Deep brain stimulation for obesity: rationale and approach to trial design

Free access

Obesity is one of the most serious public health concerns in the US. While bariatric surgery has been shown to be successful for treatment of morbid obesity for those who have undergone unsuccessful behavioral modification, its associated risks and rates of relapse are not insignificant. There exists a neurological basis for the binge-like feeding behavior observed in morbid obesity that is believed to be due to dysregulation of the reward circuitry. The authors present a review of the evidence of the neuroanatomical basis for obesity, the potential neural targets for deep brain stimulation (DBS), as well as a rationale for DBS and future trial design. Identification of an appropriate patient population that would most likely benefit from this type of therapy is essential. There are also significant cost and ethical considerations for such a neuromodulatory intervention designed to alter maladaptive behavior. Finally, the authors present a consolidated set of inclusion criteria and study end points that should serve as the basis for any trial of DBS for obesity.

ABBREVIATIONSBMI = body mass index; DBS = deep brain stimulation; DSM = Diagnostic and Statistical Manual of Mental Disorders; LH = lateral hypothalamus; NAc = nucleus accumbens; OCD = obsessive-compulsive disorder; PD = Parkinson’s disease; PWS = Prader-Willi syndrome; QALY = quality-adjusted life year; VMH = ventromedial hypothalamus; YFAS = Yale Food Addiction Scale.

Abstract

Obesity is one of the most serious public health concerns in the US. While bariatric surgery has been shown to be successful for treatment of morbid obesity for those who have undergone unsuccessful behavioral modification, its associated risks and rates of relapse are not insignificant. There exists a neurological basis for the binge-like feeding behavior observed in morbid obesity that is believed to be due to dysregulation of the reward circuitry. The authors present a review of the evidence of the neuroanatomical basis for obesity, the potential neural targets for deep brain stimulation (DBS), as well as a rationale for DBS and future trial design. Identification of an appropriate patient population that would most likely benefit from this type of therapy is essential. There are also significant cost and ethical considerations for such a neuromodulatory intervention designed to alter maladaptive behavior. Finally, the authors present a consolidated set of inclusion criteria and study end points that should serve as the basis for any trial of DBS for obesity.

Obesity is defined as a body mass index (BMI) greater than 30 kg/m2. Recent estimates suggest that the prevalence of obesity in the US is greater than 35%,19 with more than 500 million obese individuals worldwide.86 Obesity has been linked to an increased risk of hypertension, hyperlipidemia, Type 2 diabetes, stroke, coronary artery disease, and a variety of cancers.9,31 Furthermore, health care expenditures in obese individuals are as much as 45% higher than those in lean individuals, and economic-based models have estimated that health care costs related to obesity total $75 billion per year in the US alone.34,94 Based on these data, obesity is one of the most pressing public health concerns.

The standard of care, first-line treatment for obesity is lifestyle modification.40 While dietary treatment can be effective in the immediate short term, the vast majority of patients fail to achieve sustained, long-term weight loss.2 The success rate of dietary treatment is increased somewhat when combined with adjuvant group therapy, behavior modification, and/or active follow-up. However, the majority of patients ultimately relapse after an initial period of weight loss.2 Bariatric surgery is indicated in patients with BMIs greater than 40 kg/m2 (or > 35 kg/m2 in the presence of obesity-related comorbidities) who have undergone unsuccessful nonsurgical treatment.18 These procedures have been found to be superior to behavioral modification alone,13 and thus the number of bariatric procedures performed in the US has increased 10-fold over the past two decades.50 Bariatric surgery is one of the only effective treatments for long-term weight loss in morbid obesity.14 As a result, there has been an increasing demand for these surgical procedures. Failure of bariatric surgery is most often defined as less than 50% of excess weight loss at 18 months after the operation, with or without a BMI greater than 35 m/kg2.51 Nonetheless, the rates of both short-term and long-term morbidity of bariatric surgery are not insignificant.21,74 While reported failure rates are highly variable in the literature and further depend on patient selection, degree of obesity, and surgical technique,1,12 relapse of metabolic syndrome and weight regain occur in as many as 35% of surgically treated patients at long-term follow-up.28

Radiological Evidence

Both the hypothalamus and the nucleus accumbens (NAc), two brain regions with known interconnections, have been implicated in imaging studies of obese humans.32 Sweet tastes such as glucose, sucrose, and other palatable tastes, as well as images of calorically dense food, induce abnormal responses in the NAc and hypothalamus in obese patients compared with lean controls.30,52,78 Moreover, the serotonin noradrenaline reuptake inhibitor sibutramine results in attenuated hypothalamic response to high-calorie food images, and this attenuation correlates with the drug’s impact on weight and eating behavior in obese patients. Additionally, satiety after a meal attenuated the NAc response to high-calorie food images, which also correlated with eating behavior.20 Thus, change in the activity of these regions is linked to change in behavior, making them a promising target for intervention.

Murdaugh and colleagues found that a greater NAc response to high-calorie food images predicted poorer treatment response in obese patients undergoing behavioral weight-loss treatment.60 One study examining brain function in patients receiving bariatric surgery found that abnormalities in hypothalamic response to food images present in obese patients were not found more than 1 year after Roux-en-Y surgery.22 Furthermore, reduced NAc response to high-calorie food images was associated with reduced palatability and appeal of high-calorie foods and healthier eating behaviors among patients after Roux-en-Y surgery.72 Taken together, these findings further suggest these regions as promising targets for deep brain stimulation (DBS) to treat obesity, as their dysfunction appears directly associated with food stimuli and response to treatment.

Deep Brain Stimulation Targets for Obesity

Several potential DBS targets for obesity have already been identified through anatomical investigation of the appetite and reward circuitry of the brain, and studied in animal and human neuroimaging studies. Within the hypothalamus, the lateral hypothalamus (LH) and ventrome-dial hypothalamus (VMH) have long been recognized as the feeding and satiety centers of the brain, respectively. Functional MRI studies have demonstrated the complementary actions of these regions in perpetuating obesity in humans.24,80 Additionally, studies of the behavioral and psychiatric pathophysiology of morbid obesity have also identified altered reward circuitry in the brain that may play a role in food craving, which is commonly observed in these patients.41,44 Feelings of craving, reward anticipation, and reward-driven consumption all involve neural circuitry between the prefrontal cortex, which houses the inhibitory control regions, and the striatum, within which the NAc has traditionally played a prominent role in promoting reward-driven behavior.77,85 (Fig. 1)

FIG. 1.
FIG. 1.

Schematic diagram depicting the DBS targets for obesity and their role in the homeostatic pathway of energy balance in normal and PWS physiology. The cortical inhibitory control regions are located in the prefrontal cortex (PFC), and the subcortical reward circuitry in the nucleus accumbens (NAc), and lateral hypothalamus (LH). The LH is responsible for providing anabolic feedback onto the autonomie nervous system effectors. The NAc is the center of the reward pathway in the brain, integrating inputs from various higher cortical brain areas and the limbic system to reinforce certain beneficial behaviors, such as feeding. Integration of the reward pathways with feeding behavior begins with dopamine release from the ventral tegmental area (VTA) neurons that project onto the NAc. Within the NAc, there are GABAergic neurons that project to the LH, which then contains neurons that drive food intake. These nuclei also respond to various hormonal peptides such as leptin, providing one of the many links between food intake and energy metabolism in the brain. In Prader-Willi syndrome (PWS), the normal cortical inhibitory control and subcortical reward circuitry are disrupted in response to food stimuli with hypoactivation of the PFC and hyperactivation of the NAc and LH compared with healthy individuals. GABA = γ-aminobutyric acid. Modified with permission from Ho et al: Cureus 7:e259, 2015.36

