Open access peer-reviewed chapter

Dopamine and Alcohol Dependence: From Bench to Clinic

Written By

Nitya Jayaram‐Lindström, Mia Ericson, Pia Steensland and Elisabet Jerlhag

Reviewed: 16 March 2016 Published: 14 July 2016

DOI: 10.5772/63144

From the Edited Volume

Recent Advances in Drug Addiction Research and Clinical Applications

Edited by William M. Meil and Christina L. Ruby

Chapter metrics overview

2,237 Chapter Downloads

View Full Metrics

Abstract

Alcohol dependence, a chronic relapsing psychiatric disorder, is a major cause of mortality and morbidity. The role of dopamine in alcohol‐induced reward as well in the development of alcohol dependence is reviewed herein. Both preclinical and clinical studies have suggested that alcohol activates the mesolimbic dopamine system (defined as a dopamine projection from the ventral tegmental area (VTA) to the nucleus accumbens (NAc, i.e. ventral striatum)) leading to a euphoric sensation. Alcohol dependence is characterized by a disruption in the reward‐related brain areas including fewer dopamine D2 receptors in ventral striatum. Investigations of the underlying dopaminergic mechanisms involved during the development and maintenance of alcohol dependence could identify novel targets. Human and rodent experimental studies show that dopamine receptor antagonists, agonists and partial agonists as well as dopamine stabilizers influencing dopamine transmission, alter alcohol‐mediated behaviours and thus may be potential treatment targets for alcohol dependence. Although there exists promising preclinical results, the majority of placebo‐controlled randomized clinical trials with traditional dopamine antagonists and agonists have so far have been discouraging. Furthermore, the severe side-effect profiles of many of these compounds may limit their clinical use. Newer dopamine agents, such as partial agonists and dopamine stabilizers, attenuate alcohol‐mediated behaviours in rodents as well as humans. Preclinical as well as clinical studies have shown that substances indirectly targeting the mesolimbic dopamine system may be potential targets for attenuation of alcohol reward. Collectively, the data reviewed herein may contribute to further understanding the complex mechanisms involved in development of alcohol dependence and we suggest that the newer dopamine agents as well as indirect modulators of dopamine signalling deserve to be further evaluated for treatment of alcohol dependence.

Keywords

  • alcohol‐use disorder
  • mesocorticolimbic dopamine system
  • nucleus accumbens
  • dopamine stabilizer
  • antipsychotic drugs

1. Introduction

Alcohol dependence is a chronic relapsing psychiatric disorder significantly contributing to the global burden of disease [1] and affects about four percent of the world's population over the age of 15 (WHO). In the fifth edition of the diagnostic and statistical manual of mental disorders (DSM), the term alcohol use disorder was introduced and grossly defined as problem drinking that has become severe. The characteristics of this disorder include loss of control over alcohol intake, impaired cognitive functioning, negative social consequences, physical tolerance, withdrawal and craving for alcohol. To date, there are three medications approved by both the European Medicines Agency (EMA) and the Food and Drug Administration (FDA) for the treatment of alcohol dependence; disulfiram, naltrexone and acamprosate. The FDA has also approved the use of a long‐acting injectable naltrexone. More recently, the EMA granted authorization also for nalmefene, a compound intended for the reduction of alcohol consumption in adults with alcohol dependence (EMA 2012). Details regarding the mechanism of action of these compounds are outside the scope of this review. In brief, the pharmacological profile is established for disulfiram (an aldehydedehydrogenase inhibitor), naltrexone (an opioid receptor antagonist) and nalmefene (an opioid receptor modulator), whereas the mechanism of action of the anti‐alcohol relapse drug acamprosate is not fully understood. An indirect activation of mesolimbic dopamine via accumbal glycine receptors and ventral tegmental nicotinic acetylcholine receptors (nAChRs) appears likely [2, 3], but additional targets has been suggested (for review see [4]). Finally, the clinical efficacy of these agents is limited [5], possibly due to the heterogeneous nature of the disorder and the complex neurochemical mechanisms underlying alcohol dependence. Thus, the need for novel and efficacious medications remains.

Figure 1.

Representative illustration of the mesocorticolimbic dopamine system in rat brain.

The mesocorticolimbic dopamine system (or the so‐called brain reward system, Figure 1) is one of the established neurobiological systems involved during the development and maintenance of alcohol dependence and thus one potential treatment target. Here, we aim to review the animal and human data describing the role of dopamine and the mesolimbic dopamine system during acute and chronic alcohol exposure. Finally, preclinical and clinical studies evaluating the potential of available dopaminergic agents as well as indirect dopamine modulators as novel medications for alcohol dependence are discussed.

1.1. The brain reward system: the mesocorticolimbic dopamine system

The mesocorticolimbic dopamine system has an established role in driving the rewarding sensations from natural rewards such as food, sex and exercise, which are important behaviours to ensure our survival [6, 7] as well as among drugs of abuse, including alcohol (for review see [8]). The physiological importance of the mesocorticolimbic dopamine system is highlighted by its evolutionary stability and conservation in primitive invertebrates, such as, flatworms, all the way up to primates, including humans. It was identified serendipitously in the 1950s when Olds and Milner found that rats self‐administer electrical currents into certain specific brain regions [9]. These findings were later corroborated by studies showing that rats favoured electrical stimulation in the same specific brain regions, over natural rewards [10]. The primary neurotransmitter regulating the rewarding sensation was determined to be dopamine [11]. Furthermore, the specific neuronal circuitries were progressively mapped with major projections from the ventral tegmental area (VTA) to the nucleus accumbens (NAc, i.e. the ventral striatum), the prefrontal cortex (PFC) and amygdala. Collectively, this network of neurons was denominated the mesocorticolimbic dopamine system [12, 13]. The system was later divided into two distinct projections [12], modulating different dopamine‐mediated behavioural effects; the mesolimbic pathway (from the VTA to the NAc) thought to be responsible for the rewarding and pleasurable effects of natural as well as substances of abuse including alcohol (e.g. [1416]), and the mesocortical pathway (from the VTA to the PFC) believed to be responsible for the motivational and emotional effects [15]. In addition, there are dopamine projections from the VTA to the amygdala and the hippocampus, respectively, involved in reward associative learning and declarative memory formation [15, 17].

In healthy controls, alcohol consumption stimulates dopamine release mediating its reinforcing effects. Repeated bouts of intoxications will overtime downregulate the dopamine activity in the mesocorticolimbic pathway, leading to an increased risk of developing alcohol dependence and other impulse control disorders. [18, 13]. It has also been hypothesized that in vulnerable individuals (e.g. those with a family history of alcohol dependence), the proneness to develop an addiction is higher since they are born with a reduced number of dopamine D2 receptors in mesocorticolimbic pathway, leading to the alcohol dependence [18, 13]. Further, it has been speculated that this dopamine deficiency is responsible for driving craving and compulsive drinking and contributes to relapse even after a period of protracted abstinence [18, 19]. The preclinical and clinical evidence of the underlying interaction between alcohol and the dopamine D2 receptors within the mesocorticolimbic dopamine system during the acute as well as during chronic intake is reviewed below. The involvement of the dopamine D1, D3, D4 and D5 receptors falls outside the scope of the present review but has previously been reviewed elsewhere [20].

1.2. Interaction between alcohol and the mesocorticolimbic dopamine system

1.2.1. Preclinical evidence: acute alcohol exposure and dopamine

Dopamine's importance for alcohol‐induced reward was first identified in studies showing that the catecholamine‐synthesis inhibitor, α‐methyltyrosine (an agent with the ability to inhibit the formation of dopamine in the cytosol of terminals of dopaminergic neurons [21]) blocked alcohol‐induced euphoria, social interaction and talkativeness in humans [22] as well as attenuated alcohol‐induced locomotor activity in rats [23]. Ex vivo and in vivo voltammetry studies in rats found that alcohol increases the dopamine levels in NAc [24]. In addition, in vivo microdialysis studies have since shown that systemic administration of alcohol and other drugs of abuse, including amphetamine, cocaine, opiate and nicotine, increases the accumbal dopamine levels in freely moving rats [2535], strengthening the hypothesis of an association between the rewarding or euphoric sensation and dopamine release in the NAc. This hypothesis is further supported by studies showing that drugs that are not rewarding or abused by humans do not modify synaptic accumbal dopamine levels in rat [27]. In addition, voluntary alcohol consumption causes a dose‐dependent [36] release of dopamine in the NAc in rat [3739]. Finally, intravenous administration of alcohol, as well as other drugs of abuse, increases the firing rate of dopamine neurons in the VTA in rats [40, 41]. Further support for the role of dopamine D2 receptors in the reinforcing effects of alcohol is given by a study showing that dopamine receptor D2 knockout mice self‐administer less alcohol than the wild‐type mice [42]. In addition to the extensive literature showing a link between accumbal dopamine and alcohol‐induced reinforcement, it has been shown that the pure anticipation of alcohol (i.e. without alcohol being present) increases the release of dopamine in NAc in rodents trained to self‐administer alcohol [4345, 36] and that accumbal dopamine release is associated with associative learning, rather than exposure to the reward itself [46]. Moreover, this anticipation effect is more noticeable in high compared to low‐alcohol‐preferring rats [47]. Studies have also shown that the anticipation of a reward increases the firing of accumbal dopamine neurons [41]. It should, however, be mentioned that results from studies with lesion of the mesocorticolimbic dopamine pathways have shown contradicting results with both decreased [4850] and unaltered alcohol intake [5156]. These inconsistent results indicate that the role of accumbal dopamine in reinforcement is complex and highlights that the rewarding properties of alcohol may extend beyond direct or indirect effects on dopamine, involving interactions with several other neurotransmitters including acetylcholine, glutamate, GABA, serotonin (5HT), noradrenaline, taurine and opioids, as well as hormones and peptides [24, 57, 58].

To further elucidate the role of the NAc and the VTA in alcohol‐mediated dopamine regulation, extensive rodent studies, with for example intra‐cranial alcohol infusions and electrophysiological techniques, have been conducted. With regards to the NAc, rodent studies confirm that intra‐NAc alcohol perfusions increase the release of dopamine in the same brain region (e.g. [59, 38, 6062]). An effect that is suggested to be regulated via a neuronal circuitry involving glycine receptors in the NAc as well as anterior ventral tegmental nAChR [59, 63, 64]. Interestingly, the NAc is a heterogeneous region most often divided into two distinct anatomically and functionally different regions, that is the central core and the surrounding shell compartment [6569] and it has been suggested that dopaminergic innervation of the NAc core is associated with the nigrostriatal system, while that of the NAc shell is related to the mesolimbic system [70]. Alcohol has been shown to increase the release of dopamine in NAc shell, but not in the core [7173]. Studies are also emerging suggesting the need for further division of this brain region since it was demonstrated that a borderline region between the core and shell of the NAc is the region most responsive to alcohol [74].

With regards to the VTA, both in vitro and in vivo studies show that alcohol increases the firing of dopamine neurons in the VTA projecting to NAc [7579, 40]. Similarly, in a situation of synaptic transmission blockade, alcohol has been found to increase the firing of dissociated VTA dopamine neurons [76, 77] implying that alcohol activates ventral tegmental dopamine neurons independent of afferent signalling. Furthermore, studies with intra‐VTA alcohol infusions highlight that different subregions within the heterogeneous VTA might have different ability to modulate the alcohol‐induced dopamine response. Specifically, rats voluntarily self‐administer alcohol, as well as acetaldehyde (an alcohol metabolite) into the posterior, but not anterior, part of the VTA [8085], indicating that alcohol is reinforcing only within the posterior VTA. The suggestion is further supported by a study showing that intra‐cranial infusions into the posterior VTA of the D2 agonist quinpirole (in doses that activate local D2 autoreceptors, thereby reducing the firing rate of VTA dopamine neurons [86, 87]), attenuates alcohol self‐administration, which can be restored when the D2 agonist is removed or blocked with administration of a D2 antagonist [84]. In corroboration are the findings that the sensitivity of the posterior VTA to the reinforcing effects of alcohol is enhanced in alcohol‐preferring rats [88]. There are, however, some contradicting results indicating that these subregion‐specific effects might be related to the administered dose of alcohol, the use of various methods, the rat strains across the studies as well as differences in coordinates used for local injections (within the anterior VTA). For example, it has been demonstrated that perfusion of a low, but not a high dose of alcohol into the anterior, but not posterior part of the VTA increased accumbal dopamine in rats [89], and a recent study indicates that additional VTA subregions might be involved as alcohol increases the firing frequency of a subset of dopamine neurons in the medial, but not lateral, part of the VTA [90]. It should also be noted that in both outbreed as well as alcohol‐preferring rats, there are studies showing no influence on the accumbal dopamine levels regardless of dose of alcohol or location in the VTA [59, 91]. Collectively, these data suggest that VTA is a heterogeneous area that differs in morphology and topography (for review, see [92]), and the anterior/posterior and lateral/medial part have different functions regarding alcohol and its activation of the mesolimbic dopamine system.

