key: cord-0907478-iiim1tmh authors: Ryabinin, Andrey E.; Zhang, Yangmiao title: Barriers and Breakthroughs in Targeting the Oxytocin System to Treat Alcohol Use Disorder date: 2022-02-28 journal: Front Psychiatry DOI: 10.3389/fpsyt.2022.842609 sha: bc23cca374f92ac56395a05766d09e6286b44be0 doc_id: 907478 cord_uid: iiim1tmh Development of better treatments for alcohol use disorder (AUD) is urgently needed. One promising opportunity for this development is the potential of targeting the oxytocin peptide system. Preclinical studies showed that administration of exogenous oxytocin or, more recently, stimulation of neurons expressing endogenous oxytocin lead to a decreased alcohol consumption across several rodent models. Initial clinical studies also showed that administration of oxytocin decreased craving for alcohol and heavy alcohol drinking. However, several more recent clinical studies were not able to replicate these effects. Thus, although targeting the oxytocin system holds promise for the treatment of AUD, more nuanced approaches toward development and application of these treatments are needed. In this mini-review we discuss potential caveats resulting in differential success of attempts to use oxytocin for modulating alcohol use disorder-related behaviors in clinical studies and evaluate three directions in which targeting the oxytocin system could be improved: (1) increasing potency of exogenously administered oxytocin, (2) developing oxytocin receptor agonists, and (3) stimulating components of the endogenous oxytocin system. Both advances and potential pitfalls of these directions are discussed. Alcohol use disorder (AUD) is a devastating condition where the affected individuals continue to engage in drinking despite the negative experience and the harm caused by drinking. Globally, more than 5% of all deaths have been attributed to alcohol consumption (1) . In addition to alcohol-related mortality, AUD is associated with a plethora of debilitating health and societal consequences. Harmful alcohol consumption increased during the COVID-19 pandemic (2) (3) (4) . Better treatments for AUD with better access to these treatments are urgently needed. Among potential targets for such treatments, recent attention has focused on the oxytocin (OXT) neuropeptide system. OXT has a leading role in coordinating maternal behaviors, and regulates other functions, including social attachment, anxiety, food consumption, learning, body temperature, neuroinflammation, and pain (5, 6) . OXT has been suggested to modulate alcoholrelated responses including craving, tolerance, abstinence, and withdrawal-induced anxiety and social engagement (7) (8) (9) (10) . In turn, alcohol can affect OXT levels and underlying neurocircuitry (11) (12) (13) . These observations suggested that stimulating the OXT system could curb excessive alcohol use by targeting several phases of the addiction cycle: modulating alcohol's effects during the intoxication phase, decreasing alcohol's toxicity during the abstinence/withdrawal phase and/or decreasing craving during the anticipation/preoccupation phase (14) (15) (16) (17) . Indeed, OXT administration decreased alcohol self-administration in numerous preclinical studies (16, (18) (19) (20) (21) . Importantly, OXT was effective in translationally relevant animal models, including being administered via intranasal (IN) routes, in models of severe alcohol dependence (22) , in prairie voles, which share similarities in neurocircuitry regulating social behaviors (21) , and in presence of untreated peers, similarly to medication administration in outpatient clinics (23, 24) . Initial clinical studies also indicated that OXT administration in humans could decrease signs of withdrawal and craving and inhibit excessive alcohol drinking (12, 17, 25) . However, more recent studies had limited success (26) (27) (28) , calling for more nuanced approaches for the treatment of AUD with OXT. Moreover, recent basic science studies revised our understanding of organization of the OXT system (29) and exogenous OXT's penetrance into the brain (30, 31) . With these developments, the goal of this minireview is to critically analyze the potential pitfalls associated with clinical studies testing effects of OXT on signs of AUD ( Table 1 ) and to evaluate different directions of overcoming these pitfalls: by increasing potency of exogenously administered OXT, by developing OXT receptor (OXTR) agonists, and by stimulating the endogenous OXT system (Figure 1 ). So far, 11 clinical studies have been published on effects of IN OXT on alcohol drinking-related behaviors, and one study on social stress response but not drinking-related behaviors in individuals with AUD (38) ( Table 1) . These studies assessed effects of OXT treatments ranging from single to 12 weeklong daily administrations across