Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jul 9.
Published in final edited form as: Obes Surg. 2016 Aug;26(8):1659–1696. doi: 10.1007/s11695-016-2271-4

Indications for Surgery for Obesity and Weight-Related Diseases: Position Statements from the International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO)

Maurizio De Luca 1,, Luigi Angrisani 2, Jacques Himpens 3, Luca Busetto 4, Nicola Scopinaro 5, Rudolf Weiner 6, Alberto Sartori 1, Christine Stier 6, Muffazal Lakdawala 7, Aparna G Bhasker 7, Henry Buchwald 8, John Dixon 9, Sonja Chiappetta 6, Hans-Christian Kolberg 10, Gema Frühbeck 11, David B Sarwer 12, Michel Suter 13, Emanuele Soricelli 14, Mattias Blüher 15, Ramon Vilallonga 16, Arya Sharma 17, Scott Shikora 18
PMCID: PMC6037181  NIHMSID: NIHMS975025  PMID: 27412673

Final Recommendations

Obesity is a chronic disease that has already reached pandemic proportions and is becoming one of the leading causes of death and disability worldwide. A comprehensive, sustainable, and proactive strategy to deal with the challenges posed by the obesity epidemic is urgently needed. Weight loss induced by surgery has proven to be highly efficacious in treating obesity and its comorbidities.

Body mass index (BMI) and anthropometric measures, although useful, have important limitations when applied to individuals as indications for surgery for obesity and weight-related diseases. Any indication for surgery has to consider the metabolic comorbidities, particularly type 2 diabetes mellitus (T2DM), physical symptoms, psychological/psychiatric symptoms, and the limitations of functional status. For these reasons, the words “bariatric,” “obesity,” or “metabolic” surgery should be replaced by the words “surgery for obesity and weight-related diseases” which better explains the fact that these surgeries are able to dramatically improve and even cure obesity and weight-related conditions.

After a careful review of the available data concerning the safety and efficacy of surgery for obesity and weight-related diseases and its effectiveness in treating obesity and related comorbidities, this panel reached a consensus on these recommendations:

  1. Surgery for obesity and weight-related diseases may be an effective therapeutic option for the management of T2DM in patients with obesity demonstrating good results in terms of glycemic control, glycosylated hemoglobin, and diabetes medications. Furthermore, surgical weight loss treatment improves components of the metabolic syndrome (MSy). This applies to patients with class I obesity (BMI >30 kg/m2) as well as patients with obesity of higher classes. Surgery for obesity and weight-related diseases reduces cardiovascular disease risk in terms of atherosclerosis, myocardial infarction, stroke, and death.

  2. Surgical treatment of obesity may result in resolution/improvement of pulmonary diseases such as obstructive sleep apnea syndrome (OSAS) and asthma.

  3. Weight loss induced by surgery may reduce the disabilities derived from joint disease.

  4. Surgical operations for obesity and weight-related diseases, particularly Roux-en-Y gastric bypass (RYGB), may result in improvement and even cure of gastro-esophageal reflux disease (GERD).

  5. Non-alcoholic fatty liver disease (NAFLD), a condition associated with obesity that may progress to end-stage liver disease, may be improved after surgery for obesity.

  6. Mental health is a burden to candidates for surgery for obesity and weight-related diseases. Surgery is not contraindicated for patients with mood and anxiety disorders, binge eating disorder (BED), and night eating syndrome (NES) provided the patients received appropriate mental health treatment. On the contrary, surgery is a contraindication in cases of severe and untreated bipolar disorders, in cases of unstable schizophrenia and psychosis, untreated bulimia nervosa, and intractable substance and alcohol abuse.

  7. Secondary obesity caused by endocrinopathies that are inadequately treated medically represents a contraindication to surgery for obesity and weight-related diseases.

  8. Weight loss, induced or not by surgery, reduces the risks of gastrointestinal, genitourinary, reproductive, and hematopoietic malignancies.

  9. Weight loss, induced or not by surgery, is recommended for patients with idiopathic intracranial hypertension (IIH) or pseudotumor cerebri.

  10. Surgically induced weight loss improves renal function and urinary incontinence in obese patients. Moreover, surgery for obesity could be considered as a bridge to renal transplantation. Finally, surgery is not contraindicated in patients with obesity suffering from chronic renal failure requiring dialysis.

  11. Substantial weight loss following surgery for obesity and weight-related diseases may lead to bone mass loss and subsequently increases the risk of fractures. Strategies to limit lean body mass loss should be emphasized.

  12. Surgery for obesity and weight-related diseases improves quality of life proportionally to the amount of weight lost. Moreover, surgery enables patients to increase their participation in physical exercise training programs, reduces absenteeism from work, sick leave, and pension for disability.

  13. Surgery for obesity and weight-related diseases is effective for subjects under 18 years of age suffering from morbid obesity provided participation in a multidisciplinary evaluation including the patient’s pediatrician, parents’ cooperation and informed consent. Weight loss surgery is also effective in patients with obesity who are over the age of 60 years but has a higher rate of perioperative complications.

  14. Surgery for obesity and weight-related diseases is effective in patients with class I obesity accompanied by comorbidity.

Preamble

Obesity as a chronic disease that has already reached pandemic proportions and is becoming one of the leading causes of death and disability worldwide. Approximately 65 % of the world’s population inhabit countries where overweight and obesity kill more people than starvation. It is important to note that severe obesity is a rapidly growing segment of the obesity epidemic in which the detrimental effects are particularly evident and harsh. Importantly, obesity preferentially affects the socially disadvantaged members of the population, and this group has experienced the most rapid increase in obesity prevalence. As a harbinger of a multitude of disabling and fatal diseases, obesity represents one of the most challenging public health concerns of the twenty-first century.

Obesity is a progressive disease, impacting severely on individuals and society alike, and is widely acknowledged as the cause of many other disease states, including most noncommunicable diseases (NCDs). Obesity plays a central role in the development of a number of risk factors and chronic diseases such as hypertension, dyslipidemia, and T2DM inducing cardiovascular morbidity and mortality. Surgery is presently the most effective treatment and the only long-lasting option for this population. The prevention of this disease should be a top public health priority, with increased commitment for concerted, coordinated, and specific actions. A comprehensive, sustainable, and proactive strategy to deal with the challenges posed by the obesity epidemic is urgently needed. For individuals suffering from obesity and weight-related diseases, surgical and endoscopic options should be considered and offered. Surgery for obesity and weight-related diseases has proven to be highly efficacious in treating obesity and its comorbidities. Currently, surgery for obesity and weight-related diseases resolves over 75 % of morbid obesity and super obesity and is equally active on the weight-related comorbidities and complications. Ongoing research is unlocking the mechanisms of action of these procedures in terms of the metabolic parameters they influence and the hormonal and inflammatory processes they alter. Then, based on sound insights and experimental data, we may be able to modify them or even develop more efficacious or safer procedures in the future.

The indication for surgery should not be based solely on BMI. In general, a shift in the paradigm should occur focusing more on the treatment of the diseases and disability related to, and caused by, obesity rather than simply on body weight.

Surgery for obesity and weight-related diseases must now play an important role in reducing morbidity and mortality of populations worldwide. The International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO) is committed to provide the leadership, guidance, and support to national scientific societies and governments, as part of its mission of facilitating and engaging in actions that promote that surgery is an effective and safe treatment of obesity and weight-related diseaes. IFSO also supports all activities of other federations and societies worldwide to reduce the burden of unhealthy excess weight worldwide through prevention and management. However, a wider effort is still needed. The leadership of IFSO has concluded that the words “bariatric,” “obesity,” and “metabolic” surgery should be replaced by the words “surgery for obesity and weight-related diseases” which better describe the fact that this surgery is able to dramatically improve and even “cure” obesity and weight-related conditions.