1.2.2. Clinical evidence: acute alcohol consumption and dopamine

The development of positron imaging technique (PET) and the radiotracer 11C‐raclopride in the 1990s made it possible to study in vivo dopamine function in humans. A series of human imaging studies over the last decade have demonstrated that alcohol [93, 94] as well as other drugs of abuse [95] increase striatal dopamine release. This is further corroborated by the findings that self‐reported behavioural measures of stimulation, euphoria or drug wanting by alcohol correlates with the magnitude and rate of ventral striatum dopamine release [9698, 94, 99, 100]. These studies clearly substantiated the involvement of dopamine in the reinforcing effects of alcohol and closely mimicked the findings of the preclinical studies.

1.2.3. Preclinical evidence: chronic alcohol exposure and dopamine

As mentioned above, it has been hypothesized that the chronic intake of alcohol induces a dopamine deficit state in the brain reward system and that this dysfunction may drive craving and relapse to drinking [101, 18, 19]. In outbred rodents, however, the effects on the mesolimbic dopamine system following chronic alcohol treatment are inconsistent [102]. One possible explanation for these discrepancies may be that most preclinical studies to‐date have used forced alcohol administration which introduces an element of stress and artefact into the experiment, casting doubt on the applicability to our understanding of human alcohol dependence. In this review, we will therefore focus on studies with clear face validity to the human condition, that is those using voluntary self‐administration.

The dopamine deficiency hypothesis is supported by a study showing decreased dopamine receptor gene expression after several months of voluntary alcohol drinking [103]. In addition, microdialysis studies in freely moving outbred rats show a decreased dopamine output in the NAc, compared to age‐matched alcohol‐naïve controls, following 7 weeks [104] and 10 months [29] of voluntary alcohol consumption. Furthermore, after 10 months of drinking, a blunted dopamine response following a systemic alcohol challenge has been found in long‐term drinking, compared to alcohol‐naïve rats [29]. These results indicate that long‐term drinking attenuates the responsiveness of the system to external dopamine stimulation, in addition to decreasing baseline levels of dopamine. It should, however, be noted that acute administration of alcohol induces a twofold increase in dopamine output in the NAc shell in high compared to low‐alcohol‐preferring rats [105], indicating that there might be a difference in these aspects between outbred standard laboratory rats and inbred alcohol‐preferring rats.

The selectively inbred alcohol‐preferring and non‐alcohol‐preferring rat strains have been extensively used to investigate the neurochemical mechanisms underlying alcohol dependence. In line with the dopamine deficiency hypothesis, the baseline accumbal dopamine levels appear to be lower [105] and the dopamine D2 receptors in NAc are fewer [106] in high‐preferring compared to low‐preferring rats. In fact, neurochemical data show that high‐alcohol‐seeking behaviour is associated with 10–15% lower accumbal dopamine content compared with low‐alcohol‐seeking rats [107]. In addition, overexpression of accumbal dopamine D2 receptors reduces alcohol in non‐preferring as well as high‐preferring rats [108, 109]. These results highlight that not only chronic alcohol consumption, but also genetic factors, influence the dopaminergic response to alcohol. Furthermore, it has been suggested that more dorsal parts of the striatum is recruited once the dependence develops [110, 111] although until now this has been investigated only in other drugs of abuse than alcohol.

1.2.4. Clinical evidence: alcohol dependence and dopamine

As mentioned earlier, in vulnerable individuals (related to genetic and environmental factors) as well as healthy individuals, repeated administration of alcohol can lead to perturbations in the dopamine‐regulated circuitry, leading to the development of alcohol dependence. For instance, a human laboratory study has demonstrated that intravenous administration of alcohol causes an increase in dopamine in the ventral striatum in non‐treatment‐seeking alcohol‐dependent individuals [112]. Further, imaging studies have shown that the number of dopamine D2 receptors is lower in individuals with alcohol or drug dependence, compared to healthy controls [113, 114] and there is considerable evidence that the low levels of D2 receptors levels contribute to the excessive urges/craving for alcohol and subsequently to relapse [115]. In addition, decreased dopamine transmission in the mesolimbic regions, such as the ventral striatum, likely contributes to anhedonia and decreased reward sensitivity in alcohol‐dependent individuals. Further, in abstinent high‐risk drinkers as well as alcohol‐dependent individuals, alcohol‐associated cues activate the ventral striatum, which further contribute to the high risk of relapse in these individuals [116, 117].

A recent PET study [118] demonstrated for the first time that, in addition to the ventral striatum, the long‐term consumption of alcohol leads to lowered dopamine levels also in prefrontal cortical structures. These findings support the extensive clinical findings demonstrating that alcohol‐dependent individuals have significant impairments in executive functions such as working memory, impulsivity and decision‐making; functions governed by the cortical brain structures. The fact that there is also less dopamine in the prefrontal cortex, governing these executive functions, is of significance as it could impair the alcohol‐dependent individual's capacity to utilize behavioural treatment strategies, which are critical to relapse prevention.

Collectively, these data indicate that dopamine plays a central role in reward, motivation and planning. Given the relevance of dopamine in the chronic phase of alcohol use and in the development of alcohol dependence, there is considerable interest in evaluating medications that can specifically modify dopamine, thereby serving as potential pharmacotherapies to treat alcohol dependence.

1.2.5. Human genetic evidence: alcohol dependence and dopamine

The preclinical and clinical evidence presented above suggest that dopamine regulates alcohol‐mediated behaviours. Numerous human genetic studies have therefore investigated associations between alcohol dependence and genes related to dopamine function. As early as the 1990s, a polymorphism in the dopamine D2 receptor gene was found to be associated with alcohol dependence [119]. Several studies have since then tried to replicate this association, but the outcome has been inconsistent (for review, see [120]). Although associations have been found between polymorphism of the dopamine D4 gene and alcohol craving, binge drinking as well as novelty seeking (which is a known personality trait important for drinking behaviour in patients with alcohol dependence) [121123], no positive associations between dopamine D4 receptor genes and alcohol dependence per se have been established (for review, see [120]).

Released dopamine into the synaptic cleft is eliminated by catechol‐O‐methyltransferase (COMT) metabolism as well as reuptake by dopamine transporter (DAT). Studies have shown that DAT polymorphism is associated with alcohol withdrawal symptoms as well as with paternal history of alcohol dependence rather than alcohol dependence per se [124, 125]. The risk of developing late onset alcohol dependence (especially in males) as well as the co‐dependence of alcohol and nicotine is associated with polymorphism in COMT [126128]. Albeit cumulative evidence shows association between polymorphisms in various dopamine‐related genes and behaviours associated with alcohol dependence, the findings are inconclusive and therefore, the conclusions from these human genetic studies are limited and remain controversial.

Advertisement

2. The dopamine system: a potential treatment target for alcohol dependence

2.1. Dopamine D2 receptor antagonists

Traditional dopamine D2 receptor antagonists (so‐called neuroleptics, first‐generation antipsychotic drugs or typical antipsychotic drugs) are primary used for the treatment of psychosis, schizophrenia and bipolar disorder [11] based on their ability to counteract a heightened dopamine activity in the brain. It should also be mentioned that these typical antipsychotic agents might have effects on other receptors including dopamine D1, 5HT2 and alpha1 receptors. As reviewed above, the acute reinforcing effects of addictive drugs, including alcohol, could be mediated by increased dopamine release in the NAc, activating dopamine D2 receptors [71, 27, 30]. Thus, traditional dopamine D2 receptor antagonists have been evaluated as potential treatment targets for alcohol dependence based on the hypothesis that they are expected to block the rewarding effects of alcohol.

2.1.1. Preclinical evidence for the use of dopamine D2 receptor antagonists to attenuate alcohol‐mediated behaviours

The hypothesis that dopamine D2 receptor antagonists have the ability to attenuate alcohol‐mediated behaviours is supported by rodent studies showing that both haloperidol and pimozide attenuate alcohol‐induced locomotor stimulation [129] and that these compounds as well as fluphenazine, decrease alcohol‐seeking behaviour and operant self‐administration [130132]. These findings are further substantiated by the data showing that peripheral administration of the dopamine D2 receptor antagonist fluphenazine decreased responding for alcohol, without affecting responses for water in rats [133]. In addition, haloperiodol dose‐dependently reduced operant self‐administration of alcohol in rats [134] as well as decreased alcohol presentations in the self‐administration model [132]. Supportively, low doses of dopamine D2 receptor antagonists inhibit the rewarding properties of other drugs of abuse in rats [135, 42, 136]. It should be noted that some studies have shown contradicting effects [137139], indicating that the role of dopamine in alcohol‐mediated behaviours in complex.

Studies elucidating the underlying mechanism of action of the complex dopamine–alcohol interaction have been conducted. Experiments exploring the role of accumbal dopamine receptors in alcohol‐mediated behaviours showed that intra‐NAc administration of first‐generation antipsychotic drugs including fluphenazine or raclopride decreased alcohol self‐administration in rats [133] as well as the total responding for alcohol [140] and reduced the total responding by decreasing time course and response rate for alcohol self‐administration in rats [141]. On the other hand, local administration of the dopamine D2 receptor antagonist, sulpiride, into the anterior VTA did not alter alcohol nor sucrose intake in high‐alcohol‐preferring rats [142]. It should also be mentioned that accumbal dopamine D1 receptor might regulate alcohol‐induced reward. Indeed, intra‐NAc infusion of a dopamine D1 receptor antagonist (SCH23390 or ecopipam) decreased alcohol‐mediated behaviours in rats [141, 143]. Collectively, these data indicate that the dopamine D2 as well as D1 receptors within the NAc regulate alcohol reinforcement.

2.1.2. Clinical evidence for the use of dopamine D2 antagonists for the treatment of alcohol dependence

Based on the preclinical evidence of a reduction in alcohol consumption via blockade of dopamine D2 receptors, the potential of dopamine D2 antagonists as a pharmacotherapy for alcohol dependence has been investigated in clinical populations.

Dopamine D2 receptor antagonists have been studied in human laboratory studies involving alcohol administration in dependent individuals and found to be effective in reducing craving. In a laboratory study involving 16 individuals with alcohol abuse and/or dependence, the D2 antagonist haloperidol was compared to placebo. The results of this small study demonstrated that haloperidol significantly decreased measures of craving, reduced impulsivity, and the amounts of alcohol ingested [144]. The dopamine D2 antagonist flupenthixol has also been evaluated in a clinical study of 281 recently detoxified alcohol‐dependent patients [145]. The results demonstrated that treatment with the depot formulation of flupenthixol led to a significant increase in rates of relapse (85.2% on active treatment compared with 62.5% on placebo). A major concern with flupenthixol is results from studies demonstrating an increase in the risk of relapse in rodents as well as humans [146], an effect preferentially observed in males [147]. Overall, the clinical utility of atypical antipsychotics has shown to be of some benefit in patients suffering from alcohol dependence and a concomitant psychiatric diagnosis including schizophrenia [148, 149]. A major challenge, however, with the first‐generation antipsychotic drugs is their severe side effect profile including extrapyramidal symptoms, sedation, cognitive impairment, neuroleptic malignant syndrome, which have limited their use in research and in turn its clinical utility in treating alcohol dependence [150, 151].

2.2. Atypical dopamine D2 receptor antagonists

The newer generations of dopamine D2 receptor antagonists (so‐called atypical antipsychotics or second generation antipsychotic drugs) have a broader pharmacological profile since they target several dopamine receptors, including D1, D3, D4 and D5, as well as various other neurotransmitter systems including 5‐HT, muscarinic acetylcholine and histamine receptors. These atypical antipsychotics have a significantly improved side effect profile compared to the traditional first generation of dopamine D2 antagonists. Thus, there has been a renewed interest in evaluating these medications as potential treatment for alcohol dependence with the assumption that the atypical antipsychotics might reduce craving and consumption of alcohol without the substantial adverse effect profile [152]. Furthermore, they are clinically used for alcohol‐dependent patients during the acute detoxification phase to prevent agitation, hallucinations and delirium tremens [153].