In animal models, lesioning studies of the LH have revealed weight loss, and bilateral DBS in rats produced a significant weight differential compared with nonstimu-lated controls.68 A recent pilot study of DBS of the LH in 3 morbidly obese patients who had previously failed gastric bypass surgery was completed. While the study was designed to assess safety and not efficacy of DBS for obesity, stimulation parameters of the LH were optimized to induce a sustained increase in resting metabolism with stimulation, after which weight loss was observed, and no detrimental psychiatric symptoms were detected.90 In a similar fashion, lesions to the VMH have led to weight gain, increased adipose tissue, and hyperinsulinemia in animal models, with similar effects observed with high-frequency DBS in both murine and primate models.5,46 Low-frequency stimulation of this focus appeared to inhibit feeding behavior and increase weight loss in a wide variety of animal models, including in a recent study in primates in which Torres and colleagues proposed an intraventricular “floating” electrode inserted in the third ventricle contiguous to the VMH. Stimulation delivered to the VMH has been shown to evoke emotional panic attack-like behavior with autonomic disturbances in both animals and humans, which may preclude its suitability as a human DBS target for obesity.71,92 Nevertheless, VMH target refinement is already underway in some centers. Finally, lesioning studies involving the NAc have led to promising results in animal studies with elimination of food-hoarding behavior and significant weight loss.43 Furthermore, proof-of-concept DBS studies of the NAc specifically aimed at examining its effects on feeding behavior found a significant decrease in binge-eating behavior, as well as decreased caloric intake and sustained weight loss in obese mice. These authors also found that these stimulatory effects involved D2 receptors.32 Moreover, the NAc is a well-validated DBS target with a good safety profile that has been exploited for treatment of such processes such as treatment-refractory depression, obsessive-compulsive disorder (OCD), and alcoholism.27,58,70 Given the link between obesity and the mesolimbic reward circuitry, further studies targeting the NAc with DBS, specifically for treatment-refractory obesity, should be considered promising.

Cost and Ethical Considerations of DBS

Cost Considerations

In our current health care delivery paradigm, costs must be included in the consideration of any novel treatment modality. In evaluating the fiscal efficacy of DBS, it is reasonable to examine data comparing DBS to medical management of Parkinson’s disease (PD). Several studies have demonstrated the economic superiority of DBS over medical management in PD.

In terms of absolute costs, most recent cost estimates based on the Department of Veterans Affairs and Medicare databases are approximately $17,000–65,000 for initial DBS surgery, with up to an additional $17,000 in annual costs for the first 3 years. However, these data were extracted for patients with PD and encompass condition-specific costs as well.75,79 Tomaszewski and Holloway reported that DBS in PD becomes cost effective if quality of life is improved by 18% compared with best medical management.82 Meissner et al. retrospectively studied treatment costs in DBS in PD versus medical management.55 They reported an increase in costs by 32% in the first year but a decrease by 54% in the second year. They further reported a decrease in overall medication expenses and improved efficacy in patients undergoing DBS.

In contrast, in a head-to-head study of more than 3600 bariatric surgery patients and comorbidity-matched, bar-iatric-surgery eligible controls, initial costs associated with laparoscopic bariatric surgery were estimated at $30,000, but postprocedure costs savings began to accrue as soon as 3 months after the operation. The initial investment costs for the procedure were returned within 2 years, and were mainly associated with a decrease in health care utilization from obesity-related comorbidities. In addition, savings rates in the bariatric surgery cohort compared with controls increased to more than $900 per month 1 year after surgery. This analysis did not include additional quality of life and length of life benefits, nor did it include cost benefits associated with decreased disability and unemployment.15 A separate study conducting an incremental cost-effectiveness analysis for bariatric surgery found that laparoscopic bariatric surgery was highly effective, with an associated cost of slightly more than $6000 for each quality-adjusted life year (QALY) gained. This favorable cost-effectiveness result was maintained even in complete weight regain scenarios, with an incremental increase to $24,000 for each QALY gained.89 In considering DBS as an adjunct treatment following bariatric surgery failure, there are clear cost gains to be achieved as long as similar and more enduring weight-loss results are achieved. However, given the higher surgical costs associated with DBS, there will likely be a longer interval to return on investment, as well as higher costs for each QALY gained compared with bariatric surgery.

With only a few reports of DBS in obesity there are no similar studies of actual cost effectiveness. Pisapia et al. conducted a decision analysis comparing the two major surgical options for obesity: laparoscopic Roux-en-Y gastric bypass and laparoscopic adjustable gastric banding.65 Laparoscopic banding was found to not be effective at achieving successful weight loss (defined as at least 45% of excess weight), and the complication rates for gastric bypass surgery (33%) far exceeded those of DBS (19%). These reports characterize DBS in obesity as a promising therapeutic avenue from an economic standpoint. With lower complication rates compared with bariatric surgery, targeted neuromodulation could decrease long-term costs in the morbidly obese patient population, especially in patients who have already failed bariatric surgery.

Ethical Considerations

As indications for neuromodulation continue to expand, there will be an ongoing debate on the ethics of intervention. The neural circuitry of obesity overlaps with that of addiction, and this may raise concerns of patient autonomy in the context of behavioral alteration.10 Specifically, targeting the NAc may lead to alterations in reward pathways or withdrawal. A patient’s ability to experience normal pleasure may also be diminished or perturbed. The ethical argument is similar to the one for OCD in which benefit-harm ethical arguments favor DBS in patients with high-level decisional capacity.26 Moreover, autonomy may be fundamentally inadequate in individuals suffering from disorders of the reward circuitry, requiring experience-based paternalism on the part of doctors.11

An alternative perspective for patient autonomy suggests that neuromodulation may actually restore patient capacity to make choices. For example, Uusitalo argues that there is a distinction between difficulty and freedom in the ability of a patient to make a choice. Using the example of cravings in addiction, she eloquently highlights the possibility that DBS may reduce the difficulty in controlling cravings but, importantly, will not change a patient’s freedom to continue addictive behaviors.83 Müller et al. has gone further to characterize DBS as actually granting “full autonomy” to the patient.59

The ethical underpinnings of DBS treatment for obesity will be further challenged in the clinical-trial phase. Several key factors must be clarified prior to enrollment, including trial design, informed consent, and a greater discussion on the concept that DBS treatment would be a threat to patient identity.64 As such, we advocate for a multidisciplinary team of surgeons, endocrinologists, obesity specialists, neuropsychiatrists, and ethicists to provide longitudinal guidance for delivering ethical and high-level care. We believe that in an appropriately selected population of patients who are refractory to medical and surgical management of morbid obesity, there is a role for DBS. There must be stringent inclusion criteria, informed consent, and clinical equipoise in conducting clinical trials.29

Target Patient Population

A safety and feasibility trial of DBS for obesity in humans should involve those patients who have undergone unsuccessful bariatric surgery (specifically gastric bypass) due to persistent binge-eating behavior. Predictors of bariatric surgical failure have been studied extensively and include age, sex, preoperative BMI, substance abuse, compliance with follow-up appointments, eating habits, physical activity, and psychiatric status.89 With respect to the latter, there is a growing body of evidence that patients with mood or anxiety disorders have higher failure rates after bariatric surgery.16 Furthermore, there is a high prevalence of “loss of control” or binge eating in those individuals who regain weight. Interestingly, bariatric surgery has been repeatedly shown to improve binge-eating postop-eratively.53,87 However, at 2 years after surgery, Sallet et al. noted that the association between presurgery binge eating status and higher postsurgery weight regain are most clear.66 The question remains whether ongoing maladap-tive eating behaviors such as binge eating are critical factors in patients who fail bariatric surgery.23 This would be the treatment-resistant population in which we could target DBS as a multimodality treatment for weight loss.

Demonstration of treatment resistance is essential for all potential DBS trial candidates.25,93 However, for obesity, identification of a particular etiology of treatment failure is also crucial, given that DBS treatment of obesity would occur via modulation of specific neural pathways (such as the reward circuitry). The Yale Food Addiction Scale (YFAS) is the most widely used and accepted tool to measure food addiction (Table 1).23 It is based on the substance use disorder criteria in the Diagnostic and Statistical Manual of Mental Disorders-Fifth Edition (DSM-V), and is similar to other compulsive activity scales.38 There is increasing evidence in the literature to suggest that overeating may be a behavioral addiction similar to substance abuse that shares similar behavioral and neurobiological factors.84,95 Furthermore, there is evidence that scores on the YFAS are associated with brain response to anticipation of taste reward, although its relation to hypothalamic and NAc function has not yet been examined.24 In addition to weight regain, postbariatric surgery patients are at increased risk for developing alcohol and substance abuse disorders45,68 that likely represent an "addiction transfer."91 The YFAS has been validated as predictive of continued emotional and binge-eating behavior following bariatric surgery.13 Furthermore, as noted above, the presence of binge-eating behavior after bariatric surgery is clearly associated with less weight loss and more weight regain.53 Therefore, postbariatric surgery evaluation of food addiction behavior via the YFAS may aid in identifying a sub-population of patients who had unsuccessful bariatric surgery that would most benefit from DBS of the NAc, given the addiction and substance-abuse nature of their disorder.

