Abstract
Purpose:
Emerging work shows divergence in the neurobiologies of binge-eating disorder (BED) and obesity despite their frequent co-occurrence. This review highlights specific distinguishing aspects of BED, including elevated impulsivity and compulsivity possibly involving the mesocorticolimbic dopamine system, and discusses implications for differential therapeutics for BED.
Methods:
This narrative review describes epidemiological, clinical, genetic, and preclinical differences between BED and obesity. Subsequently, this review discusses human neuroimaging work showing differences in executive functioning, reward processing, and emotion reactivity in BED as compared to obesity. Finally, based on the neurobiology of BED, this review identifies existing and novel therapeutic agents that may be most promising given their specific targets based on putative mechanisms of action relevant specifically to BED.
Findings:
BED is characterized by elevated impulsivity and compulsivity compared to obesity, which is reflected in divergent neurobiological characteristics and effective pharmacotherapies. Therapeutic agents that influence both reward and executive function systems may be especially effective for BED.
Implications:
Greater attention to impulsivity-/compulsivity-related, reward-related, and emotion-reactivity-related processes may enhance conceptualization and treatment approaches for patients with BED. Consideration of these distinguishing characteristics and processes could have implications for more targeted pharmacological treatment research and interventions.
Keywords: binge-eating disorder, eating disorders, obesity, impulsive behavior, compulsive behavior, neurobiology, pharmacology
Many people struggle to regulate their eating, including those with binge-eating disorder (BED) and those with obesity. BED is a psychiatric illness characterized by frequent episodes of consuming an objectively large amount of food in a short period of time with a subjective sense of loss of control during eating without subsequent compensatory behaviors1. Obesity is a physical condition defined by elevated body mass index (BMI). Obesity and BED have high rates of comorbidity, with more than half of individuals with BED also meeting BMI criteria for overweight or obesity2, 3. Conversely, BED is highly prevalent in obese populations, especially in individuals participating in treatments targeting weight loss4, 5.
Given frequent co-occurrence between BED and obesity, and some shared behavioral features of overeating behaviors, the emerging treatment literature for BED has been influenced by the obesity field6. Indeed, some early interventions attempted for BED included treatments from the obesity literature including, for example, very-low-calorie-diets (VLCDs), multiple behavioral weight-loss methods, and weight-loss medications7–9. Until recently, there were no approved pharmacological treatments for BED10–12, and clinicians have needed to rely on off-label use of pharmacotherapy involving agents developed specifically for other medical (e.g., obesity, seizure disorders) or psychiatric (e.g., depression) conditions13–15.
While the few weight-loss medications from the obesity literature tested to date have shown some potential to enhance weight loss in patients with BED, they have generally not reduced binge eating16–18. One potential explanation for this is that BED may represent a distinct phenotype of obesity because of its unique neurobiological characteristics19–24. As compared to obesity, BED may involve greater impulsivity and compulsivity, possibly involving the mesocorticolimbic dopamine system20, 25–27. Accordingly, targeting impulsivity, compulsivity, or other features of BED may be important to treatment outcomes and have implications for differential therapeutics for BED.
Methods
This narrative review examines the neurobiology of BED as compared to obesity, with a focus on how such differences may influence pharmacotherapy treatment for BED. First, we review neurobiological systems involved in BED and obesity. Second, we discuss epidemiological, clinical, genetic, and preclinical evidence for differences between BED and obesity. Third, we review human brain imaging work suggesting divergent impulsivity/compulsivity, reward-related, and emotion-related neurobiological processes in BED and obesity. Then, we consider whether specific therapeutic agents may be beneficial for individuals with BED. We review pharmacotherapies for BED, with a focus on their mechanisms of action and related effects on weight and binge eating. Finally, we discuss how future targeted pharmacological research may help to further establish differential therapeutics for BED.
Neurobiology of Eating and Binge Eating.
Substantial research has investigated neurobiological processes underlying eating behaviors. Complex interacting systems involving physiological and neural substrates for hunger and satiety, including both homeostatic/appetitive and hedonic/reward-based processes, have been implicated in eating28. In the human brain, proposed reward circuitry includes the ventral striatum (VS), palladium, amygdala, thalamus, and midbrain neurons, and this circuit interacts with frontoparietal control systems that help to regulate appetitive responding29. Multiple neurotransmitters and neuropeptides may influence these processes, including dopamine, serotonin, opioids, orexigenic peptides, glutamate, and endocannabinoids29. Disruptions to this circuitry may promote the development of obesity28 and/or eating disorders30, 31.
The neurobiology of BED is an active area of recent investigation. Emerging work has suggested that BED is characterized by elevated sensitivity to food reward coupled with increased impulsivity and compulsivity20, 25–27, 32. Impulsivity is a multifaceted construct characterized by “actions which are poorly conceived, prematurely expressed, unduly risky or inappropriate to the situation that often results in undesirable consequences”33. In contrast, compulsivity is characterized by “perseverative behaviors that have no obvious relationship to an overall goal and often result in undesirable consequences”33. Impulsivity and compulsivity may reflect failures of “top-down” cognitive control, possibly mediated in part via interactions between prefrontal circuits and those that promote behavioral engagement like the mesocorticolimbic system32–34. A transition from impulsive, reward-driven behaviors to compulsive, habit-driven behaviors has been hypothesized to be related to both substance use disorders (SUD) and binge eating30, 35. Indeed, the degree to which eating is reported as similar to SUD (i.e., “food addiction” or addictive-like eating) may be an indicator of greater BED severity36–39. Accordingly, impulsivity-related and compulsivity-related processes may underlie BED to a greater extent than obesity20, 25–27, 32.
Epidemiological and Clinical Differences Between Obesity and BED.
Obesity is a physical condition that affects much of the population, whereas BED is a psychiatric illness (i.e., a specific eating disorder) involving recurrent episodes of loss of control while overeating (which, as reviewed below, is distinct from overeating per se). Obesity is highly prevalent, with current findings estimating that >66% of the United States adult population is overweight or obese40, 41. In comparison, BED, despite being the most prevalent eating disorder, affects only ~1–3% of the United States adult population2, 3, 42. Obesity is strongly associated with increased risk for morbidity and mortality43, and some research suggests that BED may further increase risk or lead to poorer outcomes for some conditions3, 44–46. For example, elevated rates of psychiatric conditions44, 47–49, impaired social functioning44, 50, 51, and diabetes46, 52, 53 have been observed in individuals with BED. Across epidemiological, community, and clinical samples, individuals with BED show higher levels of body image concerns and psychological distress compared to those with obesity and without BED54–57. Notably, treatment outcomes for individuals with BED are poorer than individuals with obesity alone, including worse diabetes-related complications46, 52, less weight loss58–60, and poorer outcomes following bariatric surgery among those with “loss-of-control” eating61. Accordingly, despite significant overlap, BED likely represent a distinct, more severe phenotype within a broader obesity spectrum.
The etiology and behavioral presentation of BED diverges from that of obesity. The hypothesized etiology of BED is related to risk/maintenance factors for eating-related psychopathology (e.g., body-image disturbance, dietary restraint, negative mood) which may be associated with diminished control over eating62. In contrast, the etiology of obesity is related to caloric imbalance, without necessarily-associated impairments in control over eating63. Although individuals with obesity may also show problematic eating and overeating behaviors, BED is defined based on recurring binge-eating behaviors involving perceived loss-of-control whereas obesity is not. Laboratory and ecological momentary assessment (EMA) studies suggest differences in binge eating in BED and obesity64, 65. In laboratory studies, compared to overweight controls, individuals with BED consumed more calories66, 67 and ate larger amounts of food when prompted to “lose control”68, 69. This finding contrasts with research with bulimia nervosa, where patients were observed to eat large amounts when prompted to “lose control” and eat less ad libitum68. Outside of the laboratory, EMA studies showed more binge eating70, 71, c.f. 72 and greater distress about binge eating70–72 in individuals with BED compared to overweight controls. Consistently, the experience of binge eating is not reported as pleasant73 and results in increased overall negative affect74. Nonetheless, overweight controls in these studies also reported experiences of overeating and loss-of-control over eating70–72. Accordingly, the frequency and severity of binge eating observed in BED may suggest a more problematic, compulsive pattern that leads to significant distress and could be more closely related to difficulties with impulse control than observed in obesity.
Genetics of BED and Obesity.
Genetic studies suggest BED may be distinct from obesity. BED has been found to be moderately heritable (~45%;75, 76) and aggregate in families separately from obesity77. Additionally, preliminary research suggests that individuals with BED and obesity may differ with respect to candidate genes, including variants associated with dopaminergic and serotonergic functions78.
Several genetic polymorphisms may be enriched in BED relative to obesity. Several genes investigated in BED are related to D2 receptors (D2Rs; e.g., DRD2/ANKK1) and their polymorphisms (e.g., Taq1A). These genetic polymorphisms have been associated with dopamine function, sensitivity to reward, and SUDs79. Fewer individuals with BED (relative to obesity alone) had the A1 allele of Taq1A (associated with decreased reward sensitivity80). Additionally, compared to weight-matched individuals, those with BED were more likely to be homozygous for the A2 allele of the Taq1A polymorphism and homozygous for the T genotype of the C957T marker (associated with increased reward sensitivity81). Compared to obese individuals without BED, those with BED had a higher multi-locus genetic profile on dopaminergic genes, suggesting possibly greater dopamine signaling and response to reward among those with BED82, 83. Together, this work suggests that obese individuals with BED may be genetically distinct from those with obesity alone, possibly related to hypersensitivity to reward. However, further work is needed to substantiate this, particularly given linkage disequilibrium between DRD2 and ANKK1 and nearby genes, and findings suggesting in comparison to other nearby genes, DRD2 may be less closely linked to disorders characterized by poor impulse control84, 85.
Other studies suggest potential genetic differences between individuals with BED and those with obesity. For example, serotonergic involvement in eating disorders has been widely investigated78. One study reported that the LL genotype and L allele of the serotonin transporter gene were more common in individuals with BED compared to lean individuals86. Other investigations suggest possible involvement of ghrelin87, mu-opioid receptor80, brain-derived neurotrophic-factor88, CLOCK89, and endocannabinoid90 genes in BED. However, this work has not been widely replicated, nor have genome-wide association studies with genome-wide significant findings been reported for BED91.
Animal Models of BED and Obesity.
Preclinical models of BED and obesity examine neurobiological effects of different environmental exposures related to the hypothesized etiologies of each condition92. These models find differing neurobiological characteristics associated with excess weight and binge eating in animals. In preclinical models of obesity, animals are fed a cafeteria-style diet with free access to palatable food93, 94. Under these conditions, animals gain weight gradually and show heightened sensitivity to palatable food93. Obese rats with lentivirus knockout of D2Rs engage in compulsive eating, but not when naturally exposed to palatable foods94. In contrast, there are multiple preclinical models of binge eating that involve intermittent access to palatable foods92. Unlike animals with cafeteria-style diets, these animals do not always show an increase in weight but do show differing in neurobiological characteristics, including reduced D2R density and enhanced mu-opioid receptor binding92, 95. Compulsive-like eating in these rats is modulated through VS projections in the mesocorticolimbic system96.
Overall, animal models suggest that BED may be a more severe condition along an obesity spectrum. Animal models of obesity show that exposure to hyper-palatable foods leads to dopamine-related changes associated with heightened sensitivity to reward. In contrast, animal models of binge eating with intermittent access to food lead to arguably broader structural and metabolic changes. Accordingly, BED may be characterized by a greater degree of alteration to the mesocorticolimbic system than obesity.
Human Brain Studies of BED and Obesity.