2.2.1. Preclinical evidence for the use of atypical dopamine D2 receptor antagonists (i.e. atypical antipsychotics) to attenuate alcohol‐mediated behaviours

The hypothesis that atypical antipsychotics may decrease alcohol intake are supported by two separate studies with risperidone and olanzapine in high‐alcohol‐preferring rats [154, 155]. Furthermore, remoxipride decreases the number of alcohol presentations per session in rats by inducing an early termination of the alcohol‐drinking bout during the self‐administration session [132] and repeated systemic administration of paliperidone decreased the acquisition of alcohol consumption in high‐alcohol‐preferring P rats [156]. In addition, a recent study, comparing the effect of the atypical antipsychotic drug clozapine to that of the traditional dopamine D2 receptor antagonist haloperidol, showed that clozapine but not haloperidol attenuated the initiation of alcohol drinking and development of alcohol preference in high‐alcohol‐preferring rats [157]. Neither compound had an effect on maintenance of chronic alcohol drinking [157], which is in line with a study showing that clozapine did not reduce alcohol consumption in alcohol‐preferring rats [155].

2.2.2. Clinical evidence for the use of atypical dopamine D2 antagonists for the treatment of alcohol dependence

The atypical antipsychotic tiapride has been found to be efficacious in reducing alcohol drinking two placebo‐controlled clinical trials [158, 159]. A small study in twenty alcohol‐dependent individuals, with significant levels of anxiety or depression, showed that tiapride treatment causes a reduced alcohol intake as well as prolonged periods of abstinence [158]. In the largest of the studies [159], 100 recently abstinent alcohol‐dependent patients were randomized to 300 mg of tiapride or placebo for a 3‐month treatment period. This study showed that patients receiving medication had higher rates of abstinence and improved on an array of health care outcomes.

Another atypical antipsychotic drug, quetiapine, has been evaluated in a case study [160] and an open‐label study [161] in patients with alcohol dependence and comorbid psychiatric diagnosis. Both studies demonstrated that quetiapine was well tolerated and in the latter study, the medication not only reduced alcohol consumption and overall psychiatric symptom intensity but also significantly reduced craving. A double‐blind placebo‐controlled study by Kampman and colleagues evaluated the effect of quetiapine and found that the medication was well tolerated and clinically effective in reducing drinking [162]. The effect of medication was found to be stronger in individuals with a more severe disease phenotype. It should, however, be noted that more recent clinical trials using the extended release formulation of quetiapine [163, 164] failed to replicate the clinical findings of the previous studies.

In a retrospective study of 151 schizophrenic patients with alcohol dependence, 36 patients received the atypical antipsychotic medication clozapine. At the 6‐month follow‐up, 79% of the patients on clozapine were in remission from a diagnosis of alcohol dependence, while approximately 33% of those not taking clozapine were in remission [148].

Olanzapine, another example of a second generation of antipsychotics, has been evaluated in a human cue‐craving study, where the compound reduced the urge to drink post‐exposure to alcohol cues, without affecting the rewarding effects of alcohol following the consumption of a priming dose of alcohol [152]. Based on this clinical finding and the knowledge that olanzapine also has a high affinity for the D4 receptors, it was hypothesized whether the dopamine receptor D4 gene maybe involved in meditating its clinical effects. In a subsequent pharmacogenetic, 12‐weeks placebo‐controlled trial in heavy social drinker olanzapine was evaluated in 67 individuals [165] showing that those individuals with the dopamine D4 receptor 7 repeat allele (a polymorphism of the dopamine D4 receptor gene) reported a greater reduction in cue‐induced craving and alcohol consumption compared to individuals with the short allele. These data are supported by the findings that olanzapine reduces craving for alcohol at baseline for both individuals with the DRD4 shorter and longer allele, but only reduces craving after exposure to alcohol cues and after a priming dose of alcohol for individuals with the DRD4 longer allele [166]. Overall, the results from studies evaluating olanzapine as a potential medication for alcohol dependence have provided evidence of a marginal effect restricted to a sub population of patients (with the longer dopamine D4 receptor allele).

In conclusion, although some clinical trials with atypical antipsychotics in alcohol‐dependent patients show promising results, a recent systemic review of atypical antipsychotics, a heterogeneous class of drugs [167] has demonstrated inconsistent clinical response across studies on these compounds effects on alcohol‐related parameters. The clinical use of atypical antipyschotics for treatment of alcohol dependence might also be limited by their side effects profile, even though it is substantially improved compared to the typical antipsychotics (for review see [168]).

2.3. Dopamine D2 agonists

As described previously, in vivo microdialysis studies rodent and imaging studies in individuals with alcohol dependence have demonstrated that chronic exposure to alcohol induce a dopamine deficit state. Thus, it is logical to hypothesize that a dopamine agonist would substitute for this dopaminergic dysfunction during alcohol dependence and alleviate the associated depression‐like symptoms and craving for alcohol.

2.3.1. Preclinical evidence for the use of dopamine agonists to attenuate alcohol‐mediated behaviours

The potential of dopamine D2 agonists to regulate alcohol‐mediated behaviours is supported by a study showing that apomorphine, dose‐dependently reduces operant self‐administration as well as decreases momentary response rates for alcohol in rats [134] and that SDZ‐205‐152, a synthetic‐mixed D1/D2 dopamine receptor agonist dose‐dependently reduces self‐administration of alcohol, but not water, in rats [169]. Moreover, cabergoline, a dopamine D2 receptor agonist, decreased alcohol intake, relapse drinking as well as alcohol‐seeking behaviour in rodents [170]. In addition, low doses of bromocriptine produced a significant, dose‐dependent shift in decreasing the preference for alcohol while enhancing water consumption [171], indicating that the compound at lower doses preferentially augment autoreceptor function, leading to decreased dopamine turnover with a blunted response to the rewarding effects of alcohol as a result. Studies with intra‐NAc administration of quinpirole, further indicating that D2 receptors are involved in a biphasic effect on alcohol self‐administration, by showing that low doses of the agonist increase, whereas higher doses decrease, self‐administration of alcohol [141] (but see also [140]). A study has also investigated the effect of dopamine D2 receptor agonist administration into VTA on alcohol intake. This study showed that microinjection of either quinpirole or quinelorane, into the anterior part of the VTA dose‐dependently decreased alcohol, but not sucrose, intake in alcohol‐preferring rats [142]. In support are the data showing that local administration of cabergoline into the VTA reduced alcohol‐seeking behaviour in rats [170]. These data are contradictory to the findings showing that the dopamine D2 receptor antagonist into the anterior VTA did not alter alcohol intake in high‐alcohol‐preferring rats [142]. Therefore, mechanisms regulating alcohol reinforcement might be different in selectively breed high alcohol‐consuming rats compared to outbreed rats, and this should be investigated in more detail. It should also be mentioned that infusion of the dopamine D1‐like agonist SKF 38393 into NAc had no effect on alcohol self‐administration in rats [141]. Albeit the data are somewhat contradictory, it might be hypothesized that accumbal as well as ventral tegmental dopamine D2 receptors may regulate alcohol reinforcement in rodents.

2.3.2. Clinical evidence for the use of dopamine agonists for the treatment of alcohol dependence

Bromocriptine, a dopamine agonist has been used clinically for Parkinson's disease. At low doses, bromocriptine can reduce alcohol consumption in animals [171]; it is possible that low‐dose dopamine agonists preferentially augment autoreceptor function, thereby decreasing dopamine turnover and blunting the rewarding effects of alcohol. An early double‐blinded study [172] reported that bromocriptine reduced alcohol craving in alcohol‐dependent patients with a specific genotype of the dopamine D2 receptor gene (i.e. the A1/A1 and A1/A2 genotypes). However, subsequent double‐blind placebo‐controlled trials found no effect on relapse or related behaviours [173, 174]. Currently, due to the knowledge of the addictive potential of dopamine agonists, combined with the lack of consistent findings from clinical studies, it is suggested that dopamine receptor agonists do not hold promise as a treatment for alcohol dependence.

2.4. Partial dopamine agonists

Based on the knowledge that alcohol can both stimulate dopamine activity as well as induce a hypo‐dopaminergic state, it has been suggested that partial agonists might have potential as novel medications for alcohol dependence. A partial agonist, such as aripiprazole, has a lower intrinsic activity at the receptor than a full agonist (e.g. dopamine), meaning that when it binds to the receptor, it will activate the receptor but produce a less potent biological response than the full agonist [175177]. In the presence of high levels of the full agonist, a partial agonist will have functional antagonistic activity by binding to the receptor and preventing the response from the full agonist. Partial dopamine D2 agonists, therefore, offer the opportunity to treat the dysregulated dopamine activity during acute alcohol consumption as well as alcohol dependence.

2.4.1. Preclinical evidence for the use of partial dopamine agonists to attenuate alcohol‐mediated behaviours

In line with the hypothesis that a partial dopamine D2 agonist would block the reinforcing effects of alcohol, aripiprazole attenuates alcohol's ability to increase the locomotor activity in mice [178, 179](an indirect measure of activation of the mesolimbic dopamine system). On the other hand, aripiprazole did not interfere with the alcohol‐induced impairment in motor balance as measured by rotarod test [179]. Furthermore, repeated systemic aripiprazole administration decreases alcohol intake in alcohol‐preferring rats [180], while single oral administration dose‐dependently decreases alcohol self‐administration in outbred rats [181]. In addition, aripiprazole has been shown to reverse alcohol‐induced place preference and anxiety‐like behaviour in mice [182].

2.4.2. Clinical evidence for the use of dopamine partial agonists for the treatment of alcohol dependence

Clinically, the partial dopamine D2 agonist aripiprazole has been evaluated in a few randomized placebo‐controlled trials and human laboratory studies. A pilot study showed that aripiprazole reduces the rate of relapse and craving in patients with alcohol dependence [183]. In a subsequent larger 12‐weeks, double‐blind, placebo‐controlled study of 295 alcohol‐dependent patients aripiprazole was initiated at 2 mg/day, titrated to a maximum dose of 20 mg/day [184]. This study showed that aripiprazole decreased heavy drinking days compared to placebo during week four and eight; however, the effect was lost by the maximum dose at week twelve [184]. The effects of aripiprazole were also evaluated in a human laboratory study in non‐treatment seeking alcohol‐dependent individuals (n = 30), showing that the compound was well‐tolerated and reduced drinking, especially in impulsive individuals [185]. Voronin and colleagues also showed that aripiprazole decreased the number of drinks in a bar–lab environment after consumption of a priming drink, as well as weakened the association between the priming‐induced stimulation and further drinking. In another double‐blind comparison trial, aripiprazole was shown to reduce craving [186] but to a lesser extent than the FDA‐approved medication naltrexone [187]. Finally, a brain imaging study demonstrated that aripiprazole attenuated cue‐induced activation as evidenced by a reduced activation of the right ventral striatum with a corresponding reduction in drinking in individuals with alcohol dependence [188]. Thus far, early results with aripiprazole appear promising, although whether this or similar compounds might be useful to treat alcohol dependence or be positioned as a medication with a specific profile, that is as targeted intervention in more impulsive alcohol‐dependent individuals needs to be evaluated further.

2.5. Dopamine stabilizers

As a further development of the partial agonist concept, Nobel Laureate Arvid Carlsson and co‐workers, developed a novel family of compounds based on their ability to stabilize, that is to stimulate, suppress or show no effect on the dopamine activity depending on the prevailing dopaminergic tone [189]. This stabilizing concept was postulated based on a PET study in rhesus monkeys where infusions with the compound (‐)‐OSU6162 (OSU6162) induced a dopaminergic tone‐dependent effect with a reduction in the striatal L‐[11C]DOPA influx rate in monkeys with high baseline values and an increased striatal L‐[11C]DOPA influx rate in animals with low baseline values [190]. The mechanism of action is, however, not completely understood, and although in vitro studies indicate that OSU6162, like aripiprazole, acts as a partial agonist at D2 receptors [191, 192], behavioural studies have failed to demonstrate any intrinsic activity of the compound ([195]). Instead it has been suggested that OSU6162 produces functionally opposite effects by acting as an antagonist at both presynaptic autoreceptors and postsynaptic D2 receptors [189, 193195]. Based on the hypothesis that OSU6162 can counteract both hyper‐ and hypo‐dopaminergic states, the compound has recently been evaluated in both animal models modulating alcohol‐mediated behaviours as well as in a placebo‐controlled human laboratory study in alcohol‐dependent patients.