TABLE 1.

Components and scoring of the YFAS*

Substance-Dependence Symptom from DSM-IVNo. of QuestionsNormative %
Substance taken in larger amount and for longer period than Intended321.7
Persistent desire or repeated unsuccessful attempt to quit471.3
Much time/activity to obtain, use, recover324
Important social, occupational, or recreational activities given up or reduced410.3
Use continues despite knowledge of adverse consequences (e.g., failure to fulfill role obligation, use when physically hazardous)128.3
Tolerance (marked increase in amount; marked decrease in effect)213.5
Characteristic withdrawal symptoms; substance taken to relieve withdrawal316.3
Use causes clinically significant impairment214

Adapted from Gearhardt AN, Corbin WR, Brownell KD. Yale Food Addiction Scale (YFAS). Measurement Instrument Database for the Social Sciences, 2012. Retrieved from www.midss.ie. Copyright: CC BY-NC 3.0 (http://creativecommons.org/licenses/by-nc/3.0/).

Number of questions within the YFAS addressing each symptom of substance dependence as defined by the DSM-IV, and the percentage of normal patients with each symptom (normative %).

The YFAS scoring algorithm with a median symptom count was as follows: if scoring for a specific criterion is ≥ 1, then the criterion for that symptom is met; substance dependence is considered when ≥ 3 symptom criteria are met. The normative percentage of normal patients surveyed with the scale that met YFAS criteria for substance dependence was 11.6%. association between these brain processes and sensitivity to food cues, which could be improved through DBS.62

External eating (sensitivity to external food cues) is associated with higher food addiction scores and decreases in weight loss after bariatric surgery63 and may be another measure to target in DBS trials for obesity. Passamonti and colleagues found that external eating scores modulated the connectivity between the ventral striatum and amygdala while viewing appetizing food images, suggesting an

Inclusion Criteria

Determination of appropriate inclusion criteria for a DBS obesity trial would be similar to previous studies published to date for other DBS trials for psychiatric indications.3,17,57 In most other disease trials for DBS, surgery was offered to patients with severe symptoms in whom medical therapy had failed, and who did not have any other major contraindications to DBS.47 Trial design would largely depend on the proposed DBS target, but the need for a traditional Phase I clinical trial may be obviated by the fact that a reasonable safety profile has been largely established by previous pilot trials/studies of both LH90 and NAc3,17 targets. Thus, proof-of-concept and efficacy studies would be the next logical step. Patients with general contraindications to DBS should be excluded (Table 2). DBS could be offered to adult patients with morbid obesity (BMI > 40 kg/m2, or 35 kg/m2 with comorbidi-ties) in whom medical therapy and bariatric surgery have failed, with greater consideration given to patients with obesity-related comorbidities such as Type 2 diabetes mellitus, cardiovascular disease, obstructive sleep apnea, and others.54

TABLE 2.

Contraindications for DBS

Contraindication
Recent myocardial infarction
Cardiac arrhythmia
Cardiac malformation
Epilepsy
Stroke
Degenerative disorder of the nervous system
Arterial hypertension or hypotension, not controlled by drugs
Autonomic nervous system disorder
Endocrinological illnesses
Major disturbances in electrolyte imbalance (e.g., due to renal insufficiency or hyperaldosteronism)

Given the close relationship between obesity and psychiatric disorders such as addiction, anxiety, and depression, as well as socioeconomic factors,6 patients would have to undergo extensive multidisciplinary evaluation prior to surgery including psychological and socioeconomic evaluation to ensure that appropriate periopera-tive psychological support is provided so that weight loss gains are sustained.42,61 Proposed evaluative end points of a study could include a wide range of biometric markers of obesity such as caloric consumption, resting metabolic rate, and immediate and sustained weight loss, but should also include evaluation of improvements in obesity-related comorbidities and psychological metrics, as well as improvements in overall quality of life (Table 3).

TABLE 3.

DBS for obesity: proposed inclusion criteria and study end points

DBS Study for Obesity
Inclusion criteria
 Failure of bariatric surgery w/ <50% of excess weight loss, w/ or w/o BMI >35 m/kg2, at 18 months or more after the operation52
 Diagnosis of food dependence via YFAS criteria23 (Table 1)
 Normal neurological examination
 Normal head CT scan and cerebral MRI
 Patient not pregnant
 Psychiatric evaluation
 Socioeconomic evaluation
 Ethics Committee/Institutional Review Board approval
 Patient informed and gives written consent
End points
 Immediate and sustained weight loss (BMI)
 Caloric consumption
 Resting metabolic rate
 Improvement in YFAS score
 Amelioration of obesity-related comorbidities
 Improvement in related psychiatric conditions (depression, anxiety, etc.)
 Quality of life

Prader-Willi Syndrome

Finally, given the severity of hyperphagia, the linked neural reward circuitry dysfunction, and the poor outcomes noted with bariatric surgery, patients with Prader-Willi syndrome (PWS) may represent an additional population of potential trial candidates that may most benefit from a DBS intervention tailored around neuromodulation of these very same reward circuits.36 Prader-Willi syndrome is caused by a genetic defect resulting in absent expression of several imprinted genes in the 15q11–q13 region from the paternal chromosome 15.4 The syndrome is characterized by extreme hyperphagia, obesity, and intellectual disability. Patients with PWS have insatiable appetites and are often morbidly obese.7 Nearly 1 of every 3 individuals with PWS are more than 200% of their ideal body weight, and some have even experienced stomach ruptures from over-consumption.76 The metabolic physiology of PWS differs from obesity in normal individuals, such as increased ratio of adiposity to lean mass,81 decreased total and resting energy expenditure,8 and 4 times greater fasting ghrelin levels.33 PWS is a difficult to treat syndrome that has not benefited from the most radical medical and surgical interventions. Bariatric surgery, in particular, has been applied toward PWS with limited effectiveness and concerning safety profiles given the increased overall medical comor-bidity in this population.69

Individuals with PWS consume more food and for longer periods of time than other obese individuals, suggesting a disruption of basic satiety mechanisms and a dysfunctional reward system.48 These disruptions have a basis in manifesting as postmeal hyperactivation of specific brain regions involved in the food satiety and reward circuitry, including the hypothalamus, NAc, amygdala, hippocampus, medial prefrontal cortex, orbitofrontal cortex, and insula.35,56,73 In functional MRI studies comparing individuals with PWS to obese and healthy-weight controls, individuals with PWS demonstrated higher activity in re-ward/limbic regions and lower activity in the hypothalamus before eating compared with controls. Thus, patients with PWS may be predisposed to overconsumption due to abnormal basal activity in these brain regions.37

PWS represents the pathological intersection between reward and satiety circuitry in the human brain that drives uncontrolled feeding behavior and leads to extreme obese states. Targeting this obese subgroup of individuals may be a reasonable first approach to neuromodulation for obesity given the well-known medical refractoriness of this population.36 Clinical trials targeting the LH are underway for these patients, and future trials of NAc DBS are planned. Specifically, the LH has already been targeted via DBS for obesity90 and headache,39 and the NAc for OCD, anxiety, addiction, and depression.49 We propose that these same targets may be potential targets for DBS in PWS (Fig. 1).36

Discussion

Despite the success of bariatric surgery in the treatment of obesity, there still exists a significant proportion of patients who fail surgical therapy. A neural basis for overeating exhibited in obese patients has been elucidated by both functional imaging as well as DBS studies in animals and limited DBS experience in human patients as well. Specifically, both the hypothalamus (feeding and satiety) and the NAc (reward) have been identified as crucial regulators of food eating behavior that have been demonstrated to be responsive to DBS in treating obesity in animals models. Although upfront costs of DBS are nearly twice that of bariatric surgery, similar degrees of cost savings and cost effectiveness noted in bariatric surgery can likely be obtained with DBS as long as its treatment effects endure, and especially if it is used as an adjunct to bariatric surgery in those patients who experience weight regain.

