topiramate in the treatment of antipsychotic-induced hyperprolactinemia. Med Hypotheses. 2020;138:109607.PubMed Huang Y-L, Peng M, Zhu G. Topiramate in the treatment of antipsychotic-induced hyperprolactinemia. Med Hypotheses. 2020;138:109607.PubMed
110.
Zurück zum Zitat Kranzler HR, Covault J, Feinn R, Armeli S, Tennen H, Arias AJ, et al. Topiramate treatment for heavy drinkers: moderation by a GRIK1 polymorphism. Am J Psychiatry. 2014;171(4):445–52.PubMedPubMedCentral Kranzler HR, Covault J, Feinn R, Armeli S, Tennen H, Arias AJ, et al. Topiramate treatment for heavy drinkers: moderation by a GRIK1 polymorphism. Am J Psychiatry. 2014;171(4):445–52.PubMedPubMedCentral
111.
Zurück zum Zitat Zhang X, Velumian AA, Jones OT, Carlen PL. Modulation of high-voltage-activated calcium channels in dentate granule cells by topiramate. Epilepsia. 2000;41(S1):52–60.PubMed Zhang X, Velumian AA, Jones OT, Carlen PL. Modulation of high-voltage-activated calcium channels in dentate granule cells by topiramate. Epilepsia. 2000;41(S1):52–60.PubMed
112.
Zurück zum Zitat Johnson BA, Ait-Daoud N, Bowden CL, DiClemente CC, Roache JD, Lawson K, et al. Oral topiramate for treatment of alcohol dependence: a randomised controlled trial. Lancet. 2003;361(9370):1677–85.PubMed Johnson BA, Ait-Daoud N, Bowden CL, DiClemente CC, Roache JD, Lawson K, et al. Oral topiramate for treatment of alcohol dependence: a randomised controlled trial. Lancet. 2003;361(9370):1677–85.PubMed
113.
Zurück zum Zitat Hargreaves GA, McGregor IS. Topiramate moderately reduces the motivation to consume alcohol and has a marked antidepressant effect in rats. Alcohol Clin Exp Res. 2007;31(11):1900–7.PubMed Hargreaves GA, McGregor IS. Topiramate moderately reduces the motivation to consume alcohol and has a marked antidepressant effect in rats. Alcohol Clin Exp Res. 2007;31(11):1900–7.PubMed
114.
Zurück zum Zitat Knapp CM, Mercado M, Markley TL, Crosby S, Ciraulo DA, Kornetsky C. Zonisamide decreases ethanol intake in rats and mice. Pharmacol Biochem Behav. 2007;87(1):65–72.PubMedPubMedCentral Knapp CM, Mercado M, Markley TL, Crosby S, Ciraulo DA, Kornetsky C. Zonisamide decreases ethanol intake in rats and mice. Pharmacol Biochem Behav. 2007;87(1):65–72.PubMedPubMedCentral
115.
Zurück zum Zitat Breslin FJ, Johnson BA, Lynch WJ. Effect of topiramate treatment on ethanol consumption in rats. Psychopharmacology. 2010;207(4):529–34.PubMed Breslin FJ, Johnson BA, Lynch WJ. Effect of topiramate treatment on ethanol consumption in rats. Psychopharmacology. 2010;207(4):529–34.PubMed
116.
Zurück zum Zitat Knapp CM, Ciraulo DA, Sarid-Segal O, Richardson MA, Devine E, Streeter CC, et al. Zonisamide, topiramate, and levetiracetam: efficacy and neuropsychological effects in alcohol use disorders. J Clin Psychopharmacol. 2015;35(1):34–42.PubMed Knapp CM, Ciraulo DA, Sarid-Segal O, Richardson MA, Devine E, Streeter CC, et al. Zonisamide, topiramate, and levetiracetam: efficacy and neuropsychological effects in alcohol use disorders. J Clin Psychopharmacol. 2015;35(1):34–42.PubMed
117.
Zurück zum Zitat Johnson BA, Rosenthal N, Capece JA, Wiegand F, Mao L, Beyers K, et al. Topiramate for treating alcohol dependence: a randomized controlled trial. JAMA. 2007;298(14):1641–51.PubMed Johnson BA, Rosenthal N, Capece JA, Wiegand F, Mao L, Beyers K, et al. Topiramate for treating alcohol dependence: a randomized controlled trial. JAMA. 2007;298(14):1641–51.PubMed
118.
Zurück zum Zitat Blodgett JC, Re ACD, Maisel NC, Finney JW. A meta-analysis of topiramate’s effects for individuals with alcohol use disorders. Alcohol Clin Exp Res. 2014;38(6):1481–8.PubMedPubMedCentral Blodgett JC, Re ACD, Maisel NC, Finney JW. A meta-analysis of topiramate’s effects for individuals with alcohol use disorders. Alcohol Clin Exp Res. 2014;38(6):1481–8.PubMedPubMedCentral
119.
Zurück zum Zitat Jose NA, Yadav P, Kapoor A, Mahla VP. Comparison between baclofen and topiramate in alcohol dependence: a prospective study. Ind Psychiatry J. 2019;28(1):44–50.PubMedPubMedCentral Jose NA, Yadav P, Kapoor A, Mahla VP. Comparison between baclofen and topiramate in alcohol dependence: a prospective study. Ind Psychiatry J. 2019;28(1):44–50.PubMedPubMedCentral
120.
Zurück zum Zitat Kenna GA, Lomastro TL, Schiesl A, Leggio L, Swift RM. Review of topiramate: an antiepileptic for the treatment of alcohol dependence. Curr Drug Abuse Rev. 2009;2(2):135–42.PubMed Kenna GA, Lomastro TL, Schiesl A, Leggio L, Swift RM. Review of topiramate: an antiepileptic for the treatment of alcohol dependence. Curr Drug Abuse Rev. 2009;2(2):135–42.PubMed
121.
Zurück zum Zitat Olmsted CL, Kockler DR. Topiramate for alcohol dependence. Ann Pharmacother. 2008;42(10):1475–80.PubMed Olmsted CL, Kockler DR. Topiramate for alcohol dependence. Ann Pharmacother. 2008;42(10):1475–80.PubMed
122.
Zurück zum Zitat Sills GJ. The mechanisms of action of gabapentin and pregabalin. Curr Opin Pharmacol. 2006;6(1):108–13.PubMed Sills GJ. The mechanisms of action of gabapentin and pregabalin. Curr Opin Pharmacol. 2006;6(1):108–13.PubMed
123.
Zurück zum Zitat Roberto M, Gilpin NW, O’Dell LE, Cruz MT, Morse AC, Siggins GR, et al. Cellular and behavioral interactions of gabapentin with alcohol dependence. J Neurosci. 2008;28(22):5762–71.PubMedPubMedCentral Roberto M, Gilpin NW, O’Dell LE, Cruz MT, Morse AC, Siggins GR, et al. Cellular and behavioral interactions of gabapentin with alcohol dependence. J Neurosci. 2008;28(22):5762–71.PubMedPubMedCentral
124.
Zurück zum Zitat Besheer J, Frisbee S, Randall PA, Jaramillo AA, Masciello M. Gabapentin potentiates sensitivity to the interoceptive effects of alcohol and increases alcohol self-administration in rats. Neuropharmacology. 2016;101:216–24.PubMed Besheer J, Frisbee S, Randall PA, Jaramillo AA, Masciello M. Gabapentin potentiates sensitivity to the interoceptive effects of alcohol and increases alcohol self-administration in rats. Neuropharmacology. 2016;101:216–24.PubMed
125.
Zurück zum Zitat Ozburn AR, Metten P, Potretzke S, Townsley KG, Blednov YA, Crabbe JC. Effects of pharmacologically targeting neuroimmune pathways on alcohol drinking in mice selectively bred to drink to intoxication. Alcoho Clin Exp Res. 2020;44(2):553–66. Ozburn AR, Metten P, Potretzke S, Townsley KG, Blednov YA, Crabbe JC. Effects of pharmacologically targeting neuroimmune pathways on alcohol drinking in mice selectively bred to drink to intoxication. Alcoho Clin Exp Res. 2020;44(2):553–66.
126.
Zurück zum Zitat Furieri FA, Nakamura-Palacios EM. Gabapentin reduces alcohol consumption and craving: a randomized, double-blind, placebo-controlled trial. J Clin Psychiatry. 2007;68(11):1691–700.PubMed Furieri FA, Nakamura-Palacios EM. Gabapentin reduces alcohol consumption and craving: a randomized, double-blind, placebo-controlled trial. J Clin Psychiatry. 2007;68(11):1691–700.PubMed
127.
Zurück zum Zitat Brower KJ, Myra Kim H, Strobbe S, Karam-Hage MA, Consens F, Zucker RA. A randomized double-blind pilot trial of gabapentin versus placebo to treat alcohol dependence and comorbid insomnia. Alcohol Clin Exp Res. 2008;32(8):1429–38.PubMedPubMedCentral Brower KJ, Myra Kim H, Strobbe S, Karam-Hage MA, Consens F, Zucker RA. A randomized double-blind pilot trial of gabapentin versus placebo to treat alcohol dependence and comorbid insomnia. Alcohol Clin Exp Res. 2008;32(8):1429–38.PubMedPubMedCentral
128.
Zurück zum Zitat Mason BJ, Light JM, Williams LD, Drobes DJ. Proof-of-concept human laboratory study for protracted abstinence in alcohol dependence: effects of gabapentin. Addict Biol. 2009;14(1):73–83.PubMed Mason BJ, Light JM, Williams LD, Drobes DJ. Proof-of-concept human laboratory study for protracted abstinence in alcohol dependence: effects of gabapentin. Addict Biol. 2009;14(1):73–83.PubMed
129.
Zurück zum Zitat Mason BJ, Quello S, Goodell V, Shadan F, Kyle M, Begovic A. Gabapentin treatment for alcohol dependence: a randomized clinical trial. JAMA Intern Med. 2014;174(1):70–7.PubMedPubMedCentral Mason BJ, Quello S, Goodell V, Shadan F, Kyle M, Begovic A. Gabapentin treatment for alcohol dependence: a randomized clinical trial. JAMA Intern Med. 2014;174(1):70–7.PubMedPubMedCentral
130.
Zurück zum Zitat Anton RF, Myrick H, Baros AM, Latham PK, Randall PK, Wright TM, et al. Efficacy of a combination of flumazenil and gabapentin in the treatment of alcohol dependence: relationship to alcohol withdrawal symptoms. J Clin Psychopharmacol. 2009;29(4):334–42.PubMed Anton RF, Myrick H, Baros AM, Latham PK, Randall PK, Wright TM, et al. Efficacy of a combination of flumazenil and gabapentin in the treatment of alcohol dependence: relationship to alcohol withdrawal symptoms. J Clin Psychopharmacol. 2009;29(4):334–42.PubMed
131.
Zurück zum Zitat Anton RF, Myrick H, Wright TM, Latham PK, Baros AM, Waid LR, et al. Gabapentin combined with naltrexone for the treatment of alcohol dependence. Am J Psychiatry. 2011;168(7):709–17.PubMedPubMedCentral Anton RF, Myrick H, Wright TM, Latham PK, Baros AM, Waid LR, et al. Gabapentin combined with naltrexone for the treatment of alcohol dependence. Am J Psychiatry. 2011;168(7):709–17.PubMedPubMedCentral
132.
Zurück zum Zitat Pani PP, Trogu E, Pacini M, Maremmani I. Anticonvulsants for alcohol dependence. Cochrane Database Syst Rev. 2014;13(2):CD008544. Pani PP, Trogu E, Pacini M, Maremmani I. Anticonvulsants for alcohol dependence. Cochrane Database Syst Rev. 2014;13(2):CD008544.
133.
Zurück zum Zitat Kranzler HR, Feinn R, Morris P, Hartwell EE. A meta-analysis of the efficacy of gabapentin for treating alcohol use disorder. Addiction. 2019;114(9):1547–55.PubMedPubMedCentral Kranzler HR, Feinn R, Morris P, Hartwell EE. A meta-analysis of the efficacy of gabapentin for treating alcohol use disorder. Addiction. 2019;114(9):1547–55.PubMedPubMedCentral
134.
Zurück zum Zitat Anton RF, Latham P, Voronin K, Book S, Hoffman M, Prisciandaro J, et al. Efficacy of gabapentin for the treatment of alcohol use disorder in patients with alcohol withdrawal symptoms: a randomized clinical trial. JAMA Intern Med. 2020;180(5):728–36.PubMed Anton RF, Latham P, Voronin K, Book S, Hoffman M, Prisciandaro J, et al. Efficacy of gabapentin for the treatment of alcohol use disorder in patients with alcohol withdrawal symptoms: a randomized clinical trial. JAMA Intern Med. 2020;180(5):728–36.PubMed
135.
Zurück zum Zitat Falk DE, Ryan ML, Fertig JB, Devine EG, Cruz R, Brown ES, et al. Gabapentin enacarbil extended-release for alcohol use disorder: a randomized, double-blind, placebo-controlled, multisite trial assessing efficacy and safety. Alcohol Clin Exp Res. 2019;43(1):158–69.PubMed Falk DE, Ryan ML, Fertig JB, Devine EG, Cruz R, Brown ES, et al. Gabapentin enacarbil extended-release for alcohol use disorder: a randomized, double-blind, placebo-controlled, multisite trial assessing efficacy and safety. Alcohol Clin Exp Res. 2019;43(1):158–69.PubMed
136.
Zurück zum Zitat Wiffen PJ, Derry S, Bell RF, Rice AS, Tölle TR, Phillips T, et al. Gabapentin for chronic neuropathic pain in adults. Cochrane Database Syst Rev. 2017;6:CD007938. Wiffen PJ, Derry S, Bell RF, Rice AS, Tölle TR, Phillips T, et al. Gabapentin for chronic neuropathic pain in adults. Cochrane Database Syst Rev. 2017;6:CD007938.