2.5.1. Preclinical evidence for the use of dopamine stabilizers to attenuate alcohol‐mediated behaviours

A series of experiments in outbred rats show that the dopamine stabilizer OSU6162 attenuates several alcohol‐mediated behaviours including voluntary alcohol intake, alcohol withdrawal symptoms and cue/priming‐induced reinstatement of alcohol seeking in long‐term drinking rats [196]. Furthermore, OSU6162 blunted alcohol‐induced dopamine output in the NAc of alcohol‐naïve rats [196], indicating that OSU6162 has the ability to attenuate the rewarding effects of alcohol. In contrast, a more recent microdialysis study conducted in long‐term drinking rats, showed that OSU6162, compared to vehicle‐pretreatment, had no significant effect on the alcohol‐induced dopamine peak [29]. The contrasting microdialysis results in alcohol‐drinking versus alcohol‐naïve rats highlight OSU6162´s ability to modulate the dopamine output dependent on the prevailing dopaminergic tone. Furthermore, these results indicate that OSU6162 might have the ability to attenuate alcohol‐mediated behaviours by counteracting the hypo‐dopaminergic state induced by long‐term drinking. Collectively, together with the finding that OSU6162 did not induce conditioned place preference [29] (an indication that the compound has no rewarding properties of its own), these results indicate that OSU6162 has many of the favourable characteristics of a potential medication for alcohol dependence.

2.5.2. Clinical evidence for the use of a dopamine stabilizer for the treatment of alcohol dependence

The dopamine stabilizer OSU6162 was recently evaluated in a placebo‐controlled human laboratory alcohol craving study in 56 alcohol dependent individuals [197]. Two weeks of OSU6162 treatment significantly attenuated priming‐induced craving and induced significantly lower subjective “liking” of the consumed alcohol, compared to placebo. Interestingly, the treatment effects of OSU6162 were driven by those individuals with high level of baseline impulsivity, corroborating previous results with the partial dopamine D2 agonist aripiprazole [185]. These results suggest that pharmacological stabilization of the dopamine system might prove as an effective target for alleviating some of the reward driven behaviours during alcohol dependence. Together with OSU6162's favourable side effect profile [198, 197, 199], these results render support for a larger placebo‐controlled efficacy trial in alcohol‐dependent patients to evaluate OSU6162's effect on drinking outcomes.

2.6. Pharmacological agents inducing indirect modulation of dopamine

As mentioned previously, in addition the affecting the dopamine system directly, alcohol interacts with the mesolimbic dopamine system indirectly via several other neurotransmitters. There is a wide range of such compounds, and here, we will only mention a few, specifically targeting glycine receptors and nAChRs, with a clear interaction with dopamine transmission in the mesolimbic dopamine system [64].

2.6.1. Preclinical evidence for the use of compounds that indirectly targets dopamine to attenuate alcohol‐mediated behaviours

Rodent studies exploring the potential of targeting the glycine system as a medication for alcohol dependence showed that systemic administration of the glycine transporter‐1 inhibitor Org25935 increased extracellular glycine in the NAc, which prevented alcohol‐induced dopamine release [200, 201] as well as decreased alcohol intake and prevented relapse drinking [202, 203]. These results provided rational for a randomized placebo‐controlled clinical trial in alcohol‐dependent individuals.

Emerging data suggests that the activity of dopamine neurons in the VTA projecting to the NAc is regulated by several afferents, such as, for example the cholinergic neurons projecting from the laterodorsal tegmental nucleus (LDTg) (for review see [204]). Although alcohol's direct interaction with this cholinergic‐dopaminergic reward link remains to be fully elucidated, a study show that voluntary alcohol intake in high‐alcohol‐consuming rats causes a concomitant release of ventral tegmental acetylcholine and accumbal dopamine [39]. Several rodent studies with nAChR antagonists such as mecamylamine or selective nAChRs antagonists such as alpha‐conotoxin MII highlight the potential of nAChRs as novel medications for alcohol dependence by showing that these compounds prevent alcohol from increasing dopamine and reduce alcohol consumption behaviour [28, 38, 32, 34, 35]. These nAChR antagonists are limited in a clinical setting due to low blood–brain barrier permeability and an unfavourable side effect profile. The potential of nAChR's as novel treatment target was revived with the marketing of the partial nAChR agonist varenicline as a smoking cessation agent. It has been shown that varenicline reduce alcohol intake and alcohol‐seeking behaviour in long‐term drinking rats [205] and modulate NAc dopamine after systemic administrations of alcohol alone and in combination with nicotine [206].

2.6.2. Clinical evidence for the use of indirect modulation of dopamine for the treatment of alcohol dependence

Albeit the preclinical data look promising regarding the glycine transporter‐1 inhibitor Org25935, the multicenter randomized clinical trial produced a negative outcome on alcohol intake, but did not discard the potential importance of the mechanism [207]. More promising clinical studies with varenicline show that this agent decreased alcohol consumption and craving in an experimental setting in heavy‐drinking smokers [208210]. Moreover, data from a randomized clinical trial in alcohol‐dependent individuals show that the smoking cessation agent reduced the weekly percent heavy drinking days drinks, decreased the drinks per drinking day as well as prevented alcohol craving [211]. It should, however, be noted that recent clinical trials in alcohol‐dependent individuals were unable to find a beneficial effect of varenicline based on self‐reported alcohol consumption [212, 213]. Nevertheless, when also monitoring the selective alcohol biomarker phosphatidylethanol (PEth) in the blood of the subjects in the above‐mentioned clinical trial [212], it was found that varenicline indeed had effect on this objective measure of alcohol consumption [214] strengthening the potential of varenicline as potential novel medication for alcohol dependence. Besides glycine receptors and nAChR, there are various signalling systems indirectly targeting the mesolimbic dopamine system with promising preclinical findings on alcohol‐mediated behaviours. Collectively, these data indicate that indirect modulation of dopamine signalling might be a potential target for novel treatment strategies for alcohol dependence and that these targets should be investigated in more detail in human laboratory studies as well as randomized clinical trials.

Advertisement

3. Conclusion

Extensive preclinical and clinical research support the hypothesis that alcohol's acute reinforcing effects are mediated through a dopamine surge in the mesocorticolimbic dopamine system and that the chronic and excessive alcohol consumption, in contrast, induces a dopamine deficient state driving the processes of craving and relapse. In addition, it is well substantiated that alcohol affects dopamine directly via the NAc and VTA as well as through indirect activation of the mesolimbic pathway via interaction with other reward‐related brain regions and neurotransmitters. These complex relationships need to be investigated further. Given dopamine's pivotal role in the development and maintenance of alcohol dependence, medications targeting dopamine does constitute an important area of research. Although promising preclinical results, the majority of results from the clinical studies with dopamine‐acting medications have thus far been discouraging. The side effects profile of many of the evaluated compounds, including typical antipsychotic drugs, render them clinically unfavourable. On the other hand, newer dopamine agents, without complete antagonism or agonism, especially the dopamine stabilizers show promise and deserve further investigation in alcohol‐dependent patients.

Advertisement

Acknowledgments

The study is supported by grants from the Swedish Research Council (2009‐2782, 2014‐3887 and 2015‐03219), Swedish Society for Medical Research, Swedish Alcohol Monopoly Foundation for Alcohol Research.