populations in three different countries, in subjects characterized as social drinkers, heavy drinkers or patients with post-traumatic stress disorder/AUD comorbidity, with modest sample sizes ranging from 11 to 84. Only half of the studies evaluated effects of OXT in both sexes, and most studies focused on males only. Evaluating Table 1 , one can conclude that effectiveness of OXT to decrease relevant outcomes was not overwhelming, revealing the following potential pitfalls. The majority of studies that looked at short-term outcomes failed to observe decrease in drinking. The only study that reported decreased drinking administered OXT for the longest time (25) . Importantly, decreased measures of intake in this study only started to emerge on the second week of treatment. Therefore, the usual duration of OXT treatment and observation in most current studies might be insufficient to show its effect. On the other hand, effects of OXT on measures of heavy drinking in this study appeared to be subsiding in the last weeks of treatment, suggesting either a desensitization to repeated treatment or a "floor effect" of OXT. It appears that more recent studies, showing less consistent effects, used slightly lower doses of OXT per day. One study relied on subjects' self-administration of OXT, which might have not achieved effective uptake (27) . It is important to note that in some of the studies, OXT did not decrease measures of alcohol craving or drinking despite having significant effects on measures of stress or anxiety, arguing against insufficient OXT uptake (26, 27) . On the other hand, it could be that higher doses are needed to decrease alcohol drinking than doses that can affect stress or anxiety. Another consideration is that OXT could be effective only in a subset of patients. One study indicated that OXT can even increase craving in subjects with low attachment anxiety (32) . Thus, subpopulations of subjects can be differentially sensitive to OXT treatment, requiring bigger sample sizes to achieve the power necessary to detect effects of these treatments. In studies listed in Table 1 , five studies observed decreases in craving, alcohol cue response or heavy alcohol use (12, 17, 25, 33, 37) , and the other five did not observe significant effects (26) (27) (28) (34) (35) (36) . Since only one study reported that OXT produced opposite from hypothesized effects in a subpopulation of subjects (32), we conservatively interpret this analysis that OXT can be effective in decreasing alcohol craving or intake, but its potency needs to be enhanced. While observations of positive effects of OXT on alcohol drinking in clinical studies are not consistent, this contrasts with a large body of preclinical studies showing effects. Decreased alcohol intake following OXT administration has been observed in mice, rats, and prairie voles. These studies have been previously reviewed (8, 9, 14, 15, 25, (39) (40) (41) . The success of preclinical studies may offer clues and opportunities to potentiate the effect of OXT treatment in humans. Non-human studies have explored both peripheral (e.g., subcutaneous, intraperitoneal, and IN) and central routes of administration of OXT to understand its mechanisms of action. Since OXT's half-life in plasma is estimated to be on the order of minutes (42, 43) , the observation that peripherally administered OXT had longer effects than expected from its half-life was attributed to the potential feed-forward release of OXT from magnocellular neurons (44, 45) . To overcome the difficulty interpreting results from peripherally administered OXT, some investigators resorted to study its effects following intracranial administration (12, 46, 47) . While these studies demonstrated OXT's central modulation of alcohol-related behaviors, they did not clarify how the peptide reaches the relevant intracranial sites. More recent studies indicated that exogenously administered OXT indeed reaches selected brain regions in mice, rats, monkeys and humans (30, (48) (49) (50) (51) (52) . Since OXT has additional affinity to vasopressin (AVP) receptors, it is worth noting that the exogenously administered and centrally active OXT could also exert its functions via actions that do not involve OXTRs (29, 53, 54) . The IN route results in penetration of a portion of total administered OXT to the brain via several pathways: transmission along the olfactory and trigeminal neural bundles, absorption from basal epithelia directly into cerebrospinal fluid, crossing the blood-brain barrier, and through circumventricular organs (6, 52) . The different relative size of the neural bundles vs. whole brain across species might contribute to differences in the access to specific brain regions in rodents vs. humans. Nevertheless, attempts to increase penetration of IN OXT are underway, including the