137.
Zurück zum Zitat Smith RV, Havens JR, Walsh SL. Gabapentin misuse, abuse and diversion: a systematic review. Addiction. 2016;111(7):1160–74.PubMedPubMedCentral Smith RV, Havens JR, Walsh SL. Gabapentin misuse, abuse and diversion: a systematic review. Addiction. 2016;111(7):1160–74.PubMedPubMedCentral
138.
Zurück zum Zitat Steensland P, Simms JA, Holgate J, Richards JK, Bartlett SE. Varenicline, an alpha4beta2 nicotinic acetylcholine receptor partial agonist, selectively decreases ethanol consumption and seeking. Proc Natl Acad Sci USA. 2007;104(30):12518–23.PubMedPubMedCentral Steensland P, Simms JA, Holgate J, Richards JK, Bartlett SE. Varenicline, an alpha4beta2 nicotinic acetylcholine receptor partial agonist, selectively decreases ethanol consumption and seeking. Proc Natl Acad Sci USA. 2007;104(30):12518–23.PubMedPubMedCentral
139.
Zurück zum Zitat Patkar OL, Belmer A, Tarren JR, Holgate JY, Bartlett SE. The effect of varenicline on binge-like ethanol consumption in mice is β4 nicotinic acetylcholine receptor-independent. Neurosci Lett. 2016;28(633):235–9. Patkar OL, Belmer A, Tarren JR, Holgate JY, Bartlett SE. The effect of varenicline on binge-like ethanol consumption in mice is β4 nicotinic acetylcholine receptor-independent. Neurosci Lett. 2016;28(633):235–9.
140.
Zurück zum Zitat Kamens HM, Silva C, Peck C, Miller CN. Varenicline modulates ethanol and saccharin consumption in adolescent male and female C57BL/6J mice. Brain Res Bull. 2018;138:20–5.PubMed Kamens HM, Silva C, Peck C, Miller CN. Varenicline modulates ethanol and saccharin consumption in adolescent male and female C57BL/6J mice. Brain Res Bull. 2018;138:20–5.PubMed
141.
Zurück zum Zitat Froehlich JC, Fischer SM, Dilley JE, Nicholson ER, Smith TN, Filosa NJ, et al. Combining varenicline (Chantix) with naltrexone decreases alcohol drinking more effectively than does either drug alone in a rodent model of alcoholism. Alcohol Clin Exp Res. 2016;40(9):1961–70.PubMedPubMedCentral Froehlich JC, Fischer SM, Dilley JE, Nicholson ER, Smith TN, Filosa NJ, et al. Combining varenicline (Chantix) with naltrexone decreases alcohol drinking more effectively than does either drug alone in a rodent model of alcoholism. Alcohol Clin Exp Res. 2016;40(9):1961–70.PubMedPubMedCentral
142.
Zurück zum Zitat Litten RZ, Ryan ML, Fertig JB, Falk DE, Johnson B, Dunn KE, et al. A double-blind, placebo-controlled trial assessing the efficacy of varenicline tartrate for alcohol dependence. J Addict Med. 2013;7(4):277–86.PubMedPubMedCentral Litten RZ, Ryan ML, Fertig JB, Falk DE, Johnson B, Dunn KE, et al. A double-blind, placebo-controlled trial assessing the efficacy of varenicline tartrate for alcohol dependence. J Addict Med. 2013;7(4):277–86.PubMedPubMedCentral
143.
Zurück zum Zitat de Bejczy A, Löf E, Walther L, Guterstam J, Hammarberg A, Asanovska G, et al. Varenicline for treatment of alcohol dependence: a randomized, placebo-controlled trial. Alcohol Clin Exp Res. 2015;39(11):2189–99.PubMed de Bejczy A, Löf E, Walther L, Guterstam J, Hammarberg A, Asanovska G, et al. Varenicline for treatment of alcohol dependence: a randomized, placebo-controlled trial. Alcohol Clin Exp Res. 2015;39(11):2189–99.PubMed
144.
Zurück zum Zitat Miranda R, O’Malley SS, Treloar Padovano H, Wu R, Falk DE, Ryan ML, et al. Effects of alcohol cue reactivity on subsequent treatment outcomes among treatment-seeking individuals with alcohol use disorder: a multisite randomized, double-blind, placebo-controlled clinical trial of varenicline. Alcohol Clin Exp Res. 2020;44(7):1431–43.PubMedPubMedCentral Miranda R, O’Malley SS, Treloar Padovano H, Wu R, Falk DE, Ryan ML, et al. Effects of alcohol cue reactivity on subsequent treatment outcomes among treatment-seeking individuals with alcohol use disorder: a multisite randomized, double-blind, placebo-controlled clinical trial of varenicline. Alcohol Clin Exp Res. 2020;44(7):1431–43.PubMedPubMedCentral
145.
Zurück zum Zitat Anthony JC, Echeagaray-Wagner F. Epidemiologic analysis of alcohol and tobacco use. Alcohol Res Health. 2000;24(4):201–8.PubMedPubMedCentral Anthony JC, Echeagaray-Wagner F. Epidemiologic analysis of alcohol and tobacco use. Alcohol Res Health. 2000;24(4):201–8.PubMedPubMedCentral
146.
Zurück zum Zitat McKee SA, Harrison ELR, O’Malley SS, Krishnan-Sarin S, Shi J, Tetrault JM, et al. Varenicline reduces alcohol self-administration in heavy-drinking smokers. Biol Psychiatry. 2009;66(2):185–90.PubMedPubMedCentral McKee SA, Harrison ELR, O’Malley SS, Krishnan-Sarin S, Shi J, Tetrault JM, et al. Varenicline reduces alcohol self-administration in heavy-drinking smokers. Biol Psychiatry. 2009;66(2):185–90.PubMedPubMedCentral
147.
Zurück zum Zitat Falk DE, Castle I-JP, Ryan M, Fertig J, Litten RZ. Moderators of varenicline treatment effects in a double-blind, placebo-controlled trial for alcohol dependence: an exploratory analysis. J Addict Med. 2015;9(4):296–303.PubMedPubMedCentral Falk DE, Castle I-JP, Ryan M, Fertig J, Litten RZ. Moderators of varenicline treatment effects in a double-blind, placebo-controlled trial for alcohol dependence: an exploratory analysis. J Addict Med. 2015;9(4):296–303.PubMedPubMedCentral
148.
Zurück zum Zitat Donato S, Green R, Ray LA. Alcohol use disorder severity moderates clinical response to varenicline. Alcohol Clin Exp Res. 2021;45(9):1877–87.PubMed Donato S, Green R, Ray LA. Alcohol use disorder severity moderates clinical response to varenicline. Alcohol Clin Exp Res. 2021;45(9):1877–87.PubMed
149.
Zurück zum Zitat O’Malley SS, Zweben A, Fucito LM, Wu R, Piepmeier ME, Ockert DM, et al. Effect of varenicline combined with medical management on alcohol use disorder with comorbid cigarette smoking. JAMA Psychiat. 2018;75(2):129–38. O’Malley SS, Zweben A, Fucito LM, Wu R, Piepmeier ME, Ockert DM, et al. Effect of varenicline combined with medical management on alcohol use disorder with comorbid cigarette smoking. JAMA Psychiat. 2018;75(2):129–38.
150.
Zurück zum Zitat Aripiprazole PE. Aripiprazole. Am J Hosp Palliat Care. 2017;34(2):180–5. Aripiprazole PE. Aripiprazole. Am J Hosp Palliat Care. 2017;34(2):180–5.
151.
Zurück zum Zitat Shibasaki M, Kurokawa K, Mizuno K, Ohkuma S. Effect of aripiprazole on anxiety associated with ethanol physical dependence and on ethanol-induced place preference. J Pharmacol Sci. 2012;118(2):215–24.PubMed Shibasaki M, Kurokawa K, Mizuno K, Ohkuma S. Effect of aripiprazole on anxiety associated with ethanol physical dependence and on ethanol-induced place preference. J Pharmacol Sci. 2012;118(2):215–24.PubMed
152.
Zurück zum Zitat Nirogi R, Kandikere V, Jayarajan P, Bhyrapuneni G, Saralaya R, Muddana N, et al. Aripiprazole in an animal model of chronic alcohol consumption and dopamine D2 receptor occupancy in rats. Am J Drug Alcohol Abuse. 2013;39(2):72–9.PubMed Nirogi R, Kandikere V, Jayarajan P, Bhyrapuneni G, Saralaya R, Muddana N, et al. Aripiprazole in an animal model of chronic alcohol consumption and dopamine D2 receptor occupancy in rats. Am J Drug Alcohol Abuse. 2013;39(2):72–9.PubMed
153.
Zurück zum Zitat Ingman K, Kupila J, Hyytiä P, Korpi ER. Effects of aripiprazole on alcohol intake in an animal model of high-alcohol drinking. Alcohol Alcohol. 2006;41(4):391–8.PubMed Ingman K, Kupila J, Hyytiä P, Korpi ER. Effects of aripiprazole on alcohol intake in an animal model of high-alcohol drinking. Alcohol Alcohol. 2006;41(4):391–8.PubMed
154.
Zurück zum Zitat Kranzler HR, Covault J, Pierucci-Lagha A, Chan G, Douglas K, Arias AJ, et al. Effects of aripiprazole on subjective and physiological responses to alcohol. Alcohol Clin Exp Res. 2008;32(4):573–9.PubMedPubMedCentral Kranzler HR, Covault J, Pierucci-Lagha A, Chan G, Douglas K, Arias AJ, et al. Effects of aripiprazole on subjective and physiological responses to alcohol. Alcohol Clin Exp Res. 2008;32(4):573–9.PubMedPubMedCentral
155.
Zurück zum Zitat Myrick H, Li X, Randall PK, Henderson S, Voronin K, Anton RF. The effect of aripiprazole on cue-induced brain activation and drinking parameters in alcoholics. J Clin Psychopharmacol. 2010;30(4):365–72.PubMedPubMedCentral Myrick H, Li X, Randall PK, Henderson S, Voronin K, Anton RF. The effect of aripiprazole on cue-induced brain activation and drinking parameters in alcoholics. J Clin Psychopharmacol. 2010;30(4):365–72.PubMedPubMedCentral
156.
Zurück zum Zitat Anton RF, Schacht JP, Voronin KE, Randall PK. Aripiprazole suppression of drinking in a clinical laboratory paradigm: influence of impulsivity and self-control. Alcohol Clin Exp Res. 2017;41(7):1370–80.PubMedPubMedCentral Anton RF, Schacht JP, Voronin KE, Randall PK. Aripiprazole suppression of drinking in a clinical laboratory paradigm: influence of impulsivity and self-control. Alcohol Clin Exp Res. 2017;41(7):1370–80.PubMedPubMedCentral
157.
Zurück zum Zitat Martinotti G, Di Nicola M, Di Giannantonio M, Janiri L. Aripiprazole in the treatment of patients with alcohol dependence: a double-blind, comparison trial vs. naltrexone. J Psychopharmacol. 2009;23(2):123–9.PubMed Martinotti G, Di Nicola M, Di Giannantonio M, Janiri L. Aripiprazole in the treatment of patients with alcohol dependence: a double-blind, comparison trial vs. naltrexone. J Psychopharmacol. 2009;23(2):123–9.PubMed
158.
Zurück zum Zitat Kenna GA, Leggio L, Swift RM. A safety and tolerability laboratory study of the combination of aripiprazole and topiramate in volunteers who drink alcohol. Hum Psychopharmacol. 2009;24(6):465–72.PubMed Kenna GA, Leggio L, Swift RM. A safety and tolerability laboratory study of the combination of aripiprazole and topiramate in volunteers who drink alcohol. Hum Psychopharmacol. 2009;24(6):465–72.PubMed
159.
Zurück zum Zitat Haass-Koffler CL, Goodyear K, Zywiak WH, Leggio L, Kenna GA, Swift RM. Comparing and combining topiramate and aripiprazole on alcohol-related outcomes in a human laboratory study. Alcohol Alcohol. 2018;53(3):268–76.PubMed Haass-Koffler CL, Goodyear K, Zywiak WH, Leggio L, Kenna GA, Swift RM. Comparing and combining topiramate and aripiprazole on alcohol-related outcomes in a human laboratory study. Alcohol Alcohol. 2018;53(3):268–76.PubMed
160.
Zurück zum Zitat Anton RF, Kranzler H, Breder C, Marcus RN, Carson WH, Han J. A randomized, multicenter, double-blind, placebo-controlled study of the efficacy and safety of aripiprazole for the treatment of alcohol dependence. J Clin Psychopharmacol. 2008;28(1):5–12.PubMed Anton RF, Kranzler H, Breder C, Marcus RN, Carson WH, Han J. A randomized, multicenter, double-blind, placebo-controlled study of the efficacy and safety of aripiprazole for the treatment of alcohol dependence. J Clin Psychopharmacol. 2008;28(1):5–12.PubMed
161.
Zurück zum Zitat Heitzmann E, Javelot H, Weiner L, Michel B. A case of aripiprazole-induced tardive dyskinesia with dramatic evolution. Case Rep Psychiatry. 2016;2016:7031245.PubMedPubMedCentral Heitzmann E, Javelot H, Weiner L, Michel B. A case of aripiprazole-induced tardive dyskinesia with dramatic evolution. Case Rep Psychiatry. 2016;2016:7031245.PubMedPubMedCentral
162.