References

  1. 1. Rehm J, Mathers C, Popova S, Thavorncharoensap M, Teerawattananon Y, and Patra J. Global burden of disease and injury and economic cost attributable to alcohol use and alcohol‐use disorders. Lancet. 2009;373:2223–33.
  2. 2. Chau P, Hoifodt‐Lido H, Lof E, Soderpalm B, and Ericson M. Glycine receptors in the nucleus accumbens involved in the ethanol intake‐reducing effect of acamprosate. Alcohol Clin Exp Res. 2010;34:39–45.
  3. 3. Chau P, Stomberg R, Fagerberg A, Soderpalm B, and Ericson M. Glycine receptors involved in acamprosate's modulation of accumbal dopamine levels: an in vivo microdialysis study. Alcohol Clin Exp Res. 2010;34:32–8.
  4. 4. De Witte P, Littleton J, Parot P, and Koob G. Neuroprotective and abstinence‐promoting effects of acamprosate: elucidating the mechanism of action. CNS Drugs. 2005;19:517–37.
  5. 5. Heilig M and Egli M. Pharmacological treatment of alcohol dependence: target symptoms and target mechanisms. Pharmacol Ther. 2006;111:855–76.
  6. 6. Hansen S, Harthon C, Wallin E, Lofberg L, and Svensson K. Mesotelencephalic dopamine system and reproductive behavior in the female rat: effects of ventral tegmental 6‐hydroxydopamine lesions on maternal and sexual responsiveness. Behav Neurosci. 1991;105:588–98.
  7. 7. Schultz W, Dayan P, and Montague PR. A neural substrate of prediction and reward. Science. 1997;275:1593–9.
  8. 8. Everitt BJ, Belin D, Economidou D, Pelloux Y, Dalley JW, and Robbins TW. Review. Neural mechanisms underlying the vulnerability to develop compulsive drug‐seeking habits and addiction. Philos Trans R Soc Lond B Biol Sci. 2008;363:3125–35.
  9. 9. Olds J and Milner P. Positive reinforcement produced by electrical stimulation of septal area and other regions of rat brain. J Comp Physiol Psychol. 1954;47:419–27.
  10. 10. Phillips A and Fieberger HC, Neruochemical correlates of brain‐stimulation. Untangeling the Gordian knot., in The neurochemical basis of reward, J.M. Liebman and S.J. Cooper, Editors. 1989, Clarendon Press: Oxford, England. pp. 67–104.
  11. 11. Carlsson A. Thirty years of dopamine research. Adv Neurol. 1993;60:1–10.
  12. 12. Dahlstrom A and Fuxe K. Localization of monoamines in the lower brain stem. Experientia. 1964;20:398–9.
  13. 13. Wise RA and Rompre PP. Brain dopamine and reward. Annu Rev Psychol. 1989;40:191–225.
  14. 14. Koob GF. Drugs of abuse: anatomy, pharmacology and function of reward pathways. Trends Pharmacol Sci. 1992;13:177–84.
  15. 15. Robinson TE and Berridge KC. The neural basis of drug craving: an incentive‐sensitization theory of addiction. Brain Res Brain Res Rev. 1993;18:247–91.
  16. 16. Wise RA. The role of reward pathways in the development of drug dependence. Pharmacol Ther. 1987;35:227–63.
  17. 17. Russo SJ and Nestler EJ. The brain reward circuitry in mood disorders. Nat Rev Neurosci. 2013;14:609–25.
  18. 18. Diana M. The dopamine hypothesis of drug addiction and its potential therapeutic value. Front Psychiatry. 2011;2:64.
  19. 19. Koob GF. Theoretical frameworks and mechanistic aspects of alcohol addiction: alcohol addiction as a reward deficit disorder. Curr Top Behav Neurosci. 2013;13:3–30.
  20. 20. Le Foll B, Gallo A, Le Strat Y, Lu L, and Gorwood P. Genetics of dopamine receptors and drug addiction: a comprehensive review. Behav Pharmacol. 2009;20:1–17.
  21. 21. Watanabe S, Fusa K, Takada K, Aono Y, Saigusa T, Koshikawa N, and Cools AR. Effects of alpha‐methyl‐p‐tyrosine on extracellular dopamine levels in the nucleus accumbens and the dorsal striatum of freely moving rats. J Oral Sci. 2005;47:185–90.
  22. 22. Ahlenius S, Carlsson A, Engel J, Svensson T, and Sodersten P. Antagonism by alpha methyltyrosine of the ethanol‐induced stimulation and euphoria in man. Clin Pharmacol Ther. 1973;14:586–91.
  23. 23. Engel J, Strombom U, Svensson TH, and Waldeck B. Suppression by alpha‐methyltyrosine of ethanol‐induced locomotor stimulation: partial reversal by L‐dopa. Psychopharmacologia. 1974;37:275–9.
  24. 24. Engel JA, Fahlke C, Hulthe P, Hard E, Johannessen K, Snape B, and Svensson L. Biochemical and behavioral evidence for an interaction between ethanol and calcium channel antagonists. J Neural Transm. 1988;74:181–93.
  25. 25. Blomqvist O, Engel JA, Nissbrandt H, and Soderpalm B. The mesolimbic dopamine‐activating properties of ethanol are antagonized by mecamylamine. Eur J Pharmacol. 1993;249:207–13.
  26. 26. Blomqvist O, Ericson M, Engel JA, and Soderpalm B. Accumbal dopamine overflow after ethanol: localization of the antagonizing effect of mecamylamine. Eur J Pharmacol. 1997;334:149–56.
  27. 27. Di Chiara G and Imperato A. Preferential stimulation of dopamine release in the nucleus accumbens by opiates, alcohol, and barbiturates: studies with transcerebral dialysis in freely moving rats. Ann N Y Acad Sci. 1986;473:367–81.
  28. 28. Ericson M, Blomqvist O, Engel JA, and Soderpalm B. Voluntary ethanol intake in the rat and the associated accumbal dopamine overflow are blocked by ventral tegmental mecamylamine. Eur J Pharmacol. 1998;358:189–96.
  29. 29. Feltmann K, Fredriksson I, Wirf M, Schilstrom B, and Steensland P. The monoamine stabilizer (‐)‐OSU6162 counteracts downregulated dopamine output in the nucleus accumbens of long‐term drinking Wistar rats. Addict Biol. 2016;21:438-49.
  30. 30. Imperato A and Di Chiara G. Preferential stimulation of dopamine release in the nucleus accumbens of freely moving rats by ethanol. J Pharmacol Exp Ther. 1986;239:219–28.
  31. 31. Jerlhag E, Egecioglu E, Landgren S, Salome N, Heilig M, Moechard D, Datta R, Perissoud D, Dickson SL, and Engel JA. Requirement of central ghrelin signaling for alcohol reward. PNAS. 2009;106:11318–11323.
  32. 32. Jerlhag E, Grotli M, Luthman K, Svensson L, and Engel JA. Role of the subunit composition of central nicotinic acetylcholine receptors for the stimulatory and dopamine‐enhancing effects of ethanol. Alcohol Alcohol. 2006;41:486–93.
  33. 33. Jonsson S, Adermark L, Ericson M, and Soderpalm B. The involvement of accumbal glycine receptors in the dopamine‐elevating effects of addictive drugs. Neuropharmacology. 2014;82:69–75.
  34. 34. Larsson A, Jerlhag E, Svensson L, Soderpalm B, and Engel JA. Is an alpha‐conotoxin MII‐sensitive mechanism involved in the neurochemical, stimulatory, and rewarding effects of ethanol? Alcohol. 2004;34:239–50.
  35. 35. Larsson A, Svensson L, Soderpalm B, and Engel JA. Role of different nicotinic acetylcholine receptors in mediating behavioral and neurochemical effects of ethanol in mice. Alcohol. 2002;28:157–67.
  36. 36. Weiss F, Lorang MT, Bloom FE, and Koob GF. Oral alcohol self‐administration stimulates dopamine release in the rat nucleus accumbens: genetic and motivational determinants. J Pharmacol Exp Ther. 1993;267:250–8.
  37. 37. Doyon WM, York JL, Diaz LM, Samson HH, Czachowski CL, and Gonzales RA. Dopamine activity in the nucleus accumbens during consummatory phases of oral ethanol self‐administration. Alcohol Clin Exp Res. 2003;27:1573–82.
  38. 38. Ericson M, Molander A, Lof E, Engel JA, and Soderpalm B. Ethanol elevates accumbal dopamine levels via indirect activation of ventral tegmental nicotinic acetylcholine receptors. Eur J Pharmacol. 2003;467:85–93.
  39. 39. Larsson A, Edstrom L, Svensson L, Soderpalm B, and Engel JA. Voluntary ethanol intake increases extracellular acetylcholine levels in the ventral tegmental area in the rat. Alcohol Alcohol. 2005;40:349–58.
  40. 40. Gessa GL, Muntoni F, Collu M, Vargiu L, and Mereu G. Low doses of ethanol activate dopaminergic neurons in the ventral tegmental area. Brain Res. 1985;348:201–3.
  41. 41. Martin PD and Ono T. Effects of reward anticipation, reward presentation, and spatial parameters on the firing of single neurons recorded in the subiculum and nucleus accumbens of freely moving rats. Behav Brain Res. 2000;116:23–38.
  42. 42. Risinger FO, Freeman PA, Rubinstein M, Low MJ, and Grandy DK. Lack of operant ethanol self‐administration in dopamine D2 receptor knockout mice. Psychopharmacology (Berl). 2000;152:343–50.
  43. 43. Gonzales RA and Weiss F. Suppression of ethanol‐reinforced behavior by naltrexone is associated with attenuation of the ethanol‐induced increase in dialysate dopamine levels in the nucleus accumbens. J Neurosci. 1998;18:10663–71.
  44. 44. Katner SN and Weiss F. Ethanol‐associated olfactory stimuli reinstate ethanol‐seeking behavior after extinction and modify extracellular dopamine levels in the nucleus accumbens. Alcohol Clin Exp Res. 1999;23:1751–60.
  45. 45. Melendez RI, Rodd‐Henricks ZA, Engleman EA, Li TK, McBride WJ, and Murphy JM. Microdialysis of dopamine in the nucleus accumbens of alcohol‐preferring (P) rats during anticipation and operant self‐administration of ethanol. Alcohol Clin Exp Res. 2002;26:318–25.
  46. 46. Spanagel R and Weiss F. The dopamine hypothesis of reward: past and current status. Trends Neurosci. 1999;22:521–7.
  47. 47. Katner SN, Kerr TM, and Weiss F. Ethanol anticipation enhances dopamine efflux in the nucleus accumbens of alcohol‐preferring (P) but not Wistar rats. Behav Pharmacol. 1996;7:669–674.
  48. 48. Brown ZW and Amit Z. The effects of selective catecholamine depletions by 6‐hydroxydopamine on ethanol preference in rats. Neurosci Lett. 1977;5:333–6.
  49. 49. Myers RD and Melchior CL. Alcohol drinking in the rat after destruction of serotonergic and catecholaminergic neurons in the brain. Res Commun Chem Pathol Pharmacol. 1975;10:363–78.
  50. 50. Richardson JS and Novakovski DM. Brain monoamines and free choice ethanol consumption in rats. Drug Alcohol Depend. 1978;3:253–64.
  51. 51. Fahlke C, Hansen S, Engel JA, and Hard E. Effects of ventral striatal 6‐OHDA lesions or amphetamine sensitization on ethanol consumption in the rat. Pharmacol Biochem Behav. 1994;47:345–9.
  52. 52. Hansen S, Fahlke C, Hard E, and Thomasson R. Effects of ibotenic acid lesions of the ventral striatum and the medial prefrontal cortex on ethanol consumption in the rat. Alcohol. 1995;12:397–402.
  53. 53. Kiianmaa K and Attila LM. Alcohol intake, ethanol‐induced narcosis and intoxication in rats following neonatal 6‐hydroxydopamine or 5, 7‐dihydroxytryptamine treatment. Naunyn Schmiedebergs Arch Pharmacol. 1979;308:165–70.
  54. 54. Koistinen M, Tuomainen P, Hyytia P, and Kiianmaa K. Naltrexone suppresses ethanol intake in 6‐hydroxydopamine‐treated rats. Alcohol Clin Exp Res. 2001;25:1605–12.
  55. 55. Quarfordt SD, Kalmus GW, and Myers RD. Ethanol drinking following 6‐OHDA lesions of nucleus accumbens and tuberculum olfactorium of the rat. Alcohol. 1991;8:211–7.
  56. 56. Rassnick S, Stinus L, and Koob GF. The effects of 6‐hydroxydopamine lesions of the nucleus accumbens and the mesolimbic dopamine system on oral self‐administration of ethanol in the rat. Brain Res. 1993;623:16–24.
  57. 57. Ericson M, Chau P, Clarke RB, Adermark L, and Soderpalm B. Rising taurine and ethanol concentrations in nucleus accumbens interact to produce dopamine release after ethanol administration. Addict Biol. 2011;16:377–85.
  58. 58. Little HJ. The contribution of electrophysiology to knowledge of the acute and chronic effects of ethanol. Pharmacol Ther. 1999;84:333–53.