Chapter 1: “Surgery for Obesity and Weight-Related Diseases”

The archaeologic epochs of surgery can be viewed as the layered evolution of broad disciplines. The latest epoch of surgical development is metabolic surgery, which was defined in 1978 as, “the operative manipulation of a normal organ or organ system to achieve a biological result for a potential health gain” [1]. This is in reality the definition of functional surgery, which consists of a surgically induced anatomic modification of a normal organ or organ system that produces a functional change aimed at reducing or annulling the altered function causing the disease, or at counteracting the altered function. It can be considered metabolic surgery only if the goal of the functional change is to correct a metabolic alteration, as it is the case of the partial ileal bypass mentioned below. This perspective of the role of surgery in proactive healthcare elevates the discipline beyond the older surgical crafts of incisional, extirpative, and reparative operative procedures. Peptic ulcer surgery was an early example of functional metabolic surgery, since it consisted of operations on normal stomachs and vagus nerves to achieve healing of a distal lesion left untouched by the surgeon. The Program on the Surgical Control of the Hyperlipidemias (POSCH) trial utilized an intestinal operation—partial ileal bypass—to lower plasma LDL-cholesterol and, thereby, retard and even reverse atherosclerotic cardiovascular disease. This NIH-funded trial was the first randomized controlled trial to employ functional/metabolic surgery as the intervention modality [2]. Bariatric or obesity surgery, introduced in 1953, and first mentioned in the literature in 1954 [3] is another example of functional surgery. Thus, surgery for obesity and weight-related disease did not give rise to metabolic surgery but has quite often been correlated to it [4].

Obesity per se has many metabolic implications, and there are few, if any, patients with morbid obesity who are free of concurrent metabolic diseases, which can be partially or totally cured by weight loss. This does not mean that obesity surgery is always metabolic surgery, since the primary indication for surgery is generally the weight reduction, and the metabolic results are to be considered beneficial side effects, whose absence would not influence the indication for surgery. Concentrating on only one aspect of this mosaic limits and indeed renders the treating physician ineffectual. The same is true for the bariatric surgeon who envisions the indications for surgery exclusively by weight standards. The BMI should not be the only reason for treatment. The indication for surgery should mainly be the treatment of weight-related diseases.

The surgeon for obesity and related diseases engages cognitively with the enigmas of obesity, the MSy, and, in particular, with T2DM, and searches for new gastrointestinal and other body organ procedures to mitigate these afflictions independent of weight loss. Furthermore, one must seek the acquisition of knowledge of the neurohormonal networks of these diseases and those created by the surgery. The surgeon expands the concepts of the field to new explorations. Surgeons and other practitioners of surgery for obesity and weight-related diseases should strive for a new freedom of thought and a vast extension of clinical horizons. Interestingly, recent observations demonstrated that individuals with obesity with or without MSy have the same elevated level of mortality risk [5]. This discrepancy highlights the fact that using metabolic risk factors alone as a risk stratification system may not be sufficient to identify individuals with obesity who are at elevated mortality risk, and that a unique risk-stratification system specifically for individuals with obesity is required.

Chapter 2: The Limitations of BMI and Anthropometric Measures When Applied to Individuals as Indications for Surgery

Statement 1

Surgery for obesity and weight-related diseases is a codified discipline that has proven to be effective in the treatment of obesity resulting in long-term weight loss, improvement in or resolution of comorbidities, and the lengthening of life expectancy.

(Level of evidence 1, grade of recommendation A)

In 1991 the Consensus Statement of the NIH Consensus Development Conference codified the first universally accepted guidelines for surgery for obesity and weight-related disease [6]. They assert that a candidate for surgery for obesity and weight-related diseases is an obese patient suffering from obesity with:

  • BMI >40 kg/m2

  • BMI >35 kg/m2 in the presence of specific comorbidities:

  • Hypertension

  • Ischemic heart diseases

  • Type 2 diabetes (T2DM)

  • Obstructive sleep apnea syndrome

  • Obesity syndrome/hypoventilation (Picwickian syndrome)

  • Non-alcoholic fatty liver disease and steatohepatitis

  • Dyslipidemia

  • Gastroesophageal reflux diseases

  • Asthma

  • Venous stasis diseases

  • Severe urinary incontinence

  • Disabling arthropathy

  • Severely reduced quality of life

  • Between 18 and 60 years of age

  • Longstanding obesity (>5 years)

  • Proven failure of nutritional and behavioral therapy

  • Patients must be motivated and able to provide a valid consent, are willing to undergo periodic inspections and follow an established dietary regime

  • Absence of major contraindications (very high operative risk, limited life expectancy due to illness, severe cirrhosis, alcohol abuse/drugs, etc.).

Many studies support the effectiveness of surgery. Some long-term, controlled trials have shown that patients treated surgically obtain significantly better results than patients treated with medical therapy in terms of maintaining the weight loss, the resolution or improvement of comorbidities and, ultimately, reduction of mortality.

For example, the Swedish Obese Subjects (SOS) study showed better results in patients with obesity treated by surgery, compared with patients with similar obesity treated by medical therapy [79]. These findings were described for cardiovascular, respiratory, metabolic diseases, and cancer [1015].

Other studies have revealed that surgery for obesity and weight-related diseases resulted in a significant reduction in mortality from causes related to obesity and comorbidities, particularly T2DM [1622].

While repeatedly revised and expanded over the years, the NIH 1991 guidelines have remained substantially unchanged in the cardinal principles especially regarding BMI and age of the patients.

Statement 2

The limitations BMI and anthropometric measures when applied to individual patients as indications for surgery.

(Level of evidence 2, grade of recommendation B)

In 1832, Adolphe Quetelet devised BMI as a simple mathematical equation to measure obesity. Since then, BMI has been the most widely used index to assess obesity. This is mainly because it is convenient, straightforward, economical, and usually, has a good correlation with body fat (BF) percentage.

The WHO defines obesity as a condition of excessive fat accumulation to the extent that health and well-being are affected. Obesity is characterized by excess BF which is defined conventionally as BF >25 % in males and >35 % in females [23]. Obesity is linked to an increased risk for a host of diseases such as T2DM, hypertension (HTN), dyslipidemia, coronary artery disease, OSAS, COPD, gout, joint pains, certain cancers, and infertility. In addition, BMI data does not take into account the interracial differences in fat distribution. For example, it is widely acknowledged that Asians have a higher BF percentage than their western counterparts. Wang et al. documented the differences in fat distribution between Caucasians and Asians and showed that Asians at a lower BMI have a significantly higher BF percentage than Caucasians [24]. A possible genetic predisposition to this increased adiposity in “thin” Indians was suggested by Yajnik et al. who went on to coin the term “thin fat baby syndrome” [25]. Due to this increased adiposity, Asians are at a higher risk for MSy even at relatively normal levels of BMI (22 to 23 kg/m2) [26]. This susceptibility to MSy and T2DM at lower levels of BMI in the Asian population can also be attributed to the “thrifty” gene phenotype, which historically enhanced fat storage in survival conditions, a former benefit that has turned detrimental in the present times of food surplus [27]. It can thus be concluded that Asians develop T2DM at relatively lower levels of BMI and at younger ages, and that they tend to suffer longer from the complications of diabetes.

In terms of treatment of T2DM, although lifestyle modification and pharmacotherapy are the mainstay of therapy, only 7.3 % of all patients achieve good glycemic control with medical therapies [28]. Over the years, strong evidence has emerged in favor of surgery for obesity and weight-related diseases as a treatment option for diabetics with obesity [14]. In the long-term, cause-specific mortality from T2DM has been reported to decrease after surgery for obesity and weight-related diseases [14]. In 2009, the ADA recognized surgery for obesity and weight-related diseases as an effective treatment option for diabetics with obesity [29]. Likewise, in 2011, the IDF also endorsed the use of surgery for obesity and weight-related diseases for diabetics suffering from obesity and who are uncontrolled with medications [30].

Unfortunately, since the time of its inception, patient selection criteria for surgery for obesity and weight-related diseases have been based mainly on BMI. For a considerable time, worldwide use of BMI-based NIH guidelines has denied the benefits of surgery for obesity and weight-related diseases to a large section of individuals who are metabolically obese. In recent years, various consensus meetings such as ACMOMS and ADSS have recommended that the guidelines for bariatric surgery must be more holistic and must use, in conjunction with BMI, other criteria such as waist circumference, metabolic aspects, funcional limits, psychological disorders, and the other features of obesity in conjunction with BMI [31].