Recent work in human neuroimaging has facilitated investigation of divergent neurobiological aspects of BED and obesity. PET (positron emission tomography) can use radioactive materials to measure receptor availability, and functional magnetic resonance imaging (fMRI) permits investigation of neurobiological correlates of cognitive processes. Such work helps to distinguish between neurobiological features of BED and obesity, including potential differences in receptor availability and neural correlates of psychological processes related to these conditions. Specifically, recent work has examined executive functioning (and related impulsivity/compulsivity), reward processing, and emotional reactivity in individuals with BED compared to obesity.
Receptor Availability.
In line with genetic and preclinical work, there may be differences in dopamine, serotonin, and mu-opioid receptor availabilities in individuals with BED as compared to obesity. Nonetheless, PET studies often have not found consistent differences in humans. In individuals with obesity, there are mixed findings, with some studies finding lower striatal D2/3R availability97, 98 and others unaltered D2/3R availability99, 100. A study using a D3R-preferring ligand observed increased D2/3R receptor availability in people with obesity in D3-enriched brain regions including the substantia nigra/ventral tegmental area, VS, and pallidum101. No prior work has compared D2/3R availability in individuals with BED and obesity compared to those with obesity alone, although a [11C]raclopride study suggested that methylphenidate increased food-related dopamine release in individuals with BED compared to those without102. Serotonin transporter binding may be related to BMI103, but findings are mixed in BED. One small study found reduced serotonin transporter binding in obese women with BED compared to obese women without BED104, whereas another study found enhanced serotonin transporter binding in individuals with BED compared to controls105. Other work has suggested that individuals with obesity show reduced mu-opioid availability100, which increases with weight change106. Mu-opioid receptor availability was found to be similar in obese individuals with and without BED107. Additional research involving larger samples may help to clarify these seemingly mixed findings.
Executive Functioning and Impulsivity/Compulsivity.
Executive functions are cognitive processes that underlie control of behavior, including attentional control, inhibitory control, and cognitive flexibility108. Interactions between prefrontal cortical and subcortical systems underlie these processes29, 108, 109. Difficulties in implementation of cognitive control may reflect imbalances between these systems, which may contribute to impulsive and compulsive behaviors32–34, 108, 109.
Some executive function deficits have been observed in individuals with obesity110–112. Genetic factors may link obesity with brain and cognitive measures associated with executive function113. BED and obesity also share differences in neurobehavioral circuitry with other conditions characterized by executive functioning deficits, including attention-deficit/hyperactivity disorder (ADHD)114.
Individuals with BED may experience poorer executive functioning than those with obesity, but evidence is mixed. Individuals with BED have been reported to perform worse on executive function batteries including measures of inhibitory control, attention, and cognitive flexibility115–119. However, there have been inconsistent findings, with some studies not reporting differences between individuals with BED and those with obesity alone110, 119, 120. A recent review and meta-analysis found that individuals with BED performed worse on working memory tasks compared to obese individuals but did not suggest other performance deficits119. These mixed results may reflect small samples and different measures across studies.
Despite potentially-conflicting behavioral findings, recent neuroimaging work has begun to investigate divergence in executive functioning in individuals with BED and obesity as compared to those with only obesity. Individuals with obesity and BED showed hypoactivity in brain regions associated with impulse control during a cognitive control task, including the ventromedial prefrontal cortex (vmPFC), inferior frontal gyrus (IFG) and insula, compared to obese and lean individuals121. Individuals with BED compared to overweight individuals have shown reduced recruitment of prefrontal brain systems during a food go/no-go task122. Compared to overweight individuals, those with BED have also shown more difficulty with response inhibition in behavioral tasks with food stimuli115, 123. Together, this work suggests that there are likely differences in the recruitment of prefrontal brain systems in individuals with BED as compared to obesity. This work is consistent with the hypothesis that impaired cognitive control may differentiate between BED and obesity.
Reward Processing and Food Reward.
Mesocorticolimbic pathways are involved in incentive salience and reward processing124. Aberrant reward processing, in general and in the context of food, has been implicated in obesity and BED20, 25, 26, 125. Theoretical models suggest that hyposensitivity to general rewards (reduced incentive salience) combined with hypersensitivity to food rewards (increased incentive salience) may precipitate overeating and/or binge eating62, 126.
Studies have compared individuals with BED and obesity during reward-related functional tasks. Individuals with BED have shown alterations in general reward processing during fMRI. For example, relative to lean individuals, obese individuals showed increased reward-related activity during anticipatory phases of a monetary reward task127. However, compared to obese individuals, obese individuals with BED demonstrated hypoactivation in reward-related brain regions during anticipatory reward/loss processing127. Such differences in reward-related brain activity in individuals with BED related prospectively to treatment outcomes128 and resonate with findings from people with other disorders characterized by poor impulse control (e.g., SUDs, gambling disorder)129. Additionally, compared to BMI-matched individuals, those with BED showed impaired behavioral adaptation during a reward learning task, including reduced activation associated with reward prediction error130. These results indicate a generalized pattern of diminished frontostriatal processing of non-food rewards in individuals with BED.
In the context of food, individuals with BED showed heightened reward-related reactivity compared to overweight controls19, 20. In neuroimaging studies, individuals with BED showed increased activation in reward-processing areas of the brain when shown food stimuli131–133. Studies using electroencephalography (EEG) have also found enhanced responses to food in individuals who binge eat compared to controls134, 135. Patterns of brain activation in the VS while viewing food stimuli in individuals with BED and obesity are dissociable from those in individuals with obesity only136. Further, dopamine signaling in the presence of food cues is stronger in BED compared to obesity. A [11C]raclopride PET study investigated dopamine response in the presence of food vs. neutral stimuli in the presence of oral methylphenidate (which blocks the dopamine transporter and increases dopamine signaling)102. Obese individuals with BED as compared to those without showed greater dopamine release in the caudate when exposed to food stimuli after methylphenidate102. Dopamine release was correlated with binge eating severity, but not BMI102. Collectively, this research suggests that individuals with BED have heightened neurobiological responses to food reward compared to individuals with obesity alone.
Emotional Reactivity.
Emotional reactivity, defined as the intensity and duration of emotions in response to stimuli137, is involved in eating-related psychopathology138, 139. Greater reported negative emotion is associated with subsequent binge eating140 in individuals with BED and not in overweight individuals without BED139. Further, greater negative emotion is associated with increased impulsivity34, perhaps via sensitization of the mesocorticolimbic system141. Accordingly, neurobiological differences in emotional reactivity may differentiate individuals with BED from those with obesity.
In general, individuals with BED report more frequent and severe daily negative affect than obese, overweight, or lean individuals142. Compared to controls, individuals with BED have greater reactivity to stress143 and report more severe depression and anxiety symptoms45, 131, 144. A recent review found evidence that individuals with BED show heightened responses to stress, including differences in cortisol, ghrelin, and cardiovascular measures138. For example, obese women with BED had a greater increase in negative affect and blood pressure than obese women without BED after the Trier Social Stress Task143.
Heightened emotional reactivity can reduce cognitive control and increase food-related reactivity in individuals with BED. Emotional distress may sensitize the brain’s reward system to food141 and its predictive cues145. Inducing negative affect increases activation in reward-related brain regions in chronic dieters shown food cues146. Similarly, under stress, reported desire to binge eat, the reinforcement value of food, and cortisol levels increase in people who binge eat147–149. Compared to controls and after an acute stressor, individuals with BED symptoms showed reduced inhibitory-control-related brain activity in response to food stimuli150. Further, following this acute stressor and exposure to food cues, individuals with BED ate more high calorie foods ad-libitum than controls150. Accordingly, binge eating may be precipitated by the effects of emotional reactivity on mesocorticolimbic brain systems, particularly in individuals with BED, and this possibility warrants additional direct study.
Pharmacological Treatment for BED and Obesity: Differential Therapeutics
Accumulating evidence of divergent neurobiological processes in BED and obesity may have implications for informing and refining pharmacological research and treatment for BED. To date, nearly all of the pharmacological treatment literature for BED has included testing off-label medications based on their effects on obesity and other medical or psychiatric disorders13–15. Several previous reviews have detailed pharmacological treatment outcome findings for BED13–15, 26, 151, 152. Briefly, except for randomized clinical trials (RCTs) testing lisdexamfetamine dimesylate (LDX) that led to FDA approval for “moderate-to-severe” BED (but not obesity)10–12, RCTs have generally been relatively small, of short duration, and without follow-up periods, and have produced mixed findings, which at best, despite being significantly superior to placebo, have generally been modest in their clinical effects. Additionally, several pharmacological agents have shown promise in preclinical studies, only to encounter significant problems in application to humans, including severe side effects and high rates of adverse events.
Here, we review studies testing pharmacological agents with putative mechanisms of action conceptually relevant to neurobiological factors highlighted in this review as potentially specific to BED. First, we review the application of stimulant, opioid antagonist, and serotonergic pharmacological agents to BED, with a focus on effects on impulsivity/compulsivity, reward-related, and emotion-related neurobiological processes. Subsequently, we discuss emerging evidence for novel agents for BED that may influence these processes. Consideration of these distinguishing characteristics and processes in BED could have implications for more targeted pharmacological research and treatment.
Stimulant Medications: Dual Effects on Executive Function and Food Reward.
Stimulants have been used to treat obesity and, more recently, BED. Stimulants act on the reward system by increasing catecholamine availability via reversing transport systems153, 154, which results in appetite suppression associated with elevated extracellular dopamine in the VS155 and, importantly, dose-related improvements in executive functioning156. Appetite-suppressant effects of stimulant medications led to their early applications for weight loss in patients with obesity; for example, amphetamines were used to reduce appetite beginning in the 1930s and the first stimulant to be FDA-approved for treating obesity was methamphetamine in 1947. Paralleling applications for obesity, their well-established effects on executive functioning led to widespread and long-standing use for treating ADHD in children, adolescents, and adults, with consistent findings of improvements in attentional and cognitive control157.
Stimulant medications may be especially effective for individuals with BED because of dual effects on reward and executive function systems. Indeed, the only FDA-approved pharmacotherapy for BED is LDX, a d-amphetamine prodrug158. In preclinical models of binge eating, following administration of LDX, rats stopped compulsively eating chocolate159 and reduced “binge-eating-related” impulsive choices160. In humans, RCTs found that LDX reduced binge eating and impulsivity/compulsivity symptoms10–12, 161. Notably, there is a strong correlation between compulsivity symptoms and severity/frequency of binge eating episodes observed in LDX trials162. Further, in individuals with BED, changes in prefrontal brain systems associated with LDX treatment were related to treatment outcome163. Specifically, reductions in brain activation in the vmPFC and thalamus while viewing food pictures after LDX correlated with reductions in binge eating and compulsivity symptoms163. Research investigating short-term and long-term effects of LDX, alone and in combination with cognitive behavioral therapy, is ongoing164, 165.
Research has recently tested dasotraline, an agent that inhibits dopamine and norepinephrine transporters with stimulant effects166, for treating BED167, 168. Both the flexible-dose168 and fixed-dose167 RCTs found significantly greater reductions in binge eating and associated compulsivity symptoms versus placebo. However, the manufacturer of dastroline has withdrawn their application for FDA review for approval for BED, eliminating this potential option.
Opioid Antagonists: Effects on Food Reward.
Opioid-based agents used in the treatment of SUD have recently been applied to eating and eating disorders169. Opioid receptors, particularly mu-opioid receptors, interact with the mesocorticolimbic dopamine system, which may underlie addictive properties of opioid receptor agonists and effects of opioids on appetitive behaviors including eating170. Opioid antagonists, including naltrexone, reduce craving for and consumption of substances of abuse171 and are thought to also reduce craving for food170. These agents were first applied to obesity following case studies reporting reduced weight in individuals with SUD treated with naltrexone172. In lean individuals, naltrexone results in ~20% reduction in food intake, which is associated with lower ratings of food pleasantness173. Nonetheless, monotherapy opioid antagonists neither consistently reduced food intake or weight in obese individuals174, 175, c.f. 176 nor reduced binge eating in humans169, 177, 178.