  59. 59. Ericson M, Lof E, Stomberg R, Chau P, and Soderpalm B. Nicotinic acetylcholine receptors in the anterior, but not posterior, ventral tegmental area mediate ethanol‐induced elevation of accumbal dopamine levels. J Pharmacol Exp Ther. 2008;326:76–82.
  60. 60. Lof E, Olausson P, deBejczy A, Stomberg R, McIntosh JM, Taylor JR, and Soderpalm B. Nicotinic acetylcholine receptors in the ventral tegmental area mediate the dopamine activating and reinforcing properties of ethanol cues. Psychopharmacology (Berl). 2007;195:333–43.
  61. 61. Wozniak KM and Linnoila M. Recent advances in pharmacological research on alcohol. Possible relations with cocaine. Recent Dev Alcohol. 1992;10:235–72.
  62. 62. Yoshimoto K, McBride WJ, Lumeng L, and Li TK. Alcohol stimulates the release of dopamine and serotonin in the nucleus accumbens. Alcohol. 1992;9:17–22.
  63. 63. Molander A and Soderpalm B. Glycine receptors regulate dopamine release in the rat nucleus accumbens. Alcohol Clin Exp Res. 2005;29:17–26.
  64. 64. Soderpalm B, Lof E, and Ericson M. Mechanistic studies of ethanol's interaction with the mesolimbic dopamine reward system. Pharmacopsychiatry. 2009;42 Suppl 1:S87–94.
  65. 65. Graybiel AM and Ragsdale CW, Jr. Histochemically distinct compartments in the striatum of human, monkeys, and cat demonstrated by acetylthiocholinesterase staining. Proc Natl Acad Sci USA. 1978;75:5723–6.
  66. 66. Heimer L, Zahm DS, Churchill L, Kalivas PW, and Wohltmann C. Specificity in the projection patterns of accumbal core and shell in the rat. Neuroscience. 1991;41:89–125.
  67. 67. Voorn P, Gerfen CR, and Groenewegen HJ. Compartmental organization of the ventral striatum of the rat: immunohistochemical distribution of enkephalin, substance P, dopamine, and calcium‐binding protein. J Comp Neurol. 1989;289:189–201.
  68. 68. Zahm DS. Functional‐anatomical implications of the nucleus accumbens core and shell subterritories. Ann N Y Acad Sci. 1999;877:113–28.
  69. 69. Zahm DS and Brog JS. On the significance of subterritories in the “accumbens” part of the rat ventral striatum. Neuroscience. 1992;50:751–67.
  70. 70. Deutch AY and Cameron DS. Pharmacological characterization of dopamine systems in the nucleus accumbens core and shell. Neuroscience. 1992;46:49–56.
  71. 71. Bassareo V, De Luca MA, Aresu M, Aste A, Ariu T, and Di Chiara G. Differential adaptive properties of accumbens shell dopamine responses to ethanol as a drug and as a motivational stimulus. Eur J Neurosci. 2003;17:1465–72.
  72. 72. Cadoni C, Solinas M, and Di Chiara G. Psychostimulant sensitization: differential changes in accumbal shell and core dopamine. Eur J Pharmacol. 2000;388:69–76.
  73. 73. Iyaniwura TT, Wright AE, and Balfour DJ. Evidence that mesoaccumbens dopamine and locomotor responses to nicotine in the rat are influenced by pretreatment dose and strain. Psychopharmacology (Berl). 2001;158:73–9.
  74. 74. Howard EC, Schier CJ, Wetzel JS, and Gonzales RA. The dopamine response in the nucleus accumbens core‐shell border differs from that in the core and shell during operant ethanol self‐administration. Alcohol Clin Exp Res. 2009;33:1355–65.
  75. 75. Brodie MS and Appel SB. The effects of ethanol on dopaminergic neurons of the ventral tegmental area studied with intracellular recording in brain slices. Alcohol Clin Exp Res. 1998;22:236–44.
  76. 76. Brodie MS, Pesold C, and Appel SB. Ethanol directly excites dopaminergic ventral tegmental area reward neurons. Alcohol Clin Exp Res. 1999;23:1848–52.
  77. 77. Brodie MS, Shefner SA, and Dunwiddie TV. Ethanol increases the firing rate of dopamine neurons of the rat ventral tegmental area in vitro. Brain Res. 1990;508:65–9.
  78. 78. Brodie MS, Trifunovic RD, and Shefner SA. Serotonin potentiates ethanol‐induced excitation of ventral tegmental area neurons in brain slices from three different rat strains. J Pharmacol Exp Ther. 1995;273:1139–46.
  79. 79. Bunney EB, Appel SB, and Brodie MS. Electrophysiological effects of cocaethylene, cocaine, and ethanol on dopaminergic neurons of the ventral tegmental area. J Pharmacol Exp Ther. 2001;297:696–703.
  80. 80. Gatto GJ, McBride WJ, Murphy JM, Lumeng L, and Li TK. Ethanol self‐infusion into the ventral tegmental area by alcohol‐preferring rats. Alcohol. 1994;11:557–64.
  81. 81. Ikemoto S, Murphy JM, and McBride WJ. Regional differences within the rat ventral tegmental area for muscimol self‐infusions. Pharmacol Biochem Behav. 1998;61:87–92.
  82. 82. Ikemoto S and Wise RA. Rewarding effects of the cholinergic agents carbachol and neostigmine in the posterior ventral tegmental area. J Neurosci. 2002;22:9895–904.
  83. 83. Rodd ZA, Bell RL, Zhang Y, Murphy JM, Goldstein A, Zaffaroni A, Li TK, and McBride WJ. Regional heterogeneity for the intracranial self‐administration of ethanol and acetaldehyde within the ventral tegmental area of alcohol‐preferring (P) rats: involvement of dopamine and serotonin. Neuropsychopharmacology. 2005;30:330–8.
  84. 84. Rodd ZA, Melendez RI, Bell RL, Kuc KA, Zhang Y, Murphy JM, and McBride WJ. Intracranial self‐administration of ethanol within the ventral tegmental area of male Wistar rats: evidence for involvement of dopamine neurons. J Neurosci. 2004;24:1050–7.
  85. 85. Rodd‐Henricks ZA, McKinzie DL, Crile RS, Murphy JM, and McBride WJ. Regional heterogeneity for the intracranial self‐administration of ethanol within the ventral tegmental area of female Wistar rats. Psychopharmacology (Berl). 2000;149:217–24.
  86. 86. Congar P, Bergevin A, and Trudeau LE. D2 receptors inhibit the secretory process downstream from calcium influx in dopaminergic neurons: implication of K+ channels. J Neurophysiol. 2002;87:1046–56.
  87. 87. Jeziorski M and White FJ. Dopamine agonists at repeated “autoreceptor‐selective” doses: effects upon the sensitivity of A10 dopamine autoreceptors. Synapse. 1989;4:267–80.
  88. 88. Rodd ZA, Bell RL, McQueen VK, Davids MR, Hsu CC, Murphy JM, Li TK, Lumeng L, and McBride WJ. Chronic ethanol drinking by alcohol‐preferring rats increases the sensitivity of the posterior ventral tegmental area to the reinforcing effects of ethanol. Alcohol Clin Exp Res. 2005;29:358–66.
  89. 89. Jerlhag E and Engel JA. Local infusion of low, but not high, doses of alcohol into the anterior ventral tegmental area causes release of accumbal dopamine. Open J Psychiatry. 2013;4:53-59.
  90. 90. Mrejeru A, Marti‐Prats L, Avegno EM, Harrison NL, and Sulzer D. A subset of ventral tegmental area dopamine neurons responds to acute ethanol. Neuroscience. 2015;290:649–58.
  91. 91. Tuomainen P, Patsenka A, Hyytia P, Grinevich V, and Kiianmaa K. Extracellular levels of dopamine in the nucleus accumbens in AA and ANA rats after reverse microdialysis of ethanol into the nucleus accumbens or ventral tegmental area. Alcohol. 2003;29:117–24.
  92. 92. Ikemoto S. Dopamine reward circuitry: two projection systems from the ventral midbrain to the nucleus accumbens‐olfactory tubercle complex. Brain Res Rev. 2007;56:27–78.
  93. 93. Boileau I, Assaad JM, Pihl RO, Benkelfat C, Leyton M, Diksic M, Tremblay RE, and Dagher A. Alcohol promotes dopamine release in the human nucleus accumbens. Synapse. 2003;49:226–31.
  94. 94. Urban NB, Kegeles LS, Slifstein M, Xu X, Martinez D, Sakr E, Castillo F, Moadel T, O'Malley SS, Krystal JH, and Abi‐Dargham A. Sex differences in striatal dopamine release in young adults after oral alcohol challenge: a positron emission tomography imaging study with [(1)(1)C]raclopride. Biol Psychiatry. 2010;68:689–96.
  95. 95. Volkow ND, Fowler JS, Wang GJ, and Swanson JM. Dopamine in drug abuse and addiction: results from imaging studies and treatment implications. Mol Psychiatry. 2004;9:557–69.
  96. 96. Drevets WC, Gautier C, Price JC, Kupfer DJ, Kinahan PE, Grace AA, Price JL, and Mathis CA. Amphetamine‐induced dopamine release in human ventral striatum correlates with euphoria. Biol Psychiatry. 2001;49:81–96.
  97. 97. Leyton M, Boileau I, Benkelfat C, Diksic M, Baker G, and Dagher A. Amphetamine‐induced increases in extracellular dopamine, drug wanting, and novelty seeking: a PET/[11C]raclopride study in healthy men. Neuropsychopharmacology. 2002;27:1027–35.
  98. 98. Ramchandani VA, Umhau J, Pavon FJ, Ruiz‐Velasco V, Margas W, Sun H, Damadzic R, Eskay R, Schoor M, Thorsell A, Schwandt ML, Sommer WH, George DT, Parsons LH, Herscovitch P, Hommer D, and Heilig M. A genetic determinant of the striatal dopamine response to alcohol in men. Mol Psychiatry. 2011;16:809–17.
  99. 99. Volkow ND, Wang GJ, Fowler JS, Logan J, Angrist B, Hitzemann R, Lieberman J, and Pappas N. Effects of methylphenidate on regional brain glucose metabolism in humans: relationship to dopamine D2 receptors. Am J Psychiatry. 1997;154:50–5.
  100. 100. Yoder KK, Constantinescu CC, Kareken DA, Normandin MD, Cheng TE, O'Connor SJ, and Morris ED. Heterogeneous effects of alcohol on dopamine release in the striatum: a PET study. Alcohol Clin Exp Res. 2007;31:965–73.
  101. 101. Becker HC and Mulholland PJ. Neurochemical mechanisms of alcohol withdrawal. Handb Clin Neurol. 2014;125:133–56.
  102. 102. Tupala E and Tiihonen J. Dopamine and alcoholism: neurobiological basis of ethanol abuse. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28:1221–47.
  103. 103. Jonsson S, Ericson M, and Soderpalm B. Modest long‐term ethanol consumption affects expression of neurotransmitter receptor genes in the rat nucleus accumbens. Alcohol Clin Exp Res. 2014;38:722–9.
  104. 104. Barak S, Carnicella S, Yowell QV, and Ron D. Glial cell line‐derived neurotrophic factor reverses alcohol‐induced allostasis of the mesolimbic dopaminergic system: implications for alcohol reward and seeking. J Neurosci. 2011;31:9885–94.
  105. 105. Bustamante D, Quintanilla ME, Tampier L, Gonzalez‐Lira V, Israel Y, and Herrera‐Marschitz M. Ethanol induces stronger dopamine release in nucleus accumbens (shell) of alcohol‐preferring (bibulous) than in alcohol‐avoiding (abstainer) rats. Eur J Pharmacol. 2008;591:153–8.
  106. 106. Stefanini E, Frau M, Garau MG, Garau B, Fadda F, and Gessa GL. Alcohol‐preferring rats have fewer dopamine D2 receptors in the limbic system. Alcohol Alcohol. 1992;27:127–30.
  107. 107. McBride WJ, Murphy JM, Lumeng L, and Li TK. Serotonin, dopamine and GABA involvement in alcohol drinking of selectively bred rats. Alcohol. 1990;7:199–205.
  108. 108. Thanos PK, Taintor NB, Rivera SN, Umegaki H, Ikari H, Roth G, Ingram DK, Hitzemann R, Fowler JS, Gatley SJ, Wang GJ, and Volkow ND. DRD2 gene transfer into the nucleus accumbens core of the alcohol preferring and nonpreferring rats attenuates alcohol drinking. Alcohol Clin Exp Res. 2004;28:720–8.
  109. 109. Thanos PK, Volkow ND, Freimuth P, Umegaki H, Ikari H, Roth G, Ingram DK, and Hitzemann R. Overexpression of dopamine D2 receptors reduces alcohol self‐administration. J Neurochem. 2001;78:1094–103.
  110. 110. Everitt BJ and Robbins TW. Neural systems of reinforcement for drug addiction: from actions to habits to compulsion. Nat Neurosci. 2005;8:1481–9.
  111. 111. Volkow ND, Fowler JS, Wang GJ, Swanson JM, and Telang F. Dopamine in drug abuse and addiction: results of imaging studies and treatment implications. Arch Neurol. 2007;64:1575–9.
  112. 112. Yoder KK, Albrecht DS, Dzemidzic M, Normandin MD, Federici LM, Graves T, Herring CM, Hile KL, Walters JW, Liang T, Plawecki MH, O'Connor S, and Kareken DA. Differences in IV alcohol‐induced dopamine release in the ventral striatum of social drinkers and nontreatment‐seeking alcoholics. Drug Alcohol Depend. 2016; 160:163-69.