Zurück zum Zitat Macaluso M, Flynn A, Preskorn S. Determining whether a definitive causal relationship exists between aripiprazole and tardive dyskinesia and/or dystonia in patients with major depressive disorder, part 4: case report data. J Psychiatr Pract. 2016;22(3):203–20.PubMed Macaluso M, Flynn A, Preskorn S. Determining whether a definitive causal relationship exists between aripiprazole and tardive dyskinesia and/or dystonia in patients with major depressive disorder, part 4: case report data. J Psychiatr Pract. 2016;22(3):203–20.PubMed
163.
Zurück zum Zitat Johnson BA, Ait-Daoud N. Neuropharmacological treatments for alcoholism: scientific basis and clinical findings. Psychopharmacology. 2000;149(4):327–44.PubMed Johnson BA, Ait-Daoud N. Neuropharmacological treatments for alcoholism: scientific basis and clinical findings. Psychopharmacology. 2000;149(4):327–44.PubMed
164.
Zurück zum Zitat Johnson BA. Update on neuropharmacological treatments for alcoholism: scientific basis and clinical findings. Biochem Pharmacol. 2008;75(1):34–56.PubMed Johnson BA. Update on neuropharmacological treatments for alcoholism: scientific basis and clinical findings. Biochem Pharmacol. 2008;75(1):34–56.PubMed
165.
Zurück zum Zitat Rodd ZA, Bell RL, Oster SM, Toalston JE, Pommer TJ, McBride WJ, et al. Serotonin-3 receptors in the posterior ventral tegmental area regulate ethanol self-administration of alcohol-preferring (P) rats. Alcohol. 2010;44(3):245–55.PubMedPubMedCentral Rodd ZA, Bell RL, Oster SM, Toalston JE, Pommer TJ, McBride WJ, et al. Serotonin-3 receptors in the posterior ventral tegmental area regulate ethanol self-administration of alcohol-preferring (P) rats. Alcohol. 2010;44(3):245–55.PubMedPubMedCentral
166.
Zurück zum Zitat Campbell AD, McBride WJ. Serotonin-3 receptor and ethanol-stimulated dopamine release in the nucleus accumbens. Pharmacol Biochem Behav. 1995;51(4):835–42.PubMed Campbell AD, McBride WJ. Serotonin-3 receptor and ethanol-stimulated dopamine release in the nucleus accumbens. Pharmacol Biochem Behav. 1995;51(4):835–42.PubMed
167.
Zurück zum Zitat Umathe SN, Bhutada PS, Raut VS, Jain NS, Mundhada YR. The 5-HT3 receptor antagonist, ondansetron, blocks the development and expression of ethanol-induced locomotor sensitization in mice. Behav Pharmacol. 2009;20(1):78–83.PubMed Umathe SN, Bhutada PS, Raut VS, Jain NS, Mundhada YR. The 5-HT3 receptor antagonist, ondansetron, blocks the development and expression of ethanol-induced locomotor sensitization in mice. Behav Pharmacol. 2009;20(1):78–83.PubMed
168.
Zurück zum Zitat Tomkins DM, Le AD, Sellers EM. Effect of the 5-HT3 antagonist ondansetron on voluntary ethanol intake in rats and mice maintained on a limited access procedure. Psychopharmacology. 1995;117(4):479–85.PubMed Tomkins DM, Le AD, Sellers EM. Effect of the 5-HT3 antagonist ondansetron on voluntary ethanol intake in rats and mice maintained on a limited access procedure. Psychopharmacology. 1995;117(4):479–85.PubMed
169.
Zurück zum Zitat Kostowski W, Bisaga A, Jankowska E, Krzaścik P. Studies on the effects of certain 5-HT-3 receptor antagonists on ethanol preference and withdrawal seizures in the rat. Pol J Pharmacol. 1994;46(3):133–7.PubMed Kostowski W, Bisaga A, Jankowska E, Krzaścik P. Studies on the effects of certain 5-HT-3 receptor antagonists on ethanol preference and withdrawal seizures in the rat. Pol J Pharmacol. 1994;46(3):133–7.PubMed
170.
Zurück zum Zitat Johnson BA, Ait-Daoud N, Prihoda TJ. Combining ondansetron and naltrexone effectively treats biologically predisposed alcoholics: from hypotheses to preliminary clinical evidence. Alcohol Clin Exp Res. 2000;24(5):737–42.PubMed Johnson BA, Ait-Daoud N, Prihoda TJ. Combining ondansetron and naltrexone effectively treats biologically predisposed alcoholics: from hypotheses to preliminary clinical evidence. Alcohol Clin Exp Res. 2000;24(5):737–42.PubMed
171.
Zurück zum Zitat Myrick H, Anton RF, Li X, Henderson S, Randall PK, Voronin K. Effect of naltrexone and ondansetron on alcohol cue-induced activation of the ventral striatum in alcohol-dependent people. Arch Gen Psychiatry. 2008;65(4):466–75.PubMedPubMedCentral Myrick H, Anton RF, Li X, Henderson S, Randall PK, Voronin K. Effect of naltrexone and ondansetron on alcohol cue-induced activation of the ventral striatum in alcohol-dependent people. Arch Gen Psychiatry. 2008;65(4):466–75.PubMedPubMedCentral
172.
Zurück zum Zitat Johnson BA, Roache JD, Javors MA, DiClemente CC, Cloninger CR, Prihoda TJ, et al. Ondansetron for reduction of drinking among biologically predisposed alcoholic patients: a randomized controlled trial. JAMA. 2000;284(8):963–71.PubMed Johnson BA, Roache JD, Javors MA, DiClemente CC, Cloninger CR, Prihoda TJ, et al. Ondansetron for reduction of drinking among biologically predisposed alcoholic patients: a randomized controlled trial. JAMA. 2000;284(8):963–71.PubMed
173.
Zurück zum Zitat Kranzler HR, Pierucci-Lagha A, Feinn R, Hernandez-Avila C. Effects of ondansetron in early- versus late-onset alcoholics: a prospective, open-label study. Alcohol Clin Exp Res. 2003;27(7):1150–5.PubMed Kranzler HR, Pierucci-Lagha A, Feinn R, Hernandez-Avila C. Effects of ondansetron in early- versus late-onset alcoholics: a prospective, open-label study. Alcohol Clin Exp Res. 2003;27(7):1150–5.PubMed
174.
Zurück zum Zitat Johnson BA, Roache JD, Ait-Daoud N, Zanca NA, Velazquez M. Ondansetron reduces the craving of biologically predisposed alcoholics. Psychopharmacology. 2002;160(4):408–13.PubMed Johnson BA, Roache JD, Ait-Daoud N, Zanca NA, Velazquez M. Ondansetron reduces the craving of biologically predisposed alcoholics. Psychopharmacology. 2002;160(4):408–13.PubMed
175.
Zurück zum Zitat Roila F, Tonato M, Basurto C, Bracarda S, Sassi M, Lupattelli M, et al. Ondansetron. Eur J Cancer. 1993;29A(Suppl 1):S16-21.PubMed Roila F, Tonato M, Basurto C, Bracarda S, Sassi M, Lupattelli M, et al. Ondansetron. Eur J Cancer. 1993;29A(Suppl 1):S16-21.PubMed
176.
Zurück zum Zitat Shen WW. Anticraving therapy for alcohol use disorder: a clinical review. Neuropsychopharmacol Rep. 2018;38(3):105–16.PubMedPubMedCentral Shen WW. Anticraving therapy for alcohol use disorder: a clinical review. Neuropsychopharmacol Rep. 2018;38(3):105–16.PubMedPubMedCentral
177.
Zurück zum Zitat Simms JA, Haass-Koffler CL, Bito-Onon J, Li R, Bartlett SE. Mifepristone in the central nucleus of the amygdala reduces yohimbine stress-induced reinstatement of ethanol-seeking. Neuropsychopharmacology. 2012;37(4):906–18.PubMed Simms JA, Haass-Koffler CL, Bito-Onon J, Li R, Bartlett SE. Mifepristone in the central nucleus of the amygdala reduces yohimbine stress-induced reinstatement of ethanol-seeking. Neuropsychopharmacology. 2012;37(4):906–18.PubMed
178.
Zurück zum Zitat Vendruscolo LF, Estey D, Goodell V, Macshane LG, Logrip ML, Schlosburg JE, et al. Glucocorticoid receptor antagonism decreases alcohol seeking in alcohol-dependent individuals. J Clin Invest. 2015;125(8):3193–7.PubMedPubMedCentral Vendruscolo LF, Estey D, Goodell V, Macshane LG, Logrip ML, Schlosburg JE, et al. Glucocorticoid receptor antagonism decreases alcohol seeking in alcohol-dependent individuals. J Clin Invest. 2015;125(8):3193–7.PubMedPubMedCentral
179.
Zurück zum Zitat Jimenez VA, Walter NAR, Shnitko TA, Newman N, Diem K, Vanderhooft L, et al. Mifepristone decreases chronic voluntary ethanol consumption in rhesus macaques. J Pharmacol Exp Ther. 2020;375(2):258–67.PubMedPubMedCentral Jimenez VA, Walter NAR, Shnitko TA, Newman N, Diem K, Vanderhooft L, et al. Mifepristone decreases chronic voluntary ethanol consumption in rhesus macaques. J Pharmacol Exp Ther. 2020;375(2):258–67.PubMedPubMedCentral
180.
Zurück zum Zitat Holtyn AF, Weerts EM. Evaluation of mifepristone effects on alcohol-seeking and self-administration in baboons. Exp Clin Psychopharmacol. 2019;27(3):227–35.PubMed Holtyn AF, Weerts EM. Evaluation of mifepristone effects on alcohol-seeking and self-administration in baboons. Exp Clin Psychopharmacol. 2019;27(3):227–35.PubMed
181.
Zurück zum Zitat Donoghue K, Rose A, Coulton S, Milward J, Reed K, Drummond C, et al. Double-blind, 12 month follow-up, placebo-controlled trial of mifepristone on cognition in alcoholics: the MIFCOG trial protocol. BMC Psychiatry. 2016;24(16):40. Donoghue K, Rose A, Coulton S, Milward J, Reed K, Drummond C, et al. Double-blind, 12 month follow-up, placebo-controlled trial of mifepristone on cognition in alcoholics: the MIFCOG trial protocol. BMC Psychiatry. 2016;24(16):40.
182.
Zurück zum Zitat Donoghue K, Rose A, Coulton S, Coleman R, Milward J, Philips T, et al. Double-blind, placebo-controlled trial of mifepristone on cognition and depression in alcohol dependence. Trials. 2020;21(1):796.PubMedPubMedCentral Donoghue K, Rose A, Coulton S, Coleman R, Milward J, Philips T, et al. Double-blind, placebo-controlled trial of mifepristone on cognition and depression in alcohol dependence. Trials. 2020;21(1):796.PubMedPubMedCentral
183.
Zurück zum Zitat Gibson LCD, Hastings SF, McPhee I, Clayton RA, Darroch CE, Mackenzie A, et al. The inhibitory profile of Ibudilast against the human phosphodiesterase enzyme family. Eur J Pharmacol. 2006;538(1–3):39–42.PubMed Gibson LCD, Hastings SF, McPhee I, Clayton RA, Darroch CE, Mackenzie A, et al. The inhibitory profile of Ibudilast against the human phosphodiesterase enzyme family. Eur J Pharmacol. 2006;538(1–3):39–42.PubMed
184.
Zurück zum Zitat Cho Y, Crichlow GV, Vermeire JJ, Leng L, Du X, Hodsdon ME, et al. Allosteric inhibition of macrophage migration inhibitory factor revealed by ibudilast. Proc Natl Acad Sci USA. 2010;107(25):11313–8.PubMedPubMedCentral Cho Y, Crichlow GV, Vermeire JJ, Leng L, Du X, Hodsdon ME, et al. Allosteric inhibition of macrophage migration inhibitory factor revealed by ibudilast. Proc Natl Acad Sci USA. 2010;107(25):11313–8.PubMedPubMedCentral
185.
Zurück zum Zitat Mizuno T, Kurotani T, Komatsu Y, Kawanokuchi J, Kato H, Mitsuma N, et al. Neuroprotective role of phosphodiesterase inhibitor ibudilast on neuronal cell death induced by activated microglia. Neuropharmacology. 2004;46(3):404–11.PubMed Mizuno T, Kurotani T, Komatsu Y, Kawanokuchi J, Kato H, Mitsuma N, et al. Neuroprotective role of phosphodiesterase inhibitor ibudilast on neuronal cell death induced by activated microglia. Neuropharmacology. 2004;46(3):404–11.PubMed
186.
Zurück zum Zitat Mayfield J, Harris RA. The neuroimmune basis of excessive alcohol consumption. Neuropsychopharmacology. 2017;42(1):376.PubMed Mayfield J, Harris RA. The neuroimmune basis of excessive alcohol consumption. Neuropsychopharmacology. 2017;42(1):376.PubMed
187.
Zurück zum Zitat Johnson K, Matsuda K, Iwaki Y. Ibudilast for the treatment of drug addiction and other neurological conditions. Clin Investig. 2014;4(3):269–79. Johnson K, Matsuda K, Iwaki Y. Ibudilast for the treatment of drug addiction and other neurological conditions. Clin Investig. 2014;4(3):269–79.
188.
Zurück zum Zitat Bell RL, Lopez MF, Cui C, Egli M, Johnson KW, Franklin KM, et al. Ibudilast reduces alcohol drinking in multiple animal models of alcohol dependence. Addict Biol. 2015;20(1):38–42.PubMed Bell RL, Lopez MF, Cui C, Egli M, Johnson KW, Franklin KM, et al. Ibudilast reduces alcohol drinking in multiple animal models of alcohol dependence. Addict Biol. 2015;20(1):38–42.PubMed
189.