Combining opioid-based agents (which may target food reward) with complementary agents may improve the efficacy of pharmacotherapy for eating and binge eating. Specifically, the combination of naltrexone+bupropion, an FDA-approved weight-loss medication, seems conceptually relevant for BED given a number of findings that follow.
Bupropion, a selective reuptake inhibitor of dopamine and norepinephrine, has been found to be effective for smoking cessation179, 180 and to reduce cravings for various substances181. Although some reports suggest reduced food cravings and weight loss with bupropion182, the literature is mixed. In a RCT with overweight women with BED, bupropion did not significantly reduce binge eating nor food cravings relative to placebo, although patients receiving bupropion lost significantly more weight than those on placebo183. Some preliminary work suggests that bupropion may be associated with improvements in executive function184, 185 which, as reviewed above, seems related to disordered eating.
Importantly, bupropion is thought to work synergistically with other agents180, 186; for review,187 which stimulated a series of large-scale RCTs demonstrating the efficacy of combined naltrexone+bupropion for weight-loss186, 188, 189; for review,190 and subsequent FDA approval for obesity. Preliminary research suggests that naltrexone+bupropion may reduce food cue reactivity and enhance cognitive control191. Preliminary open-label trials of naltrexone+bupropion reported reductions in binge eating in adolescents with eating disorders192 and in individuals with self-reported depressive and binge-eating symptoms193. One pilot placebo-controlled double-blind RCT reported preliminary findings that naltrexone+bupropion showed promise relative to placebo for reducing weight in patients with BED; the observed trends for binge-eating reductions, although not statistically significant, suggested the need for larger-scale and adequately-powered RCT194. There are two ongoing NIH-funded RCTs testing naltrexone+bupropion, alone and in combination with behavioral treatment, for BED195, 196, which were designed to examine potential mechanisms (e.g., changes in impulsivity/compulsivity) in addition to testing the short- and longer-term effects of this agent.
Serotonergic Agents: Effects on Emotion Reactivity?
Serotonergic agents, long-prescribed for anxiety and mood-related psychiatric conditions, have historically been tested for obesity, given animal research indicating that serotonin influences eating by modulating satiety197–201. Overall, these agents produced very modest weight loss in patients with obesity202 and those effects were poorly maintained16, 203, 204. This broad class of agents has, quite unfortunately, had a troubled history when applied to obesity, with several serotonergic drugs receiving FDA approval only to be subsequently withdrawn from the market due to emergent safety concerns. Medications with such concerns include sibutramine (for cardiovascular complications205), fenfluramine (for pulmonary hypertension205), and most recently lorcaserin (for cancer risk206).
Despite poor results for treatment of obesity, serotonergic agents were initially thought to hold promise for BED for several reasons. Fluoxetine, a selective serotonin reuptake inhibitor (SSRI), is the sole FDA-approved medication for bulimia nervosa (another eating disorder characterized by recurrent binge eating) following a series of RCTs demonstrating efficacy relative to placebo207–209. Additionally, SSRIs have been found to reduce emotional reactivity210 and symptoms of obsessive-compulsive disorder211. However, RCTs for serotonergic agents have consistently showed weak to non-significant effects in RCTs with BED for both binge eating and weight outcomes15, 212. Despite the suggestion that SSRIs could reduce emotional reactivity that may precipitate or maintain binge eating, individuals with greater depressive symptoms showed poorer outcomes on fluoxetine than in cognitive behavioral therapy213, and depressive symptoms did not predict response to fluoxetine in patients with bulimia nervosa214. Accordingly, although individuals with BED may show heightened emotional reactivity, serotonergic agents are likely not effective as either stand-alone treatments or as additive treatments for BED212, 213, see 18.
Novel Therapeutic Agents.
Early, emerging work has tested novel therapeutic agents that may reduce eating and binge eating. Some of these agents influence neurobiological processes that may be relatively specific to BED, including impulsivity. However, several agents tested in preclinical settings have encountered challenges, including significant side effects, when translated to humans. Thus, this brief overview is offered preliminarily and should be considered cautiously given the early stages of the research.
Orexin Receptor Antagonists.
Recent preclinical work has investigated the effects of orexin-based agents215–217 on impulsive behavior and binge eating. In animals, selective OX1 receptor antagonists (SB-334867 and GSK1059865) reduced palatable food eating217, 218 and a dual OX1/OX2 receptor antagonist (SB-649868) reduced binge eating219. In rats, OX1 receptor antagonists reduced impulsive behavior under baseline and cocaine conditions220, 221, c.f. 222. Given effects on impulsive behavior in animals, an important next step will be to determine whether these agents influence impulsive behavior and binge eating in humans.
Endocannabinoid Antagonists.
The endocannabinoid system modulates the transmission of other neurotransmitters in mesocorticolimbic pathways223. CB1 receptor inverse agonists or antagonists (SR141716A, AM 251 and rimonabant) decreased eating and weight in rodents224–228, decreased food consumption in primates229, reduced binge eating in animals226, 230, 231, and reduced impulsivity during delay discounting tasks in overweight rats232, 233. However, in humans, despite preliminary effects on weight234–237 and binge eating238, CB1 receptor agonists had clear and significant side effects. These agents are not approved for use in humans because of concerns that the therapeutic dose and dose that generates seizures are similar, and concerns about psychiatric side effects, including severe depression237.
Glutamatergic Agents.
Glutamatergic pharmacotherapies modulate mesocorticolimbic pathways239 and are involved in eating240–244. For example, topiramate, an anticonvulsant, reduces binge eating and weight compared to placebo15, 245, 246. Notably, a double-blind RCT found that topiramate was superior to placebo for enhancing CBT outcomes for both binge eating and weight247, which has not been shown for any other pharmacological agent tested to date for BED. It has been posited that topiramate may target impulsivity and therefore be especially effective for individuals with greater impulsivity and SUDs248, and this might also account for some of its effects in patients with BED. Despite these promising findings for BED, topiramate is associated with high rates of adverse events and dropout (e.g., 28–68% dropout; 245, 249 , 250) and this limits its wider use.
Novel glutamatergic agents are being tested for BED. Memantine is an NDMA receptor antagonist that is FDA-approved for treatment of Alzheimer’s disease and has been used off-label for psychiatric disorders, including SUDs and BED. Very early and preliminary uncontrolled reports suggest that memantine may reduce binge eating in humans251, 252, as shown in animal models253–255. However, animal work found that memantine increased impulsive choice256, 257, potentially limiting efficacy for BED.
GABA Receptor Agonists.
GABAB receptors are found on GABA and dopamine neurons in the mesocorticolimbic system258, 259. Preliminary trials of baclofen, a GABA agonist used to treat muscle spasms, showed reductions in body weight in ten obese subjects260 and reduced binge eating and food craving in a total of twenty-two individuals with BED in two early pilot studies261, 262. Baclofen generates sedation in humans and has not been shown to reduce impulsivity, limiting its clinical utility for BED.
Conclusions & Future Directions
BED and obesity often co-occur and clinicians have employed similar behavioral and pharmacological treatment approaches to treat these conditions. However, given emerging evidence of divergent neurobiological characteristics of BED as compared to obesity, adopting a neurobiologically-informed, mechanism-focused approach to selecting pharmacological treatment for BED may prove to be advantageous. Indeed, pharmacotherapies that target both impulsivity/compulsivity and reward processing, such as LDX, have been shown to be effective for BED. Increased attention to the effects of pharmacological treatments on neurobiological processes related to BED, including impulsivity/compulsivity, reward processing, and emotional reactivity, may be relevant to enriching treatment conceptualizations and prescriptions, and for informing treatment research. Future work should investigate changes in impulsivity, compulsivity, and reward and emotion processing in preclinical and human trials of pharmacotherapies for BED. Such contributions could improve BED treatment and contribute to broader, transdiagnostic approaches to conceptualizing BED and other forms of impulsivity-related and compulsivity-related psychopathologies.
Funding
This work was supported by the National Institute of Health R01 DK121551. Dr. Grilo was also supported, in part, by National Institutes of Health (NIH) Grants R01 DK49587, R01 DK114075 and R01 DK112771. The funding agencies had no role or influence on the content of this paper or in the decision to submit the paper for publication.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflicts of Interest
No external commercial entities sponsored or provided assistance in the creation of this review. The authors report the following disclosures. Dr. Potenza has consulted for and advised pharmaceutical and healthcare entities including AXA, Idorsia and Opiant/Lakelight Therapeutics. He has also consulted for and advised other entities including Game Day Data and the Addiction Policy Forum; received research support from the Mohegan Sun Casino and the National Center for Responsible Gaming (now the International Center for Responsible Gaming); participated in surveys, mailings, or telephone consultations related to drug addiction, impulse-control disorders, or other health topics; consulted for legal and gambling entities on issues related to impulse-control and addictive disorders; performed grant reviews for the National Institutes of Health and other agencies; edited journals and journal sections; given academic lectures in grand rounds, CME events, and other clinical/scientific venues; and generated books or chapters for publishers of mental health texts. Dr. Grilo has consulted to Sunovion and Weight Watchers; has received honoraria for lectures, CME activities, and presentations at scientific conferences; and has received royalties from Guilford Press and Taylor & Francis Publishers for academic books. Dr. Boswell does not have broader interests to disclose.