use of mucoadhesives, nanoemulsions and liposomal formulations (6, 55) . Another development is the recent recognition that brain penetrance of OXT is aided by the participation of the Receptor for Advanced Glycation Endproducts (RAGE) (31, 56) . Development of allosteric modulators of RAGE could be an interesting approach to enhance the potency of exogenous OXT. Finally, as mentioned, OXT could be differentially effective across different populations of subjects. OXT was found to differentially affect alcohol craving in subjects with low and high attachment anxiety (32) . Several indicators suggest that it could be more effective in males vs. females (57) . Moreover, subjects carrying polymorphisms in OXTR and OXT show differences in alcohol consumption patterns (58, 59) . Carriers of these polymorphisms could be differentially sensitive to OXT treatments. Studies targeting these subpopulations could be a step toward development of individualized AUD treatments. OXT's lack of stability, low brain penetrance and non-specific effects on AVP receptors prompted researchers to consider developing OXTR agonists (14, 60) . The majority of developed and tested OXTR agonist are peptides, with presumably minimal penetrance of the blood-brain barrier. Because of this, peptidebased OXTR agonist PF-06655075 was used to test whether effects from manipulating the OXT system are through central or peripheral OXTRs (22) . This is an important consideration as stimulation of peripheral OXTR can affect certain behaviors, for example fear conditioning (61) . However, central, but not peripheral administration of PF-06655075 decreased alcohol drinking in dependent rats, indicating that stimulation of the central OXT system is crucial for moderating alcohol consumption (22) . Carbetocin is a highly stable OXT analog used in clinics to prevent postpartum hemorrhage. It has been demonstrated to modulate central effects of morphine (62, 63) . In relation to alcohol, however, reported effects are mixed. While the same dose of repeated carbetocin inhibited acquisition, enhanced extinction and suppressed reinstatement of ethanol-induced conditioned place preference in a mouse study (64) , it enhanced ethanolinduced conditioned place preference in another study (65) . We are not aware of any reports on carbetocin's effects on alcohol drinking. Other peptide OXTR agonists include Lipo-oxytocin-1 and [Thr 4 ,Gly 7 ]OXT (66) (67) (68) . Their effect on alcohol-related behaviors have not been reported, but the strong possibility of effects on various peripheral systems could be a matter of concern. Another possibility is to explore the effects of OXT metabolites-OXT fragments composed of shorter chained peptides. As smaller molecules, they should have better penetrance into the brain and, therefore, higher potency. In contrast to carbetocin, OXT (4-9) improved social preference in the BALBc/J model of autism spectrum disorder-like social deficits (69) . In early studies, the tripeptide OXT (6-9) was shown to attenuate tolerance to hypothermic effects of repeated ethanol injections in mice (70) . However, whether OXT metabolites can affect alcohol drinking or alcohol reward is still to be discovered. Development of non-peptide OXTR agonists continues to progress. Three small-molecule agonists have been reported (14, 60, 71, 72) . TC-OT-39 has an EC 50 of ∼100 nM at OXTR as an agonist, but also a moderate affinity at V1a AVP receptors as an antagonist. Therefore, its neurochemical mode of action in vivo is difficult to estimate. In any case, it was not effective in studies of BALBc/J model of autism spectrum disorder-like social deficits (69) . Comparing to TC-OT-39, WAY-267464 is a level of magnitude more potent OXTR agonist, but with a similar affinity to V1a AVP receptors (71) , again making it difficult to evaluate specificity of its effects after systemic administration. Promisingly, however, administration of this compound into amygdala facilitated extinction of fear conditioning (73) whereas systemic injection of this compound rescued not only social deficits in Shank3b mutant mice (74) , but also social deficits caused by adolescent alcohol exposure in rats (75) . LIT-001, the most recently developed small molecule, has an agonist activity on OXTR similar to that of WAY-267464 in in vitro studies, but several magnitudes lower affinity to AVP receptors, for the first time allowing receptor-specific systemic pharmacology of OXTR (71) . Systemic administration of LIT-001 rescued deficits in social interaction in the Oprm1 knockout model of autism spectrum disorder (71) and inhibited inflammation-induced hyperalgesia in rats (76) . Studies testing LIT-001 in models of AUD are underway. Development of small-molecule OXTR agonists is a promising avenue for future research. Though striving to develop compounds