These patients in whom bariatric surgery ultimately fails likely represent a subpopulation of morbidly obese individuals with a psychiatric basis for their continued excessive food consumption that could lend itself to DBS therapy. Patients with PWS may represent an additional treatment indication for DBS given the overlap between the obesity secondary to hyperphagia and the dysregu-lated reward circuitry observed in this disorder that may make them ideal candidates for DBS. Finally, the efficacy of DBS for obesity will have to be definitively evaluated with a clinical trial that will benefit most from stringent and thoughtful inclusion criteria.

Author Contributions

Conception and design: Halpern, Ho. Acquisition of data: Ho, Sussman, Pendharkar, Bohon. Analysis and interpretation of data: Halpern, Ho, Azagury, Bohon. Drafting the article: Ho, Sussman, Pendharkar, Bohon. Critically revising the article: all authors. Reviewed submitted version of manuscript: Halpern, Ho, Azagury. Approved the final version of the manuscript on behalf of all authors: Halpern.

References

  • 1

    Al-Bader IKhoursheed MAl Sharaf KMouzannar DAAshraf AFingerhut A: Revisional laparoscopic gastric pouch resizing for inadequate weight loss after Roux-en-Y gastric bypass. Obes Surg epub ahead of print2015

  • 2

    Ayyad CAndersen T: Long-term efficacy of dietary treatment of obesity: a systematic review of studies published between 1931 and 1999. Obes Rev 1:1131192000

  • 3

    Bewernick BHKayser SSturm VSchlaepfer TE: Long-term effects of nucleus accumbens deep brain stimulation in treatment-resistant depression: evidence for sustained efficacy. Neuropsychopharmacology 37:197519852012

  • 4

    Bittel DCButler MG: Prader-Willi syndrome: clinical genetics, cytogenetics and molecular biology. Expert Rev Mol Med 7:1202005

  • 5

    Brobeck JR: Mechanism of the development of obesity in animals with hypothalamic lesions. Physiol Rev 26:5415591946

  • 6

    Brunner EJMarmot MGNanchahal KShipley MJStans-feld SAJuneja M: Social inequality in coronary risk: central obesity and the metabolic syndrome. Evidence from the Whitehall II study. Diabetologia 40:134113491997

  • 7

    Butler MG: Prader-Willi syndrome: current understanding of cause and diagnosis. Am J Med Genet 35:3193321990

  • 8

    Butler MGTheodoro MFBittel DCDonnelly JE: Energy expenditure and physical activity in Prader-Willi syndrome: comparison with obese subjects. Am J Med Genet A 143A:4494592007

  • 9

    Calle EERodriguez CWalker-Thurmond KThun MJ: Overweight, obesity, and mortality from cancer in a prospec-tively studied cohort of U.S. adults. N Engl J Med 348:162516382003

  • 10

    Caplan A: Denying autonomy in order to create it: the paradox of forcing treatment upon addicts. Addiction 103:191919212008

  • 11

    Caplan AL: Why autonomy needs help. J Med Ethics 40:3013022014

  • 12

    Chang SHStoll CRSong JVarela JEEagon CJColditz GA: The effectiveness and risks of bariatric surgery: an updated systematic review and meta-analysis, 2003–2012. JAMA Surg 149:2752872014

  • 13

    Clark SMSaules KK: Validation of the Yale Food Addiction Scale among a weight-loss surgery population. Eat Behav 14:2162192013

  • 14

    Colquitt JLPickett KLoveman EFrampton GK: Surgery for weight loss in adults. Cochrane Database Syst Rev 8:CD0036412014

  • 15

    Cremieux PYBuchwald HShikora SAGhosh AYang HEBuessing M: A study on the economic impact of bariatric surgery. Am J Manag Care 14:5895962008

  • 16

    de Zwaan MEnderle JWagner SMühlhans BDitzen BGefeller O: Anxiety and depression in bariatric surgery patients: a prospective, follow-up study using structured clinical interviews. J Affect Disord 133:61682011

  • 17

    Denys DMantione MFigee Mvan den Munckhof PKoerselman FWestenberg H: Deep brain stimulation of the nucleus accumbens for treatment-refractory obsessive-compulsive disorder. Arch Gen Psychiatry 67:106110682010

  • 18

    Expert Panel on the Identification Evaluation and Treatment of Overweight in Adults: Clinical guidelines on the identification, evaluation, and treatment of overweight and obesity in adults: executive summary. Am J Clin Nutr 68:8999171998

  • 19

    Flegal KMCarroll MDKit BKOgden CL: Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999–2010. JAMA 307:4914972012

  • 20

    Fletcher PCNapolitano ASkeggs AMiller SRDelafont BCambridge VC: Distinct modulatory effects of satiety and sibutramine on brain responses to food images in humans: a double dissociation across hypothalamus, amygdala, and ventral striatum. J Neurosci 30:14346143552010

  • 21

    Flum DRBelle SHKing WCWahed ASBerk PChapman W: Perioperative safety in the longitudinal assessment of bariatric surgery. N Engl J Med 361:4454542009

  • 22

    Frank SWilms BVeit RErnst BThurnheer MKullmann S: Altered brain activity in severely obese women may recover after Roux-en Y gastric bypass surgery. Int J Obes (Lond) 38:3413482014

  • 23

    Gearhardt ANCorbin WRBrownell KD: Preliminary validation of the Yale Food Addiction Scale. Appetite 52:4304362009

  • 24

    Gearhardt ANYokum SOrr PTStice ECorbin WRBrownell KD: Neural correlates of food addiction. Arch Gen Psychiatry 68:8088162011

  • 25

    Giacobbe PMayberg HSLozano AM: Treatment resistant depression as a failure of brain homeostatic mechanisms: implications for deep brain stimulation. Exp Neurol 219:44522009

  • 26

    Glannon W: Consent to deep brain stimulation for neurological and psychiatric disorders. J Clin Ethics 21:1041112010

  • 27

    Goodman WKFoote KDGreenberg BDRicciuti NBauer RWard H: Deep brain stimulation for intractable obsessive compulsive disorder: pilot study using a blinded, staggered-onset design. Biol Psychiatry 67:5355422010

  • 28

    Gracia-Solanas JAElia MAguilella VRamirez JMMartínez JBielsa MA: Metabolic syndrome after bariatric surgery. Results depending on the technique performed. Obes Surg 21:1791852011

  • 29

    Grant RAHalpern CHBaltuch GHO’Reardon JPCaplan A: Ethical considerations in deep brain stimulation for psychiatric illness. J Clin Neurosci 21:152014

  • 30

    Green EJacobson AHaase LMurphy C: Reduced nucleus accumbens and caudate nucleus activation to a pleasant taste is associated with obesity in older adults. Brain Res 1386:1091172011

  • 31

    Guh DPZhang WBansback NAmarsi ZBirmingham CLAnis AH: The incidence of co-morbidities related to obesity and overweight: a systematic review and meta-analysis. BMC Public Health 9:882009

  • 32

    Halpern CHTekriwal ASantollo JKeating JGWolf JADaniels D: Amelioration of binge eating by nucleus accumbens shell deep brain stimulation in mice involves D2 receptor modulation. J Neurosci 33:712271292013

  • 33

    Haqq AMFarooqi ISO’Rahilly SStadler DDRosenfeld RGPratt KL: Serum ghrelin levels are inversely correlated with body mass index, age, and insulin concentrations in normal children and are markedly increased in Prader-Willi syndrome. J Clin Endocrinol Metab 88:1741782003

  • 34

    Hartman MMartin ABBenson JCatlin A: National health spending in 2011: overall growth remains low, but some payers and services show signs of acceleration. Health Aff (Millwood) 32:87992013

  • 35

    Hinton ECHolland AJGellatly MSSoni SPatterson MGhatei MA: Neural representations of hunger and satiety in Prader-Willi syndrome. Int J Obes (Lond) 30:3133212006