The NIH 1991 [6] guidelines have been repeatedly revised and expanded over the years, while remaining substantially unchanged in the cardinal principles especially regarding BMI and age of the patients. At this stage the indications for surgery should be adjusted according to the present actual knowledge and should include new criteria for indications for surgery for obesity and weight-related diseases. The present statement may be the basis on which to develop evidence-based guidelines as directed by the World of Obesity (WOF), the International Diabetes Federation (IDF) and under guidance and leadership of the IFSO. The basic conditions, that (1) the patients have to be able to provide a valid consent, to be willing to undergo periodic inspections, and to follow an established dietary regime and (2) major contraindications such as very high operative risk, limited life expectancy due to illness, severe cirrhosis, alcohol abuse, drugs, etc.) are absent, remain unchanged.

In the literature, there are numerous studies supporting the effectiveness of surgery for significantly reducing mortality from obesity and related comorbidities. Some of these studies provide controlled, long-term evidence that patients treated surgically obtain significantly better results than patients treated with medical therapy in terms of maintaining the weight loss, resolution or improvement of comorbidities, and ultimately reduction of mortality.

The indications for surgery for obesity and weight-related diseases should be based on the overall assessment of the current health status of the patient with obesity and on the identification of disease risk factors that are not addressed by only the calculation of BMI. These factors should include the following:

  • Distribution of adipose tissue as an important cardiovascular and metabolic risk factor; a high amount of visceral fat is often associated with increased liver, muscle, and pancreatic fat and represents a significant BMI independent risk factor with a causal relationship to the MSy (sub-chapter 4.4) [3235].

  • Different body composition related to gender; at equal BMI, the percentage of adipose tissue is greater in females than in males.

  • Individual fat distribution; at equal BMI, distribution between adipose tissue and non-adipose tissue may vary.

  • Body composition related to age; a positive correlation between age, visceral fat, and abnormal lipid and glucose metabolism has been demonstrated.

  • Body composition linked to race; at equal BMl, the risk of developing T2DM and MSy is greater in individuals originating from Asian countries. In fact, when compared to Western standards, these populations and some other ethnic groups are at high risk and a reduction of the threshold value by 2.5 kg/m2 compared to Western standards is recommended for obesity classification, [2427, 36].

  • Psychiatric and psycological symptoms (sub-chapter 3.7)

  • Limitation of functional aspects (sub-chapter 3.13) [3739].

It can be concluded from the above that determining a more accurate “phenotype” of the obese patient with obesity is recommended. There needs to be a more careful determination of the percentage of adipose tissue, its body distribution and identification of established clinical parameters, so as to allow for an overall evaluation of the patient with obesity as a candidate for surgery for obesity and weight-related diseases.

It is therefore important to identify new criteria for classification of obesity allowing for the selection of patients as candidates for surgery for obesity and weight-related diseases. New classifications, presently and in the future, should quantitatively represent the health condition of each individual.

A recent method based on the clinical and functional classification of obesity has been proposed, namely the Edmonton Obesity Staging System (EOSS). EOSS classifies obesity in five stages, from 0 to 4, considering physical symptoms, psychological/psychiatric symptoms, and limitation of functional aspects [3740]. The EOSS stages are as follows:

  • Stage 0: Patient has no apparent obesity-related risk factors (e.g., blood pressure, serum lipids, fasting glucose, etc. within normal range), no physical symptoms, no psychopathology, no functional limitations or impairment of well-being.

  • Stage 1: Patient has one or more obesity-related sub-clinical risk factors (e.g., elevated blood pressure, impaired fasting glucose, elevated liver enzymes, etc.), mild physical symptoms (e.g., dyspnea on moderate exertion, occasional aches and pains, fatigue, etc.), mild psychopathology, mild functional limitations and/or mild impairment of well-being.

  • Stage 2: Patient has one or more established obesity-related chronic diseases requiring medical treatment (e.g., hypertension, type 2 diabetes, sleep apnea, osteoarthritis, reflux disease, polycystic ovary syndrome, anxiety disorder, etc.), moderate functional limitations and/or moderate impairment of well-being.

  • Stage 3: Patient has clinically significant end-organ damage such as myocardial infarction, heart failure, diabetic complications, incapacitating osteoarthritis, significant psychopathology, significant functional limitations and/or significant impairment of well-being.

  • Stage 4: Patient has severe (potentially end-stage) disabilities from obesity-related chronic diseases, severe disabling psychopathology, severe functional limitations and/or severe impairment of well-being.

In large epidemiological databases, EOSS staging has been shown to be a more accurate predictor of total mortality than BMI levels in large epidemiological databases and its application for the selection/prioritization of patients with obesity for obesity and weight-related diseases has been suggested.

In parallel with proposals for new classifications, reports have been published that suggest a more careful definition of the predictive parameters of overall operative risk is in order [4143]. Better clinical diagnosis of patients with obesity and a more careful definition of the operative risk may be of help to reach a therapeutic decision that can be more logical and thoughtful than the simple value of BMI.

The validation and application of EOSS or other alternative staging systems for the selection/prioritization of patients with obesity to surgery for obesity and weight-related diseases beyond BMI values should be a focus of future clinical research in the field.

Chapter 3: Specific Considerations and Indications for Surgery in the Presence of Weight-Related Diseases

Sub-chapter 3.1 Diabetes and metabolic syndrome

Statement 3.1.1

Surgery for obesity and weight-related diseases is a safe and effective therapeutic option for the management of T2DM in patients with obesity. Along with optimal medical treatment and lifestyle adjustment, it has been demonstrated that surgery for obesity and weight-related diseases can achieve a better glycemic control, lower glycosylated hemoglobin, and reduction of diabetes medications than optimal medical and lifestyle treatment alone.

(Level of evidence 1, grade of recommendation A)

In 2013, Guo et al. performed a systematic review and meta-analysis of randomized controlled trials (RCT) in order to assess the effectiveness and the safety of surgery for obesity and weight-related diseases and optimal medical and lifestyle treatment versus optimal medical and lifestyle treatment alone for the management of T2DM. They were able to demonstrate superior results in the surgical group in terms of the reduction of fasting plasma glucose (FDG), glycosylated hemoglobin (HbA1c) and diabetes medications [44]. Three RCTs had been selected, accounting for 270 patients overall, 170 in the surgical and medical therapy group and 100 in the medical therapy group [10, 45, 46]. The surgical procedures performed in these 3 studies were laparoscopic adjustable gastric banding (LAGB), sleeve gastrectomy (SG), gastric bypass (GBP) and biliopancreatic diversion (BPD). Follow-up ranged from 12 to 24 months. The results of the analysis showed that not only could surgery for obesity and weight-related diseases significantly decrease the levels of HbA1c, fasting plasma glucose (FPG) and other metabolites, they also had significantly higher rates of diabetes remission when compared to medical therapy alone. Meanwhile, there were no statistical differences in the serious adverse events between the surgical and medical groups. These data have been confirmed by more recent RCTs. Ikramuddin et al. showed that, at 12 months, 60 randomized patients assigned to intensive lifestyle-medical management and GBP procedure were more likely to achieve the composite goal of HbA1c less than 7.0 %, low-density lipoprotein cholesterol less than 100 mg/dL, and systolic blood pressure less than 130 mm/Hg when compared to those 60 patients who received intensive lifestyle-medical management alone (49 % versus 19 %) [13]. However, since the incidence of serious adverse events was higher in the surgical group than in the medical group (22 versus 15), the authors claimed that the potential metabolic benefits should be carefully weighted with the risks of the surgical procedure [13]. The durability of the metabolic effect of bariatric procedures has been addressed in two RCTs with a follow-up of 3 years. In the first one Schauer et al. randomized 150 patients with obesity and uncontrolled T2DM to receive either intensive medical therapy alone or intensive medical therapy following GBP or SG [10]. The primary endpoint was to reach a Hb1Ac level of 6 % or less at 3 years. This was achieved in a significantly higher percentage of GBP and SG patients (38 % and 24 % respectively), when compared to the medical therapy group. There were more rapid and more sustained reductions in levels of Hb1Ac and FPG and in the use of medications in the two surgical groups compared with the medical therapy group. At 3 years, the daily intake of medications was lower in the GBP group than in the SG group (0.48 ± 0.80 versus 1.02 ± 1.01) and the proportion of patients who were not taking any glucose-lowering medications was significantly higher in the GBP than in the SG group. In the entire cohort, the BMI reduction was the only criteria to be significantly related to the achievement of the primary end point. In contrast with the two surgical groups, for the medical group both the reduction of the BMI and a history of diabetes less than 8 years have shown predictive values for T2DM remission [10]. These data were confirmed by the results of the RCT by Courcoulas et al., where patients were randomized to either an intensive lifestyle weight loss intervention for 1 year followed by a low-level lifestyle intervention for 2 years or surgical treatments (GBP or LAGB) followed by low-level lifestyle intervention in years 2 and 3 [47]. Partial or complete T2DM remission was achieved by 40 % of GBP, 29 % of LAGB, and none of the intensive lifestyle weight loss intervention participants (P = 0.004). The use of diabetes medications was reduced more (defined as participants going from using insulin or oral medication at baseline to no medication at year 3) in the surgical groups than the lifestyle intervention–alone group, 65 % of GBP, 33 % of LAGB, and none of the intensive lifestyle weight loss intervention, respectively (P < 0.001) [47].