Zurück zum Zitat Blednov YA, Benavidez JM, Black M, Harris RA. Inhibition of phosphodiesterase 4 reduces ethanol intake and preference in C57BL/6J mice. Front Neurosci. 2014;8:129.PubMedPubMedCentral Blednov YA, Benavidez JM, Black M, Harris RA. Inhibition of phosphodiesterase 4 reduces ethanol intake and preference in C57BL/6J mice. Front Neurosci. 2014;8:129.PubMedPubMedCentral
190.
Zurück zum Zitat Logrip ML, Vendruscolo LF, Schlosburg JE, Koob GF, Zorrilla EP. Phosphodiesterase 10A regulates alcohol and saccharin self-administration in rats. Neuropsychopharmacology. 2014;39(7):1722–31.PubMedPubMedCentral Logrip ML, Vendruscolo LF, Schlosburg JE, Koob GF, Zorrilla EP. Phosphodiesterase 10A regulates alcohol and saccharin self-administration in rats. Neuropsychopharmacology. 2014;39(7):1722–31.PubMedPubMedCentral
191.
Zurück zum Zitat Wen R-T, Zhang M, Qin W-J, Liu Q, Wang W-P, Lawrence AJ, et al. The phosphodiesterase-4 (PDE4) inhibitor rolipram decreases ethanol seeking and consumption in alcohol-preferring Fawn-Hooded rats. Alcohol Clin Exp Res. 2012;36(12):2157–67.PubMedPubMedCentral Wen R-T, Zhang M, Qin W-J, Liu Q, Wang W-P, Lawrence AJ, et al. The phosphodiesterase-4 (PDE4) inhibitor rolipram decreases ethanol seeking and consumption in alcohol-preferring Fawn-Hooded rats. Alcohol Clin Exp Res. 2012;36(12):2157–67.PubMedPubMedCentral
192.
Zurück zum Zitat Ray LA, Bujarski S, Shoptaw S, Roche DJ, Heinzerling K, Miotto K. Development of the neuroimmune modulator ibudilast for the treatment of alcoholism: a randomized, placebo-controlled, human laboratory trial. Neuropsychopharmacology. 2017;42(9):1776–88.PubMedPubMedCentral Ray LA, Bujarski S, Shoptaw S, Roche DJ, Heinzerling K, Miotto K. Development of the neuroimmune modulator ibudilast for the treatment of alcoholism: a randomized, placebo-controlled, human laboratory trial. Neuropsychopharmacology. 2017;42(9):1776–88.PubMedPubMedCentral
193.
Zurück zum Zitat Grodin EN, Bujarski S, Towns B, Burnette E, Nieto S, Lim A, et al. Ibudilast, a neuroimmune modulator, reduces heavy drinking and alcohol cue-elicited neural activation: a randomized trial. Transl Psychiatry. 2021;11(1):355.PubMedPubMedCentral Grodin EN, Bujarski S, Towns B, Burnette E, Nieto S, Lim A, et al. Ibudilast, a neuroimmune modulator, reduces heavy drinking and alcohol cue-elicited neural activation: a randomized trial. Transl Psychiatry. 2021;11(1):355.PubMedPubMedCentral
194.
Zurück zum Zitat Burnette EM, Ray LA, Irwin MR, Grodin EN. Ibudilast attenuates alcohol cue-elicited frontostriatal functional connectivity in alcohol use disorder. Alcohol Clin Exp Res. 2021;45(10):2017–28.PubMed Burnette EM, Ray LA, Irwin MR, Grodin EN. Ibudilast attenuates alcohol cue-elicited frontostriatal functional connectivity in alcohol use disorder. Alcohol Clin Exp Res. 2021;45(10):2017–28.PubMed
195.
Zurück zum Zitat Rolan P, Gibbons JA, He L, Chang E, Jones D, Gross MI, et al. Ibudilast in healthy volunteers: safety, tolerability and pharmacokinetics with single and multiple doses. Br J Clin Pharmacol. 2008;66(6):792–801.PubMedPubMedCentral Rolan P, Gibbons JA, He L, Chang E, Jones D, Gross MI, et al. Ibudilast in healthy volunteers: safety, tolerability and pharmacokinetics with single and multiple doses. Br J Clin Pharmacol. 2008;66(6):792–801.PubMedPubMedCentral
196.
Zurück zum Zitat Fox RJ, Coffey CS, Conwit R, Cudkowicz ME, Gleason T, Goodman A, et al. Phase 2 trial of ibudilast in progressive multiple sclerosis. N Engl J Med. 2018;379(9):846–55.PubMedPubMedCentral Fox RJ, Coffey CS, Conwit R, Cudkowicz ME, Gleason T, Goodman A, et al. Phase 2 trial of ibudilast in progressive multiple sclerosis. N Engl J Med. 2018;379(9):846–55.PubMedPubMedCentral
197.
Zurück zum Zitat Sommermeyer H, Frielingsdorf J, Knorr A. Effects of prazosin on the dopaminergic neurotransmission in rat brain. Eur J Pharmacol. 1995;276(3):267–70.PubMed Sommermeyer H, Frielingsdorf J, Knorr A. Effects of prazosin on the dopaminergic neurotransmission in rat brain. Eur J Pharmacol. 1995;276(3):267–70.PubMed
198.
Zurück zum Zitat Leggio L, Kenna GA. Doxasozin for alcoholism. Alcohol Clin Exp Res. 2013;37(2):191–3.PubMed Leggio L, Kenna GA. Doxasozin for alcoholism. Alcohol Clin Exp Res. 2013;37(2):191–3.PubMed
199.
Zurück zum Zitat Perez DM. α1-Adrenergic receptors in neurotransmission, synaptic plasticity, and cognition. Front Pharmacol. 2020;11:581098.PubMedPubMedCentral Perez DM. α1-Adrenergic receptors in neurotransmission, synaptic plasticity, and cognition. Front Pharmacol. 2020;11:581098.PubMedPubMedCentral
200.
Zurück zum Zitat Haass-Koffler CL, Swift RM, Leggio L. Noradrenergic targets for the treatment of alcohol use disorder. Psychopharmacology. 2018;235(6):1625–34.PubMedPubMedCentral Haass-Koffler CL, Swift RM, Leggio L. Noradrenergic targets for the treatment of alcohol use disorder. Psychopharmacology. 2018;235(6):1625–34.PubMedPubMedCentral
201.
Zurück zum Zitat Walker BM, Rasmussen DD, Raskind MA, Koob GF. alpha1-noradrenergic receptor antagonism blocks dependence-induced increases in responding for ethanol. Alcohol. 2008;42(2):91–7.PubMedPubMedCentral Walker BM, Rasmussen DD, Raskind MA, Koob GF. alpha1-noradrenergic receptor antagonism blocks dependence-induced increases in responding for ethanol. Alcohol. 2008;42(2):91–7.PubMedPubMedCentral
202.
Zurück zum Zitat Funk D, Coen K, Tamadon S, Li Z, Loughlin A, Lê AD. Effects of prazosin and doxazosin on yohimbine-induced reinstatement of alcohol seeking in rats. Psychopharmacology. 2016;233(11):2197–207.PubMed Funk D, Coen K, Tamadon S, Li Z, Loughlin A, Lê AD. Effects of prazosin and doxazosin on yohimbine-induced reinstatement of alcohol seeking in rats. Psychopharmacology. 2016;233(11):2197–207.PubMed
203.
Zurück zum Zitat Froehlich JC, Hausauer B, Fischer S, Wise B, Rasmussen DD. Prazosin reduces alcohol intake in an animal model of alcohol relapse. Alcohol Clin Exp Res. 2015;39(8):1538–46.PubMedPubMedCentral Froehlich JC, Hausauer B, Fischer S, Wise B, Rasmussen DD. Prazosin reduces alcohol intake in an animal model of alcohol relapse. Alcohol Clin Exp Res. 2015;39(8):1538–46.PubMedPubMedCentral
204.
Zurück zum Zitat O’Neil ML, Beckwith LE, Kincaid CL, Rasmussen DD. The α1-adrenergic receptor antagonist, doxazosin, reduces alcohol drinking in alcohol-preferring (P) rats. Alcohol Clin Exp Res. 2013;37(2):202–12.PubMed O’Neil ML, Beckwith LE, Kincaid CL, Rasmussen DD. The α1-adrenergic receptor antagonist, doxazosin, reduces alcohol drinking in alcohol-preferring (P) rats. Alcohol Clin Exp Res. 2013;37(2):202–12.PubMed
205.
Zurück zum Zitat Lopez MF, Reasons SE, Carper BA, Nolen TL, Williams RL, Becker HC. Evaluation of the effect of doxasozin and zonisamide on voluntary ethanol intake in mice that experienced chronic intermittent ethanol exposure and stress. Alcohol. 2020;89:37–42.PubMedPubMedCentral Lopez MF, Reasons SE, Carper BA, Nolen TL, Williams RL, Becker HC. Evaluation of the effect of doxasozin and zonisamide on voluntary ethanol intake in mice that experienced chronic intermittent ethanol exposure and stress. Alcohol. 2020;89:37–42.PubMedPubMedCentral
206.
Zurück zum Zitat Simpson TL, Saxon AJ, Meredith CW, Malte CA, McBride B, Ferguson LC, et al. A pilot trial of the alpha-1 adrenergic antagonist, prazosin, for alcohol dependence. Alcohol Clin Exp Res. 2009;33(2):255–63.PubMed Simpson TL, Saxon AJ, Meredith CW, Malte CA, McBride B, Ferguson LC, et al. A pilot trial of the alpha-1 adrenergic antagonist, prazosin, for alcohol dependence. Alcohol Clin Exp Res. 2009;33(2):255–63.PubMed
207.
Zurück zum Zitat Simpson TL, Saxon AJ, Stappenbeck C, Malte CA, Lyons R, Tell D, et al. Double-blind randomized clinical trial of the α−1 noradrenergic antagonist prazosin for alcohol use disorder. Am J Psychiatry. 2018;175(12):1216–24.PubMedPubMedCentral Simpson TL, Saxon AJ, Stappenbeck C, Malte CA, Lyons R, Tell D, et al. Double-blind randomized clinical trial of the α−1 noradrenergic antagonist prazosin for alcohol use disorder. Am J Psychiatry. 2018;175(12):1216–24.PubMedPubMedCentral
208.
Zurück zum Zitat Kenna GA, Haass-Koffler CL, Zywiak WH, Edwards SM, Brickley MB, Swift RM, et al. Role of the α1 blocker doxazosin in alcoholism: a proof-of-concept randomized controlled trial. Addict Biol. 2016;21(4):904–14.PubMed Kenna GA, Haass-Koffler CL, Zywiak WH, Edwards SM, Brickley MB, Swift RM, et al. Role of the α1 blocker doxazosin in alcoholism: a proof-of-concept randomized controlled trial. Addict Biol. 2016;21(4):904–14.PubMed
209.
Zurück zum Zitat Haass-Koffler CL, Goodyear K, Zywiak WH, Magill M, Eltinge SE, Wallace PM, et al. Higher pretreatment blood pressure is associated with greater alcohol drinking reduction in alcohol-dependent individuals treated with doxazosin. Drug Alcohol Depend. 2017;1(177):23–8. Haass-Koffler CL, Goodyear K, Zywiak WH, Magill M, Eltinge SE, Wallace PM, et al. Higher pretreatment blood pressure is associated with greater alcohol drinking reduction in alcohol-dependent individuals treated with doxazosin. Drug Alcohol Depend. 2017;1(177):23–8.
211.
Zurück zum Zitat Vanderkam P, Solinas M, Ingrand I, Doux N, Ebrahimighavam S, Jaafari N, et al. Effectiveness of drugs acting on adrenergic receptors in the treatment for tobacco or alcohol use disorders: systematic review and meta-analysis. Addiction. 2021;116(5):1011–20.PubMed Vanderkam P, Solinas M, Ingrand I, Doux N, Ebrahimighavam S, Jaafari N, et al. Effectiveness of drugs acting on adrenergic receptors in the treatment for tobacco or alcohol use disorders: systematic review and meta-analysis. Addiction. 2021;116(5):1011–20.PubMed
212.
Zurück zum Zitat Stoops WW, Strickland JC, Hays LR, Rayapati AO, Lile JA, Rush CR. Influence of n-acetylcysteine maintenance on the pharmacodynamic effects of oral ethanol. Pharmacol Biochem Behav. 2020;198:173037.PubMedPubMedCentral Stoops WW, Strickland JC, Hays LR, Rayapati AO, Lile JA, Rush CR. Influence of n-acetylcysteine maintenance on the pharmacodynamic effects of oral ethanol. Pharmacol Biochem Behav. 2020;198:173037.PubMedPubMedCentral
213.
Zurück zum Zitat Chen X, Michaelis ML, Michaelis EK. Effects of chronic ethanol treatment on the expression of calcium transport carriers and NMDA/glutamate receptor proteins in brain synaptic membranes. J Neurochem. 1997;69(4):1559–69.PubMed Chen X, Michaelis ML, Michaelis EK. Effects of chronic ethanol treatment on the expression of calcium transport carriers and NMDA/glutamate receptor proteins in brain synaptic membranes. J Neurochem. 1997;69(4):1559–69.PubMed
214.
Zurück zum Zitat Dahchour A, De Witte P. Excitatory and inhibitory amino acid changes during repeated episodes of ethanol withdrawal: an in vivo microdialysis study. Eur J Pharmacol. 2003;459(2–3):171–8.PubMed Dahchour A, De Witte P. Excitatory and inhibitory amino acid changes during repeated episodes of ethanol withdrawal: an in vivo microdialysis study. Eur J Pharmacol. 2003;459(2–3):171–8.PubMed
215.