  • 36

    Ho ALSussman ESZhang MPendharkar AVAzagury DEBohon C: Deep brain stimulation for obesity. Cureus 7:e2592015

  • 37

    Holsen LMSavage CRMartin LEBruce ASLepping RJKo E: Importance of reward and prefrontal circuitry in hunger and satiety: Prader-Willi syndrome vs simple obesity. Int J Obes (Lond) 36:6386472012

  • 38

    Hone-Blanchet AFecteau S: Overlap of food addiction and substance use disorders definitions: analysis of animal and human studies. Neuropharmacology 85:81902014

  • 39

    Jenkins BTepper SJ: Neurostimulation for primary headache disorders: Part 2, review of central neurostimulators for primary headache, overall therapeutic efficacy, safety, cost, patient selection, and future research in headache neuromodulation. Headache 51:140814182011

  • 40

    Jensen MDRyan DHApovian CMArd JDComuzzie AGDonato KA: 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. J Am Coll Cardiol 63:25 Pt B298530232014

  • 41

    Johnson PMKenny PJ: Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats. NatNeurosci 13:6356412010

  • 42

    Karlsson JTaft CRydén ASjöström LSullivan M: Ten-year trends in health-related quality of life after surgical and conventional treatment for severe obesity: the SOS intervention study. Int J Obes (Lond) 31:124812612007

  • 43

    Kelley AEStinus L: Disappearance of hoarding behavior after 6-hydroxydopamine lesions of the mesolimbic dopamine neurons and its reinstatement with L-dopa. Behav Neurosci 99:5315451985

  • 44

    Kenny PJ: Reward mechanisms in obesity: new insights and future directions. Neuron 69:6646792011

  • 45

    King WCChen JYMitchell JEKalarchian MASteffen KJEngel SG: Prevalence of alcohol use disorders before and after bariatric surgery. JAMA 307:251625252012

  • 46

    Laćan GDe Salles AAGorgulho AAKrahl SEFrighetto LBehnke EJ: Modulation of food intake following deep brain stimulation of the ventromedial hypothalamus in the vervet monkey. Laboratory investigation. J Neurosurg 108:3363422008

  • 47

    Lang AEHoueto JLKrack PKubu CLyons KEMoro E: Deep brain stimulation: preoperative issues. Mov Disord 21:Suppl 14S171S1962006

  • 48

    Lindgren ACBarkeling BHägg ARitzén EMMarcus CRössner S: Eating behavior in Prader-Willi syndrome, normal weight, and obese control groups. J Pediatr 137:50552000

  • 49

    Lucas-Neto LMourato BNeto DOliveira EMartins HCorreia F: The nucleus accumbens beyond the anterior commissure: implications for psychosurgery. Stereotact Funct Neurosurg 92:2912992014

  • 50

    Maggard MAShugarman LRSuttorp MMaglione MSug-erman HJLivingston EH: Meta-analysis: surgical treatment of obesity. Ann Intern Med 142:5475592005

  • 51

    Mann JPJakes ADHayden JDBarth JH: Systematic review of definitions of failure in revisional bariatric surgery. Obes Surg 25:5715742014

  • 52

    Matsuda MLiu YMahankali SPu YMahankali AWang J: Altered hypothalamic function in response to glucose ingestion in obese humans. Diabetes 48:180118061999

  • 53

    Meany GConceição EMitchell JE: Binge eating, binge eating disorder and loss of control eating: effects on weight outcomes after bariatric surgery. Eur Eat Disord Rev 22:87912014

  • 54

    Mechanick JIYoudim AJones DBGarvey WTHurley DLMcMahon MM: Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient—2013 update: cosponsored by American Association of Clinical Endocrinologists, The Obesity Society, and American Society for Metabolic & Bariatric Surgery. Obesity (Silver Spring) 21:Suppl 1S1S272013

  • 55

    Meissner WSchreiter DVolkmann JTrottenberg TSchneider GHSturm V: Deep brain stimulation in late stage Parkinson’s disease: a retrospective cost analysis in Germany. J Neurol 252:2182232005

  • 56

    Miller JLJames GAGoldstone APCouch JAHe GDriscoll DJ: Enhanced activation of reward mediating prefrontal regions in response to food stimuli in Prader-Willi syndrome. J Neurol Neurosurg Psychiatry 78:6156192007

  • 57

    Mink JWWalkup JFrey KAComo PCath DDelong MR: Patient selection and assessment recommendations for deep brain stimulation in Tourette syndrome. Mov Disord 21:183118382006

  • 58

    Müller UJSturm VVoges JHeinze HJGalazky IHel-dmann M: Successful treatment of chronic resistant alcoholism by deep brain stimulation of nucleus accumbens: first experience with three cases. Pharmacopsychiatry 42:2882912009

  • 59

    Müller UJVoges JSteiner JGalazky IHeinze HJMöller M: Deep brain stimulation of the nucleus accumbens for the treatment of addiction. Ann N Y Acad Sci 1282:1191282013

  • 60

    Murdaugh DLCox JECook EW IIIWeller RE: fMRI reactivity to high-calorie food pictures predicts short- and long-term outcome in a weight-loss program. Neuroimage 59:270927212012

  • 61

    Nicolai AIppoliti CPetrelli MD: Laparoscopic adjustable gastric banding: essential role of psychological support. Obes Surg 12:8578632002

  • 62

    Passamonti LRowe JBSchwarzbauer CEwbank MPvon dem Hagen ECalder AJ: Personality predicts the brain’s response to viewing appetizing foods: the neural basis of a risk factor for overeating. J Neurosci 29:43512009

  • 63

    Pepino MYStein RIEagon JCKlein S: Bariatric surgeryinduced weight loss causes remission of food addiction in extreme obesity. Obesity (Silver Spring) 22:179217982014

  • 64

    Pisapia JMHalpern CHMuller UJVinai PWolf JAWhiting DM: Ethical considerations in deep brain stimulation for the treatment of addiction and overeating associated with obesity. AJOB Neurosci 4:35462013

  • 65

    Pisapia JMHalpern CHWilliams NNWadden TABaltuch GHStein SC: Deep brain stimulation compared with bariatric surgery for the treatment of morbid obesity: a decision analysis study. Neurosurg Focus 29:2E152010

  • 66

    Sallet PCSallet JADixon JBCollis EPisani CELevy A: Eating behavior as a prognostic factor for weight loss after gastric bypass. Obes Surg 17:4454512007

  • 67

    Sani SJobe KSmith AKordower JHBakay RA: Deep brain stimulation for treatment of obesity in rats. J Neurosurg 107:8098132007

  • 68

    Saules KKWiedemann AIvezaj VHopper JAFoster-Hartsfield JSchwarz D: Bariatric surgery history among substance abuse treatment patients: prevalence and associated features. Surg Obes Relat Dis 6:6156212010

  • 69

    Scheimann AOButler MGGourash LCuffari CKlish W: Critical analysis of bariatric procedures in Prader-Willi syndrome. J Pediatr Gastroenterol Nutr 46:80832008

  • 70

    Schlaepfer TECohen MXFrick CKosel MBrodesser DAxmacher N: Deep brain stimulation to reward circuitry alleviates anhedonia in refractory major depression. Neuropsychopharmacology 33:3683772008

  • 71

    Schoenen JDi Clemente LVandenheede MFumal ADe Pasqua VMouchamps M: Hypothalamic stimulation in chronic cluster headache: a pilot study of efficacy and mode of action. Brain 128:9409472005

  • 72

    Scholtz SMiras ADChhina NPrechtl CGSleeth MLDaud NM: Obese patients after gastric bypass surgery have lower brain-hedonic responses to food than after gastric banding. Gut 63:8919022014

  • 73

    Shapira NALessig MCHe AGJames GADriscoll DJLiu Y: Satiety dysfunction in Prader-Willi syndrome demonstrated by fMRI. J Neurol Neurosurg Psychiatry 76:2602622005

  • 74

    Stenberg ESzabo EAgren GNäslund EBoman LBylund A: Early complications after laparoscopic gastric bypass surgery: results from the Scandinavian Obesity Surgery Registry. Ann Surg 260:104010472014