with more specific and potent effects than OXT, one has to keep in mind the potential side effects. For example, a compound that has potent anxiolytic and analgesic properties must be evaluated for its own potential addictive properties. OXT is an evolutionary old peptide. Therefore, its mixed affinities toward OXTR and AVP receptors, as well as its poor brainpenetrance, could have advantages for fitness. It might be, therefore, more prudent to stimulate the endogenous OXT system to produce beneficial effects instead of manipulating the system exogenously. As a proof of this concept, a recent study by King et al. used a chemogenetic approach to activate OXT neurons of the mouse paraventricular nucleus of hypothalamus (PVN) (77) . This activation significantly decreased alcohol consumption in a mouse model of binge drinking. This finding implicates the need to explore the feasibility to develop effective means of stimulating the endogenous central OXT system for AUD treatment. The importance of OXT in regulating social attachments is well-demonstrated across vertebrate species (78) (79) (80) (81) (82) (83) . Positive social interactions are known to stimulate OXT neurons (84) (85) (86) (87) . Therefore, social interactions could stimulate the OXT system and hence, decrease alcohol consumption. However, the relationship between social interaction and activity of OXT system is more complex. OXT neurons are also activated during social isolation and after exposure to other stressors (88) (89) (90) . Stressful experiences are well-known to increase alcohol consumption (7, 91, 92) . Therefore, it would be more relevant to selectively stimulate OXT system in a way that mimics the effect of positive social interactions to regulate AUDrelated phenotypes. The latter task is extremely difficult because there are currently no methods allowing to evaluate when specific components of the central OXT system are activated in humans. Our understanding of the relationship between the activation of subpopulations of OXT neurons and OXT's peripheral levels continues to change, and knowledge of projections sites of parvocellular and magnocellular OXT neurons are being revised (29) . Additionally, it is difficult to estimate the relationship between levels of OXT in plasma vs. levels in the brain. A meta-analysis across 17 studies found no significant associations between peripheral and central-in either cerebrospinal fluid or specific brain regions-OXT levels under basal conditions in both humans and non-human animals (93) . On the other hand, this metaanalysis confirmed significant positive associations between these levels after IN OXT in humans and rodents (49, 94, 95) and after experimental stress in rodents (93, (96) (97) (98) , indicating that correspondence in OXT levels can be established. Recent studies also complicate the interpretation of previous studies showing that alcohol consumption decreases OXT levels (9) . For instance, while one study showed significantly higher OXT levels in blood of patients with AUD (99), another study found significantly lower OXT levels in AUD patients (100). The reason for such opposite results needs to be further studied. Taken together, stimulation of endogenous OXT system could contribute to treatments of AUD by modulating social relationships or adherence to therapy. This potential could be especially relevant to group-based approaches to treatment. However, direct behavioral manipulation of this system to moderate excessive alcohol intake in AUD patients at this point is still a theoretical goal. Its realization relies on better understanding of the OXT system. While recent clinical studies show mixed results in the effectiveness of IN OXT to decrease excessive drinking, more sophisticated approaches targeting the OXT system are needed. One possibility is to enhance access of exogenously administered OXT to relevant brain regions either through development of better delivery systems or through enhancement of transporters. Another possibility is to further explore the development of OXTR agonists. The potential of behavioral stimulation of parts of the endogenous OXT system should also be investigated. The better we understand the organization of the OXT system, the greater chance we have of harnessing it for the treatment of AUD. AR and YZ conceptualized and wrote the manuscript. All authors contributed to the article and approved the submitted version. This work was supported by NIH R01 grants (AA019793, AA025024, and AA028680) to AR. Changes in alcohol use during COVID-19 and associations with contextual and individual difference variables: a systematic review and metaanalysis Eating behaviour changes during the COVID-19 pandemic: a systematic review of longitudinal studies Associations between substance use problems and stress during COVID-19 Is oxytocin "nature's medicine"? Brain oxytocin: how puzzle stones from animal studies translate into psychiatry Drug addiction: hyperkatifeia/negative reinforcement as a framework for medications development Oxytocin for the treatment of drug and alcohol use disorders Alcohol and oxytocin: scrutinizing the relationship The inhibitory effect of ethanol on the release of oxytocin during parturition in the rabbit Oxytocin reduces alcohol cue-reactivity in alcoholdependent rats and humans Alcohol consumption decreases oxytocin neurons in the anterior paraventricular nucleus of the hypothalamus in prairie voles Oxytocin signaling in the treatment of drug addiction: therapeutic opportunities and challenges Oxytocin and rodent models of addiction Breaking the loop: oxytocin as a potential treatment for drug addiction Intranasal oxytocin blocks alcohol withdrawal in human subjects Oxytocin reduces ethanol self-administration in mice Peripheral oxytocin administration reduces ethanol consumption in rats Differential effects of baclofen and oxytocin on the increased ethanol consumption following chronic psychosocial stress in mice Oxytocin reduces alcohol consumption in prairie voles Oxytocin blocks enhanced motivation for alcohol in alcohol dependence and blocks alcohol effects on GABAergic transmission in the central amygdala Temporal analysis of individual ethanol consumption in socially housed mice and the effects of oxytocin Assessing effects of oxytocin on alcohol consumption in socially housed prairie voles using radio frequency tracking Oxytocin, tolerance, and the dark side of addiction Effects of oxytocin on stress reactivity and craving in veterans with co-occurring PTSD and alcohol use disorder Efficacy of self-administered intranasal oxytocin on alcohol use and craving after detoxification in patients with alcohol dependence. A double-blind placebo-controlled trial Intranasal oxytocin does not modulate responses to alcohol in social drinkers The multiple faces of the oxytocin and vasopressin systems in the brain Labeled oxytocin administered via the intranasal route reaches the brain in rhesus macaques RAGE regulates oxytocin transport into the brain Intranasal oxytocin selectively modulates social perception, craving, and approach behavior in subjects with alcohol use disorder Oxytocin modulates alcohol-cue induced functional connectivity in the nucleus accumbens of social drinkers Oxytocin-enhanced motivational interviewing group therapy for methamphetamine use disorder in men who have sex with men: study protocol for a randomized controlled trial Effect of intranasal oxytocin on alcohol withdrawal syndrome: a randomized placebocontrolled double-blind clinical trial Actigraphy assessment of motor activity and sleep in patients with alcohol withdrawal syndrome and the effects of intranasal oxytocin Oxytocin attenuates neural response to emotional faces in social drinkers: an fMRI study Craving moderates the effects of intranasal oxytocin on anger in response to social stress among veterans with co-occurring posttraumatic stress disorder and alcohol use disorder Rebalancing the addicted brain: oxytocin interference with the neural substrates of addiction Oxytocin signaling as a target to block social defeat-induced increases in drug abuse reward The potential role of oxytocin in addiction: what is the target process? Penetration of neurohypophyseal hormones from plasma into cerebrospinal fluid (CSF): half-times of disappearance of these neuropeptides from CSF Half-life of oxytoxin in blood of pregnant and non-pregnant woman Adolescent oxytocin exposure causes persistent reductions in anxiety and alcohol consumption and enhances sociability in rats Central effects of oxytocin Oxytocin attenuates stress-induced reinstatement of alcohol seeking behavior in male and female mice Oxytocin inhibits ethanol consumption and ethanol-induced dopamine release in the nucleus accumbens Oxytocin by intranasal and intravenous routes reaches the cerebrospinal fluid in rhesus macaques: determination using a novel oxytocin assay Increased brain and plasma oxytocin after nasal and peripheral administration in rats and mice A spatiotemporal profile of in vivo cerebral blood flow changes following intranasal oxytocin in humans Oxytocin delivered nasally or intraperitoneally reaches the brain and plasma of normal and oxytocin knockout mice Nasal oxytocin for the treatment of psychiatric disorders and pain: achieving meaningful brain concentrations Oxytocin prevents ethanol actions at delta subunit-containing GABAA receptors and attenuates ethanol-induced motor impairment in rats Comparative perspectives on oxytocin and vasopressin receptor research in rodents and primates: translational implications Intranasal administration for pain: oxytocin and other polypeptides Vascular RAGE transports oxytocin into the