Zurück zum Zitat Lebourgeois S, González-Marín MC, Jeanblanc J, Naassila M, Vilpoux C. Effect of N-acetylcysteine on motivation, seeking and relapse to ethanol self-administration. Addict Biol. 2018;23(2):643–52.PubMed Lebourgeois S, González-Marín MC, Jeanblanc J, Naassila M, Vilpoux C. Effect of N-acetylcysteine on motivation, seeking and relapse to ethanol self-administration. Addict Biol. 2018;23(2):643–52.PubMed
216.
Zurück zum Zitat Lebourgeois S, González-Marín MC, Antol J, Naassila M, Vilpoux C. Evaluation of N-acetylcysteine on ethanol self-administration in ethanol-dependent rats. Neuropharmacology. 2019;15(150):112–20. Lebourgeois S, González-Marín MC, Antol J, Naassila M, Vilpoux C. Evaluation of N-acetylcysteine on ethanol self-administration in ethanol-dependent rats. Neuropharmacology. 2019;15(150):112–20.
217.
Zurück zum Zitat Mocelin R, Marcon M, da Rosa Araujo AS, Herrmann AP, Piato A. Withdrawal effects following repeated ethanol exposure are prevented by N-acetylcysteine in zebrafish. Prog Neuropsychopharmacol Biol Psychiatry. 2019;13(93):161–70. Mocelin R, Marcon M, da Rosa Araujo AS, Herrmann AP, Piato A. Withdrawal effects following repeated ethanol exposure are prevented by N-acetylcysteine in zebrafish. Prog Neuropsychopharmacol Biol Psychiatry. 2019;13(93):161–70.
218.
Zurück zum Zitat Morais-Silva G, Alves GC, Marin MT. N-Acetylcysteine treatment blocks the development of ethanol-induced behavioural sensitization and related ΔFosB alterations. Neuropharmacology. 2016;110(Pt A):135–42.PubMed Morais-Silva G, Alves GC, Marin MT. N-Acetylcysteine treatment blocks the development of ethanol-induced behavioural sensitization and related ΔFosB alterations. Neuropharmacology. 2016;110(Pt A):135–42.PubMed
219.
Zurück zum Zitat Schneider R, Santos CF, Clarimundo V, Dalmaz C, Elisabetsky E, Gomez R. N-Acetylcysteine prevents behavioral and biochemical changes induced by alcohol cessation in rats. Alcohol. 2015;49(3):259–63.PubMed Schneider R, Santos CF, Clarimundo V, Dalmaz C, Elisabetsky E, Gomez R. N-Acetylcysteine prevents behavioral and biochemical changes induced by alcohol cessation in rats. Alcohol. 2015;49(3):259–63.PubMed
220.
Zurück zum Zitat Schneider R, Bandiera S, Souza DG, Bellaver B, Caletti G, Quincozes-Santos A, et al. N-Acetylcysteine prevents alcohol related neuroinflammation in rats. Neurochem Res. 2017;42(8):2135–41.PubMed Schneider R, Bandiera S, Souza DG, Bellaver B, Caletti G, Quincozes-Santos A, et al. N-Acetylcysteine prevents alcohol related neuroinflammation in rats. Neurochem Res. 2017;42(8):2135–41.PubMed
221.
Zurück zum Zitat Israel Y, Quintanilla ME, Ezquer F, Morales P, Santapau D, Berríos-Cárcamo P, Ezquer M, Olivares B, Herrera-Marschitz M. Aspirin and N-acetylcysteine co-administration markedly inhibit chronic ethanol intake and block relapse binge drinking: Role of neuroinflammation-oxidative stress self-perpetuation. Addict Biol. 2021;26(1):e12853.PubMed Israel Y, Quintanilla ME, Ezquer F, Morales P, Santapau D, Berríos-Cárcamo P, Ezquer M, Olivares B, Herrera-Marschitz M. Aspirin and N-acetylcysteine co-administration markedly inhibit chronic ethanol intake and block relapse binge drinking: Role of neuroinflammation-oxidative stress self-perpetuation. Addict Biol. 2021;26(1):e12853.PubMed
222.
Zurück zum Zitat Duailibi MS, Cordeiro Q, Brietzke E, Ribeiro M, LaRowe S, Berk M, et al. N-Acetylcysteine in the treatment of craving in substance use disorders: Systematic review and meta-analysis. Am J Addict. 2017;26(7):660–6.PubMed Duailibi MS, Cordeiro Q, Brietzke E, Ribeiro M, LaRowe S, Berk M, et al. N-Acetylcysteine in the treatment of craving in substance use disorders: Systematic review and meta-analysis. Am J Addict. 2017;26(7):660–6.PubMed
223.
Zurück zum Zitat Squeglia LM, Tomko RL, Baker NL, McClure EA, Book GA, Gray KM. The effect of N-acetylcysteine on alcohol use during a cannabis cessation trial. Drug Alcohol Depend. 2018;01(185):17–22. Squeglia LM, Tomko RL, Baker NL, McClure EA, Book GA, Gray KM. The effect of N-acetylcysteine on alcohol use during a cannabis cessation trial. Drug Alcohol Depend. 2018;01(185):17–22.
224.
Zurück zum Zitat Back SE, Gray K, Ana ES, Jones JL, Jarnecke AM, Joseph JE, et al. N-Acetylcysteine for the treatment of comorbid alcohol use disorder and posttraumatic stress disorder: design and methodology of a randomized clinical trial. Contemp Clin Trials. 2020;91:105961.PubMedPubMedCentral Back SE, Gray K, Ana ES, Jones JL, Jarnecke AM, Joseph JE, et al. N-Acetylcysteine for the treatment of comorbid alcohol use disorder and posttraumatic stress disorder: design and methodology of a randomized clinical trial. Contemp Clin Trials. 2020;91:105961.PubMedPubMedCentral
225.
Zurück zum Zitat Hammond CJ. The role of pharmacotherapy in the treatment of adolescent substance use disorders. Child Adolesc Psychiatr Clin N Am. 2016;25(4):685–711.PubMedPubMedCentral Hammond CJ. The role of pharmacotherapy in the treatment of adolescent substance use disorders. Child Adolesc Psychiatr Clin N Am. 2016;25(4):685–711.PubMedPubMedCentral
226.
Zurück zum Zitat Walker LC, Ch’ng SS, Lawrence AJ. Role of lateral hypothalamic orexin (hypocretin) neurons in alcohol use and abuse: recent advances. Curr Pharmacol Rep. 2016;2(6):241–52. Walker LC, Ch’ng SS, Lawrence AJ. Role of lateral hypothalamic orexin (hypocretin) neurons in alcohol use and abuse: recent advances. Curr Pharmacol Rep. 2016;2(6):241–52.
227.
Zurück zum Zitat Korotkova TM, Sergeeva OA, Eriksson KS, Haas HL, Brown RE. Excitation of ventral tegmental area dopaminergic and nondopaminergic neurons by orexins/hypocretins. J Neurosci. 2003;23(1):7–11.PubMedPubMedCentral Korotkova TM, Sergeeva OA, Eriksson KS, Haas HL, Brown RE. Excitation of ventral tegmental area dopaminergic and nondopaminergic neurons by orexins/hypocretins. J Neurosci. 2003;23(1):7–11.PubMedPubMedCentral
228.
Zurück zum Zitat Nakamura T, Uramura K, Nambu T, Yada T, Goto K, Yanagisawa M, et al. Orexin-induced hyperlocomotion and stereotypy are mediated by the dopaminergic system. Brain Res. 2000;873(1):181–7.PubMed Nakamura T, Uramura K, Nambu T, Yada T, Goto K, Yanagisawa M, et al. Orexin-induced hyperlocomotion and stereotypy are mediated by the dopaminergic system. Brain Res. 2000;873(1):181–7.PubMed
229.
Zurück zum Zitat Lei K, Kwok C, Darevsky D, Wegner SA, Yu J, Nakayama L, et al. Nucleus accumbens shell orexin-1 receptors are critical mediators of binge intake in excessive-drinking individuals. Front Neurosci. 2019;13:88.PubMedPubMedCentral Lei K, Kwok C, Darevsky D, Wegner SA, Yu J, Nakayama L, et al. Nucleus accumbens shell orexin-1 receptors are critical mediators of binge intake in excessive-drinking individuals. Front Neurosci. 2019;13:88.PubMedPubMedCentral
230.
Zurück zum Zitat Moorman DE, James MH, Kilroy EA, Aston-Jones G. Orexin/hypocretin-1 receptor antagonism reduces ethanol self-administration and reinstatement selectively in highly-motivated rats. Brain Res. 2017;1654(Pt A):34–42.PubMed Moorman DE, James MH, Kilroy EA, Aston-Jones G. Orexin/hypocretin-1 receptor antagonism reduces ethanol self-administration and reinstatement selectively in highly-motivated rats. Brain Res. 2017;1654(Pt A):34–42.PubMed
231.
Zurück zum Zitat Brown RM, Kim AK, Khoo SY-S, Kim JH, Jupp B, Lawrence AJ. Orexin-1 receptor signalling in the prelimbic cortex and ventral tegmental area regulates cue-induced reinstatement of ethanol-seeking in iP rats. Addict Biol. 2016;21(3):603–12. Brown RM, Kim AK, Khoo SY-S, Kim JH, Jupp B, Lawrence AJ. Orexin-1 receptor signalling in the prelimbic cortex and ventral tegmental area regulates cue-induced reinstatement of ethanol-seeking in iP rats. Addict Biol. 2016;21(3):603–12.
232.
Zurück zum Zitat Ubaldi M, Giordano A, Severi I, Li H, Kallupi M, de Guglielmo G, et al. Activation of hypocretin-1/orexin-a neurons projecting to the bed nucleus of the stria terminalis and paraventricular nucleus is critical for reinstatement of alcohol seeking by neuropeptide S. Biol Psychiatry. 2016;79(6):452–62.PubMed Ubaldi M, Giordano A, Severi I, Li H, Kallupi M, de Guglielmo G, et al. Activation of hypocretin-1/orexin-a neurons projecting to the bed nucleus of the stria terminalis and paraventricular nucleus is critical for reinstatement of alcohol seeking by neuropeptide S. Biol Psychiatry. 2016;79(6):452–62.PubMed
233.
Zurück zum Zitat Lawrence AJ, Cowen MS, Yang H-J, Chen F, Oldfield B. The orexin system regulates alcohol-seeking in rats. Br J Pharmacol. 2006;148(6):752–9.PubMedPubMedCentral Lawrence AJ, Cowen MS, Yang H-J, Chen F, Oldfield B. The orexin system regulates alcohol-seeking in rats. Br J Pharmacol. 2006;148(6):752–9.PubMedPubMedCentral
234.
Zurück zum Zitat Shoblock JR, Welty N, Aluisio L, Fraser I, Motley ST, Morton K, et al. Selective blockade of the orexin-2 receptor attenuates ethanol self-administration, place preference, and reinstatement. Psychopharmacology. 2011;215(1):191–203.PubMed Shoblock JR, Welty N, Aluisio L, Fraser I, Motley ST, Morton K, et al. Selective blockade of the orexin-2 receptor attenuates ethanol self-administration, place preference, and reinstatement. Psychopharmacology. 2011;215(1):191–203.PubMed
235.
Zurück zum Zitat Brown RM, Khoo SY-S, Lawrence AJ. Central orexin (hypocretin) 2 receptor antagonism reduces ethanol self-administration, but not cue-conditioned ethanol-seeking, in ethanol-preferring rats. Int J Neuropsychopharmacol. 2013;16(9):2067–79.PubMed Brown RM, Khoo SY-S, Lawrence AJ. Central orexin (hypocretin) 2 receptor antagonism reduces ethanol self-administration, but not cue-conditioned ethanol-seeking, in ethanol-preferring rats. Int J Neuropsychopharmacol. 2013;16(9):2067–79.PubMed
236.
Zurück zum Zitat Barson JR, Ho HT, Leibowitz SF. Anterior thalamic paraventricular nucleus is involved in intermittent access ethanol drinking: role of orexin receptor 2. Addict Biol. 2015;20(3):469–81.PubMed Barson JR, Ho HT, Leibowitz SF. Anterior thalamic paraventricular nucleus is involved in intermittent access ethanol drinking: role of orexin receptor 2. Addict Biol. 2015;20(3):469–81.PubMed
237.
Zurück zum Zitat Anderson RI, Becker HC, Adams BL, Jesudason CD, Rorick-Kehn LM. Orexin-1 and orexin-2 receptor antagonists reduce ethanol self-administration in high-drinking rodent models. Front Neurosci. 2014;8:33.PubMedPubMedCentral Anderson RI, Becker HC, Adams BL, Jesudason CD, Rorick-Kehn LM. Orexin-1 and orexin-2 receptor antagonists reduce ethanol self-administration in high-drinking rodent models. Front Neurosci. 2014;8:33.PubMedPubMedCentral
238.
Zurück zum Zitat Srinivasan S, Simms JA, Nielsen CK, Lieske SP, Bito-Onon JJ, Yi H, et al. The dual orexin/hypocretin receptor antagonist, almorexant, in the ventral tegmental area attenuates ethanol self-administration. PLoS ONE. 2012;7(9):e44726.PubMedPubMedCentral Srinivasan S, Simms JA, Nielsen CK, Lieske SP, Bito-Onon JJ, Yi H, et al. The dual orexin/hypocretin receptor antagonist, almorexant, in the ventral tegmental area attenuates ethanol self-administration. PLoS ONE. 2012;7(9):e44726.PubMedPubMedCentral
239.
Zurück zum Zitat Campbell EJ, Norman A, Bonomo Y, Lawrence AJ. Suvorexant to treat alcohol use disorder and comorbid insomnia: plan for a phase II trial. Brain Res. 2020;1(1728):146597. Campbell EJ, Norman A, Bonomo Y, Lawrence AJ. Suvorexant to treat alcohol use disorder and comorbid insomnia: plan for a phase II trial. Brain Res. 2020;1(1728):146597.
240.