  • 75

    Stephen JHHalpern CHBarrios CJBalmuri UPisapia JMWolf JA: Deep brain stimulation compared with methadone maintenance for the treatment of heroin dependence: a threshold and cost-effectiveness analysis. Addiction 107:6246342012

  • 76

    Stevenson DAHeinemann JAngulo MButler MGLoker JRupe N: Gastric rupture and necrosis in Prader-Willi syndrome. J Pediatr Gastroenterol Nutr 45:2722742007

  • 77

    Stice EYokum SBohon CMarti NSmolen A: Reward circuitry responsivity to food predicts future increases in body mass: moderating effects of DRD2 and DRD4. Neuroimage 50:161816252010

  • 78

    Stoeckel LEWeller RECook EW IIITwieg DBKnowlton RCCox JE: Widespread reward-system activation in obese women in response to pictures of high-calorie foods. Neuroimage 41:6366472008

  • 79

    Stroupe KTWeaver FMCao LIppolito DBarton BRBurnett-Zeigler IE: Cost of deep brain stimulation for the treatment of Parkinson’s disease by surgical stimulation sites. Mov Disord 29:166616742014

  • 80

    Tataranni PADelParigi A: Functional neuroimaging: a new generation of human brain studies in obesity research. Obes Rev 4:2292382003

  • 81

    Theodoro MFTalebizadeh ZButler MG: Body composition and fatness patterns in Prader-Willi syndrome: comparison with simple obesity. Obesity (Silver Spring) 14:168516902006

  • 82

    Tomaszewski KJHolloway RG: Deep brain stimulation in the treatment of Parkinson’s disease: a cost-effectiveness analysis. Neurology 57:6636712001

  • 83

    Uusitalo S: Autonomy and DBS treatment for addicts. AJOB Neurosci 4:49502013

  • 84

    Volkow NDWang GJFowler JSTelang F: Overlapping neuronal circuits in addiction and obesity: evidence of systems pathology. Philos Trans R Soc Lond B Biol Sci 363:319132002008

  • 85

    Volkow NDWang GJTelang FFowler JSLogan JChil-dress AR: Cocaine cues and dopamine in dorsal stria-tum: mechanism of craving in cocaine addiction. J Neurosci 26:658365882006

  • 86

    Vucenik IStains JP: Obesity and cancer risk: evidence, mechanisms, and recommendations. Ann N Y Acad Sci 1271:37432012

  • 87

    Wadden TAFaulconbridge LFJones-Corneille LRSarwer DBFabricatore ANThomas JG: Binge eating disorder and the outcome of bariatric surgery at one year: a prospective, observational study. Obesity (Silver Spring) 19:122012282011

  • 88

    Wang BCWong ESAlfonso-Cristancho RHe HFlum DRArterburn DE: Cost-effectiveness of bariatric surgical procedures for the treatment of severe obesity. Eur J Health Econ 15:2532632014

  • 89

    Wedin SMadan ACorrell JCrowley NMalcolm RKarl Byrne T: Emotional eating, marital status and history of physical abuse predict 2-year weight loss in weight loss surgery patients. Eat Behav 15:6196242014

  • 90

    Whiting DMTomycz NDBailes Jde Jonge LLecoultre VWilent B: Lateral hypothalamic area deep brain stimulation for refractory obesity: a pilot study with preliminary data on safety, body weight, and energy metabolism. J Neurosurg 119:56632013

  • 91

    Wiedemann AASaules KKIvezaj V: Emergence of new onset substance use disorders among post-weight loss surgery patients. Clin Obes 3:1942012013

  • 92

    Wilent WBOh MYBueteflsch CMBailes JECantella DAngle C: Induction of panic attack by stimulation of the ventromedial hypothalamus. J Neurosurg 112:129512982010

  • 93

    Williams NROkun MS: Deep brain stimulation (DBS) at the interface of neurology and psychiatry. J Clin Invest 123:454645562013

  • 94

    Withrow DAlter DA: The economic burden of obesity worldwide: a systematic review of the direct costs of obesity. Obes Rev 12:1311412011

  • 95

    Ziauddeen HFarooqi ISFletcher PC: Obesity and the brain: how convincing is the addiction model?. Nat Rev Neurosci 13:2792862012

If the inline PDF is not rendering correctly, you can download the PDF file here.

Article Information

Correspondence Casey H. Halpern, Department of Neurosurgery, Stanford University, 300 Pasteur Dr., Edwards Bldg. R-293, Stanford, CA 94305. email: chalpern@stanford.edu.

INCLUDE WHEN CITING DOI: 10.3171/2015.3.FOCUS1538.

DISCLOSURE The authors report no conflict of interest concerning the materials or methods used in this study or the findings specified in this paper.

© AANS, except where prohibited by US copyright law.

Headings

Figures

  • View in gallery

    Schematic diagram depicting the DBS targets for obesity and their role in the homeostatic pathway of energy balance in normal and PWS physiology. The cortical inhibitory control regions are located in the prefrontal cortex (PFC), and the subcortical reward circuitry in the nucleus accumbens (NAc), and lateral hypothalamus (LH). The LH is responsible for providing anabolic feedback onto the autonomie nervous system effectors. The NAc is the center of the reward pathway in the brain, integrating inputs from various higher cortical brain areas and the limbic system to reinforce certain beneficial behaviors, such as feeding. Integration of the reward pathways with feeding behavior begins with dopamine release from the ventral tegmental area (VTA) neurons that project onto the NAc. Within the NAc, there are GABAergic neurons that project to the LH, which then contains neurons that drive food intake. These nuclei also respond to various hormonal peptides such as leptin, providing one of the many links between food intake and energy metabolism in the brain. In Prader-Willi syndrome (PWS), the normal cortical inhibitory control and subcortical reward circuitry are disrupted in response to food stimuli with hypoactivation of the PFC and hyperactivation of the NAc and LH compared with healthy individuals. GABA = γ-aminobutyric acid. Modified with permission from Ho et al: Cureus 7:e259, 2015.36

References

1

Al-Bader IKhoursheed MAl Sharaf KMouzannar DAAshraf AFingerhut A: Revisional laparoscopic gastric pouch resizing for inadequate weight loss after Roux-en-Y gastric bypass. Obes Surg epub ahead of print2015

2

Ayyad CAndersen T: Long-term efficacy of dietary treatment of obesity: a systematic review of studies published between 1931 and 1999. Obes Rev 1:1131192000

3

Bewernick BHKayser SSturm VSchlaepfer TE: Long-term effects of nucleus accumbens deep brain stimulation in treatment-resistant depression: evidence for sustained efficacy. Neuropsychopharmacology 37:197519852012

4

Bittel DCButler MG: Prader-Willi syndrome: clinical genetics, cytogenetics and molecular biology. Expert Rev Mol Med 7:1202005

5

Brobeck JR: Mechanism of the development of obesity in animals with hypothalamic lesions. Physiol Rev 26:5415591946

6

Brunner EJMarmot MGNanchahal KShipley MJStans-feld SAJuneja M: Social inequality in coronary risk: central obesity and the metabolic syndrome. Evidence from the Whitehall II study. Diabetologia 40:134113491997

7

Butler MG: Prader-Willi syndrome: current understanding of cause and diagnosis. Am J Med Genet 35:3193321990

8

Butler MGTheodoro MFBittel DCDonnelly JE: Energy expenditure and physical activity in Prader-Willi syndrome: comparison with obese subjects. Am J Med Genet A 143A:4494592007

9

Calle EERodriguez CWalker-Thurmond KThun MJ: Overweight, obesity, and mortality from cancer in a prospec-tively studied cohort of U.S. adults. N Engl J Med 348:162516382003

10

Caplan A: Denying autonomy in order to create it: the paradox of forcing treatment upon addicts. Addiction 103:191919212008

11

Caplan AL: Why autonomy needs help. J Med Ethics 40:3013022014

12

Chang SHStoll CRSong JVarela JEEagon CJColditz GA: The effectiveness and risks of bariatric surgery: an updated systematic review and meta-analysis, 2003–2012. JAMA Surg 149:2752872014

13

Clark SMSaules KK: Validation of the Yale Food Addiction Scale among a weight-loss surgery population. Eat Behav 14:2162192013