brain to elicit its maternal bonding behaviour in mice No changes in the oxytocin system in alcohol-dependent female rodents and humans: towards a sexspecific psychopharmacology in alcoholism Oxytocin receptor gene variation rs53576 and alcohol abuse in a longitudinal population representative study Association between oxytocin and receptor genetic polymorphisms and aggression in a northern Chinese Han population with alcohol dependence Role of oxytocin and vasopressin in neuropsychiatric disorders: therapeutic potential of agonists and antagonists Peripheral administration of a long-acting peptide oxytocin receptor agonist inhibits fear-induced freezing The oxytocin analogue carbetocin prevents priminginduced reinstatement of morphine-seeking: involvement of dopaminergic, noradrenergic and MOPr systems The oxytocin analogue carbetocin prevents emotional impairment and stress-induced reinstatement of opioidseeking in morphine-abstinent mice The oxytocin receptor impairs ethanol reward in mice Environmental enrichment enhances conditioned place preference to ethanol via an oxytocinergic-dependent mechanism in male mice Selective and potent agonists and antagonists for investigating the role of mouse oxytocin receptors Effects of three lipidated oxytocin analogs on behavioral deficits in CD38 knockout mice Mice heterozygous for the oxytocin receptor gene (Oxtr(+/-)) show impaired social behaviour but not increased aggression or cognitive inflexibility: evidence of a selective haploinsufficiency gene effect Prosocial effects of an oxytocin metabolite, but not synthetic oxytocin receptor agonists, in a mouse model of autism C-terminal fragments of oxytocin (prolyl-leucyl-glycinamide and Z-prolyl-D-leucine) attenuate the development of tolerance to ethanol LIT-001, the first nonpeptide oxytocin receptor agonist that improves social interaction in a mouse model of autism Oxytocinergic manipulations in corticolimbic circuit differentially affect fear acquisition and extinction Social stimuli induce activation of oxytocin neurons within the paraventricular nucleus of the hypothalamus to promote social behavior in male mice Oxytocin and vasopressin modulation of social anxiety following adolescent intermittent ethanol exposure A nonpeptide oxytocin receptor agonist for a durable relief of inflammatory pain Activation of hypothalamic oxytocin neurons reduces binge-like alcohol drinking through signaling at central oxytocin receptors Life in groups: the roles of oxytocin in mammalian sociality Titi monkeys as a novel non-human primate model for the neurobiology of pair bonding Oxytocin and social relationships: from attachment to bond disruption Oxytocin, vasopressin, and the neurogenetics of sociality Evolving nonapeptide mechanisms of gregariousness and social diversity in birds The neurobiology of pair bonding Oxytocin in the nucleus accumbens shell reverses CRFR2-evoked passive stress-coping after partner loss in monogamous male prairie voles Central expression of c-Fos in neonatal male and female prairie voles in response to treatment with oxytocin A role for oxytocin and 5-HT(1A) receptors in the prosocial effects of 3,4 methylenedioxymethamphetamine Physiological and endocrine effects of social contact Social isolation induces behavioral and neuroendocrine disturbances relevant to depression in female and male prairie voles Post-weaning social isolation alters anxiety-related behavior and neurochemical gene expression in the brain of male prairie voles Forced swimminginduced oxytocin release into blood and brain: effects of adrenalectomy and corticosterone treatment Influence of stress associated with chronic alcohol exposure on drinking Interaction of ethanol and stress: research with experimental animals-an update The correlation between central and peripheral oxytocin concentrations: a systematic review and meta-analysis Elevated cerebrospinal fluid and blood concentrations of oxytocin following its intranasal administration in humans The interaction between the oxytocin and pain modulation in headache patients GABA selectively controls the secretory activity of oxytocin neurons in the rat supraoptic nucleus Maternal contact differentially modulates central and peripheral oxytocin in rat pups during a brief regime of mother-pup interaction that induces a filial huddling preference Chronic cocaine exposure during pregnancy increases postpartum neuroendocrine stress responses Oxytocin blood concentrations in alcohol use disorder: a cross-sectional, longitudinal, and sex-separated study Beta-endorphin and oxytocin in patients with alcohol use disorder and comorbid depression The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.Publisher's Note: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.Copyright © 2022 Ryabinin and Zhang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.