Zurück zum Zitat Sun H, Yee KL, Gill S, Liu W, Li X, Panebianco D, et al. Psychomotor effects, pharmacokinetics and safety of the orexin receptor antagonist suvorexant administered in combination with alcohol in healthy subjects. J Psychopharmacol. 2015;29(11):1159–69.PubMed Sun H, Yee KL, Gill S, Liu W, Li X, Panebianco D, et al. Psychomotor effects, pharmacokinetics and safety of the orexin receptor antagonist suvorexant administered in combination with alcohol in healthy subjects. J Psychopharmacol. 2015;29(11):1159–69.PubMed
241.
Zurück zum Zitat Zhou Y, Colombo G, Carai MAM, Ho A, Gessa GL, Kreek MJ. Involvement of arginine vasopressin and V1b receptor in alcohol drinking in Sardinian alcohol-preferring rats. Alcohol Clin Exp Res. 2011;35(10):1876–83.PubMedPubMedCentral Zhou Y, Colombo G, Carai MAM, Ho A, Gessa GL, Kreek MJ. Involvement of arginine vasopressin and V1b receptor in alcohol drinking in Sardinian alcohol-preferring rats. Alcohol Clin Exp Res. 2011;35(10):1876–83.PubMedPubMedCentral
242.
Zurück zum Zitat Edwards S, Guerrero M, Ghoneim OM, Roberts E, Koob GF. Evidence that vasopressin V1b receptors mediate the transition to excessive drinking in ethanol-dependent rats. Addict Biol. 2012;17(1):76–85.PubMed Edwards S, Guerrero M, Ghoneim OM, Roberts E, Koob GF. Evidence that vasopressin V1b receptors mediate the transition to excessive drinking in ethanol-dependent rats. Addict Biol. 2012;17(1):76–85.PubMed
243.
Zurück zum Zitat Katz DA, Locke C, Liu W, Zhang J, Achari R, Wesnes KA, et al. Single-dose interaction study of the arginine vasopressin type 1B receptor antagonist ABT-436 and alcohol in moderate alcohol drinkers. Alcohol Clin Exp Res. 2016;40(4):838–45.PubMed Katz DA, Locke C, Liu W, Zhang J, Achari R, Wesnes KA, et al. Single-dose interaction study of the arginine vasopressin type 1B receptor antagonist ABT-436 and alcohol in moderate alcohol drinkers. Alcohol Clin Exp Res. 2016;40(4):838–45.PubMed
244.
Zurück zum Zitat Ryan ML, Falk DE, Fertig JB, Rendenbach-Mueller B, Katz DA, Tracy KA, et al. A phase 2, double-blind, placebo-controlled randomized trial assessing the efficacy of ABT-436, a novel V1b receptor antagonist, for alcohol dependence. Neuropsychopharmacology. 2017;42(5):1012–23.PubMed Ryan ML, Falk DE, Fertig JB, Rendenbach-Mueller B, Katz DA, Tracy KA, et al. A phase 2, double-blind, placebo-controlled randomized trial assessing the efficacy of ABT-436, a novel V1b receptor antagonist, for alcohol dependence. Neuropsychopharmacology. 2017;42(5):1012–23.PubMed
245.
Zurück zum Zitat Tomasini MC, Borelli AC, Beggiato S, Tanganelli S, Loche A, Cacciaglia R, et al. GET73 prevents ethanol-induced neurotoxicity in primary cultures of rat hippocampal neurons. Alcohol Alcohol. 2016;51(2):128–35.PubMed Tomasini MC, Borelli AC, Beggiato S, Tanganelli S, Loche A, Cacciaglia R, et al. GET73 prevents ethanol-induced neurotoxicity in primary cultures of rat hippocampal neurons. Alcohol Alcohol. 2016;51(2):128–35.PubMed
246.
Zurück zum Zitat Loche A, Simonetti F, Lobina C, Carai MAM, Colombo G, Castelli MP, et al. Anti-alcohol and anxiolytic properties of a new chemical entity, GET73. Front Psychiatry. 2012;3:8.PubMedPubMedCentral Loche A, Simonetti F, Lobina C, Carai MAM, Colombo G, Castelli MP, et al. Anti-alcohol and anxiolytic properties of a new chemical entity, GET73. Front Psychiatry. 2012;3:8.PubMedPubMedCentral
247.
Zurück zum Zitat Colombo G, Carai MAM, Gessa GL. Suppression by γ-hydroxybutyric acid of “alcohol deprivation effect” in rats: preclinical evidence of its anti-relapse properties. Front Psychiatry. 2012;3:95.PubMedPubMedCentral Colombo G, Carai MAM, Gessa GL. Suppression by γ-hydroxybutyric acid of “alcohol deprivation effect” in rats: preclinical evidence of its anti-relapse properties. Front Psychiatry. 2012;3:95.PubMedPubMedCentral
248.
Zurück zum Zitat Beggiato S, Borelli AC, Tomasini MC, Castelli MP, Pintori N, Cacciaglia R, et al. In vitro functional characterization of GET73 as possible negative allosteric modulator of metabotropic glutamate receptor 5. Front Pharmacol. 2018;9:327.PubMedPubMedCentral Beggiato S, Borelli AC, Tomasini MC, Castelli MP, Pintori N, Cacciaglia R, et al. In vitro functional characterization of GET73 as possible negative allosteric modulator of metabotropic glutamate receptor 5. Front Pharmacol. 2018;9:327.PubMedPubMedCentral
249.
Zurück zum Zitat Haass-Koffler CL, Goodyear K, Loche A, Long VM, Lobina C, Tran HH, et al. Administration of the metabotropic glutamate receptor subtype 5 allosteric modulator GET 73 with alcohol: a translational study in rats and humans. J Psychopharmacol. 2018;32(2):163–73.PubMedPubMedCentral Haass-Koffler CL, Goodyear K, Loche A, Long VM, Lobina C, Tran HH, et al. Administration of the metabotropic glutamate receptor subtype 5 allosteric modulator GET 73 with alcohol: a translational study in rats and humans. J Psychopharmacol. 2018;32(2):163–73.PubMedPubMedCentral
250.
Zurück zum Zitat Haass-Koffler CL, Goodyear K, Long VM, Tran HH, Loche A, Cacciaglia R, et al. A phase I randomized clinical trial testing the safety, tolerability and preliminary pharmacokinetics of the mGluR5 negative allosteric modulator GET 73 following single and repeated doses in healthy volunteers. Eur J Pharm Sci. 2017;15(109):78–85. Haass-Koffler CL, Goodyear K, Long VM, Tran HH, Loche A, Cacciaglia R, et al. A phase I randomized clinical trial testing the safety, tolerability and preliminary pharmacokinetics of the mGluR5 negative allosteric modulator GET 73 following single and repeated doses in healthy volunteers. Eur J Pharm Sci. 2017;15(109):78–85.
251.
Zurück zum Zitat Haass-Koffler CL, Perciballi R, Magill M, et al. An inpatient human laboratory study assessing the safety and tolerability, pharmacokinetics, and biobehavioral effect of GET 73 when co-administered with alcohol in individuals with alcohol use disorder. Psychopharmacology (Berl). 2022;239:35–46. Haass-Koffler CL, Perciballi R, Magill M, et al. An inpatient human laboratory study assessing the safety and tolerability, pharmacokinetics, and biobehavioral effect of GET 73 when co-administered with alcohol in individuals with alcohol use disorder. Psychopharmacology (Berl). 2022;239:35–46.
252.
Zurück zum Zitat Maccioni P, Colombo G. Potential of gabab receptor positive allosteric modulators in the treatment of alcohol use disorder. CNS Drugs. 2019;33(2):107–23.PubMed Maccioni P, Colombo G. Potential of gabab receptor positive allosteric modulators in the treatment of alcohol use disorder. CNS Drugs. 2019;33(2):107–23.PubMed
253.
Zurück zum Zitat Urwyler S. Allosteric modulation of family C G-protein-coupled receptors: from molecular insights to therapeutic perspectives. Pharmacol Rev. 2011;63(1):59–126.PubMed Urwyler S. Allosteric modulation of family C G-protein-coupled receptors: from molecular insights to therapeutic perspectives. Pharmacol Rev. 2011;63(1):59–126.PubMed
254.
Zurück zum Zitat Gjoni T, Desrayaud S, Imobersteg S, Urwyler S. The positive allosteric modulator GS39783 enhances GABA(B) receptor-mediated inhibition of cyclic AMP formation in rat striatum in vivo. J Neurochem. 2006;96(5):1416–22.PubMed Gjoni T, Desrayaud S, Imobersteg S, Urwyler S. The positive allosteric modulator GS39783 enhances GABA(B) receptor-mediated inhibition of cyclic AMP formation in rat striatum in vivo. J Neurochem. 2006;96(5):1416–22.PubMed
255.
Zurück zum Zitat Agabio R, Colombo G. GABAB receptor ligands for the treatment of alcohol use disorder: preclinical and clinical evidence. Front Neurosci. 2014;8:140.PubMedPubMedCentral Agabio R, Colombo G. GABAB receptor ligands for the treatment of alcohol use disorder: preclinical and clinical evidence. Front Neurosci. 2014;8:140.PubMedPubMedCentral
256.
Zurück zum Zitat Augier E, Dulman RS, Damadzic R, Pilling A, Hamilton JP, Heilig M. The GABAB positive allosteric modulator ADX71441 attenuates alcohol self-administration and relapse to alcohol seeking in rats. Neuropsychopharmacology. 2017;42(9):1789–99.PubMedPubMedCentral Augier E, Dulman RS, Damadzic R, Pilling A, Hamilton JP, Heilig M. The GABAB positive allosteric modulator ADX71441 attenuates alcohol self-administration and relapse to alcohol seeking in rats. Neuropsychopharmacology. 2017;42(9):1789–99.PubMedPubMedCentral
257.
Zurück zum Zitat Vengeliene V, Takahashi TT, Dravolina OA, Belozertseva I, Zvartau E, Bespalov AY, et al. Efficacy and side effects of baclofen and the novel GABAB receptor positive allosteric modulator CMPPE in animal models for alcohol and cocaine addiction. Psychopharmacology. 2018;235(7):1955–65.PubMed Vengeliene V, Takahashi TT, Dravolina OA, Belozertseva I, Zvartau E, Bespalov AY, et al. Efficacy and side effects of baclofen and the novel GABAB receptor positive allosteric modulator CMPPE in animal models for alcohol and cocaine addiction. Psychopharmacology. 2018;235(7):1955–65.PubMed
258.
Zurück zum Zitat Maccioni P, Colombo G, Lorrai I, Zaru A, Carai MAM, Gessa GL, et al. Suppressing effect of COR659 on alcohol, sucrose, and chocolate self-administration in rats: involvement of the GABAB and cannabinoid CB1 receptors. Psychopharmacology. 2017;234(17):2525–43.PubMed Maccioni P, Colombo G, Lorrai I, Zaru A, Carai MAM, Gessa GL, et al. Suppressing effect of COR659 on alcohol, sucrose, and chocolate self-administration in rats: involvement of the GABAB and cannabinoid CB1 receptors. Psychopharmacology. 2017;234(17):2525–43.PubMed
259.
Zurück zum Zitat Murai N, Kondo Y, Akuzawa S, Mihara T, Shiraishi N, Kakimoto S, et al. A novel GABAB receptor positive allosteric modulator, ASP8062, exerts analgesic effects in a rat model of fibromyalgia. Eur J Pharmacol. 2019;865:172750.PubMed Murai N, Kondo Y, Akuzawa S, Mihara T, Shiraishi N, Kakimoto S, et al. A novel GABAB receptor positive allosteric modulator, ASP8062, exerts analgesic effects in a rat model of fibromyalgia. Eur J Pharmacol. 2019;865:172750.PubMed
260.
Zurück zum Zitat Walzer M, Marek GJ, Wu R, Nagata M, Han D. Single- and multiple-dose safety, tolerability, and pharmacokinetic profiles of ASP8062: results from 2 phase 1 studies. Clin Pharmacol Drug Dev. 2020;9(3):297–306.PubMed Walzer M, Marek GJ, Wu R, Nagata M, Han D. Single- and multiple-dose safety, tolerability, and pharmacokinetic profiles of ASP8062: results from 2 phase 1 studies. Clin Pharmacol Drug Dev. 2020;9(3):297–306.PubMed
261.
Zurück zum Zitat Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999;402(6762):656–60.PubMed Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999;402(6762):656–60.PubMed
262.
Zurück zum Zitat Müller TD, Nogueiras R, Andermann ML, Andrews ZB, Anker SD, Argente J, et al. Ghrelin. Mol Metab. 2015;4(6):437–60.PubMedPubMedCentral Müller TD, Nogueiras R, Andermann ML, Andrews ZB, Anker SD, Argente J, et al. Ghrelin. Mol Metab. 2015;4(6):437–60.PubMedPubMedCentral
263.
Zurück zum Zitat Leggio L. Role of the ghrelin system in alcoholism: acting on the growth hormone secretagogue receptor to treat alcohol-related diseases. Drug News Perspect. 2010;23(3):157–66.PubMed Leggio L. Role of the ghrelin system in alcoholism: acting on the growth hormone secretagogue receptor to treat alcohol-related diseases. Drug News Perspect. 2010;23(3):157–66.PubMed
264.
Zurück zum Zitat Leggio L, Zywiak WH, Fricchione SR, Edwards SM, de la Monte SM, Swift RM, et al. Intravenous ghrelin administration increases alcohol craving in alcohol-dependent heavy drinkers: a preliminary investigation. Biol Psychiatry. 2014;76(9):734–41.PubMedPubMedCentral Leggio L, Zywiak WH, Fricchione SR, Edwards SM, de la Monte SM, Swift RM, et al. Intravenous ghrelin administration increases alcohol craving in alcohol-dependent heavy drinkers: a preliminary investigation. Biol Psychiatry. 2014;76(9):734–41.PubMedPubMedCentral
265.