References
- 1.APA. Diagnostic and Statistical Manual of Mental Disorders (5th ed). Arlington, VA: American Psychiatric Association; 2013. [Google Scholar]
- 2.Hudson JI, Hiripi E, Pope HG, Kessler RM. The prevalence and correlates of eating disorders in the National Comorbidity Survey Replication. Biol Psychiatry. 2007;61:348–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Udo T, Grilo CM. Prevalence and Corrleates of DSM-5-Defined Eating Disorders in a Nationally Representative Sample of U.S. Adults. Biol Psychiatry 2018;84:345–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Spitzer RL, Yanovski SZ, Wadden TA, et al. Binge eating disorder: its further validation in a multisite study. Int J Eat Disord. 1993;13:137–153. [PubMed] [Google Scholar]
- 5.Spitzer RL, Devlin MJ, Walsh BT, et al. Binge eating disorder: A multisite field trial of the diagnostic criteria. Int J Eat Disord. 1992;11:191–203. [Google Scholar]
- 6.Allison KC, Grilo CM, Masheb RM, Stunkard AJ. Binge eating disorder and night eating syndrome: a comparative study of disordered eating. J Consult Clin Psychol. 2005;73:1007–1015. [DOI] [PubMed] [Google Scholar]
- 7.Wilson GT, Grilo CM, Vitousek KM. Psychological treatment of eating disorders. Am Psychol. 2007;62:199–216. [DOI] [PubMed] [Google Scholar]
- 8.Yanovski SZ, Gormally JF, Leser MS, Gwirtsman HE, Yanovski JA. Binge eating disorder affects outcome of comprehensive very-low-calorie diet treatment. Obes Res. 1994;2:205–212. [DOI] [PubMed] [Google Scholar]
- 9.Stunkard AJ, Berkowitz R, Tanrikut C, Reiss E, Young L. d-fenfluramine treatment of binge eating disorder. Am J Psychiatry. 1996;153:1455–1459. [DOI] [PubMed] [Google Scholar]
- 10.McElroy SL, Hudson J, Ferreira-Cornwell MC, Radewonuk J, Whitaker T, Gasior M. Lisdexamfetamine Dimesylate for Adults with Moderate to Severe Binge Eating Disorder: Results of Two Pivotal Phase 3 Randomized Controlled Trials. Neuropsychopharmacology. 2016;41:1251–1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hudson JI, McElroy SL, Ferreira-Cornwell MC, Radewonuk J, Gasior M. Efficacy of Lisdexamfetamine in Adults With Moderate to Severe Binge-Eating Disorder: A Randomized Clinical Trial. JAMA Psychiatry. 2017;74:903–910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.McElroy SL, Hudson JI, Mitchell JE, et al. Efficacy and Safety of Lisdexamfetamine for Treatment of Adults With Moderate to Severe Binge-Eating Disorder: A Randomized Clinical Trial. JAMA Psychiatry. 2015;72:235–246. [DOI] [PubMed] [Google Scholar]
- 13.Grilo CM, Reas DL, Mitchell JE. Combining Pharmacological and Psychological Treatments for Binge Eating Disorder: Current Status, Limitations, and Future Directions. Curr Psychiatry Rep. 2016;18:55. [DOI] [PubMed] [Google Scholar]
- 14.Appolinario JC, Nardi AE, McElroy SL. Investigational drugs for the treatment of binge eating disorder (BED): an update. Expert Opin Investig Drugs. 2019;28:1081–1094. [DOI] [PubMed] [Google Scholar]
- 15.Reas DL, Grilo CM. Pharmacological treatment for binge eating disorder: updated review and synthesis. Expert Opin Pharmacother. 2015;16:1463–1478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Grilo CM, Masheb RM, White MA, et al. Treatment of binge eating disorder in racially and ethnically diverse obese patients in primary care: randomized placebo-controlled clinical trial of self-help and medication. Behav Res Ther. 2014;58:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Grilo CM, White MA. Orlistat with behavioral weight loss for obesity with versus without binge eating disorder: randomized placebo-controlled trial at a community mental health center serving educationally and economically disadvantaged Latino/as.. Behav Res Ther. 2013;51:167–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Grilo CM, Masheb RM, Salant SL. Cognitive behavioral therapy guided self-help and orlistat for the treatment of binge eating disorder: a randomized, double-blind, placebo-controlled trial Biol Psychiatry. 2005;57:1193–1201. [DOI] [PubMed] [Google Scholar]
- 19.Schag K, Schonleber J, Teufel M, Zipfel S, Giel KE. Food-related impulsivity in obesity and binge eating disorder--a systematic review. Obes Rev. 2013;14:477–495. [DOI] [PubMed] [Google Scholar]
- 20.Kober H, Boswell RG. Potential psychological & neural mechanisms in binge eating disorder: Implications for treatment. Clin Psychol Rev. 2018;60:32–44. [DOI] [PubMed] [Google Scholar]
- 21.Carnell S, Gibson C, Benson L, Ochner CN, Geliebter A. Neuroimaging and obesity: current knowledge and future directions. Obes Rev. 2012;13:43–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Devlin MJ. Is there a place for obesity in DSM-V? Int J Eat Disord. 2007;40 Suppl:S83–88. [DOI] [PubMed] [Google Scholar]
- 23.Kessler RM, Zald DH, Ansari MS, Li R, Cowan RL. Changes in dopamine release and dopamine D2/3 receptor levels with the development of mild obesity. Synapse. 2014;68:317–320. [DOI] [PubMed] [Google Scholar]
- 24.Leehr EJ, Schag K, Dresler T, et al. Food specific inhibitory control under negative mood in binge-eating disorder: Evidence from a multimethod approach. Int J Eat Disord. 2018;51:112–123. [DOI] [PubMed] [Google Scholar]
- 25.Balodis IM, Grilo CM, Potenza MN. Neurobiological features of binge eating disorder. CNS Spectr. 2015;20:557–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hutson PH, Balodis IM, Potenza MN. Binge-eating disorder: Clinical and therapeutic advances. Pharmacol Ther. 2018;182:15–27. [DOI] [PubMed] [Google Scholar]
- 27.Kessler RM, Hutson PH, Herman BK, Potenza MN. The neurobiological basis of binge-eating disorder. Neurosci Biobehav Rev. 2016;63:223–238. [DOI] [PubMed] [Google Scholar]
- 28.Clifton PG. Neural circuits of eating behaviour: Opportunities for therapeutic development. J Psychopharmacol. 2017;31:1388–1402. [DOI] [PubMed] [Google Scholar]
- 29.Haber SN. Anatomy and Connectivity of the Reward Circuit. 2017:3–19. [Google Scholar]
- 30.Berridge KC. “Liking” and “wanting” food rewards: Brain substrates and roles in eating disorders. Physiol Behav. 2009;97:537–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kaye WH, Wagner A, Fudge JL, Paulus M. Neurocircuity of eating disorders. Curr Top Behav Neurosci. 2011;6:37–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Robbins TW, Gillan CM, Smith DG, de Wit S, Ersche KD. Neurocognitive endophenotypes of impulsivity and compulsivity: towards dimensional psychiatry. Trends Cogn Sci. 2012;16:81–91. [DOI] [PubMed] [Google Scholar]
- 33.Dalley JW, Everitt BJ, Robbins TW. Impulsivity, compulsivity, and top-down cognitive control. Neuron. 2011;69:680–694. [DOI] [PubMed] [Google Scholar]
- 34.Bari A, Robbins TW. Inhibition and impulsivity: behavioral and neural basis of response control. Prog Neurobiol. 2013;108:44–79. [DOI] [PubMed] [Google Scholar]
- 35.Berridge KC, Robinson TE. Liking, wanting, and the incentive-sensitization theory. American Psychologist. 2016;71:670–679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Gearhardt AN, White MA, Masheb RM, Morgan PT, Crosby RD, Grilo CM. An examination of the food addiction construct in obese patients with binge eating disorder. Int J Eat Disord. 2011;45:657–663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gearhardt AN, White MA, Potenza MN. Binge Eating Disorder and Food Addiction. Curr Drug Abuse Rev. 2011;4:201–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Gearhardt AN, White MA, Masheb RM, Grilo CM. An examination of food addiction in a racially diverse sample of obese patients with binge eating disorder in primary care settings. Compr Psychiatry. 2013;54:500–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Schulte EM, Grilo CM, Gearhardt AN. Shared and unique mechanisms underlying binge eating disorder and addictive disorders. Clin Psychol Rev. 2016;44:125–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ogden CL, Carroll MD, Fryar CD, Flegal KM. Prevalence of obesity among adults and youth: United States, 2011–2014. NCHS Data Brief. 2015;219. [PubMed] [Google Scholar]
- 41.Organization WH. Obesity and overweight 2016. [Google Scholar]
- 42.Striegel-Moore RH, Franko DL. Should binge eating disorder be included in the DSM-V? A critical review of the state of the evidence. Annu Rev Clin Psychol. 2008;4:305–324. [DOI] [PubMed] [Google Scholar]
- 43.Collaboration PS. Body-mass index and cause-specific mortality in 900000 adults: collaborative analyses of 57 prospective studies. Lancet. 2009;373:1083–1096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kessler RC, Berglund PA, Chiu WT, et al. The prevalence and correlates of binge eating disorder in the World Health Organization World Mental Health Surveys. Biol Psychiatry. 2013;73:904–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Udo T, Grilo CM. Psychiatric and medical correlates of DSM-5 eating disorders in a nationally representative sample of adults in the United States. Int J Eat Disord. 2019;52:42–50. [DOI] [PubMed] [Google Scholar]
- 46.Hudson JI, Lalonde JK, Coit CE, et al. Longitudinal study of the diagnosis of components of the metabolic syndrome in individuals with binge-eating disorder. Am J Clin Nutr. 2010;91:1568–1573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Udo T, Bitley S, Grilo CM. Suicide attempts in U.S. adults with lifetime DSM-5 eating disorders. BMC Med. 2019;17:120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Javaras KN, Pope HG, Lalonde JK, et al. Co-occurence of binge eating disorder with psychiatric and medical disorders. J Clin Psychiatry. 2008;69:266–273. [DOI] [PubMed] [Google Scholar]
- 49.Bulik CM, Sullivan PF, Kendler KS. Medical and psychiatric morbidity in obese women with and without binge eating. Int J Eat Disord. 2002;32:72–78. [DOI] [PubMed] [Google Scholar]
- 50.Whisman MA, Dementyeva A, Baucom DH, Bulik CM. Marital functioning and binge eating disorder in married women. Int J Eat Disord. 2012;45:385–389. [DOI] [PubMed] [Google Scholar]
- 51.Kessler RC, Shahly V, Hudson JI, et al. A comparative analysis of role attainment and impairment in binge-eating disorder and bulimia nervosa: results from the WHO World Mental Health Surveys.. Epidemiological Psychiatric Science. 2014;23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Raevouri A, Suokas J, Haukka J, et al. Highly increased risk of type 2 diabetes in patients with binge eating disorder and bulimia nervosa. Int J Eat Disord. 2014;48. [DOI] [PubMed] [Google Scholar]