Statement 3.1.2

Surgery for obesity and weight-related diseases has proven to be a cost-effective and, in some instances, a cost-saving approach for the management of patients suffering from obesity and T2DM.

(Level of evidence 2, grade of recommendation C)

T2DM entails substantial lifetime costs, including both direct medical costs related to diabetes and its complications and indirect costs caused by absence from work, reduced productivity at work, disability and premature death. These costs are estimated at 172.000 US$ direct costs and 305.000 US$ indirect costs per patient when diabetes is diagnosed at the age of 50 and 30 years respectively. Over 60 % of the costs are incurred within 10 years of diagnosis [48]. Surgery for obesity and weight-related diseases has demonstrated to be a cost-effective and, in some instances, cost-saving approach for the management of patients suffering from obesity, with or without T2DM [4951]. It has been calculated that, while the costs for surgery for obesity are substantial in the period from 1 month before surgery to 2 months following surgery, cost savings start accruing from the third month after surgery. Consequently, surgical costs are fully recovered 25 months after the operation [52]. Moreover, in order to specifically assess the economic impact of surgery for obesity in diabetic patients, Warren et al. have proposed a model that applied the population cost estimates for medical care of T2DM to a retrospective cohort of patients with morbid obesity [53]. They compared differences in 10-year medical costs between those having surgery for obesity and weight-related diseases and controls, assuming an incidence of T2DM resolution of 40 % in the bariatric cohort. Considering only the direct medical costs of T2DM, the 10-year aggregate cost savings compared with a control group is $2.7 million/1000 patients; the total (direct and indirect) cost savings is $5.4 million/1000 patients [53].

Statement 3.1.3

Diabetic obese patients with obesity undergoing bilipancreatic diversion/duodenal switch (BPD/DS) achieve the greatest rate of T2DM resolution when compared to the other surgical procedures. GBP and SG have a similar short- to midterm effectiveness on the improvement of glycaemic control, while the antidiabetic effects of LAGB are lower.

(Level of evidence 2, grade of recommendation: B)

A systematic review of the literature by Buchwald et al., accounting for 621 studies with 135,246 patients, reported a T2DM remission in 78.1 % of patients undergoing surgery for obesity, while the incidence of resolution and improvement of diabetic disease was 86.6 % [21]. When considering those studies reporting only diabetic patients, 79.3 % of the participants had resolution of their clinical and laboratory (biochemical) manifestations of type 2 diabetes and 98.9 % had resolution or improvement. Moreover the proportion of patients with T2DM remission or improvement was fairly constant at time points less or more than 2 years [21]. Diabetes resolution was greater for patients undergoing BPD/DS (95.1 %), followed by GBP (80.3 %), gastroplasty (79.7 %) and then LAGB (56.7 %). This review was limited by the quality of the papers with different follow-up, inhomogeneus definition of remission and disparate biochemical measures. [21].

Recently we witnessed the dramatic worldwide adoption of laparoscopic SG. Consequently several clinical randomized and non-randomized trials were performed in order to compare the effectiveness of SG on weight loss and comorbities resolution to that of other bariatric procedures, in particular GBP. Published meta-analysis studies have shown that despite the greater weight loss achieved with the GBP compared with the SG, there are no differences between the two procedures in terms of remission of T2DM [5459].

Statement 3.1.4

Surgery for obesity and weight-related diseases demonstrated an excellent short- and midterm risk/benefit ratio in patients with class I obesity (BMI 30–35 kg/m2) suffering from T2DM and/or other comorbidities.

(Level of evidence 1, grade of recommendation A)

The IFSO and the American Society for Metabolic and Bariatric Surgery (ASMBS) recently stated that class I obesity is a well-defined condition deserving treatment because it causes or exacerbates multiple other diseases and decreases both the duration and the quality of life. Current options for the nonsurgical treatment for class I obesity showed little efficacy in the majority of patients in achieving a substantial and durable weight reduction. Both ASMBS and IFSO concluded that surgery for obesity and weight-related diseases should be an available option for patients with class I obesity who do not achieve substantial and durable weight loss and comorbidity improvement with nonsurgical options [60, 61]. These statements are supported by a robust body of literature, including 5 RCTs with high level of evidence [10, 13, 45, 62, 63] and two large meta-analysis/systematic reviews analyzing the outcomes of several prospective and retrospective studies conducted in patients with class I obesity, and T2DM and other comorbidities [64, 65]. The comprehensive evaluation of these studies demonstrated that subjects with class I obesity undergoing surgery for obesity and weight-related diseases achieve an excellent weight loss and positive effects on glycemic control and diabetes remission. These benefits occur after all of the most established surgical procedures and without substantial differences in respect to the outcomes observed in patients with BMI > 35 kg/m2.

Statement 3.1.5

Surgery for obesity and weight-related diseases is not effective in patients with Latent Autoimmune Diabetes in Adults (LADA). The diagnosis of LADA patients should be carefully evaluated before considering surgery.

(Level of evidence 3, grade of recommendation C)

The failure to achieve diabetes remission after surgery for obesity and weight-related diseases can occur in patients with longstanding, poorly controlled diabetes or insufficient weight loss, i.e. inadequate loss or regain of weight [6669]. However, up to 10 % of patients with adult-onset noninsulin-requiring-dependent diabetes are thought to suffer from a form of autoimmune diabetes called latent autoimmune diabetes of adult-onset (LADA). LADA can initially mimic a T2DM but results in a slow destruction of pancreatic beta cells [70]. LADA is characterized by the presence of specific auto-antibodies in the peripheral blood, (in particular anti-glutamic acid decarboxylase autoantibodies ((GADA)), low plasma insulin and very low fasting and meal-stimulated C-peptide. LADA usually affects normal weight or overweight patients, aged from 30 to 70, with a diabetes history < 5 years and who start exogenous insulin less than 6 months after the diagnosis [70]. After surgery, patients with LADA may initially respond to oral antidiabetic medication while there are still 20 % of the beta cells functioning, but these patients will progress to insulin-dependent diabetes despite achieving substantial weight loss. The literature concerning the true impact of LADA on diabetic outcomes after surgery is sparse [71, 72]. This is a consequence of the fact that in most of cases, subjects with adult-onset noninsulin-dependent diabetes are assumed to be type 2 diabetics. Conversely, in adults, it is important to differentiate between type 1 and type 2 diabetes when bariatric surgery is being considered, particularly for class I obesity patients [73]. This consideration is supported by the study of Lee et al., who reported that after SG, low fasting C-peptide has been associated with failure to achieve resolution/remission of diabetes [74].