Zurück zum Zitat Farokhnia M, Grodin EN, Lee MR, Oot EN, Blackburn AN, Stangl BL, et al. Exogenous ghrelin administration increases alcohol self-administration and modulates brain functional activity in heavy-drinking alcohol-dependent individuals. Mol Psychiatry. 2018;23(10):2029–38.PubMed Farokhnia M, Grodin EN, Lee MR, Oot EN, Blackburn AN, Stangl BL, et al. Exogenous ghrelin administration increases alcohol self-administration and modulates brain functional activity in heavy-drinking alcohol-dependent individuals. Mol Psychiatry. 2018;23(10):2029–38.PubMed
266.
Zurück zum Zitat Jerlhag E, Egecioglu E, Landgren S, Salomé N, Heilig M, Moechars D, et al. Requirement of central ghrelin signaling for alcohol reward. Proc Natl Acad Sci USA. 2009;106(27):11318–23.PubMedPubMedCentral Jerlhag E, Egecioglu E, Landgren S, Salomé N, Heilig M, Moechars D, et al. Requirement of central ghrelin signaling for alcohol reward. Proc Natl Acad Sci USA. 2009;106(27):11318–23.PubMedPubMedCentral
267.
Zurück zum Zitat Kaur S, Ryabinin AE. Ghrelin receptor antagonism decreases alcohol consumption and activation of perioculomotor urocortin-containing neurons. Alcohol Clin Exp Res. 2010;34(9):1525–34.PubMedPubMedCentral Kaur S, Ryabinin AE. Ghrelin receptor antagonism decreases alcohol consumption and activation of perioculomotor urocortin-containing neurons. Alcohol Clin Exp Res. 2010;34(9):1525–34.PubMedPubMedCentral
268.
Zurück zum Zitat Gomez JL, Cunningham CL, Finn DA, Young EA, Helpenstell LK, Schuette LM, et al. Differential effects of ghrelin antagonists on alcohol drinking and reinforcement in mouse and rat models of alcohol dependence. Neuropharmacology. 2015;97:182–93.PubMedPubMedCentral Gomez JL, Cunningham CL, Finn DA, Young EA, Helpenstell LK, Schuette LM, et al. Differential effects of ghrelin antagonists on alcohol drinking and reinforcement in mouse and rat models of alcohol dependence. Neuropharmacology. 2015;97:182–93.PubMedPubMedCentral
269.
Zurück zum Zitat Landgren S, Simms JA, Hyytiä P, Engel JA, Bartlett SE, Jerlhag E. Ghrelin receptor (GHS-R1A) antagonism suppresses both operant alcohol self-administration and high alcohol consumption in rats. Addict Biol. 2012;17(1):86–94.PubMed Landgren S, Simms JA, Hyytiä P, Engel JA, Bartlett SE, Jerlhag E. Ghrelin receptor (GHS-R1A) antagonism suppresses both operant alcohol self-administration and high alcohol consumption in rats. Addict Biol. 2012;17(1):86–94.PubMed
270.
Zurück zum Zitat Stevenson JR, Francomacaro LM, Bohidar AE, Young KA, Pesarchick BF, Buirkle JM, et al. Ghrelin receptor (GHS-R1A) antagonism alters preference for ethanol and sucrose in a concentration-dependent manner in prairie voles. Physiol Behav. 2016;1(155):231–6. Stevenson JR, Francomacaro LM, Bohidar AE, Young KA, Pesarchick BF, Buirkle JM, et al. Ghrelin receptor (GHS-R1A) antagonism alters preference for ethanol and sucrose in a concentration-dependent manner in prairie voles. Physiol Behav. 2016;1(155):231–6.
271.
Zurück zum Zitat Kong J, Chuddy J, Stock IA, Loria PM, Straub SV, Vage C, et al. Pharmacological characterization of the first in class clinical candidate PF-05190457: a selective ghrelin receptor competitive antagonist with inverse agonism that increases vagal afferent firing and glucose-dependent insulin secretion ex vivo. Br J Pharmacol. 2016;173(9):1452–64.PubMedPubMedCentral Kong J, Chuddy J, Stock IA, Loria PM, Straub SV, Vage C, et al. Pharmacological characterization of the first in class clinical candidate PF-05190457: a selective ghrelin receptor competitive antagonist with inverse agonism that increases vagal afferent firing and glucose-dependent insulin secretion ex vivo. Br J Pharmacol. 2016;173(9):1452–64.PubMedPubMedCentral
273.
Zurück zum Zitat Denney WS, Sonnenberg GE, Carvajal-Gonzalez S, Tuthill T, Jackson VM. Pharmacokinetics and pharmacodynamics of PF-05190457: the first oral ghrelin receptor inverse agonist to be profiled in healthy subjects. Br J Clin Pharmacol. 2017;83(2):326–38.PubMed Denney WS, Sonnenberg GE, Carvajal-Gonzalez S, Tuthill T, Jackson VM. Pharmacokinetics and pharmacodynamics of PF-05190457: the first oral ghrelin receptor inverse agonist to be profiled in healthy subjects. Br J Clin Pharmacol. 2017;83(2):326–38.PubMed
274.
Zurück zum Zitat Laprairie RB, Bagher AM, Kelly MEM, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br J Pharmacol. 2015;172(20):4790–805.PubMedPubMedCentral Laprairie RB, Bagher AM, Kelly MEM, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br J Pharmacol. 2015;172(20):4790–805.PubMedPubMedCentral
275.
Zurück zum Zitat Martínez-Pinilla E, Varani K, Reyes-Resina I, Angelats E, F F, Ferreiro-Vera C, et al. Binding and signaling studies disclose a potential allosteric site for cannabidiol in cannabinoid CB 2 receptors. Frontiers in pharmacology [Internet]. 2017 [cited 2021 Aug 30];8. https://pubmed.ncbi.nlm.nih.gov/29109685/. Martínez-Pinilla E, Varani K, Reyes-Resina I, Angelats E, F F, Ferreiro-Vera C, et al. Binding and signaling studies disclose a potential allosteric site for cannabidiol in cannabinoid CB 2 receptors. Frontiers in pharmacology [Internet]. 2017 [cited 2021 Aug 30];8. https://​pubmed.​ncbi.​nlm.​nih.​gov/​29109685/​.
276.
Zurück zum Zitat Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br J Pharmacol. 2008;153(2):199–215.PubMed Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br J Pharmacol. 2008;153(2):199–215.PubMed
277.
Zurück zum Zitat Russo EB, Burnett A, Hall B, Parker KK. Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochem Res. 2005;30(8):1037–43.PubMed Russo EB, Burnett A, Hall B, Parker KK. Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochem Res. 2005;30(8):1037–43.PubMed
278.
Zurück zum Zitat Kathmann M, Flau K, Redmer A, Tränkle C, Schlicker E. Cannabidiol is an allosteric modulator at mu- and delta-opioid receptors. Naunyn Schmiedebergs Arch Pharmacol. 2006;372(5):354–61.PubMed Kathmann M, Flau K, Redmer A, Tränkle C, Schlicker E. Cannabidiol is an allosteric modulator at mu- and delta-opioid receptors. Naunyn Schmiedebergs Arch Pharmacol. 2006;372(5):354–61.PubMed
279.
Zurück zum Zitat De Ternay J, Naassila M, Nourredine M, Louvet A, Bailly F, Sescousse G, et al. Therapeutic prospects of cannabidiol for alcohol use disorder and alcohol-related damages on the liver and the brain. Front Pharmacol. 2019;10:627.PubMedPubMedCentral De Ternay J, Naassila M, Nourredine M, Louvet A, Bailly F, Sescousse G, et al. Therapeutic prospects of cannabidiol for alcohol use disorder and alcohol-related damages on the liver and the brain. Front Pharmacol. 2019;10:627.PubMedPubMedCentral
280.
Zurück zum Zitat Turna J, Syan SK, Frey BN, Rush B, Costello MJ, Weiss M, et al. Cannabidiol as a novel candidate alcohol use disorder pharmacotherapy: a systematic review. Alcohol Clin Exp Res. 2019;43(4):550–63.PubMedPubMedCentral Turna J, Syan SK, Frey BN, Rush B, Costello MJ, Weiss M, et al. Cannabidiol as a novel candidate alcohol use disorder pharmacotherapy: a systematic review. Alcohol Clin Exp Res. 2019;43(4):550–63.PubMedPubMedCentral
281.
Zurück zum Zitat Hurd YL, Spriggs S, Alishayev J, Winkel G, Gurgov K, Kudrich C, et al. Cannabidiol for the reduction of cue-induced craving and anxiety in drug-abstinent individuals with heroin use disorder: a double-blind randomized placebo-controlled trial. Am J Psychiatry. 2019;176(11):911–22.PubMed Hurd YL, Spriggs S, Alishayev J, Winkel G, Gurgov K, Kudrich C, et al. Cannabidiol for the reduction of cue-induced craving and anxiety in drug-abstinent individuals with heroin use disorder: a double-blind randomized placebo-controlled trial. Am J Psychiatry. 2019;176(11):911–22.PubMed
282.
Zurück zum Zitat Freeman TP, Hindocha C, Baio G, Shaban NDC, Thomas EM, Astbury D, et al. Cannabidiol for the treatment of cannabis use disorder: a phase 2a, double-blind, placebo-controlled, randomised, adaptive Bayesian trial. Lancet Psychiatry. 2020;7(10):865–74.PubMedPubMedCentral Freeman TP, Hindocha C, Baio G, Shaban NDC, Thomas EM, Astbury D, et al. Cannabidiol for the treatment of cannabis use disorder: a phase 2a, double-blind, placebo-controlled, randomised, adaptive Bayesian trial. Lancet Psychiatry. 2020;7(10):865–74.PubMedPubMedCentral
283.
Zurück zum Zitat Litten RZ, Falk DE, Ryan ML, Fertig JB. Discovery, development, and adoption of medications to treat alcohol use disorder: goals for the phases of medications development. Alcohol Clin Exp Res. 2016;40(7):1368–79.PubMedPubMedCentral Litten RZ, Falk DE, Ryan ML, Fertig JB. Discovery, development, and adoption of medications to treat alcohol use disorder: goals for the phases of medications development. Alcohol Clin Exp Res. 2016;40(7):1368–79.PubMedPubMedCentral
284.
Zurück zum Zitat Venegas A, Donato S, Meredith LR, Ray LA. Understanding low treatment seeking rates for alcohol use disorder: A narrative review of the literature and opportunities for improvement. Am J Drug Alcohol Abuse. 2021;31:1–16. Venegas A, Donato S, Meredith LR, Ray LA. Understanding low treatment seeking rates for alcohol use disorder: A narrative review of the literature and opportunities for improvement. Am J Drug Alcohol Abuse. 2021;31:1–16.
285.
Zurück zum Zitat Volkow ND, Gordon JA, Koob GF. Choosing appropriate language to reduce the stigma around mental illness and substance use disorders. Neuropsychopharmacology. 2021;46(13):2230–2.PubMed Volkow ND, Gordon JA, Koob GF. Choosing appropriate language to reduce the stigma around mental illness and substance use disorders. Neuropsychopharmacology. 2021;46(13):2230–2.PubMed
286.
Zurück zum Zitat Falk DE, O’Malley SS, Witkiewitz K, Anton RF, Litten RZ, Slater M, et al. Evaluation of drinking risk levels as outcomes in alcohol pharmacotherapy trials: a secondary analysis of 3 randomized clinical trials. JAMA Psychiat. 2019;76(4):374–81. Falk DE, O’Malley SS, Witkiewitz K, Anton RF, Litten RZ, Slater M, et al. Evaluation of drinking risk levels as outcomes in alcohol pharmacotherapy trials: a secondary analysis of 3 randomized clinical trials. JAMA Psychiat. 2019;76(4):374–81.
287.
Zurück zum Zitat Witkiewitz K, Kranzler HR, Hallgren KA, O’Malley SS, Falk DE, Litten RZ, et al. Drinking risk level reductions associated with improvements in physical health and quality of life among individuals with alcohol use disorder. Alcohol Clin Exp Res. 2018;42(12):2453–65.PubMedPubMedCentral Witkiewitz K, Kranzler HR, Hallgren KA, O’Malley SS, Falk DE, Litten RZ, et al. Drinking risk level reductions associated with improvements in physical health and quality of life among individuals with alcohol use disorder. Alcohol Clin Exp Res. 2018;42(12):2453–65.PubMedPubMedCentral
288.
Zurück zum Zitat Hasin DS, Wall M, Witkiewitz K, Kranzler HR, Falk D, Litten R, et al. Change in non-abstinent WHO drinking risk levels and alcohol dependence: a 3-year follow-up study in the US general population. Lancet Psychiatry. 2017;4(6):469–76.PubMedPubMedCentral Hasin DS, Wall M, Witkiewitz K, Kranzler HR, Falk D, Litten R, et al. Change in non-abstinent WHO drinking risk levels and alcohol dependence: a 3-year follow-up study in the US general population. Lancet Psychiatry. 2017;4(6):469–76.PubMedPubMedCentral
289.
Zurück zum Zitat Mann K, Vollstädt-Klein S, Reinhard I, Leménager T, Fauth-Bühler M, Hermann D, et al. Predicting naltrexone response in alcohol-dependent patients: the contribution of functional magnetic resonance imaging. Alcohol Clin Exp Res. 2014;38(11):2754–62.PubMed Mann K, Vollstädt-Klein S, Reinhard I, Leménager T, Fauth-Bühler M, Hermann D, et al. Predicting naltrexone response in alcohol-dependent patients: the contribution of functional magnetic resonance imaging. Alcohol Clin Exp Res. 2014;38(11):2754–62.PubMed
290.