- 53.de Jonge P, Alonso J, Stein DJ, et al. Associations between DSM-IV mental disorders and diabetes mellitus: a role for impulse control disorders and depression. Diabetologia. 2014;57:699–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Coffino JA, Udo T, Grilo CM. The Significance of Overvaluation of Shape or Weight in Binge-Eating Disorder: Results from a National Sample of U.S. Adults.. Obesity (Silver Spring). 2019;27:1367–1371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Grilo CM, Masheb RM, White MA. Significance of overvaluation of shape/weight in binge-eating disorder: comparative study with overweight and bulimia nervosa. Obesity (Silver Spring). 2010;18:499–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Grilo CM, Crosby RD, Masheb RM, et al. Overvaluation of shape and weight in binge eating disorder, bulimia nervosa, and sub-threshold bulimia nervosa. Behav Res Ther. 2009;47:692–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Grilo CM, Hrabosky JI, White MA, Allison KC, Stunkard AJ, Masheb RM. Overvaluation of shape and weight in binge eating disorder and overweight controls: refinement of a diagnostic construct.. J Abnorm Psychol. 2008;117:414–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Blaine B, Rodman J. Responses to weight loss treatment among obese individuals with and without BED: a matched-study meta-analysis. Eat Weight Disord. 2007;12:54–60. [DOI] [PubMed] [Google Scholar]
- 59.Sherwood NE, Jeffery RW, Wing RR. Binge status as a predictor of weight loss treatment outcome. Int J Obes (Lond). 1999;23:485–493. [DOI] [PubMed] [Google Scholar]
- 60.Marcus MD, Wing RR, Hopkins J. Obese binge eaters: affect, cognitions, and response to behavioral weight control. J Consult Clin Psychol. 1988;56:433–439. [DOI] [PubMed] [Google Scholar]
- 61.White MA, Kalarchian MA, Masheb RM, Marcus MD, Grilo CM. Loss of Control Over Eating Predicts Outcomes in Bariatric Surgery Patients: A Prospective, 24-Month Follow-Up Study. J Clin Psychiatry. 2010;71:175–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Fairburn CG. Overcoming Binge Eating. New York: The Guilford Press; 2013. [Google Scholar]
- 63.Hill JO, Wyatt H, Reed GW, Peters JC. Obesity and the Environment: Where Do We Go from Here? Science. 2003;299:853–855. [DOI] [PubMed] [Google Scholar]
- 64.Wonderlich SA, Gordon KH, Mitchell JE, Crosby RD, Engel SG. The validity and clinical utility of binge eating disorder. Int J Eat Disord. 2009;42:687–705. [DOI] [PubMed] [Google Scholar]
- 65.Walsh BT, Devlin MJ. Binge-Eating Disorder: Unanswered Questions In: Frank G, Berner LA, eds. Binge Eating: a Transdiagnostic Psychopathology: Springer; In Press. [Google Scholar]
- 66.Yanovski SZ, Leet M, Yanovski JA, et al. Food selection and intake of obese women with binge-eating disorder. Am J Clin Nutr. 1992;56:975–980. [DOI] [PubMed] [Google Scholar]
- 67.Guss JL, Kissilejf HR, Walsh BT, Devlin MJ. Binge Eating Behavior in Patients with Eating Disorders. Obes Res. 1994;2:355–363. [DOI] [PubMed] [Google Scholar]
- 68.Walsh BT, Boudreau G. Laboratory studies of binge eating disorder. Int J Eat Disord. 2003;34 Suppl:S30–38. [DOI] [PubMed] [Google Scholar]
- 69.Walsh BT. The importance of eating behavior in eating disorders. Physiol Behav. 2011;104:525–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Greeno CG, Wing RR, Shiffman S. Binge antecedents in obese women with and without binge eating disorder. J Consult Clin Psychol. 2000;68:95–102. [PubMed] [Google Scholar]
- 71.Goldschmidt AB, Engel SG, Wonderlich SA, et al. Momentary affect surrounding loss of control and overeating in obese adults with and without binge eating disorder. Obesity (Silver Spring). 2012;20:1206–12011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Le Grange D, Gorin A, Catley D, Stone AA. Does momentary assessment detect binge eating in overweight women that is denied at interview? Eur Eat Disord Rev. 2001;9:309–324. [Google Scholar]
- 73.Deaver CM, Miltenberger RG, Smyth J, Meidinger A, Crosby RD. An evaluation of affect and binge eating. Behav Mod. 2003;27:578–599. [DOI] [PubMed] [Google Scholar]
- 74.Haedt-Matt AA, Keel PK. Revisiting the Affect Regulation Model of Binge Eating : A Meta-Analysis of Studies Using Ecological Momentary Assessment. Psychological Bulletin. 2011;137:660–681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Javaras KN, Laird NM, Reichborn-Kjennerud T, Bulik CM, Pope HG Jr., Hudson JI. Familiality and heritability of binge eating disorder: results of a case-control family study and a twin study. Int J Eat Disord. 2008;41:174–179. [DOI] [PubMed] [Google Scholar]
- 76.Mitchell KS, Neale MC, Bulik CM, Aggen SH, Kendler KS, Mazzeo SE. Binge eating disorder: a symptom-level investigation of genetic and environmental influences on liability. Psychol Med. 2010;40:1899–1906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Hudson JI, Lalonde JK, Berry JK, et al. Binge-Eating Disorder as a Distinct Familial Phenotype in Obese Individuals. Arch Gen Psychiatry. 2006;63:313–319. [DOI] [PubMed] [Google Scholar]
- 78.Trace SE, Baker JH, Penas-Lledo E, Bulik CM. The genetics of eating disorders. Annu Rev Clin Psychol. 2013;9:589–620. [DOI] [PubMed] [Google Scholar]
- 79.Le Foll B, Gallo A, Le Strat Y, Lu L, Gorwood P. Genetics of dopamine receptors and drug addiction: a comprehensive review. Behav Pharmacol. 2009;20:1–17. [DOI] [PubMed] [Google Scholar]
- 80.Davis C, Levitan RD, Reid C, et al. Dopamine for “wanting” and opioids for “liking”: a comparison of obese adults with and without binge eating. Obesity (Silver Spring). 2009;17:1220–1225. [DOI] [PubMed] [Google Scholar]
- 81.Davis C, Levitan RD, Yilmaz Z, Kaplan AS, Carter JC, Kennedy JL. Binge eating disorder and the dopamine D2 receptor: genotypes and sub-phenotypes. Prog Neuropsychopharmacol Biol Psychiatry. 2012;38:328–335. [DOI] [PubMed] [Google Scholar]
- 82.Davis C, Levitan RD, Carter JC. Binge eating disorder and “food addiction”: a multi-locus genetic profile study In: Society PpatamotEDR, ed. Porto, Portugal: 2012. [Google Scholar]
- 83.Nikolova YS, Ferrell RE, Manuck SB, Hariri AR. Multilocus genetic profile for dopamine signaling predicts ventral striatum reactivity. Neuropsychopharmacology. 2011;36:140–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Gelernter J, Yu Y, Weiss R, et al. Haplotype spanning TTC12 and ANKK1, flanked by the DRD2 and NCAM1 loci, is strongly associated to nicotine dependence in two distinct American populations Human Molecular Genetics. 2006;15:3498–3507. [DOI] [PubMed] [Google Scholar]
- 85.Yang B, Kranzler HR, Zhao H, Gruen JR, Luo X, Gelernter J. Association of haplotypic variants in DRD2, ANKK1, TTC12 and NCAM1 to alcohol dependence in independent case–control and family samples. Human Molecular Genetics. 2007;16:2844–2853. [DOI] [PubMed] [Google Scholar]
- 86.Monteleone P, Tortorella A, Castaldo E, Maj M. Association of a functional serotonin transporter gene polymorphism with binge eating disorder. Am J Med Genet B Neuropsychiatr Genet. 2006;141B:7–9. [DOI] [PubMed] [Google Scholar]
- 87.Monteleone P, Tortorella A, Castaldo E, Di Filippo C, Maj M. The Leu72Met polymorphism of the ghrelin gene is significantly associated with binge eating disorder. Psychiatric Genetics. 2007;17:13–17. [DOI] [PubMed] [Google Scholar]
- 88.Monteleone P, Zanardini R, Tortorella A, et al. The 196G/A (val66met) polymorphism of the BDNF gene is significantly associated with binge eating behavior in women with bulimia nervosa or binge eating disorder. Neurosci Lett. 2006;406:133–137. [DOI] [PubMed] [Google Scholar]
- 89.Monteleone P, Tortorella A, Docimo L, et al. Investigation of 3111T/C polymorphism of the CLOCK gene in obese individuals with or without binge eating disorder: association with higher body mass index. Neurosci Lett. 2008;435:30–33. [DOI] [PubMed] [Google Scholar]
- 90.Monteleone P, Tortorella A, Martiadis V, Di Filippo C, Canestrelli B, Maj M. The cDNA 385C to A missense polymorphism of the endocannabinoid degrading enzyme fatty acid amide hydrolase (FAAH) is associated with overweight/obesity but not with binge eating disorder in overweight/obese women. Psychoneuroendocrinology. 2008;33:546–550. [DOI] [PubMed] [Google Scholar]
- 91.Himmerich H, Bentley J, Kan C, Treasure J. Genetic risk factors for eating disorders: an update and insights into pathophysiology. Therapeutic Advances in Pharmacology. 2019;9:1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Corwin RL, Avena NM, Boggiano MM. Feeding and reward: perspectives from three rat models of binge eating. Physiol Behav. 2011;104:87–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Geiger BM, Haburcak M, Avena NM, Moyer MC, Hoebel BG, Pothos EN. Deficits of mesolimbic dopamine neurotransmission in rat dietary obesity. Neuroscience. 2009;159:1193–1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Johnson PM, Kenny PJ. Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats. Nat Neurosci. 2010;13:635–641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Avena NM. The study of food addiction using animal models of binge eating. Appetite. 2010;55:734–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Spierling S, de Guglielmo G, Kirson D, et al. Insula to ventral striatal projections mediate compulsive eating produced by intermittent access to palatable food. Neuropsychopharmacology. 2020;45:579–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Wang G-J, Volkow ND, Logan J, et al. Brain dopamine and obesity. The Lancet. 2001;357:354–357. [DOI] [PubMed] [Google Scholar]
- 98.de Weijer BA, van de Geissen E, van Amelsvoort TA, et al. Lower striatal dopamine D2/3 receptor availability in obese compared with non-obese subjects. EJNMMI Res. 2011;1:1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Haltia LT, Rinne JO, Merisaari H, et al. Effects of intravenous glucose on dopaminergic function in the human brain in vivo. Synapse. 2007;61:748–756. [DOI] [PubMed] [Google Scholar]
- 100.Karlsson HK, Tuominen L, Tuulari JJ, et al. Obesity is associated with decreased mu-opioid but unaltered dopamine D2 receptor availability in the brain. J Neurosci. 2015;35:3959–3965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Gaiser EC, Gallezot J, Worhunsky PD, et al. Elevated Dopamine D 2/3 Receptor Availability in Obese Individuals: A PET Imaging Study with [11 C](+)PHNO. Neuropsychopharmacology. 2016;41:3042–3050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wang GJ, Geliebter A, Volkow ND, et al. Enhanced striatal dopamine release during food stimulation in binge eating disorder. Obesity (Silver Spring). 2011;19:1601–1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Erritzoe D, Frokjaer VG, Haahr MT, et al. Cerebral serotonin transporter binding is inversely related to body mass index. Neuroimage. 2010;52:284–289. [DOI] [PubMed] [Google Scholar]
- 104.Kuikka JT, Tammela L, Karhunen L, et al. Reduced serotonin transporter binding in binge eating women. Psychopharmacology (Berl). 2001;155:310–314. [DOI] [PubMed] [Google Scholar]
- 105.Majuri J, Joutsa J, Johansson J, et al. Serotonin transporter density in binge eating disorder and pathological gambling: A PET study with [(11)C]MADAM. Eur Neuropsychopharmacol. 2017;27:1281–1288. [DOI] [PubMed] [Google Scholar]