Statement 3.1.6

Type 1 Diabetes Mellitus (T1DM) in combination with morbid obesity is an indication for surgery for obesity and weight-related diseases. Even if there will be no recovery of ß-cell function itself, patients with T1DM will still benefit from the positive effects on all of the other weight-related diseases, as well as reduction in the daily insulin requirements as a result of the decrease in insulin resistance, that is seen with weight loss.

(Level of evidence 4, grade of recommendation C)

To date, fewer than 10 cases of surgery for obesity in patients with T1DM have been reported in the literature. These studies have shown that surgery resulted in a significant weight reduction and an improvement in glycemic control [7577]. A recent experimental study in a rat model of spontaneous development of T1DM has also shown that a particular type of gastrointestinal bypass (duodenal-jejunal bypass) lowers blood glucose concentration within 2 days after surgery [78]. The aim of the study was to assess the metabolic outcomes, including the glycemic status of patients with T1DM after weight loss surgery. The findings of this study indicate that surgery leads to a remarkable and sustained weight loss in patients with severe obesity and with T1DM. Furthermore, surgery results in significant improvements in their glycemic status and comorbid conditions, despite having prolonged diabetes and undetectable C-peptide. The favorable metabolic effects of surgery for obesity and weight-related diseases may facilitate medical management of T1DM in the setting of morbid obesity.

Statement 3.1.7

Many studies comparing surgery for obesity and weight-related diseases with nonsurgical weight loss therapies have demonstrated greater improvement in the components of the Metabolic Syndrome in the surgically treated patients.

(Level of evidence 2, grade of recommendation A)

The MSy, initially named by Gerald Reaven as syndrome X [79], describes a cluster of conditions including insulin resistance, hypertension, impaired glucose tolerance or T2DM, dyslipidemia characterized by high plasma triglycerides, low high density lipoprotein cholesterol levels, and central weight distribution. Many definitions of MSy have subsequently been developed to define the syndrome across different age groups and ethnicities [80]. Over the past decade the term MSy has lost favor due to a lack of evidence that the cluster of factors represent an additional risk when compared to the individual components [81, 82]. Many randomized trials comparing the effects of weight-related metabolic surgery versus nonsurgical weight loss therapies have demonstrated greater improvement in the components of the MSy in the surgically treated patients (except in the specific domain of elevated blood pressure) [62, 83, 84].

The MSy is also associated with many other features of obesity-related diseases and insulin resistance including nonalcoholic fatty liver disease, obstructive sleep apnea, and the polycystic ovary syndrome. Various forms of an expanded organ- or disease-specific syndrome have been proposed. However, it is now recommended that predictors of individual diseases such as diabetes, sleep apnea, and cardiovascular disease should be assessed individually rather than trying to combine disease predictors into one common entity or syndrome [85].

Sub-chapter 3.2 Cardiovascular diseases

Statement 3.2.1

Obesity, and visceral obesity in particular, is a major modifiable risk factor for cardiovascular diseases (CVD). Weight loss induced by surgery has been shown to reduce CVD risk, with the most relevant reductions in risk observed in the group of patients having the higher CVD risk before surgery. These patients obtain the most significant metabolic improvements thereafter.

(Level of evidence 1, grade of recommendation A)

Obesity is considered a major modifiable risk factor for cardiovascular diseases (CVD). The large prospective studies collaboration epidemiologic survey, including more than 900,0000 adults enrolled in more than 50 prospective longitudinal studies, confirms a strong association between BMI levels and mortality rates, with a particular reference to cardiovascular deaths [86]. However, cardiovascular risk is not homogeneous within the obese population, and it can vary substantially even in patients with the same BMI levels. CVD in obese patients with obesity varies according to age, gender, family history, genetic background, metabolic status, and behavioral factors (e.g. smoking), but the most relevant factor in determining CVD risk in patients with obesity is probably fat distribution. The importance of fat distribution as a CVD risk determinant has been well-documented in the INTERHEART database, a case-control standardized study enrolling 27,0098 participants from 52 different countries [87]. In INTERHEART, the association between BMI levels and the risk of myocardial infarction tends to attenuate after adjustment for the other known classic CVD risk factors, whereas the association between the risk of myocardial infarction and the waist circumference value, an anthropometric marker of visceral fat deposition, remains highly significant even after adjustment [87].

Weight loss induced by surgery has been shown to reduce CVD risk, as estimated by the use of risk calculators or algorithms that combined the effects of single CVD risk factors in an estimation of the 10-year probability of suffering a fatal or nonfatal cardiovascular event (Framingham Risk Score or Prospective Cardiovascular Munster Heart Study PROCAM score). Some reviews and systematic analyses reported in long-term studies improvements in CVD risk after surgery for obesity and weight-related diseases. However, these studies included different types of surgical procedures and some of them did not have a nonsurgical control group [3, 4]. Whereas CVD risk tended to increase in the control groups, a marked and significant reduction of CVD risk estimation has been found in all the surgical groups [88, 89]. The most relevant reduction in CVD risk has been observed in the group of patients with the higher CVD risk who demonstrated the most significant metabolic improvements postoperatively [89, 90].

Statement 3.2.2

Weight loss induced by surgery for obesity and weight-related diseases is associated to a reduction in the incidence of major cardiovascular events in patients with obesity, including myocardial infarction and stroke. Event reductions are more relevant in patients with a high cardiovascular risk before surgery.

(Level of evidence 1, grade of recommendation A)

The SOS study was the first controlled intervention trial to demonstrate that weight loss in subjects with obesity was associated with a reduction of major cardiovascular events [9]. In the SOS study, surgery for obesity and weight-related diseases was associated with a reduced number of cardiovascular deaths (28 events among 2010 patients in the surgery group versus 49 events among 2037 patients in the control group; adjusted hazard ratio [HR], 0.47; 95 % CI, 0.29–0.76; P = 0.002). The number of total first time (fatal or nonfatal) cardiovascular events (myocardial infarction or stroke, whichever came first) was lower in the surgery group (199 events among 2010 patients) than in the control group (234 events among 2037 patients; adjusted HR, 0.67; 95 % CI, 0.54–0.83; P < 0.001) [9]. One of the more interesting aspects of the SOS study was that both in the surgical group and in the control group the rate of cardiovascular events were not related to the baseline BMI but to the fasting insulin baseline levels. Most of the difference in the events rate between surgical and control group was observed in patients with the highest insulin levels at baseline [9].

More recently, Kwok et al. performed a systematic review and meta-analysis of the studies analyzing the impact of surgery for obesity and weight-related diseases on cardiovascular disease and mortality [91]. Fourteen studies were included, enrolling a total of 29,208 patients undergoing surgery for obesity and weight-related diseases and 166,200 nonsurgical controls. Compared to nonsurgical controls, there was more than a 50 % reduction in mortality amongst patients who had surgery (OR 0.48 95 % CI 0.35–0.64). In a pooled analysis of four studies with adjusted data, surgery was associated with a significantly reduced risk of composite cardiovascular adverse events (OR 0.54 95 % CI 0.41–0.70). Surgery for obesity and weight-related diseases was also associated with a significant reduction in specific endpoints of myocardial infarction (OR 0.46 95 % CI 0.30–0.69, 4 studies) and stroke (OR 0.49 95 % CI 0.32–0.75, 4 studies) [91].

Finally, from a large administrative health database, Johnson et al. retrospectively analyzed the rates of macro- and micro-vascular events observed in patients with T2DM having had surgery for obesity and weight-related diseases (2,580 cases) or not (13,371 cases) [92]. The authors concluded that surgery for obesity and weight-related diseases was associated with a 65 % reduction of major macro- and micro-vascular events in patients with obesity and T2DM [92].

Statement 3.2.3

Weight loss induced by surgery for obesity and weight-related diseases is associated with a regression or improvement of early structural markers of atherosclerosis (carotid intima-media thickness, brachial flow-mediated dilation, coronary artery calcium score).