14

Colquitt JLPickett KLoveman EFrampton GK: Surgery for weight loss in adults. Cochrane Database Syst Rev 8:CD0036412014

15

Cremieux PYBuchwald HShikora SAGhosh AYang HEBuessing M: A study on the economic impact of bariatric surgery. Am J Manag Care 14:5895962008

16

de Zwaan MEnderle JWagner SMühlhans BDitzen BGefeller O: Anxiety and depression in bariatric surgery patients: a prospective, follow-up study using structured clinical interviews. J Affect Disord 133:61682011

17

Denys DMantione MFigee Mvan den Munckhof PKoerselman FWestenberg H: Deep brain stimulation of the nucleus accumbens for treatment-refractory obsessive-compulsive disorder. Arch Gen Psychiatry 67:106110682010

18

Expert Panel on the Identification Evaluation and Treatment of Overweight in Adults: Clinical guidelines on the identification, evaluation, and treatment of overweight and obesity in adults: executive summary. Am J Clin Nutr 68:8999171998

19

Flegal KMCarroll MDKit BKOgden CL: Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999–2010. JAMA 307:4914972012

20

Fletcher PCNapolitano ASkeggs AMiller SRDelafont BCambridge VC: Distinct modulatory effects of satiety and sibutramine on brain responses to food images in humans: a double dissociation across hypothalamus, amygdala, and ventral striatum. J Neurosci 30:14346143552010

21

Flum DRBelle SHKing WCWahed ASBerk PChapman W: Perioperative safety in the longitudinal assessment of bariatric surgery. N Engl J Med 361:4454542009

22

Frank SWilms BVeit RErnst BThurnheer MKullmann S: Altered brain activity in severely obese women may recover after Roux-en Y gastric bypass surgery. Int J Obes (Lond) 38:3413482014

23

Gearhardt ANCorbin WRBrownell KD: Preliminary validation of the Yale Food Addiction Scale. Appetite 52:4304362009

24

Gearhardt ANYokum SOrr PTStice ECorbin WRBrownell KD: Neural correlates of food addiction. Arch Gen Psychiatry 68:8088162011

25

Giacobbe PMayberg HSLozano AM: Treatment resistant depression as a failure of brain homeostatic mechanisms: implications for deep brain stimulation. Exp Neurol 219:44522009

26

Glannon W: Consent to deep brain stimulation for neurological and psychiatric disorders. J Clin Ethics 21:1041112010

27

Goodman WKFoote KDGreenberg BDRicciuti NBauer RWard H: Deep brain stimulation for intractable obsessive compulsive disorder: pilot study using a blinded, staggered-onset design. Biol Psychiatry 67:5355422010

28

Gracia-Solanas JAElia MAguilella VRamirez JMMartínez JBielsa MA: Metabolic syndrome after bariatric surgery. Results depending on the technique performed. Obes Surg 21:1791852011

29

Grant RAHalpern CHBaltuch GHO’Reardon JPCaplan A: Ethical considerations in deep brain stimulation for psychiatric illness. J Clin Neurosci 21:152014

30

Green EJacobson AHaase LMurphy C: Reduced nucleus accumbens and caudate nucleus activation to a pleasant taste is associated with obesity in older adults. Brain Res 1386:1091172011

31

Guh DPZhang WBansback NAmarsi ZBirmingham CLAnis AH: The incidence of co-morbidities related to obesity and overweight: a systematic review and meta-analysis. BMC Public Health 9:882009

32

Halpern CHTekriwal ASantollo JKeating JGWolf JADaniels D: Amelioration of binge eating by nucleus accumbens shell deep brain stimulation in mice involves D2 receptor modulation. J Neurosci 33:712271292013

33

Haqq AMFarooqi ISO’Rahilly SStadler DDRosenfeld RGPratt KL: Serum ghrelin levels are inversely correlated with body mass index, age, and insulin concentrations in normal children and are markedly increased in Prader-Willi syndrome. J Clin Endocrinol Metab 88:1741782003

34

Hartman MMartin ABBenson JCatlin A: National health spending in 2011: overall growth remains low, but some payers and services show signs of acceleration. Health Aff (Millwood) 32:87992013

35

Hinton ECHolland AJGellatly MSSoni SPatterson MGhatei MA: Neural representations of hunger and satiety in Prader-Willi syndrome. Int J Obes (Lond) 30:3133212006

36

Ho ALSussman ESZhang MPendharkar AVAzagury DEBohon C: Deep brain stimulation for obesity. Cureus 7:e2592015

37

Holsen LMSavage CRMartin LEBruce ASLepping RJKo E: Importance of reward and prefrontal circuitry in hunger and satiety: Prader-Willi syndrome vs simple obesity. Int J Obes (Lond) 36:6386472012

38

Hone-Blanchet AFecteau S: Overlap of food addiction and substance use disorders definitions: analysis of animal and human studies. Neuropharmacology 85:81902014

39

Jenkins BTepper SJ: Neurostimulation for primary headache disorders: Part 2, review of central neurostimulators for primary headache, overall therapeutic efficacy, safety, cost, patient selection, and future research in headache neuromodulation. Headache 51:140814182011

40

Jensen MDRyan DHApovian CMArd JDComuzzie AGDonato KA: 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. J Am Coll Cardiol 63:25 Pt B298530232014

41

Johnson PMKenny PJ: Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats. NatNeurosci 13:6356412010

42

Karlsson JTaft CRydén ASjöström LSullivan M: Ten-year trends in health-related quality of life after surgical and conventional treatment for severe obesity: the SOS intervention study. Int J Obes (Lond) 31:124812612007

43

Kelley AEStinus L: Disappearance of hoarding behavior after 6-hydroxydopamine lesions of the mesolimbic dopamine neurons and its reinstatement with L-dopa. Behav Neurosci 99:5315451985

44

Kenny PJ: Reward mechanisms in obesity: new insights and future directions. Neuron 69:6646792011

45

King WCChen JYMitchell JEKalarchian MASteffen KJEngel SG: Prevalence of alcohol use disorders before and after bariatric surgery. JAMA 307:251625252012

46

Laćan GDe Salles AAGorgulho AAKrahl SEFrighetto LBehnke EJ: Modulation of food intake following deep brain stimulation of the ventromedial hypothalamus in the vervet monkey. Laboratory investigation. J Neurosurg 108:3363422008

47

Lang AEHoueto JLKrack PKubu CLyons KEMoro E: Deep brain stimulation: preoperative issues. Mov Disord 21:Suppl 14S171S1962006

48

Lindgren ACBarkeling BHägg ARitzén EMMarcus CRössner S: Eating behavior in Prader-Willi syndrome, normal weight, and obese control groups. J Pediatr 137:50552000

49

Lucas-Neto LMourato BNeto DOliveira EMartins HCorreia F: The nucleus accumbens beyond the anterior commissure: implications for psychosurgery. Stereotact Funct Neurosurg 92:2912992014

50

Maggard MAShugarman LRSuttorp MMaglione MSug-erman HJLivingston EH: Meta-analysis: surgical treatment of obesity. Ann Intern Med 142:5475592005

51

Mann JPJakes ADHayden JDBarth JH: Systematic review of definitions of failure in revisional bariatric surgery. Obes Surg 25:5715742014

52

Matsuda MLiu YMahankali SPu YMahankali AWang J: Altered hypothalamic function in response to glucose ingestion in obese humans. Diabetes 48:180118061999

53

Meany GConceição EMitchell JE: Binge eating, binge eating disorder and loss of control eating: effects on weight outcomes after bariatric surgery. Eur Eat Disord Rev 22:87912014

54

Mechanick JIYoudim AJones DBGarvey WTHurley DLMcMahon MM: Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient—2013 update: cosponsored by American Association of Clinical Endocrinologists, The Obesity Society, and American Society for Metabolic & Bariatric Surgery. Obesity (Silver Spring) 21:Suppl 1S1S272013

55

Meissner WSchreiter DVolkmann JTrottenberg TSchneider GHSturm V: Deep brain stimulation in late stage Parkinson’s disease: a retrospective cost analysis in Germany. J Neurol 252:2182232005