- 106.Karlsson HK, Tuulari JJ, Tuominen L, et al. Weight loss after bariatric surgery normalizes brain opioid receptors in morbid obesity. Mol Psychiatry. 2016;21:1057–1062. [DOI] [PubMed] [Google Scholar]
- 107.Joutsa J, Karlsson HK, Majuri J, et al. Binge eating disorder and morbid obesity are associated with lowered mu-opioid receptor availability in the brain. Psychiatry Res Neuroimaging. 2018;276:41–45. [DOI] [PubMed] [Google Scholar]
- 108.Diamond A Executive functions. Annu Rev Psychol. 2013;64:135–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Casey BJ. Beyond simple models of self-control to circuit-based accounts of adolescent behavior. Annu Rev Psychol. 2015;66:295–319. [DOI] [PubMed] [Google Scholar]
- 110.Lavagnino L, Arnone D, Cao B, Soares JC, Selvaraj S. Inhibitory control in obesity and binge eating disorder: A systematic review and meta-analysis of neurocognitive and neuroimaging studies. Neurosci Biobehav Rev. 2016;68:714–726. [DOI] [PubMed] [Google Scholar]
- 111.Coppin G, Nolan-Poupart S, Jones-Gotman M, Small DM. Working memory and reward association learning impairments in obesity. Neuropsychologia. 2014;65:146–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Prickett C, Brennan L, Stolwyk R. Examining the relationship between obesity and cognitive function: a systematic literature review. Obes Res Clin Pract. 2015;9:93–113. [DOI] [PubMed] [Google Scholar]
- 113.Vainik U, Baker TE, Dadar M, et al. Neurobehavioral correlates of obesity are largely heritable. PNAS. 2018;115:9312–9317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Seymour KE, Reinblatt SP, Benson L, Carnell S. Overlapping neurobehavioral circuits in ADHD, obesity, and binge eating: evidence from neuroimaging research. CNS Spectr. 2015;20:401–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Svaldi J, Naumann E, Trentowska M, Schmitz F. General and food-specific inhibitory deficits in binge eating disorder. Int J Eat Disord. 2014;47:534–542. [DOI] [PubMed] [Google Scholar]
- 116.Boeka AG, Lokken KL. Prefrontal systems involvement in binge eating. Eating and Weight Disorders- Studies on Anorexia, Bulimia, and Obesity. 2011;16:e131–e126. [DOI] [PubMed] [Google Scholar]
- 117.Duchesne M, Mattos P, Appolinario JC, et al. Assessment of executive functions in obese individuals with binge eating disorder. Braz J Psychiatry. 2010;32:381–388. [DOI] [PubMed] [Google Scholar]
- 118.Mobbs O, Iglesias K, Golay A, Van der Linden M. Cognitive deficits in obese persons with and without binge eating disorder. Investigation using a mental flexibility task. Appetite. 2011;57:263–271. [DOI] [PubMed] [Google Scholar]
- 119.Cury MEG, Berberian A, Scarpato BS, Kerr-Gaffney J, Santos FH, Claudino AM. Scrutinizing Domains of Executive Function in Binge Eating Disorder: A Systematic Review and Meta-Analysis. Front Psychiatry. 2020;11:288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wu M, Hartmann M, Skunde M, Herzog W, Friederich HC. Inhibitory control in bulimic-type eating disorders: a systematic review and meta-analysis. PLoS One. 2013;8:e83412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Balodis IM, Molina ND, Kober H, et al. Divergent neural substrates of inhibitory control in binge eating disorder relative to other manifestations of obesity. Obesity (Silver Spring). 2013;21:367–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hege MA, Stingl KT, Kullmann S, et al. Attentional impulsivity in binge eating disorder modulates response inhibition performance and frontal brain networks. Int J Obes (Lond). 2015;39:353–360. [DOI] [PubMed] [Google Scholar]
- 123.Manasse SM, Goldstein SP, Wyckoff E, et al. Slowing down and taking a second look: Inhibitory deficits associated with binge eating are not food-specific. Appetite. 2016;96:555–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Berridge KC, Kringelbach ML. Pleasure systems in the brain. Neuron. 2015;86:646–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Volkow ND, Wang GJ, Tomasi D, Baler RD. Obesity and addiction: neurobiological overlaps. Obes Rev. 2013;14:2–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Boswell RG, Kober H. Food cue reactivity and craving predict eating and weight gain: a meta-analytic review. Obesity Reviews. 2016;17:159–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Balodis IM, Kober H, Worhunsky PD, et al. Monetary reward processing in obese individuals with and without binge eating disorder. Biol Psychiatry. 2013;73:877–886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Balodis IM, Grilo CM, Kober H, et al. A pilot study linking reduced fronto-Striatal recruitment during reward processing to persistent bingeing following treatment for binge-eating disorder. Int J Eat Disord. 2014;47:376–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Luijten M, Schellekens AF, Kuhn S, Machielse MWJ, Sescousse G. Disruption of Reward Processing in Addiction : An Image-Based Meta-analysis of Functional Magnetic Resonance Imaging Studies. JAMA Psychiatry. 2017;74:387–398. [DOI] [PubMed] [Google Scholar]
- 130.Reiter AM, Heinze HJ, Schlagenhauf F, Deserno L. Impaired Flexible Reward-Based Decision-Making in Binge Eating Disorder: Evidence from Computational Modeling and Functional Neuroimaging. Neuropsychopharmacology. 2017;42:628–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Schienle A, Schafer A, Hermann A, Vaitl D. Binge-eating disorder: reward sensitivity and brain activation to images of food. Biol Psychiatry. 2009;65:654–661. [DOI] [PubMed] [Google Scholar]
- 132.Lee JE, Namkoong K, Jung YC. Impaired prefrontal cognitive control over interference by food images in binge-eating disorder and bulimia nervosa. Neurosci Lett. 2017;651:95–101. [DOI] [PubMed] [Google Scholar]
- 133.Karhunen LJ, Vanninen EJ, Kuikka JT, Lappalainen R, Tiihonen J, Uusitupa M. Regional cerebral blood flow during exposure to food in obese binge eating women. Psychiatry Research: Neuroimaging. 2000;99:29–42. [DOI] [PubMed] [Google Scholar]
- 134.Wolz I, Sauvaget A, Granero R, et al. Subjective craving and event-related brain response to olfactory and visual chocolate cues in binge-eating and healthy individuals. Sci Rep. 2017;7:41736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Svaldi J, Tuschen-Caffier B, Peyk P, Blechert J. Information processing of food pictures in binge eating disorder. Appetite. 2010;55:685–694. [DOI] [PubMed] [Google Scholar]
- 136.Weygandt M, Schaefer A, Schienle A, Haynes JD. Diagnosing different binge-eating disorders based on reward-related brain activation patterns. Hum Brain Mapp. 2012;33:2135–2146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Nock MK, Wedig MM, Holmberg EB, Hooley JM. The Emotion Reactivity Scale: Development, Evaluation, and Relation to Self-Injurious Thoughts and Behaviors. Behavior Therapy. 2008;39:107–116. [DOI] [PubMed] [Google Scholar]
- 138.Naish KR, Laliberte M, MacKillop J, Balodis IM. Systematic review of the effects of acute stress in binge eating disorder. Eur J Neurosci. 2019;50:2415–2429. [DOI] [PubMed] [Google Scholar]
- 139.Leehr EJ, Krohmer K, Schag K, Dresler T, Zipfel S, Giel KE. Emotion regulation model in binge eating disorder and obesity - a systematic review. Neuroscience & Biobehavioral Reviews. 2015;49:125–134. [DOI] [PubMed] [Google Scholar]
- 140.Haedt-Matt AA, Keel PK. Revisiting the affect regulation model of binge eating: a meta-analysis of studies using ecological momentary assessment. Psychol Bull. 2011;137:660–681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Adam TC, Epel ES. Stress, eating and the reward system. Physiol Behav. 2007;91:449–458. [DOI] [PubMed] [Google Scholar]
- 142.Zeeck A, Stelzer N, Linster HW, Joos A, Hartmann A. Emotion and eating in binge eating disorder and obesity. Eur Eat Disord Rev. 2011;19:426–437. [DOI] [PubMed] [Google Scholar]
- 143.Klatzkin RR, Gaffney S, Cyrus K, Bigus E, Brownley KA. Binge eating disorder and obesity: Preliminary evidence for distinct cardiovascular and psychological phenotypes. Physiology & Behavior. 2015;142:20–27. [DOI] [PubMed] [Google Scholar]
- 144.Schag K, Teufel M, Junne F, et al. Impulsivity in binge eating disorder: food cues elicit increased reward responses and disinhibition. PLoS One. 2013;8:e76542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Pecina S, Smith KS, Berridge KC. Hedonic hot spots in the brain. Neuroscientist. 2006;12:500–511. [DOI] [PubMed] [Google Scholar]
- 146.Wagner DD, Boswell RG, Kelley WM, Heatherton TF. Inducing Negative Affect Increases the Reward Value of Appetizing Foods in Dieters. Journal of Cognitive Neuroscience. 2012;24:1625–1633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Gluck ME, Geliebter A, Hung J, Yahav E. Cortisol, hunger, and desire to binge eat following a cold stress test in obese women with binge eating disorder. Psychosom Med. 2004;66:876–881. [DOI] [PubMed] [Google Scholar]
- 148.Gluck ME. Stress response and binge eating disorder. Appetite. 2006;46:26–30. [DOI] [PubMed] [Google Scholar]
- 149.Goldfield GS, Adamo KB, Rutherford J, Legg C. Stress and the relative reinforcing value of food in female binge eaters. Physiol Behav. 2008;93:579–587. [DOI] [PubMed] [Google Scholar]
- 150.Lyu Z, Jackson T. Acute Stressors Reduce Neural Inhibition to Food Cues and Increase Eating Among Binge Eating Disorder Symptomatic Women. Front Behav Neurosci. 2016;10:188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.McElroy SL, Guerdjikova AI, Mori N, Keck PE Jr. Psychopharmacologic treatment of eating disorders: emerging findings. Curr Psychiatry Rep. 2015;17:35. [DOI] [PubMed] [Google Scholar]
- 152.McElroy SL, Guerdjikova AI, Mori N, Munoz MR, Keck PE. Overview of the treatment of binge eating disorder. CNS Spectr. 2015;20:546–556. [DOI] [PubMed] [Google Scholar]
- 153.Madras BK, Miller GM, Fischman AJ. The dopamine transporter and attention-deficit/hyperactivity disorder. Biol Psychiatry. 2005;57:1397–1409. [DOI] [PubMed] [Google Scholar]
- 154.Kuczenski R, Segal DS. Effects of Methyiphenidate on Extracellular Dopamine, Serotonin, and Norepinephrine: Comparison with Amphetamine. Journal of Neurochemistry. 1997;58:2032–2037. [DOI] [PubMed] [Google Scholar]
- 155.Poulton AS, Hibbert EJ, Champion BL, Nanan RK. Stimulants for the Control of Hedonic Appetite. Front Pharmacol. 2016;7:105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Gamo NJ, Wang M, Arnsten AFT. Methylphenidate and Atomoxetine Enhance Prefrontal Function Through a2-Adrenergic and Dopamine D1 Receptors. Journal of the American Academy of Child and Adolescent Psychiatry. 2010;49:1011–1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Swanson J, Baler RD, Volkow ND. Understanding the effects of stimulant medications on cognition in individuals with attention-deficit hyperactivity disorder: a decade of progress. Neuropsychopharmacology. 2011;36:207–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Hutson PH, Pennick M, Secker R. Preclinical pharmacokinetics, pharmacology and toxicology of lisdexamfetamine: a novel d-amphetamine pro-drug. Neuropharmacology. 2014;87:41–50. [DOI] [PubMed] [Google Scholar]