(Level of evidence 3, grade of recommendation B)

Thus far, only a few, mostly short-term, uncontrolled studies have demonstrated a regression of early structural markers of atherosclerosis after surgery for obesity and weight-related diseases. Both a reduction of carotid intima-media thickness and an improvement of brachial flow-mediated dilation have been reported 18 months after surgery in 37 patients with obesity treated with GBP or LGB [93]. These findings were confirmed in a long-term observation performed 5 years later in the same patients [94]. Similar results have been reported by Habib et al. in 50 patients 12 months after GBP [95] and by Saleh et al. in 47 patients 10 months after GBP [96]. Sarmiento et al. confirmed a reduction of carotid intima-media thickness in 18 patients 12 months after GBP [97]. Finally, Nerla et al. reported an improvement of brachial flow-mediated dilation 3 months after surgery for obesity in 50 patients [98].

Elevated coronary artery calcium (CAC) is another marker of coronary atherosclerosis that is strongly predictive of cardiovascular events. Priester et al. evaluated CAC with electrocardiographic-gated, non-contrast, CT axial scanning of the chest in 65 GBP patients and 84 nonsurgical patients. At 6 years after surgery, CAC score was significantly lower in patients who underwent GBP than those patients who did not undergo surgery (P < 0.01. Additionaly GBP subjects had a lower likelihood of having measurable coronary calcium (odds ratio of CAC > 0 = 0.39; 95%CI of (0.17, 0.90) [99].

Statement 3.2.4

Weight loss induced by surgery for obesity and weight-related diseases is associated with improvement of functional status and symptoms in patients with morbid obesity and with pre-existing ischemic heart disease or heart failure, but the effects on long-term prognosis are not known.

(Level of evidence 3, grade of recommendation B)

Surgery for obesity and weight-related diseases can be performed safely in patients with morbid obesity and with preexisting ischemic heart disease (IHD) [100, 101]. However, at present, the long-term effect of surgery for obesity and weight-related diseases on the fate of established IHD is difficult to evaluate at present, given the paucity of specific data collected in these particular patients. Only a very few subjects with preexisting IHD were included in the SOS study (21 in the surgical and 14 in the control group). Weight loss in the surgical group was satisfactory and clinical conditions, defined as the presence of chest pain or shortness of breath, were improved. However, the number of new cardiovascular adverse events, including myocardial infarction, coronary revascularization, and cardiovascular deaths, was not different in the two groups [102].

Similarly, in a small retrospective controlled study performed in patients with obesity and severe heart failure, surgical weight loss was associated with a significant improvement in functional status, but with mixed results for the ejection fraction, with some patients gaining function and other patients not experiencing any change [103]. Hospital readmission at 1 year was, however, significantly lower in the surgical patients than in the controls [103]. Along the same lines, Miranda et al. reported improvements in the quality of life, frequency of exertional dyspnea, and leg edema in 13 patients with heart failure treated with surgery when compared with six nonsurgical controls [104]. Despite these positive results, convincing data demonstrating that weight loss induced by surgery may change the fate of patients with severe heart failure are still missing. In addition, the fact that in epidemiologic studies weight loss in patients with heart failure has been found to be associated with higher mortality rates should be taken into account [105].

Statement 3.2.5

Preliminary results suggest that surgery for obesity and weight-related diseases maybe useful as a bridge to successful heart transplantation in patients with severe obesity and end-stage heart failure.

(Level of evidence 3, grade of recommendation C)

Some small series and case reports have suggested that surgery for obesity and weight-related diseases may be useful in patients with morbid obesity and end-stage heart failure who are not suitable for heart transplantation because of their severe obesity. Reduced body weight permits successful organ transplantation [106108]. Despite promising results, the experience on this use of surgery for obesity and weight-related diseases as a bridge to successful heart transplantation in patients with severe obesity and end-stage heart failure remains very limited.

Sub-chapter 3.3 Pulmonary diseases

Statement 3.3.1

Surgery for obesity and weight-related diseases may result in resolution/improvement of obstructive sleep apnea syndrome (OSAS).

(Level of evidence 1, grade of recommendation A)

Obesity is a major risk factor for development of OSAS, reported to be up 1.14 times the relative risk for each per unit increase in BMI [109111]. In turn, OSAS is associated with a higher incidence of MSy and an increased cardiovascular risk [112]. This correlation is supported by the discovery, in several published studies, of the improvement in metabolic homeostasis and blood pressure in patients undergoing treatment of OSAS with positive pressure ventilation [113, 114]. A meta-analysis was conducted by Greenburg et al. Twelve case series (342 patients) were included: five prospective and seven retrospective studies [19]. There was no evidence of publication bias. The pooled mean BMI (10 studies) decreased from 55.3 at baseline to 37 after surgery (LAGB and laparoscopic GBP). The mean apnea-hypopnea index (AHI) decreased from 54.7 events per hour (95 % CI 49.0 to 60.3) to 15.8 events per hour (95 % CI 12.6 to 19.0). The authors concluded that surgery for obesity and weight-related diseases significantly improves obstructive sleep apnea, as measured by the AHI [19].

In contrast, Dixon et al. conducted a 2-year randomized controlled trial involving 60 patients with severe obesity comparing LAGB (30 pts) with conventional weight loss therapy (30 pts) for the management of moderate to severe OSAS [115]. Patients lost a mean of 5.1 kg in the conventional weight loss program compared with 27.8 kg of the surgical group (P < 0.001). The AHI decreased by 14.0 events/h (95 % CI, 3.3 to 24.6 events/h) in the conventional weight loss group and by 25.5 events/h (95 % CI, 14.2 to 36.7 events/h) in the surgery for obesity and weight-related diseases group. The between-group difference was −11.5 events/h (95 % CI, −28.3 to 5.3 events/h; P = 0.18). Continuous positive airway pressure (CPAP) adherence did not differ between the groups. The authors concluded that in a group of obese patients with severe obesity and OSAS, the use of surgery (LAGB) compared with conventional weight loss therapy did not result in a statistically greater reduction in AHI despite major differences in weight loss [115].

Statement 3.3.2

The respiratory function of the candidate for surgery for obesity and weight-related diseases should be carefully examined by clinical and instrumental investigations such as chest X-ray, pulmonary function tests, arterial blood gas, and by specific questionnaires. If the diagnosis of sleep apnea syndrome is suspected, a nocturnal oximetry or polysomnographic examination is suggested to assess whether a respiratory therapy device such as CPAP, should be used perioperatively.

(Level of evidence 3, grade of recommendation C)

OSAS patients show higher rates of postoperative complications and, in particular, after abdominal surgery [116, 117]. Preoperative respiratory therapy with CPAP has been shown to reduce the frequency of episodes of respiratory failure after extubation and reduce the rate of postoperative complications [118]. However, several studies have demonstrated that there is a close correlation between the extent of the symptoms detected by the specific questionnaires and the severity of the polysomnography framework [119122]. Consequently, a significant number of patients undergoing surgical procedures are likely to face surgery with an undiagnosed or inadequately treated OSAS. Many different calculation models have been proposed without success. They were based on the use of specific anthropometric and/or clinical parameters. However, it is possible to estimate the severity of OSAS and therefore the need to subject the patient to a polysomnographic examination [123, 124].

Statement 3.3.3

In asthmatic patients, weight loss after surgery for obesity and weight-related diseases results in significant improvement of asthma management, defined as symptoms, level of lung function, and use of medication.

(Level of evidence 3, grade of recommendation C)

The risk of asthma in patients suffering from obesity is two to three times higher than in normal-weight individuals [125]. In addition, obesity significantly impairs asthma management, in terms of symptoms, lung function, and use of medications. A systematic review by Juel et al. demonstrated that weight loss in individuals with obesity and asthma is associated with a 48– 100 % remission of asthma symptoms and use of asthma medication [126].

Some retrospective and prospective studies have reported that after surgery for obesity and weight-related diseases, there is a high symptomatic remission rate and significant improvements in asthma management, l, as assessed by symptoms, use of medication, lung function, and hospitalizations [127132].

Whether this improvement is the result of better mechanical properties of the airways or decreased systemic and bronchial inflammation remains unclear. Van Huisstede et al. reported that surgery for obesity and weight-related diseases in asthmatic patients with obesity improves small airway function and decreases systemic inflammation and the number of mast cells in the airways [133].