Zurück zum Zitat Witkiewitz K, Roos CR, Mann K, Kranzler HR. Advancing precision medicine for alcohol use disorder: replication and extension of reward drinking as a predictor of naltrexone response. Alcohol Clin Exp Res. 2019;43(11):2395–405.PubMedPubMedCentral Witkiewitz K, Roos CR, Mann K, Kranzler HR. Advancing precision medicine for alcohol use disorder: replication and extension of reward drinking as a predictor of naltrexone response. Alcohol Clin Exp Res. 2019;43(11):2395–405.PubMedPubMedCentral
291.
Zurück zum Zitat Litten RZ, Falk DE, Ryan ML, Fertig J, Leggio L. Advances in pharmacotherapy development: human clinical studies. Handb Exp Pharmacol. 2018;248:579–613.PubMed Litten RZ, Falk DE, Ryan ML, Fertig J, Leggio L. Advances in pharmacotherapy development: human clinical studies. Handb Exp Pharmacol. 2018;248:579–613.PubMed
292.
Zurück zum Zitat Litten RZ, Ryan ML, Falk DE, Reilly M, Fertig JB, Koob GF. Heterogeneity of alcohol use disorder: understanding mechanisms to advance personalized treatment. Alcohol Clin Exp Res. 2015;39(4):579–84.PubMed Litten RZ, Ryan ML, Falk DE, Reilly M, Fertig JB, Koob GF. Heterogeneity of alcohol use disorder: understanding mechanisms to advance personalized treatment. Alcohol Clin Exp Res. 2015;39(4):579–84.PubMed
293.
Zurück zum Zitat Agabio R, Pisanu C, Gessa GL, Franconi F. Sex differences in alcohol use disorder. Curr Med Chem. 2017;24(24):2661–70.PubMed Agabio R, Pisanu C, Gessa GL, Franconi F. Sex differences in alcohol use disorder. Curr Med Chem. 2017;24(24):2661–70.PubMed
294.
Zurück zum Zitat Heilig M, Leggio L. What the alcohol doctor ordered from the neuroscientist: theragnostic biomarkers for personalized treatments. Prog Brain Res. 2016;224:401–18.PubMed Heilig M, Leggio L. What the alcohol doctor ordered from the neuroscientist: theragnostic biomarkers for personalized treatments. Prog Brain Res. 2016;224:401–18.PubMed
295.
Zurück zum Zitat Castillo-Carniglia A, Keyes KM, Hasin DS, Cerdá M. Psychiatric comorbidities in alcohol use disorder. Lancet Psychiatry. 2019;6(12):1068–80.PubMedPubMedCentral Castillo-Carniglia A, Keyes KM, Hasin DS, Cerdá M. Psychiatric comorbidities in alcohol use disorder. Lancet Psychiatry. 2019;6(12):1068–80.PubMedPubMedCentral
296.
Zurück zum Zitat Leggio L, Lee MR. Treatment of alcohol use disorder in patients with alcoholic liver disease. Am J Med. 2017;130(2):124–34.PubMed Leggio L, Lee MR. Treatment of alcohol use disorder in patients with alcoholic liver disease. Am J Med. 2017;130(2):124–34.PubMed
297.
Zurück zum Zitat Guillot A, Ren T, Jourdan T, Pawlosky RJ, Han E, Kim S-J, et al. Targeting liver aldehyde dehydrogenase-2 prevents heavy but not moderate alcohol drinking. Proc Natl Acad Sci USA. 2019;116(51):25974–81.PubMedPubMedCentral Guillot A, Ren T, Jourdan T, Pawlosky RJ, Han E, Kim S-J, et al. Targeting liver aldehyde dehydrogenase-2 prevents heavy but not moderate alcohol drinking. Proc Natl Acad Sci USA. 2019;116(51):25974–81.PubMedPubMedCentral
298.
Zurück zum Zitat Tampier L, Quintanilla ME, Israel Y. Tolerance to disulfiram induced by chronic alcohol intake in the rat. Alcohol Clin Exp Res. 2008;32(6):937–41.PubMed Tampier L, Quintanilla ME, Israel Y. Tolerance to disulfiram induced by chronic alcohol intake in the rat. Alcohol Clin Exp Res. 2008;32(6):937–41.PubMed
299.
Zurück zum Zitat Spanagel R, Hölter SM, Allingham K, Landgraf R, Zieglgänsberger W. Acamprosate and alcohol: I. Effects on alcohol intake following alcohol deprivation in the rat. Eur J Pharmacol. 1996;305(1–3):39–44.PubMed Spanagel R, Hölter SM, Allingham K, Landgraf R, Zieglgänsberger W. Acamprosate and alcohol: I. Effects on alcohol intake following alcohol deprivation in the rat. Eur J Pharmacol. 1996;305(1–3):39–44.PubMed
300.
Zurück zum Zitat Spanagel R, Putzke J, Stefferl A, Schöbitz B, Zieglgänsberger W. Acamprosate and alcohol: II. Effects on alcohol withdrawal in the rat. Eur J Pharmacol. 1996;305(1–3):45–50.PubMed Spanagel R, Putzke J, Stefferl A, Schöbitz B, Zieglgänsberger W. Acamprosate and alcohol: II. Effects on alcohol withdrawal in the rat. Eur J Pharmacol. 1996;305(1–3):45–50.PubMed
301.
Zurück zum Zitat Chester JA, Grahame NJ, Li TK, Lumeng L, Froehlich JC. Effects of acamprosate on sensitization to the locomotor-stimulant effects of alcohol in mice selectively bred for high and low alcohol preference. Behav Pharmacol. 2001;12(6–7):535–43.PubMed Chester JA, Grahame NJ, Li TK, Lumeng L, Froehlich JC. Effects of acamprosate on sensitization to the locomotor-stimulant effects of alcohol in mice selectively bred for high and low alcohol preference. Behav Pharmacol. 2001;12(6–7):535–43.PubMed
302.
Zurück zum Zitat Kurokawa K, Mizuno K, Shibasaki M, Higashioka M, Oka M, Hirouchi M, et al. Acamprosate suppresses ethanol-induced place preference in mice with ethanol physical dependence. J Pharmacol Sci. 2013;122(4):289–98.PubMed Kurokawa K, Mizuno K, Shibasaki M, Higashioka M, Oka M, Hirouchi M, et al. Acamprosate suppresses ethanol-induced place preference in mice with ethanol physical dependence. J Pharmacol Sci. 2013;122(4):289–98.PubMed
303.
Zurück zum Zitat Jacobsen JHW, Buisman-Pijlman FTA, Mustafa S, Rice KC, Hutchinson MR. The efficacy of (+)-Naltrexone on alcohol preference and seeking behaviour is dependent on light-cycle. Brain Behav Immun. 2018;1(67):181–93. Jacobsen JHW, Buisman-Pijlman FTA, Mustafa S, Rice KC, Hutchinson MR. The efficacy of (+)-Naltrexone on alcohol preference and seeking behaviour is dependent on light-cycle. Brain Behav Immun. 2018;1(67):181–93.
304.
Zurück zum Zitat Jacobsen JHW, Buisman-Pijlman FT, Mustafa S, Rice KC, Hutchinson MR. Antagonising TLR4-TRIF signalling before or after a low-dose alcohol binge during adolescence prevents alcohol drinking but not seeking behaviour in adulthood. Neuropharmacology. 2018;1(128):460–73. Jacobsen JHW, Buisman-Pijlman FT, Mustafa S, Rice KC, Hutchinson MR. Antagonising TLR4-TRIF signalling before or after a low-dose alcohol binge during adolescence prevents alcohol drinking but not seeking behaviour in adulthood. Neuropharmacology. 2018;1(128):460–73.
305.
Zurück zum Zitat Wu Y, Lousberg EL, Moldenhauer LM, Hayball JD, Coller JK, Rice KC, et al. Inhibiting the TLR4-MyD88 signalling cascade by genetic or pharmacological strategies reduces acute alcohol-induced sedation and motor impairment in mice. Br J Pharmacol. 2012;165(5):1319–29.PubMedPubMedCentral Wu Y, Lousberg EL, Moldenhauer LM, Hayball JD, Coller JK, Rice KC, et al. Inhibiting the TLR4-MyD88 signalling cascade by genetic or pharmacological strategies reduces acute alcohol-induced sedation and motor impairment in mice. Br J Pharmacol. 2012;165(5):1319–29.PubMedPubMedCentral
306.
Zurück zum Zitat Montesinos J, Gil A, Guerri C. nalmefene prevents alcohol-induced neuroinflammation and alcohol drinking preference in adolescent female mice: role of TLR4. Alcohol Clin Exp Res. 2017;41(7):1257–70.PubMed Montesinos J, Gil A, Guerri C. nalmefene prevents alcohol-induced neuroinflammation and alcohol drinking preference in adolescent female mice: role of TLR4. Alcohol Clin Exp Res. 2017;41(7):1257–70.PubMed
307.
Zurück zum Zitat Calleja-Conde J, Echeverry-Alzate V, Giné E, Bühler K-M, Nadal R, Maldonado R, et al. Nalmefene is effective at reducing alcohol seeking, treating alcohol-cocaine interactions and reducing alcohol-induced histone deacetylases gene expression in blood. Br J Pharmacol. 2016;173(16):2490–505.PubMedPubMedCentral Calleja-Conde J, Echeverry-Alzate V, Giné E, Bühler K-M, Nadal R, Maldonado R, et al. Nalmefene is effective at reducing alcohol seeking, treating alcohol-cocaine interactions and reducing alcohol-induced histone deacetylases gene expression in blood. Br J Pharmacol. 2016;173(16):2490–505.PubMedPubMedCentral
308.
Zurück zum Zitat Foo JC, Vengeliene V, Noori HR, Yamaguchi I, Morita K, Nakamura T, et al. drinking levels and profiles of alcohol addicted rats predict response to nalmefene. Front Pharmacol. 2019;10:471.PubMedPubMedCentral Foo JC, Vengeliene V, Noori HR, Yamaguchi I, Morita K, Nakamura T, et al. drinking levels and profiles of alcohol addicted rats predict response to nalmefene. Front Pharmacol. 2019;10:471.PubMedPubMedCentral
309.
Zurück zum Zitat Colombo G, Gessa GL. Suppressing effect of baclofen on multiple alcohol-related behaviors in laboratory animals. Front Psychiatry. 2018;9:475.PubMedPubMedCentral Colombo G, Gessa GL. Suppressing effect of baclofen on multiple alcohol-related behaviors in laboratory animals. Front Psychiatry. 2018;9:475.PubMedPubMedCentral
310.
Zurück zum Zitat Colombo G, Vacca G, Serra S, Brunetti G, Carai MAM, Gessa GL. Baclofen suppresses motivation to consume alcohol in rats. Psychopharmacology. 2003;167(3):221–4.PubMed Colombo G, Vacca G, Serra S, Brunetti G, Carai MAM, Gessa GL. Baclofen suppresses motivation to consume alcohol in rats. Psychopharmacology. 2003;167(3):221–4.PubMed
311.
Zurück zum Zitat Maccioni P, Lorrai I, Contini A, Leite-Morris K, Colombo G. Microinjection of baclofen and CGP7930 into the ventral tegmental area suppresses alcohol self-administration in alcohol-preferring rats. Neuropharmacology. 2018;136(Pt A):146–58.PubMed Maccioni P, Lorrai I, Contini A, Leite-Morris K, Colombo G. Microinjection of baclofen and CGP7930 into the ventral tegmental area suppresses alcohol self-administration in alcohol-preferring rats. Neuropharmacology. 2018;136(Pt A):146–58.PubMed
312.
Zurück zum Zitat Colombo G, Addolorato G, Agabio R, Carai MAM, Pibiri F, Serra S, et al. Role of GABA(B) receptor in alcohol dependence: reducing effect of baclofen on alcohol intake and alcohol motivational properties in rats and amelioration of alcohol withdrawal syndrome and alcohol craving in human alcoholics. Neurotox Res. 2004;6(5):403–14.PubMed Colombo G, Addolorato G, Agabio R, Carai MAM, Pibiri F, Serra S, et al. Role of GABA(B) receptor in alcohol dependence: reducing effect of baclofen on alcohol intake and alcohol motivational properties in rats and amelioration of alcohol withdrawal syndrome and alcohol craving in human alcoholics. Neurotox Res. 2004;6(5):403–14.PubMed
313.
Zurück zum Zitat Maccioni P, Pes D, Fantini N, Carai MAM, Gessa GL, Colombo G. gamma-Hydroxybutyric acid (GHB) suppresses alcohol’s motivational properties in alcohol-preferring rats. Alcohol. 2008;42(2):107–13.PubMed Maccioni P, Pes D, Fantini N, Carai MAM, Gessa GL, Colombo G. gamma-Hydroxybutyric acid (GHB) suppresses alcohol’s motivational properties in alcohol-preferring rats. Alcohol. 2008;42(2):107–13.PubMed
Metadaten
Titel
Novel Agents for the Pharmacological Treatment of Alcohol Use Disorder
verfasst von
Elizabeth M. Burnette
Steven J. Nieto
Erica N. Grodin
Lindsay R. Meredith
Brian Hurley
Karen Miotto
Artha J. Gillis
Lara A. Ray
Publikationsdatum
01.02.2022
Verlag
Springer International Publishing
Erschienen in
Drugs / Ausgabe 3/2022
Print ISSN: 0012-6667
Elektronische ISSN: 1179-1950
DOI
https://doi.org/10.1007/s40265-021-01670-3

Weitere Artikel der Ausgabe 3/2022

Drugs 3/2022 Zur Ausgabe

AdisInsight Report

Maribavir: First Approval