- 159.Vickers SP, Hackett D, Murray F, Hutson PH, Heal DJ. Effects of lisdexamfetamine in a rat model of binge-eating. J Psychopharmacol. 2015;29:1290–1307. [DOI] [PubMed] [Google Scholar]
- 160.Vickers SP, Goddard S, Brammer RJ, Hutson PH, Heal DJ. Investigation of impulsivity in binge-eating rats in a delay-discounting task and its prevention by the d-amphetamine prodrug, lisdexamfetamine. J Psychopharmacol. 2017;31:784–797. [DOI] [PubMed] [Google Scholar]
- 161.McElroy SL, Mitchell JE, Wilfley D, et al. Lisdexamfetamine Dimesylate Effects on Binge Eating Behavior and Obsessive-Compulsive and Impulsive Features in Adults with Binge Eating Disorder. Eur Eat Disord Rev. 2016;24:223–231. [DOI] [PubMed] [Google Scholar]
- 162.Citrome L, Kando JC, Bliss C. Relationships between clinical scales and binge eating days in adults with moderate to severe binge eating disorder in two Phase III studies. Neuropsychiatr Dis Treat. 2018;15:537–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Fleck DE, Eliassen JC, Guerdjikova AI, et al. Effect of lisdexamfetamine on emotional network brain dysfunction in binge eating disorder. Psychiatry Res Neuroimaging. 2019;286:53–59. [DOI] [PubMed] [Google Scholar]
- 164.ClinicalTrials.Gov. Cognitive-Behavioral and Pharmacologic (LDX) Treatment of Binge-Eating Disorder and Obesity: Acute Phase (NCT03924193) In: Medicine USNLo, ed. Vol 2020. [Google Scholar]
- 165.ClinicalTrials.Gov. Cognitive-Behavioral and Pharmacologic (LDX) Treatment of Binge-Eating Disorder and Obesity: Maintenance Treatment (NCT03926052) In: Medicine USNLo, ed. Vol 2020. [Google Scholar]
- 166.Hopkins SC, Sunkaraneni S, Skende E, et al. Dasotraline in the Treatment of Attention-Deficit/Hyperactivity Disorder in Adults. Clinical Drug Investigation. 2016;36:137–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Grilo CM, McElroy SL, Hudson JI, et al. Efficacy and safety of dasotraline in adults with binge-eating disorder: a randomized, placebo-controlled, fixed-dose clinical trial. CNS Spectr. 2020:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.McElroy SL, Hudson JI, Grilo CM, et al. Efficacy and Safety of Dasotraline in Adults With Binge-Eating Disorder: A Randomized, Placebo-Controlled, Flexible-Dose Clinical Trial. J Clin Psychiatry. 2020;81:19m13068. [DOI] [PubMed] [Google Scholar]
- 169.O’Malley SS, Sinha R, Grilo CM, et al. Naltrexone and cognitive behavioral coping skills therapy for the treatment of alcohol drinking and eating disorder features in alcohol-dependent women: a randomized controlled trial. Alcohol Clin Exp Res. 2007;31:625–634. [DOI] [PubMed] [Google Scholar]
- 170.Yeomans MR, Gray RW. Opioid peptides and the control of human ingestive behavior. Neurosci Biobehav Rev. 2002;26:713–728. [DOI] [PubMed] [Google Scholar]
- 171.O’Malley SS, Krishnan-Sarin S, Farren C, Sinha R, Kreek M. Naltrexone decreases craving and alcohol self-administration in alcohol-dependent subjects and activates the hypothalamo–pituitary–adrenocortical axis. Psychopharmacology (Berl). 2002;160:19–29. [DOI] [PubMed] [Google Scholar]
- 172.Sternbach HA, Annitto W, Pottash ALC, Gold MS. Anorexic effects of naltrexone in man. Lancet. 1982;1:388–389. [DOI] [PubMed] [Google Scholar]
- 173.Bertino M, Beauchamp GK, Engelman K. Naltrexone, an opioid blocker, changes taste perception and nutrient intake in humans. Am J Physiol. 1991;261:R59–63. [DOI] [PubMed] [Google Scholar]
- 174.Spiegel TA, Stunkard AJ, Shrager EE, O’Brien CP, Morrison MF, Stellar E. Effect of naltrexone on food intake, hunger, and satiety in obese men. Physiology & Behavior. 1996;40:135–141. [DOI] [PubMed] [Google Scholar]
- 175.Atkinson RL, Berke LK, Drake CR, Bibbs ML, Williams FL, Kaiser DL. Effects of Long-Term Therapy With Naltrexone on Body Weight in Obesity. Clin Pharmacol Ther. 1985;38:419–422. [DOI] [PubMed] [Google Scholar]
- 176.Maggio CA, Presta E, Bracco EF, et al. Naltrexone and human eating behavior: a dose-ranging inpatient trial in moderately obese men. Brain Research Bulletin. 1985;14. [DOI] [PubMed] [Google Scholar]
- 177.McElroy SL, Guerdjikova AI, Blom TJ, et al. A placebo-controlled pilot study of the novel opioid receptor antagonist ALKS-33 in binge eating disorder. Int J Eat Disord. 2013;46:239–245. [DOI] [PubMed] [Google Scholar]
- 178.Ziauddeen H, Chamberlain SR, Nathan PJ, et al. Effects of the mu-opioid receptor antagonist GSK1521498 on hedonic and consummatory eating behaviour: a proof of mechanism study in binge-eating obese subjects. Mol Psychiatry. 2013;18:1287–1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Hurt RD, Sachs DPL, Glover ED, et al. A Comparison of Sustained-Release Bupropion and Placebo for Smoking Cessation. N Engl J Med. 1997;337:1195–1202. [DOI] [PubMed] [Google Scholar]
- 180.Jorenby DE, Leischo SJ, Nides MA, et al. A controlled trial of sustained-release bupropion, a nicotine patch, or both for smoking cessation. N Engl J Med. 1999;340:685–691. [DOI] [PubMed] [Google Scholar]
- 181.Newton TF, Roache JD, De La Garza R, et al. Bupropion Reduces Methamphetamine-Induced Subjective Effects and Cue-Induced Craving. Neuropsychopharmacology. 2005;31:1537–1544. [DOI] [PubMed] [Google Scholar]
- 182.Jain AK, Kaplan RA, Gadde KM, et al. Bupropion SR vs. placebo for weight loss in obese patients with depressive symptoms Obes Res. 2002;10:1049–1056. [DOI] [PubMed] [Google Scholar]
- 183.White MA, Grilo CM. Bupropion for Overweight Women with Binge Eating Disorder: Randomized Double-blind Placebo-controlled Trial. J Clin Psychiatry. 2014;74:400–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Butler K, Le Foll B. Impact of Substance Use Disorder Pharmacotherapy on Executive Function: A Narrative Review. Front Psychiatry. 2019;10:98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Maneeton N, Naneeton B, Srisurapanont M, Martin SD. Bupropion for adults with attention-deficit hyperactivity disorder: Meta-analysis of randomized, placebo-controlled trials. Psychiatry and Clinical Neuroscience. 2011;65:611–617. [DOI] [PubMed] [Google Scholar]
- 186.Greenway FL, Whitehouse MK, Guttadauria M, Anderson JW, Atkinson RL, Fujioka K. Rational design of a combination medication for the treatment of obesity. Obesity (Silver Spring). 2009;17:30–39. [DOI] [PubMed] [Google Scholar]
- 187.Billes SK, Greenway FL. Combination therapy with naltrexone and bupropion for obesity.. Expert Opin Pharmacother. 2011;12:1813–1826. [DOI] [PubMed] [Google Scholar]
- 188.Greenway FL, Dunayevich E, Tollefson GD, et al. Comparison of Combined Bupropion and Naltrexone Therapy for Obesity with Monotherapy and Placebo. J Clin Endocrinol Metab. 2009;94:4898–4906. [DOI] [PubMed] [Google Scholar]
- 189.Greenway FL, Fujioka K, Plodkowski RA, et al. Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (COR-I): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet. 2010;376:21–27. [DOI] [PubMed] [Google Scholar]
- 190.Yanovski SZ, Yanovski JA. Naltrexone Extended-Release Plus Bupropion Extended-Release for Treatment of Obesity. JAMA. 2015;313:1213–1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Wang GJ, Tomasi D, Volkow ND, et al. Effect of combined naltrexone and bupropion therapy on the brain’s reactivity to food cues. Int J Obes (Lond). 2014;38:682–688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Carbone EA, Caroleo M, Rania M, et al. An open-label trial on the efcacy and tolerability of naltrexone/bupropion SR for treating altered eating behaviours and weight loss in binge eating disorder. Eating and Weight Disorders- Studies on Anorexia, Bulimia, and Obesity. 2020. [DOI] [PubMed] [Google Scholar]
- 193.Guerdjikova AI, Walsh B, Shan K, Halesth AE, Dunayevich E, McElroy SL. Concurrent Improvement in Both Binge Eating and Depressive Symptoms with Naltrexone/Bupropion Therapy in Overweight or Obese Subjects with Major Depressive Disorder in an Open-Label, Uncontrolled Study. Advances in Therapy. 2017;34:2307–2315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Grilo CM, Lydecker JA, Morgan PT, Gueorguieva R. A Randomized Controlled Pilot Study Testing Naltrexone and Bupropion Combination for the Teratment of Binge-Eating Disorder with Obesity. Clin Therapeutics. Under Review. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.ClinicalTrials.Gov. Behavioral and Pharmacologic Treatment of Binge Eating and Obesity (NCT03045341; NCT03063606; NCT03047005) In: Medicine USNLo, ed. Vol 2020. [Google Scholar]
- 196.ClinicalTrials.Gov. Efficacy and Mechanisms of Naltrexone+Bupropion for Binge Eating Disorder (NCT03539900) In: Medicine USNLo, ed. [Google Scholar]
- 197.Blundell J, Halford JCG. Serotonin and appetite regulation. CNS Drugs. 1998;9:473–495. [Google Scholar]
- 198.Blundell JE, Latham CJ. Pharmacological manipulation of feeding behaviour: possible influences of serotonin and dopamine In: Garattini S, R S, eds. Central mechanisms of anorextic drugs. New York, NY: Raven Press; 1978:83–109. [Google Scholar]
- 199.Clifton PG, Barnfield AMC, Philcox L. A behavioural profile of fluoxetine-induced anorexia. Psychopharmacology (Berl). 1989;97:89–95. [DOI] [PubMed] [Google Scholar]
- 200.McGuirk J, Muscat R, Willner P. Effects of chronically administered fluoxetine and fenfluramine on food intake, body weight and the behavioural satiety sequence. Psychopharmacology (Berl). 1992;106:401–407. [DOI] [PubMed] [Google Scholar]
- 201.Halford JCG, Harrold JA, Lawton CL, Blundell JE. Serotonin (5-HT) Drugs: Effects on Appetite Expression and Use for the Treatment of Obesity. Curr Drug Targets. 2005;6:201–213. [DOI] [PubMed] [Google Scholar]
- 202.Haddock CK, Poston WSC, Dill PL, Foreyt JP, Ericsson M. Pharmacotherapy for obesity: a quantitative analysis of four decades of published randomized clinical trials. International Journal of Obesity. 2002;26:262–273. [DOI] [PubMed] [Google Scholar]
- 203.Padwal RS, Rucker D, Li SK, Curioni C, Lau DCW. Long-term pharmacotherapy for obesity and overweight. Cochrane Database of Systematic Reviews. 2009;4. [DOI] [PubMed] [Google Scholar]
- 204.Goldstein DJ, Rampey AH, Roback PJ, et al. Efficacy and safety of long-term fluoxetine treatment of obesity-maximizing success. Obes Res. 1995;3:481S–490S. [DOI] [PubMed] [Google Scholar]
- 205.Abenhaim L, Moride Y, Brenot F, et al. Appetite-suppressant drugs and the risk of primary pulmonary hypertension. NEJM. 1996;335:609–616. [DOI] [PubMed] [Google Scholar]
- 206.Pharmaceuticals A FDA Issues Complete Response Letter for Lorcaserin New Drug Application 2010.