A recent study by Dandona et al. demonstrated that following GBP and weight loss, there is a reduction in the expression of key genes involved in asthma pathogenesis including IL-4, MMP-9, LIGHT, LTBR, and ADAM-33 [134]. Because of these changes in gene expression, GBP may provide a potential therapeutic approach to asthma in the morbidly obese.

Sub-chapter 3.4 Osteoarthritis

Statement 3.4.1

In patients undergoing surgery for obesity and weight-related diseases, weight loss results in a substantial improvement in pain and a reduction of disability derived from joint disease.

(Level of evidence 1, grade of recommendation A)

Excess weight is a major “adjustable” risk factor for the development of osteoarthritis. Different epidemiological studies have shown that the risk of developing knee osteoarthritis is 13 to 20 times more likely for patients with obesity when compared to normal-weight individuals. Conversely, the risk of coxoarthritis is 1.7 times higher in individuals with a BMI >28 kg/m2 compared to subjects with BMI <24.9 kg/m2 [135138]. A meta-analysis of randomized clinical trials demonstrated an overall reduction in osteoarthritis after a weekly weight loss of at least 5 % of body weight. Weight loss produced a significant reduction in pain and disability from knee and hip osteoarthritis [139].

Statement 3.4.2

Surgery for obesity and weight-related diseases reduces the incidence of peri- and postoperative complications in patients undergoing knee and hip replacement and reduces the surgical risk for revision of the prosthesis.

(Level of Evidence 3, grade of recommendation C)

Excess weight is an important negative prognostic factor for outcomes of prosthetic joint replacement operations. There is an increased incidence of infections, dislocations, and revision surgery in patients with obesity compared with those of normal weight [140, 141]. However, to date, some published studies have shown that the primary surgical treatment of obesity does not improve the outcome of a subsequent prosthetic replacement procedure [142, 143]. On the contrary, Inacio et al. compared the results of knee or hip arthroplasties in patients who previously underwent surgery for obesity and weight-related diseases with those subjects still awaiting surgery for obesity. They reported a lower postoperative hospitalization rate and a lower incidence of revisional surgery in patients who had undergone surgical treatment of obesity and weight-related diseases compared to those who did not yet have surgery [144]. However, for patients with obesity, who are candidates for prosthetic replacement surgery, preoperative weight loss is still desirable in order to improve the overall health status and the management of diseases associated with obesity.

Sub-chapter 3.5 Gastroesophageal reflux disease (GERD)

Statement 3.5.1

Surgery for obesity and weight-related diseases is effective in controlling GERD and is therefore indicated in patients with morbid obesity who have signs and/or clinical symptoms of severe reflux disease.

(Level of evidence 2, grade of recommendation B)

In the overweight or obese population, the risk for GERD and its complications (erosive esophagitis, Barrett’s esophagus, esophageal adenocarcinoma) is 1.5–3 times higher than in normal-weight individuals with an increased risk directly proportional to the increase in BMI [145150]. Patients with obesity, in particular patients with the so-called central obesity, show signs and clinical symptoms of pathological reflux caused by the increase of abdominal pressure, esophageal and/or gastric motility disorders, and the increased incidence of hiatal hernias [151, 152]. It has been shown that weight loss contributes significantly to an improvement and even resolution of GERD and also increases the sensitivity of the patient to medical therapy with proton pump inhibitors (PPI) [153, 154]. In patients with obesity, surgery for obesity and weight-related diseases, particularly GBP, has shown efficacy similar to that of traditional laparoscopic antireflux surgery (Nissen fundoplication or Toupet fundoplication) in the control of GERD [155157]. This effect is due not only to the reduction in the proportion of visceral fat following the procedure but also to the abolition of some pathophysiological mechanisms that underly the pathological reflux [157, 158].

Statement 3.5.2

In patients undergoing surgery for morbid obesity, GBP is the procedure of choice for patients with obesity and severe GERD.

(Level of evidence 2, grade of recommendation B)

In a recent review of a prospective US database which analyzed the effectiveness of three surgical procedures (GBP, LAGB, and SG) on GERD in a sample of 22,870 patients, an improvement of reflux symptoms was recorded in a significantly higher percentage of patients undergoing GBP than those undergoing LAGB or SG [159]. GBP exerts an intrinsic “antireflux” action, independent from weight loss, that is mediated by the reduction of acid secretion due to the creation of the small gastric pouch and the diversion of bile flow due to the creation of a Roux limb [157, 158]. This results in a rapid and sustained postoperative control of the symptoms associated with reflux disease [160162] and in a significant improvement of the complications related to it (eg., Barrett’s esophagus) [163, 164]. At present there is only one randomized clinical trial demonstrating the efficacy of GBP as being significantly greater than SG in the resolution of GERD [165].

Statement 3.5.3

GBP is the best option for patients with morbid obesity presenting recurrence of symptoms of GERD after traditional antireflux surgery.

(Level of evidence 2, grade of recommendation C)

In patients with morbid obesity, who suffer from recurrent symptoms of GERD after antireflux surgery, the GBP is a revisional procedure that can result in a rapid and complete resolution of symptoms of reflux in the great majority of patients. However, because of its high technical complexity and increased incidence of peri- and postoperative complications, it is advisable for this procedure to be performed by surgeons with proven experience in surgery for obesity and weight-related diseases [166, 167].

Statement 3.5.4

GERD symptoms are not a contraindication to LAGB. However, the presence of esophageal motility disorders at the time of surgery may reduce the efficacy in terms of reflux symptoms.

(Level of evidence 2, grade of recommendation B)

In a systematic review of 20 short follow-up studies with a total sample of 3307 patients operated by LAGB [168], improvement in GERD symptoms and pH-metric parameters as well as reduction in the prevalence of esophagitis were recorded. However, in the long-term, worsening of reflux symptoms or their de novo appearance were documented. This worsening appears to be related to the progressive inflation of the band and to the development of gastric pouch dilation. An increased incidence of esophageal motility disorders was also highlighted that in some cases has been associated with an increased acid exposure [168, 169].

Statement 3.5.5

GERD symptoms are not a contraindication to sleeve gastrectomy.

(Level of evidence 3, grade of recommendation C)

GERD is the most frequent surgical complication after SG surgery with a mean incidence from 7 to 9 % [170]. However, the influence of SG on symptoms of GERD is still a topic of discussion. Existing studies are only short-term, mostly based on symptoms without objective criteria, and have reported very conflictual results. [171, 172]. The discrepancy can be attributed to the pathophysiological mechanisms related to certain anatomical changes after SG surgery. Some of them seem to promote reflux, while others could exert an antireflux action. [157, 158, 173, 174].

Sub-chapter 3.6 Hepatopbiliary disease

Statement 3.6.1

Weight loss after surgery for obesity and weight-related diseases provides improvement or resolution of nonalcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH).

(Level of evidence 2, grade of recommendation B)

NAFLD is increasingly recognized as a condition associated with overweight or obesity that may progress to end-stage liver disease. NAFLD histology resembles alcohol-induced liver injury, but occurs in patients without a history of alcohol abuse. NAFLD has a broad spectrum of clinical and histological signs, ranging from simple fatty liver to hepatic steatosis with inflammation, advanced fibrosis, and cirrhosis. The inflammatory stage is known as non-alcoholic steatohepatitis (NASH) [175].

Recent reports indicate that weight loss induced by surgery for obesity and weight-related diseases could be beneficial for NASH treatment. Chavez-Tapia et al. searched The Cochrane Hepato-Biliary Group Controlled Trials Register, the Cochrane Central Register of Controlled Trials (CENTRAL) and The Cochrane Library, MEDLINE, EMBASE, and Science Citation Index Expanded for randomized clinical trials evaluating any surgical procedure versus no intervention, placebo (sham procedure), or other interventions in patients with NASH regardless of publication status, number of patients randomized, language, or blinding. It appeared that only four studies described hepatic deterioration in the degree of fibrosis. The lack of randomized clinical trials and quasi-randomized clinical studies precludes the correct assesment of the benefits of surgery for obesity and weight-related diseases as a therapeutic approach for patients with NASH. Along the same lines, the limitations of the other studies due to inferior design, do not allow any unbiased conclusions to be drawn concerning surgery for obesity and weight-related diseases as the treatment of NASH [176].