  113. 113. Dettling M, Heinz A, Dufeu P, Rommelspacher H, Graf KJ, and Schmidt LG. Dopaminergic responsivity in alcoholism: trait, state, or residual marker? Am J Psychiatry. 1995;152:1317–21.
  114. 114. Volkow ND, Wang GJ, Fowler JS, Logan J, Ding YS, Gatley J, Hitzemann R, and Pappas N. Decreases in dopamine receptors but not in dopamine transporters in alcoholics. J Nucl Med. 1996;37:122–122.
  115. 115. Heinz A, Siessmeier T, Wrase J, Hermann D, Klein S, Grusser SM, Flor H, Braus DF, Buchholz HG, Grunder G, Schreckenberger M, Smolka MN, Rosch F, Mann K, and Bartenstein P. Correlation between dopamine D(2) receptors in the ventral striatum and central processing of alcohol cues and craving. Am J Psychiatry. 2004;161:1783–9.
  116. 116. Braus DF, Wrase J, Grusser S, Hermann D, Ruf M, Flor H, Mann K, and Heinz A. Alcohol‐associated stimuli activate the ventral striatum in abstinent alcoholics. J Neural Transm. 2001;108:887–94.
  117. 117. Kareken DA, Claus ED, Sabri M, Dzemidzic M, Kosobud AE, Radnovich AJ, Hector D, Ramchandani VA, O'Connor SJ, Lowe M, and Li TK. Alcohol‐related olfactory cues activate the nucleus accumbens and ventral tegmental area in high‐risk drinkers: preliminary findings. Alcohol Clin Exp Res. 2004;28:550–7.
  118. 118. Narendran R, Mason NS, Paris J, Himes ML, Douaihy AB, and Frankle WG. Decreased prefrontal cortical dopamine transmission in alcoholism. Am J Psychiatry. 2014;171:881–8.
  119. 119. Blum K, Noble EP, Sheridan PJ, Montgomery A, Ritchie T, Jagadeeswaran P, Nogami H, Briggs AH, and Cohn JB. Allelic association of human dopamine D2 receptor gene in alcoholism. JAMA. 1990;263:2055–60.
  120. 120. Kimura M and Higuchi S. Genetics of alcohol dependence. Psychiatry Clin Neurosci. 2011;65:213–25.
  121. 121. Hutchison KE, McGeary J, Smolen A, Bryan A, and Swift RM. The DRD4 VNTR polymorphism moderates craving after alcohol consumption. Health Psychol. 2002;21:139–46.
  122. 122. Laucht M, Becker K, Blomeyer D, and Schmidt MH. Novelty seeking involved in mediating the association between the dopamine D4 receptor gene exon III polymorphism and heavy drinking in male adolescents: results from a high‐risk community sample. Biol Psychiatry. 2007;61:87–92.
  123. 123. Vaughn MG, Beaver KM, DeLisi M, Howard MO, and Perron BE. Dopamine D4 receptor gene exon III polymorphism associated with binge drinking attitudinal phenotype. Alcohol. 2009;43:179–84.
  124. 124. Gorwood P, Limosin F, Batel P, Hamon M, Ades J, and Boni C. The A9 allele of the dopamine transporter gene is associated with delirium tremens and alcohol‐withdrawal seizure. Biol Psychiatry. 2003;53:85–92.
  125. 125. Vaske J, Beaver KM, Wright JP, Boisvert D, and Schnupp R. An interaction between DAT1 and having an alcoholic father predicts serious alcohol problems in a sample of males. Drug Alcohol Depend. 2009;104:17–22.
  126. 126. Enoch MA, Waheed JF, Harris CR, Albaugh B, and Goldman D. Sex differences in the influence of COMT Val158Met on alcoholism and smoking in plains American Indians. Alcohol Clin Exp Res. 2006;30:399–406.
  127. 127. Tiihonen J, Hallikainen T, Lachman H, Saito T, Volavka J, Kauhanen J, Salonen JT, Ryynanen OP, Koulu M, Karvonen MK, Pohjalainen T, Syvalahti E, and Hietala J. Association between the functional variant of the catechol‐O‐methyltransferase (COMT) gene and type 1 alcoholism. Mol Psychiatry. 1999;4:286–9.
  128. 128. Wang T, Franke P, Neidt H, Cichon S, Knapp M, Lichtermann D, Maier W, Propping P, and Nothen MM. Association study of the low‐activity allele of catechol‐O‐methyltransferase and alcoholism using a family‐based approach. Mol Psychiatry. 2001;6:109–11.
  129. 129. Liljequist S, Berggren U, and Engel J. The effect of catecholamine receptor antagonists on ethanol‐induced locomotor stimulation. J Neural Transm. 1981;50:57–67.
  130. 130. Czachowski CL, Chappell AM, and Samson HH. Effects of raclopride in the nucleus accumbens on ethanol seeking and consumption. Alcohol Clin Exp Res. 2001;25:1431–40.
  131. 131. Czachowski CL, Santini LA, Legg BH, and Samson HH. Separate measures of ethanol seeking and drinking in the rat: effects of remoxipride. Alcohol. 2002;28:39–46.
  132. 132. Files FJ, Denning CE, and Samson HH. Effects of the atypical antipsychotic remoxipride on alcohol self‐administration. Pharmacol Biochem Behav. 1998;59:281–5.
  133. 133. Rassnick S, Pulvirenti L, and Koob GF. SDZ‐205,152, a novel dopamine receptor agonist, reduces oral ethanol self‐administration in rats. Alcohol. 1993;10:127–32.
  134. 134. Pfeffer AO and Samson HH. Haloperidol and apomorphine effects on ethanol reinforcement in free feeding rats. Pharmacol Biochem Behav. 1988;29:343–50.
  135. 135. Maldonado R, Saiardi A, Valverde O, Samad TA, Roques BP, and Borrelli E. Absence of opiate rewarding effects in mice lacking dopamine D2 receptors. Nature. 1997;388:586–9.
  136. 136. Wise RA. Neurobiology of addiction. Curr Opin Neurobiol. 1996;6:243–51.
  137. 137. Brown ZW, Gill K, Abitbol M, and Amit Z. Lack of effect of dopamine receptor blockade on voluntary ethanol consumption in rats. Behav Neural Biol. 1982;36:291–4.
  138. 138. Linseman MA. Effects of dopaminergic agents on alcohol consumption by rats in a limited access paradigm. Psychopharmacology (Berl). 1990;100:195–200.
  139. 139. Pfeffer AO and Samson HH. Effect of pimozide on home cage ethanol drinking in the rat: dependence on drinking session length. Drug Alcohol Depend. 1986;17:47–55.
  140. 140. Samson HH, Hodge CW, Tolliver GA, and Haraguchi M. Effect of dopamine agonists and antagonists on ethanol‐reinforced behavior: the involvement of the nucleus accumbens. Brain Res Bull. 1993;30:133–41.
  141. 141. Hodge CW, Samson HH, and Chappelle AM. Alcohol self‐administration: further examination of the role of dopamine receptors in the nucleus accumbens. Alcohol Clin Exp Res. 1997;21:1083–91.
  142. 142. Nowak KL, McBride WJ, Lumeng L, Li TK, and Murphy JM. Involvement of dopamine D2 autoreceptors in the ventral tegmental area on alcohol and saccharin intake of the alcohol‐preferring P rat. Alcohol Clin Exp Res. 2000;24:476–83.
  143. 143. Price KL and Middaugh LD. The dopamine D1 antagonist reduces ethanol reward for C57BL/6 mice. Alcohol Clin Exp Res. 2004;28:1666–75.
  144. 144. Modell JG, Mountz JM, Glaser FB, and Lee JY. Effect of haloperidol on measures of craving and impaired control in alcoholic subjects. Alcohol Clin Exp Res. 1993;17:234–40.
  145. 145. Wiesbeck GA, Weijers HG, Lesch OM, Glaser T, Toennes PJ, and Boening J. Flupenthixol decanoate and relapse prevention in alcoholics: results from a placebo‐controlled study. Alcohol Alcohol. 2001;36:329–34.
  146. 146. Walter H, Ramskogler K, Semler B, Lesch OM, and Platz W. Dopamine and alcohol relapse: D1 and D2 antagonists increase relapse rates in animal studies and in clinical trials. J Biomed Sci. 2001;8:83–8.
  147. 147. Wiesbeck GA, Weijers HG, Wodarz N, Lesch OM, Glaser T, and Boening J. Gender‐related differences in pharmacological relapse prevention with flupenthixol decanoate in detoxified alcoholics. Arch Womens Ment Health. 2003;6:259–62.
  148. 148. Drake RE, Xie H, McHugo GJ, and Green AI. The effects of clozapine on alcohol and drug use disorders among patients with schizophrenia. Schizophr Bull. 2000;26:441–9.
  149. 149. Soyka M, Aichmuller C, v Bardeleben U, Beneke M, Glaser T, Hornung‐Knobel S, and Wegner U. Flupenthixol in relapse prevention in schizophrenics with comorbid alcoholism: results from an open clinical study. Eur Addict Res. 2003;9:65–72.
  150. 150. Osser DN, Najarian DM, and Dufresne RL. Olanzapine increases weight and serum triglyceride levels. J Clin Psychiatry. 1999;60:767–70.
  151. 151. Sussman N. The implications of weight changes with antipsychotic treatment. J Clin Psychopharmacol. 2003;23:S21–6.
  152. 152. Hutchison KE, Swift R, Rohsenow DJ, Monti PM, Davidson D, and Almeida A. Olanzapine reduces urge to drink after drinking cues and a priming dose of alcohol. Psychopharmacology (Berl). 2001;155:27–34.
  153. 153. Peters DH and Faulds D. Tiapride. A review of its pharmacology and therapeutic potential in the management of alcohol dependence syndrome. Drugs. 1994;47:1010–32.
  154. 154. Ingman K, Honkanen A, Hyytia P, Huttunen MO, and Korpi ER. Risperidone reduces limited access alcohol drinking in alcohol‐preferring rats. Eur J Pharmacol. 2003;468:121–7.
  155. 155. Ingman K and Korpi ER. Alcohol drinking of alcohol‐preferring AA rats is differentially affected by clozapine and olanzapine. Eur J Pharmacol. 2006;534:133–40.
  156. 156. Chau DT, Khokhar JY, Gulick D, Dawson R, and Green AI. Desipramine enhances the ability of paliperidone to decrease alcohol drinking. J Psychiatr Res. 2015;69:9–18.
  157. 157. Chau DT, Khokhar JY, Dawson R, Ahmed J, Xie H, and Green AI. The comparative effects of clozapine versus haloperidol on initiation and maintenance of alcohol drinking in male alcohol‐preferring P rat. Alcohol. 2013;47:611–8.
  158. 158. Shaw GK, Majumdar SK, Waller S, MacGarvie J, and Dunn G. Tiapride in the long‐term management of alcoholics of anxious or depressive temperament. Br J Psychiatry. 1987;150:164–8.
  159. 159. Shaw GK, Waller S, Majumdar SK, Alberts JL, Latham CJ, and Dunn G. Tiapride in the prevention of relapse in recently detoxified alcoholics. Br J Psychiatry. 1994;165:515–23.
  160. 160. Croissant B, Klein O, Gehrlein L, Kniest A, Hermann D, Diehl A, and Mann K. Quetiapine in relapse prevention in alcoholics suffering from craving and affective symptoms: a case series. Eur Psychiatry. 2006;21:570–3.
  161. 161. Martinotti G, Di Nicola M, Romanelli R, Andreoli S, Pozzi G, Moroni N, and Janiri L. High and low dosage oxcarbazepine versus naltrexone for the prevention of relapse in alcohol‐dependent patients. Hum Psychopharmacol. 2007;22:149–56.
  162. 162. Kampman KM, Pettinati HM, Lynch KG, Whittingham T, Macfadden W, Dackis C, Tirado C, Oslin DW, Sparkman T, and O'Brien CP. A double‐blind, placebo‐controlled pilot trial of quetiapine for the treatment of Type A and Type B alcoholism. J Clin Psychopharmacol. 2007;27:344–51.
  163. 163. Brown ES, Davila D, Nakamura A, Carmody TJ, Rush AJ, Lo A, Holmes T, Adinoff B, Caetano R, Swann AC, Sunderajan P, and Bret ME. A randomized, double‐blind, placebo‐controlled trial of quetiapine in patients with bipolar disorder, mixed or depressed phase, and alcohol dependence. Alcohol Clin Exp Res. 2014;38:2113–8.
  164. 164. Litten RZ, Fertig JB, Falk DE, Ryan ML, Mattson ME, Collins JF, Murtaugh C, Ciraulo D, Green AI, Johnson B, Pettinati H, Swift R, Afshar M, Brunette MF, Tiouririne NA, Kampman K, Stout R, and Group NS. A double‐blind, placebo‐controlled trial to assess the efficacy of quetiapine fumarate XR in very heavy‐drinking alcohol‐dependent patients. Alcohol Clin Exp Res. 2012;36:406–16.
  165. 165. Hutchison KE, Ray L, Sandman E, Rutter MC, Peters A, Davidson D, and Swift R. The effect of olanzapine on craving and alcohol consumption. Neuropsychopharmacology. 2006;31:1310–7.
  166. 166. Hutchison KE, Wooden A, Swift RM, Smolen A, McGeary J, Adler L, and Paris L. Olanzapine reduces craving for alcohol: a DRD4 VNTR polymorphism by pharmacotherapy interaction. Neuropsychopharmacology. 2003;28:1882–8.