- 207.Nal. Fluoxetine in the treatment of bulimia nervosa. A multicenter, placebo-controlled, double-blind trial. Fluoxetine Bulimia Nervosa Collaborative Study Group.. Arch Gen Psychiatry. 1992;49:139–142. [PubMed] [Google Scholar]
- 208.Goldstein DJ, Wilson MG, Thompson VL, Potvin JH, Rampey AH. Long-term fluoxetine treatment of bulimia nervosa. Fluoxetine Bulimia Nervosa Research Group.. Br J Psychiatry. 1995;166:660–666. [DOI] [PubMed] [Google Scholar]
- 209.Romano SJ, Halmi KA, Sarkar NP, Koke SC, Lee JS. A placebo-controlled study of fluoxetine in continued treatment of bulimia nervosa after successful acute fluoxetine treatment.. Am J Psychiatry. 2002;159:96–102. [DOI] [PubMed] [Google Scholar]
- 210.Gorka SM, Young CB, Klumpp H, et al. Emotion-based brain mechanisms and predictors for SSRI and CBT treatment of anxiety and depression: a randomized trial. Neuropsychopharmacology. 2019;44:1639–1648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Eddy KT, Dutra L, Bradley R, Westen D. A multidimensional meta-analysis of psychotherapy and pharmacotherapy for obsessive-compulsive disorder. Clinical Psychology Review. 2004;24:1011–1030. [DOI] [PubMed] [Google Scholar]
- 212.Grilo CM, Masheb RM, Wilson GT. Efficacy of cognitive behavioral therapy and fluoxetine for the treatment of binge eating disorder: a randomized double-blind placebo-controlled comparison. Journal of Consulting and Clinical Psychology. 2005;80:1108–1113. [Google Scholar]
- 213.Grilo CM, Masheb RM, Crosby RD. Predictors and moderators of response to cognitive behavioral therapy and medication for the treatment of binge eating disorder. J Consult Clin Psychol. 2012;80:897–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Goldbloom DS, Olmsted MP. Pharmacotherapy of bulimia nervosa with fluoxetine: assessment of clinically significant attitudinal change. Am J Psychiatry. 1993;150:770–774. [DOI] [PubMed] [Google Scholar]
- 215.Aston-Jones G, Smith RJ, Sartor GC, et al. Lateral hypothalamic orexin/hypocretin neurons: A role in reward-seeking and addiction. Brain Res. 2010;1314:74–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Dalley JW, Mar AC, Economidou D, Robbins TW. Neurobehavioral mechanisms of impulsivity: fronto-striatal systems and functional neurochemistry. Pharmacol Biochem Behav. 2008;90:250–260. [DOI] [PubMed] [Google Scholar]
- 217.Barson JR. Orexin/hypocretin and dysregulated eating: Promotion of foraging behavior. Brain Res. 2020;1731:145915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Piccoli L, Micioni Di Bonaventura MV, Cifani C, et al. Role of orexin-1 receptor mechanisms on compulsive food consumption in a model of binge eating in female rats. Neuropsychopharmacology. 2012;37:1999–2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Alcaraz-Iborra M, Carvajal F, Lerma-Cabrera JM, Valor LM, Cubero I. Binge-like consumption of caloric and non-caloric palatable substances in ad libitum-fed C57BL/6J mice: pharmacological and molecular evidence of orexin involvement. Behav Brain Res. 2014;272:93–99. [DOI] [PubMed] [Google Scholar]
- 220.Muschamp JW, Hollander JA, Thompson JL, et al. Hypocretin (orexin) facilitates reward by attenuating the antireward effects of its cotransmitter dynorphin in ventral tegmental area. Proc Natl Acad Sci U S A 2014;111:E1648–1655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Gentile TA, Simmons SJ, Watson MN, et al. Effects of Suvorexant, a Dual Orexin/Hypocretin Receptor Antagonist, on Impulsive Behavior Associated with Cocaine. Neuropsychopharmacology. 2018;43:1001–1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Wiskerke J, James MH, Aston-Jones G. The orexin-1 receptor antagonist SB-334867 reduces motivation, but not inhibitory control, in a rat stop signal task. Brain Res. 2020;1731:146222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Di Marzo V, Ligresti A, Cristino L. The endocannabinoid system as a link between homoeostatic and hedonic pathways involved in energy balance regulation. Int J Obes (Lond). 2009;33 Suppl 2:S18–24. [DOI] [PubMed] [Google Scholar]
- 224.Carai MAM, Colombo G, MAccioni P, Gessa GL. Efficacy of Rimonabant and Other Cannabinoid CB1 Receptor Antagonists in Reducing Food Intake and Body Weight: Preclinical and Clinical Data. CNS Drug Review. 2006;12:91–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.McLaughlin PJ, Winston K, Swezey L, et al. The cannabinoid CB1 antagonists SR 141716A and AM 251 suppress food intake and food-reinforced behavior in a variety of tasks in rats. Behavioral Pharmacology. 2003;14:583–588. [DOI] [PubMed] [Google Scholar]
- 226.Parylak SL, Cottone P, Sabino V, Rice KC, Zorrilla EP. Effects of CB1 and CRF1 receptor antagonists on binge-like eating in rats with limited access to a sweet fat diet: lack of withdrawal-like responses. Physiol Behav. 2012;107:231–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Rasmussen EB, Reilly W, Buckley J, Boomhower SR. Rimonabant reduces the essential value of food in the genetically obese Zucker rat: an exponential demand analysis. Physiol Behav. 2012;105:734–741. [DOI] [PubMed] [Google Scholar]
- 228.Addy C, Wright H, Van Laere K, et al. The acyclic CB1R inverse agonist taranabant mediates weight loss by increasing energy expenditure and decreasing caloric intake. Cell Metab. 2008;7:68–78. [DOI] [PubMed] [Google Scholar]
- 229.Foltin RW, Haney M. Effects of the cannabinoid antagonist SR141716 (rimonabant) and d-amphetamine on palatable food and food pellet intake in non-human primates. Pharmacol Biochem Behav. 2007;86:766–773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Scherma M, Fattore L, Buscino F, et al. Pharmacological modulation of the endocannabinoid signalling alters binge-type eating behaviour in female rats. British Journal of Pharmacology. 2013;169:820–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Dore R, Valenza M, Wang X, Rice KC, Sabino V, Cottone P. The inverse agonist of CB1 receptor SR141716 blocks compulsive eating of palatable food. Addict Biol. 2014;19:849–861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Hernandez G, Oleson EB, Gentry RN, et al. Endocannabinoids promote cocaine-induced impulsivity and its rapid dopaminergic correlates. Biol Psychiatry. 2014;75:487–498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Boomhower SR, Rasmussen EB. Haloperidol and rimonabant increase delay discounting in rats fed high-fat and standard-chow diets. Behav Pharmacol. 2014;25:705–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Van Gaal LF, Rissanen AM, Scheen AJ, Ziegler O, Rössner S. Effects of the cannabinoid-1 receptor blocker rimonabant on weight reduction and cardiovascular risk factors in overweight patients: 1-year experience from the RIO-Europe study. The Lancet. 2005;365:1389–1397. [DOI] [PubMed] [Google Scholar]
- 235.Despres JP, Golay A, Sjostrom L. Effects of Rimonabant on Metabolic Risk Factors in Overweight Patients with Dyslipidemia. NEJM. 2005;353:2121–2134. [DOI] [PubMed] [Google Scholar]
- 236.Pi-Sunyer FX, Aronne LJ, Heshmati HM, Devin J, Rosenstock J. Effect of Rimonabant, a Cannabinoid-1 Receptor Blocker, on Weight and Cardiometabolic Risk Factors in Overweight or Obese Patients RIO-North America: A Randomized Controlled Trial. JAMA. 2005;295:761–776. [DOI] [PubMed] [Google Scholar]
- 237.Scheen AJ, Finer N, Hollander P, Jensen MD, Van Gaal LF. Efficacy and tolerability of rimonabant in overweight or obese patients with type 2 diabetes: a randomised controlled study. The Lancet. 2006;368:1660–1672. [DOI] [PubMed] [Google Scholar]
- 238.Pataky Z, Gasteyger C, Ziegler O, Rissanen A, Hanotin C, Golay A. Efficacy of rimonabant in obese patients with binge eating disorder. Exp Clin Endocrinol Diabetes. 2013;121:20–26. [DOI] [PubMed] [Google Scholar]
- 239.Yamaguchi T, Wang HL, Li X, Ng TH, Morales M. Mesocorticolimbic glutamatergic pathway. J Neurosci. 2011;31:8476–8490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Bednar I, Qian M, Qureshi GA, et al. Glutamate inhibits Ingestive Behaviour. Journal of Neuroendocrinology. 1994;6:403–408. [DOI] [PubMed] [Google Scholar]
- 241.Stanley BG, Urstadt KR, Charles JR, Kee T. Glutamate and GABA in lateral hypothalamic mechanisms controlling food intake. Physiol Behav. 2011;104:40–46. [DOI] [PubMed] [Google Scholar]
- 242.Duva MA, Tomkins EM, Moranda LM, et al. Reverse microdialysis of N-methyl-D-aspartic acid into the lateral hypothalamus of rats: effects on feeding and other behaviors. Brain Res. 2001;921:122–132. [DOI] [PubMed] [Google Scholar]
- 243.Hung CY, Covasa M, Ritter RC, Burns GA. Hindbrain administration of NMDA receptor antagonist AP-5 increases food intake in the rat. Am J Physiol Regul Integr Comp Physiol. 2006;290:R642–651. [DOI] [PubMed] [Google Scholar]
- 244.Lee SW, Stanley BG. NMDA receptors mediate feeding elicited by neuropeptide Y in the lateral and perifornical hypothalamus. Brain Res. 2005;1063:1–8. [DOI] [PubMed] [Google Scholar]
- 245.McElroy SL, Hudson JI, Capece JA, et al. Topiramate for the Treatment of Binge Eating Disorder Associated With Obesity: A Placebo-Controlled Study. Biol Psychiatry. 2007;61:1039–1048. [DOI] [PubMed] [Google Scholar]
- 246.Nourredine M, Jurek L, Auffret M, et al. Efficacy and safety of topiramate in binge eating disorder: a systematic review and meta-analysis. CNS Spectr. 2020. [DOI] [PubMed] [Google Scholar]
- 247.Claudino AM, de Oliveira IR, Appolinario JC, et al. Double-blind, randomized, placebo-controlled trial of topiramate plus cognitive-behavior therapy in binge-eating disorder. J Clin Psychiatry. 2007;68:1324–1332. [DOI] [PubMed] [Google Scholar]
- 248.Blevins D, Wang X, Sharma S, Ait-Daoud N. Impulsiveness as a predictor of topiramate response for cocaine use disorder. Am J Addictions. 2019;28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.McElroy SL, Shapira NA, Arnold LM, et al. Topiramate in the Long-Term Treatment of Binge-Eating Disorder Associated With Obesity. J Clin Psychiatry. 2004;65:1463–1469. [DOI] [PubMed] [Google Scholar]
- 250.McElroy SL, Arnold LM, Shapira NA, et al. Topiramate in the treatment of binge eating disorder associated with obesity: a randomized, placebo-controlled trial. Am J Psychiatry. 2003;160:255–261. [DOI] [PubMed] [Google Scholar]
- 251.Brennan BP, Roberts JL, Fogarty KV, Reynolds KA, Jonas JM, Hudson JI. Memantine in the treatment of binge eating disorder: an open-label, prospective trial. Int J Eat Disord. 2008;41:520–526. [DOI] [PubMed] [Google Scholar]
- 252.Hermanussen M, Tresguerres JA. A new anti-obesity drug treatment: first clinical evidence that, antagonising glutamate-gated Ca2+ ion channels with memantine normalises binge-eating disorders. Econ Hum Biol. 2005;3:329–337. [DOI] [PubMed] [Google Scholar]
- 253.Bisaga A, Danysz W, Foltin RW. Antagonism of glutamatergic NMDA and mGluR5 receptors decreases consumption of food in baboon model of binge-eating disorder. Eur Neuropsychopharmacol. 2008;18:794–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Popik P, Kos T, Zhang Y, Bisaga A. Memantine reduces consumption of highly palatable food in a rat model of binge eating. Amino Acids. 2011;40:477–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Smith KL, Rao RR, Velazquez-Sanchez C, et al. The uncompetitive N-methyl-D-aspartate antagonist memantine reduces binge-like eating, food-seeking behavior, and compulsive eating: role of the nucleus accumbens shell. Neuropsychopharmacology. 2015;40:1163–1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Cottone P, Iemolo A, Narayan AR, Kwak J, Momaney D, Sabino V. The uncompetitive NMDA receptor antagonists ketamine and memantine preferentially increase the choice for a small, immediate reward in low-impulsive rats. Psychopharmacology (Berl). 2013;226:127–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Murphy ER, Fernando AB, Urcelay GP, et al. Impulsive behaviour induced by both NMDA receptor antagonism and GABAA receptor activation in rat ventromedial prefrontal cortex. Psychopharmacology (Berl). 2012;219:401–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Cruz HG, Ivanova T, Lunn ML, Stoffel M, Slesinger PA, Luscher C. Bi-directional effects of GABA(B) receptor agonists on the mesolimbic dopamine system. Nat Neurosci. 2004;7:153–159. [DOI] [PubMed] [Google Scholar]
- 259.Caprioli D, Sawiak SJ, Merlo E, et al. Gamma aminobutyric acidergic and neuronal structural markers in the nucleus accumbens core underlie trait-like impulsive behavior. Biol Psychiatry. 2014;75:115–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Arima H, Oiso Y. Positive effect of baclofen on body weight reduction in obese subjects: a pilot study. Intern Med. 2010;49:2043–2047. [DOI] [PubMed] [Google Scholar]
- 261.Broft AI, Spanos A, Corwin RL, et al. Baclofen for binge eating: an open-label trial. Int J Eat Disord. 2007;40:687–691. [DOI] [PubMed] [Google Scholar]
- 262.Corwin RL, Boan J, Peters KF, Ulbrecht JS. Baclofen reduces binge eating in a double-blind, placebo-controlled, crossover study. Behav Pharmacol. 2012;23:616–625. [DOI] [PubMed] [Google Scholar]