56

Miller JLJames GAGoldstone APCouch JAHe GDriscoll DJ: Enhanced activation of reward mediating prefrontal regions in response to food stimuli in Prader-Willi syndrome. J Neurol Neurosurg Psychiatry 78:6156192007

57

Mink JWWalkup JFrey KAComo PCath DDelong MR: Patient selection and assessment recommendations for deep brain stimulation in Tourette syndrome. Mov Disord 21:183118382006

58

Müller UJSturm VVoges JHeinze HJGalazky IHel-dmann M: Successful treatment of chronic resistant alcoholism by deep brain stimulation of nucleus accumbens: first experience with three cases. Pharmacopsychiatry 42:2882912009

59

Müller UJVoges JSteiner JGalazky IHeinze HJMöller M: Deep brain stimulation of the nucleus accumbens for the treatment of addiction. Ann N Y Acad Sci 1282:1191282013

60

Murdaugh DLCox JECook EW IIIWeller RE: fMRI reactivity to high-calorie food pictures predicts short- and long-term outcome in a weight-loss program. Neuroimage 59:270927212012

61

Nicolai AIppoliti CPetrelli MD: Laparoscopic adjustable gastric banding: essential role of psychological support. Obes Surg 12:8578632002

62

Passamonti LRowe JBSchwarzbauer CEwbank MPvon dem Hagen ECalder AJ: Personality predicts the brain’s response to viewing appetizing foods: the neural basis of a risk factor for overeating. J Neurosci 29:43512009

63

Pepino MYStein RIEagon JCKlein S: Bariatric surgeryinduced weight loss causes remission of food addiction in extreme obesity. Obesity (Silver Spring) 22:179217982014

64

Pisapia JMHalpern CHMuller UJVinai PWolf JAWhiting DM: Ethical considerations in deep brain stimulation for the treatment of addiction and overeating associated with obesity. AJOB Neurosci 4:35462013

65

Pisapia JMHalpern CHWilliams NNWadden TABaltuch GHStein SC: Deep brain stimulation compared with bariatric surgery for the treatment of morbid obesity: a decision analysis study. Neurosurg Focus 29:2E152010

66

Sallet PCSallet JADixon JBCollis EPisani CELevy A: Eating behavior as a prognostic factor for weight loss after gastric bypass. Obes Surg 17:4454512007

67

Sani SJobe KSmith AKordower JHBakay RA: Deep brain stimulation for treatment of obesity in rats. J Neurosurg 107:8098132007

68

Saules KKWiedemann AIvezaj VHopper JAFoster-Hartsfield JSchwarz D: Bariatric surgery history among substance abuse treatment patients: prevalence and associated features. Surg Obes Relat Dis 6:6156212010

69

Scheimann AOButler MGGourash LCuffari CKlish W: Critical analysis of bariatric procedures in Prader-Willi syndrome. J Pediatr Gastroenterol Nutr 46:80832008

70

Schlaepfer TECohen MXFrick CKosel MBrodesser DAxmacher N: Deep brain stimulation to reward circuitry alleviates anhedonia in refractory major depression. Neuropsychopharmacology 33:3683772008

71

Schoenen JDi Clemente LVandenheede MFumal ADe Pasqua VMouchamps M: Hypothalamic stimulation in chronic cluster headache: a pilot study of efficacy and mode of action. Brain 128:9409472005

72

Scholtz SMiras ADChhina NPrechtl CGSleeth MLDaud NM: Obese patients after gastric bypass surgery have lower brain-hedonic responses to food than after gastric banding. Gut 63:8919022014

73

Shapira NALessig MCHe AGJames GADriscoll DJLiu Y: Satiety dysfunction in Prader-Willi syndrome demonstrated by fMRI. J Neurol Neurosurg Psychiatry 76:2602622005

74

Stenberg ESzabo EAgren GNäslund EBoman LBylund A: Early complications after laparoscopic gastric bypass surgery: results from the Scandinavian Obesity Surgery Registry. Ann Surg 260:104010472014

75

Stephen JHHalpern CHBarrios CJBalmuri UPisapia JMWolf JA: Deep brain stimulation compared with methadone maintenance for the treatment of heroin dependence: a threshold and cost-effectiveness analysis. Addiction 107:6246342012

76

Stevenson DAHeinemann JAngulo MButler MGLoker JRupe N: Gastric rupture and necrosis in Prader-Willi syndrome. J Pediatr Gastroenterol Nutr 45:2722742007

77

Stice EYokum SBohon CMarti NSmolen A: Reward circuitry responsivity to food predicts future increases in body mass: moderating effects of DRD2 and DRD4. Neuroimage 50:161816252010

78

Stoeckel LEWeller RECook EW IIITwieg DBKnowlton RCCox JE: Widespread reward-system activation in obese women in response to pictures of high-calorie foods. Neuroimage 41:6366472008

79

Stroupe KTWeaver FMCao LIppolito DBarton BRBurnett-Zeigler IE: Cost of deep brain stimulation for the treatment of Parkinson’s disease by surgical stimulation sites. Mov Disord 29:166616742014

80

Tataranni PADelParigi A: Functional neuroimaging: a new generation of human brain studies in obesity research. Obes Rev 4:2292382003

81

Theodoro MFTalebizadeh ZButler MG: Body composition and fatness patterns in Prader-Willi syndrome: comparison with simple obesity. Obesity (Silver Spring) 14:168516902006

82

Tomaszewski KJHolloway RG: Deep brain stimulation in the treatment of Parkinson’s disease: a cost-effectiveness analysis. Neurology 57:6636712001

83

Uusitalo S: Autonomy and DBS treatment for addicts. AJOB Neurosci 4:49502013

84

Volkow NDWang GJFowler JSTelang F: Overlapping neuronal circuits in addiction and obesity: evidence of systems pathology. Philos Trans R Soc Lond B Biol Sci 363:319132002008

85

Volkow NDWang GJTelang FFowler JSLogan JChil-dress AR: Cocaine cues and dopamine in dorsal stria-tum: mechanism of craving in cocaine addiction. J Neurosci 26:658365882006

86

Vucenik IStains JP: Obesity and cancer risk: evidence, mechanisms, and recommendations. Ann N Y Acad Sci 1271:37432012

87

Wadden TAFaulconbridge LFJones-Corneille LRSarwer DBFabricatore ANThomas JG: Binge eating disorder and the outcome of bariatric surgery at one year: a prospective, observational study. Obesity (Silver Spring) 19:122012282011

88

Wang BCWong ESAlfonso-Cristancho RHe HFlum DRArterburn DE: Cost-effectiveness of bariatric surgical procedures for the treatment of severe obesity. Eur J Health Econ 15:2532632014

89

Wedin SMadan ACorrell JCrowley NMalcolm RKarl Byrne T: Emotional eating, marital status and history of physical abuse predict 2-year weight loss in weight loss surgery patients. Eat Behav 15:6196242014

90

Whiting DMTomycz NDBailes Jde Jonge LLecoultre VWilent B: Lateral hypothalamic area deep brain stimulation for refractory obesity: a pilot study with preliminary data on safety, body weight, and energy metabolism. J Neurosurg 119:56632013

91

Wiedemann AASaules KKIvezaj V: Emergence of new onset substance use disorders among post-weight loss surgery patients. Clin Obes 3:1942012013

92

Wilent WBOh MYBueteflsch CMBailes JECantella DAngle C: Induction of panic attack by stimulation of the ventromedial hypothalamus. J Neurosurg 112:129512982010

93

Williams NROkun MS: Deep brain stimulation (DBS) at the interface of neurology and psychiatry. J Clin Invest 123:454645562013

94

Withrow DAlter DA: The economic burden of obesity worldwide: a systematic review of the direct costs of obesity. Obes Rev 12:1311412011

95

Ziauddeen HFarooqi ISFletcher PC: Obesity and the brain: how convincing is the addiction model?. Nat Rev Neurosci 13:2792862012

TrendMD

Metrics

Metrics

All Time Past Year Past 30 Days
Abstract Views 0 0 0
Full Text Views 232 232 44
PDF Downloads 242 242 50
EPUB Downloads 0 0 0

PubMed

Google Scholar