  167. 167. Kishi T, Sevy S, Chekuri R, and Correll CU. Antipsychotics for primary alcohol dependence: a systematic review and meta‐analysis of placebo‐controlled trials. J Clin Psychiatry. 2013;74:e642–54.
  168. 168. Ucok A and Gaebel W. Side effects of atypical antipsychotics: a brief overview. World Psychiatry. 2008;7:58–62.
  169. 169. Rassnick S, Krechman J, and Koob GF. Chronic ethanol produces a decreased sensitivity to the response‐disruptive effects of GABA receptor complex antagonists. Pharmacol Biochem Behav. 1993;44:943–50.
  170. 170. Carnicella S, Ahmadiantehrani S, He DY, Nielsen CK, Bartlett SE, Janak PH, and Ron D. Cabergoline decreases alcohol drinking and seeking behaviors via glial cell line‐derived neurotrophic factor. Biol Psychiatry. 2009;66:146–53.
  171. 171. Weiss F, Mitchiner M, Bloom FE, and Koob GF. Free‐choice responding for ethanol versus water in alcohol preferring (P) and unselected Wistar rats is differentially modified by naloxone, bromocriptine, and methysergide. Psychopharmacology (Berl). 1990;101:178–86.
  172. 172. Lawford BR, Young RM, Rowell JA, Qualichefski J, Fletcher BH, Syndulko K, Ritchie T, and Noble EP. Bromocriptine in the treatment of alcoholics with the D2 dopamine receptor A1 allele. Nat Med. 1995;1:337–41.
  173. 173. Naranjo CA, Dongier M, and Bremner KE. Long‐acting injectable bromocriptine does not reduce relapse in alcoholics. Addiction. 1997;92:969–78.
  174. 174. Powell BJ, Campbell JL, Landon JF, Liskow BI, Thomas HM, Nickel EJ, Dale TM, Penick EC, Samuelson SD, and Lacoursiere RB. A double‐blind, placebo‐controlled study of nortriptyline and bromocriptine in male alcoholics subtyped by comorbid psychiatric disorders. Alcohol Clin Exp Res. 1995;19:462–8.
  175. 175. Burris KD, Molski TF, Xu C, Ryan E, Tottori K, Kikuchi T, Yocca FD, and Molinoff PB. Aripiprazole, a novel antipsychotic, is a high‐affinity partial agonist at human dopamine D2 receptors. J Pharmacol Exp Ther. 2002;302:381–9.
  176. 176. Carlsson A, Waters N, Holm‐Waters S, Tedroff J, Nilsson M, and Carlsson ML. Interactions between monoamines, glutamate, and GABA in schizophrenia: new evidence. Annu Rev Pharmacol Toxicol. 2001;41:237–60.
  177. 177. Yokoi F, Grunder G, Biziere K, Stephane M, Dogan AS, Dannals RF, Ravert H, Suri A, Bramer S, and Wong DF. Dopamine D2 and D3 receptor occupancy in normal humans treated with the antipsychotic drug aripiprazole (OPC 14597): a study using positron emission tomography and [11C]raclopride. Neuropsychopharmacology. 2002;27:248–59.
  178. 178. Jerlhag E. The antipsychotic aripiprazole antagonizes the ethanol‐ and amphetamine‐induced locomotor stimulation in mice. Alcohol. 2008;42:123–7.
  179. 179. Viana TG, Almeida‐Santos AF, Aguiar DC, and Moreira FA. Effects of aripiprazole, an atypical antipsychotic, on the motor alterations induced by acute ethanol administration in mice. Basic Clin Pharmacol Toxicol. 2013;112:319–24.
  180. 180. Ingman K, Kupila J, Hyytia P, and Korpi ER. Effects of aripiprazole on alcohol intake in an animal model of high‐alcohol drinking. Alcohol Alcohol. 2006;41:391–8.
  181. 181. Nirogi R, Kandikere V, Jayarajan P, Bhyrapuneni G, Saralaya R, Muddana N, and Abraham R. Aripiprazole in an animal model of chronic alcohol consumption and dopamine D(2) receptor occupancy in rats. Am J Drug Alcohol Abuse. 2013;39:72–9.
  182. 182. Shibasaki M, Kurokawa K, Mizuno K, and Ohkuma S. Effect of aripiprazole on anxiety associated with ethanol physical dependence and on ethanol‐induced place preference. J Pharmacol Sci. 2012;118:215–24.
  183. 183. Janiri L, Martinotti G, and Di Nicola M. Aripiprazole for relapse prevention and craving in alcohol‐dependent subjects: results from a pilot study. J Clin Psychopharmacol. 2007;27:519–20.
  184. 184. Anton RF, Kranzler H, Breder C, Marcus RN, Carson WH, and 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;28:5–12.
  185. 185. Voronin K, Randall P, Myrick H, and Anton R. Aripiprazole effects on alcohol consumption and subjective reports in a clinical laboratory paradigm—possible influence of self‐control. Alcohol Clin Exp Res. 2008;32:1954–61.
  186. 186. Martinotti G, Di Nicola M, and Janiri L. Efficacy and safety of aripiprazole in alcohol dependence. Am J Drug Alcohol Abuse. 2007;33:393–401.
  187. 187. Martinotti G, Di Nicola M, Di Giannantonio M, and Janiri L. Aripiprazole in the treatment of patients with alcohol dependence: a double‐blind, comparison trial vs. naltrexone. J Psychopharmacol. 2009;23:123–9.
  188. 188. Myrick H, Li X, Randall PK, Henderson S, Voronin K, and Anton RF. The effect of aripiprazole on cue‐induced brain activation and drinking parameters in alcoholics. J Clin Psychopharmacol. 2010;30:365–72.
  189. 189. Carlsson ML, Carlsson A, and Nilsson M. Schizophrenia: from dopamine to glutamate and back. Curr Med Chem. 2004;11:267–77.
  190. 190. Tedroff J, Torstenson R, Hartvig P, Sonesson C, Waters N, Carlsson A, Neu H, Fasth KJ, and Langstrom B. Effects of the substituted (S)‐3‐phenylpiperidine (‐)‐OSU6162 on PET measurements in subhuman primates: evidence for tone‐dependent normalization of striatal dopaminergic activity. Synapse. 1998;28:280–7.
  191. 191. Kara E, Lin H, Svensson K, Johansson AM, and Strange PG. Analysis of the actions of the novel dopamine receptor‐directed compounds (S)‐OSU6162 and ACR16 at the D2 dopamine receptor. Br J Pharmacol. 2010;161:1343–50.
  192. 192. Seeman P and Guan HC. Dopamine partial agonist action of (‐)OSU6162 is consistent with dopamine hyperactivity in psychosis. Eur J Pharmacol. 2007;557:151–3.
  193. 193. Lahti RA, Tamminga CA, and Carlsson A. Stimulating and inhibitory effects of the dopamine “stabilizer” (‐)‐OSU6162 on dopamine D2 receptor function in vitro. J Neural Transm (Vienna). 2007;114:1143–6.
  194. 194. Rung JP, Rung E, Helgeson L, Johansson AM, Svensson K, Carlsson A, and Carlsson ML. Effects of (‐)‐OSU6162 and ACR16 on motor activity in rats, indicating a unique mechanism of dopaminergic stabilization. J Neural Transm (Vienna). 2008;115:899–908.
  195. 195. Sonesson C, Lin CH, Hansson L, Waters N, Svensson K, Carlsson A, Smith MW, and Wikstrom H. Substituted (S)‐phenylpiperidines and rigid congeners as preferential dopamine autoreceptor antagonists: synthesis and structure‐activity relationships. J Med Chem. 1994;37:2735–53.
  196. 196. Steensland P, Fredriksson I, Holst S, Feltmann K, Franck J, Schilstrom B, and Carlsson A. The monoamine stabilizer (‐)‐OSU6162 attenuates voluntary ethanol intake and ethanol‐induced dopamine output in nucleus accumbens. Biol Psychiatry. 2012; 72:823-31
  197. 197. Khemiri L, Steensland P, Guterstam J, Beck O, Carlsson A, Franck J, and Jayaram‐Lindstrom N. The effects of the monoamine stabilizer (‐)‐OSU6162 on craving in alcohol dependent individuals: a human laboratory study. Eur Neuropsychopharmacol. 2015;25:2240–51.
  198. 198. Johansson B, Carlsson A, Carlsson ML, Karlsson M, Nilsson MK, Nordquist‐Brandt E, and Ronnback L. Placebo‐controlled cross‐over study of the monoaminergic stabiliser (‐)‐OSU6162 in mental fatigue following stroke or traumatic brain injury. Acta Neuropsychiatr. 2012;24:266–74.
  199. 199. Kloberg A, Constantinescu R, Nilsson MK, Carlsson ML, Carlsson A, Wahlstrom J, and Haghighi S. Tolerability and efficacy of the monoaminergic stabilizer (‐)‐OSU6162 (PNU‐96391A) in Huntington's disease: a double‐blind cross‐over study. Acta Neuropsychiatr. 2014;26:298–306.
  200. 200. Lido HH, Ericson M, Marston H, and Soderpalm B. A role for accumbal glycine receptors in modulation of dopamine release by the glycine transporter‐1 inhibitor org25935. Front Psychiatry. 2011;2:8.
  201. 201. Lido HH, Stomberg R, Fagerberg A, Ericson M, and Soderpalm B. The glycine reuptake inhibitor org 25935 interacts with basal and ethanol‐induced dopamine release in rat nucleus accumbens. Alcohol Clin Exp Res. 2009;33:1151–7.
  202. 202. Molander A, Lido HH, Lof E, Ericson M, and Soderpalm B. The glycine reuptake inhibitor Org 25935 decreases ethanol intake and preference in male wistar rats. Alcohol Alcohol. 2007;42:11–8.
  203. 203. Vengeliene V, Leonardi‐Essmann F, Sommer WH, Marston HM, and Spanagel R. Glycine transporter‐1 blockade leads to persistently reduced relapse‐like alcohol drinking in rats. Biol Psychiatry. 2010;68:704–11.
  204. 204. Larsson A and Engel JA. Neurochemical and behavioral studies on ethanol and nicotine interactions. Neurosci Biobehav Rev. 2004;27:713–720.
  205. 205. Steensland P, Simms JA, Holgate J, Richards JK, and Bartlett SE. Varenicline, an alpha4beta2 nicotinic acetylcholine receptor partial agonist, selectively decreases ethanol consumption and seeking. Proc Natl Acad Sci USA. 2007;104:12518–23.
  206. 206. Ericson M, Lof E, Stomberg R, and Soderpalm B. The smoking cessation medication varenicline attenuates alcohol and nicotine interactions in the rat mesolimbic dopamine system. J Pharmacol Exp Ther. 2009;329:225–30.
  207. 207. de Bejczy A, Nations KR, Szegedi A, Schoemaker J, Ruwe F, and Soderpalm B. Efficacy and safety of the glycine transporter‐1 inhibitor org 25935 for the prevention of relapse in alcohol‐dependent patients: a randomized, double‐blind, placebo‐controlled trial. Alcohol Clin Exp Res. 2014;38:2427–35.
  208. 208. Fucito LM, Toll BA, Wu R, Romano DM, Tek E, and O'Malley SS. A preliminary investigation of varenicline for heavy drinking smokers. Psychopharmacology (Berl). 2011;215:655–63.
  209. 209. McKee SA, Harrison EL, O'Malley SS, Krishnan‐Sarin S, Shi J, Tetrault JM, Picciotto MR, Petrakis IL, Estevez N, and Balchunas E. Varenicline reduces alcohol self‐administration in heavy‐drinking smokers. Biol Psychiatry. 2009;66:185–90.
  210. 210. Mitchell JM, Teague CH, Kayser AS, Bartlett SE, and Fields HL. Varenicline decreases alcohol consumption in heavy‐drinking smokers. Psychopharmacology (Berl). 2012;223:299–306.
  211. 211. Litten RZ, Ryan ML, Fertig JB, Falk DE, Johnson B, Dunn KE, Green AI, Pettinati HM, Ciraulo DA, Sarid‐Segal O, Kampman K, Brunette MF, Strain EC, Tiouririne NA, Ransom J, Scott C, and Stout R. A double‐blind, placebo‐controlled trial assessing the efficacy of varenicline tartrate for alcohol dependence. J Addict Med. 2013;7:277-86.
  212. 212. de Bejczy A, Lof E, Walther L, Guterstam J, Hammarberg A, Asanovska G, Franck J, Isaksson A, and Soderpalm B. Varenicline for treatment of alcohol dependence: a randomized, placebo‐controlled trial. Alcohol Clin Exp Res. 2015;39:2189–99.
  213. 213. Schacht JP, Anton RF, Randall PK, Li X, Henderson S, and Myrick H. Varenicline effects on drinking, craving and neural reward processing among non‐treatment‐seeking alcohol‐dependent individuals. Psychopharmacology (Berl). 2014;231:3799–807.
  214. 214. Walther L, de Bejczy A, Lof E, Hansson T, Andersson A, Guterstam J, Hammarberg A, Asanovska G, Franck J, Soderpalm B, and Isaksson A. Phosphatidylethanol is superior to carbohydrate‐deficient transferrin and gamma‐glutamyltransferase as an alcohol marker and is a reliable estimate of alcohol consumption level. Alcohol Clin Exp Res. 2015;39:2200–8.

Written By

Nitya Jayaram‐Lindström, Mia Ericson, Pia Steensland and Elisabet Jerlhag

Reviewed: 16 March 2016 Published: 14 July 2016