A systematic review of published studies reporting preoperative and postoperative plasma liver enzymes or liver histology was done in patients with obesity and NAFLD undergoing surgery for obesity and weight-related diseases. The data were meta-analyzed using random-effects modeling. The authors revealed that surgery is associated with a significant reduction in the weighted incidence of a number of histological features of NAFLD including steatosis (50.2 and 95 %CI of 35.5–65.0), fibrosis (11.9 and 95 %CI of 7.4–16.3 %), hepatocyte ballooning (67.7 and 95 %CI 56.9–78.5) and lobular inflammation (50.7 and 95 %CI 26.6–74.8 %). Surgery is also associated with a reduction in plasma liver enzyme levels, with statistically significant reductions in ALT (11.36 u/l, 95 %CI 8.36–14.39), AST (3.91 u/l, 95 %CI 2.23–5.59), ALP (10.55 u/l, 95 %CI 4.40–16.70) and gamma-GT (18.39 u/l, 95 %CI 12.62–24.16). However, the heterogeneity of the results was high [177].

In the Practice Guidelines of the American Gastroenterological Association, American Association for the Study of Liver Diseases, and American College of Gastroenterology, Chalasani N et al., recommend that (a) foregut bariatric surgery is not contraindicated in otherwise eligible individuals with obesity who suffer from NAFLD or NASH (but without established cirrhosis); (b) the type, safety, and efficacy of foregut bariatric surgery in otherwise eligible obese individuals with obesity and established cirrhosis due to NAFLD are not established; and (c) it is premature to consider foregut bariatric surgery as an established option to specifically treat NASH [178].

In any case in the majority of studies, weight loss after surgery appeared to provide improvement or resolution of obesity and MSy associated abnormal liver histological features in subjects with severe obesity [179].

Statement 3.6.2

Cholecystectomy performed during surgery for obesity and weight-related diseases is suggested only for patients with biliary symptoms or patients with evidence of gallstones as documented during preoperative ultrasonography.

(Level of evidence 2, grade of recommendation B)

Obesity is a risk factor for cholelithiasis. It increases biliary secretion of cholesterol and decreases gallbladder contractility [180, 181]. Other risk factors for cholelithiasis after surgery for obesity and weight-related diseases include rapid weight loss and the malabsorption that occurs postoperatively. Gallstone formation appears to occur most commonly in the first 6 months following GBP, as documented in a study by Nagem [180]. The debate regarding routine versus selective cholecystectomy has been extensively studied in patients undergoing GBP, but there is limited data for patients undergoing LAGB, SG, and BPD [182]. Some authors suggest routinary cholecystectomy; others suggest cholecystectomy only for patients with biliary symptoms or those with documented evidence of gallstones during preoperative ultrasonography [183]. The argument against removing the gallbladder at time of surgery for obesity and weight-related diseases was based on the assumption that the risk of developing symptomatic cholelithiasis is not significantly higher after GBP than in the general population, particularly with the routine use of prophylactic ursodeoxycolic acid after surgery [184]. Additionally, laparoscopic cholecystectomy is technically more challenging to perform in patients with morbid obesity, which may result in higher complication and open conversion rates [185]. Conversely, if biliary symptoms occur, laparoscopic cholecystectomy would be easier and safer to perform after the initial weight loss. It should however be mentioned that conventional ERCP may not be possible after RYGBP which implies that prophylactic cholecystectomy might be considered more favorably for GBP patients versus those patients having other procedures.

Statement 3.7 Mental health

Statement 3.7.1

Obesity is associated with a significant psychosocial burden. Some candidates for surgery for obesity and weight-related diseases present with significant psychopathology which may impact the outcome of surgery, and, in some cases, represent a contraindication to surgery.

(Level of evidence 2, grade of recommendation C)

Several studies have identified the presence of psychopathology according to the Diagnostic and Statistical Manual of Mental Disorders V (DSM-V) in candidates for surgery for obesity and weight-related diseases [186189]. Current American and European guidelines, as well as some reviews [190192] have emphasized that the presence of specific psychiatric disorders are considered risk factors for suboptimal outcomes after surgical treatment. These disorders include schizophrenia and schizoaffective disorders, psychosis, bipolar disorder, substance abuse disorders, eating disorders (bulimia, BED, and NES), neurocognitive disorders and personality disorders. Treatment with certain psychotropic medications, including antipsychotics and mood stabilizers, previous psychiatric hospitalizations, as well as history of suicide attempts or other self-injurious behaviors also are widely considered risk factors for poor postoperative outcomes. Some of these conditions, in particular severe, uncontrolled psychosis, bipolar disorder, and substance abuse are widely considered contraindications to surgery [193, 194].

Statement 3.7.2

Mood and anxiety disorders are considered negative predictors for the outcome of surgery for obesity and weight-related diseases, but not a contraindication for treatment, provided the patient is receiving appropriate mental health treatment.

(Level of evidence 2, grade of recommendation A)

Studies have suggested that individuals diagnosed with mood and anxiety disorders prior to surgery for obesity and weight-related diseases experience smaller weight losses postoperatively [195198]. The causes for this are not well understood, but could be both behavioral or biochemical in nature. Some studies have suggested that eating foods with high fat content promotes negative emotional states that activate brain circuits of the rewarding systems [199203]. Weight reduction activates specific receptors, inducing less sensitivity to stress and results in improvement of the MSy [204]. However, mood and anxiety disorders are not considered contraindications for bariatric surgery unless the conditions are severe and untreated. Mood and anxiety symptoms should be monitored carefully both before and after surgery.

Statement 3.7.3

Severe and untreated bipolar disorder is generally considered a contraindication to surgery for obesity and weight-related diseases both for its symptoms and for the difficulty in stabilizing drug treatment in the postoperative period. Similarly, severe and unstable schizophrenia and psychosis also are contraindications to surgery for obesity and weight-related diseases.

(Level of evidence 2, grade of recommendation C)

Disturbances of the “bipolar spectrum” are characterized by mood swings with alternating phases of hyperthymia and inhibition. This functional dysregulation results in the development of alterations in the thymus (psychopathology mood), the thought processes (alteration of the form and content of thought), motility and behavioral initiative, as well as in neurodegenerative manifestations (abnormal levels of energy, appetite, libido, the sleep-wake cycle, etc.). Bipolar disorders typically require long-term pharmacotherapeutic treatment. The neuroleptics, in particular, and most antidepressants, are known to induce weight gain as a side effect [205]. In addition, the absorption of these drugs is believed to be impacted after GBP surgery. The impact of the SG and LAGB procedures on absoption of these medications is less well established [205212]. There are few studies of postoperative outcomes for individuals with schizophrenia or acute psychosis. However, the severity of these conditions, the difficulty that many patients have with medical and pharmacological compliance, and the concerns about absorption of the medication used to treat the conditions have led experts to conclude that these conditions, when not optimally controlled, are contraindications to surgery for obesity and weight-related diseases.

Statement 3.7.4

Active or recent substance abuse and dependence, including alcohol abuse, is a contraindication to surgery for obesity and weight-related diseases.

(Level of evidence 3, grade of recommendation C)

Several studies have suggested that alcohol dependence is a negative prognostic factor for postoperative outcomes [212216]. Several studies have found an increase in the consumption of alcohol following surgery for obesity and weight-related diseases [217220]. Moreover, as demonstrated by studies on the effects of alcohol on the brain, there is clear evidence for a greater sensitivity to ethanol after surgery, especially after GBP. This greater sensitivity to alcohol seems to be less in LAGB patients [221224]. Moreover, vitamin deficiencies after GBP, especially thiamine, can result in the onset of acute Wernicke’s encephalopathy and/or Korsakoff syndrome [225]. Studies on the effects of cognitive behavioral therapy before surgery for obesity to reduce dependence on alcohol showed no significant effect in the long-term follow-up of patients with substance dependence [226]. Furthermore, there is a concern that some individuals with a history of substance abuse or dependence prior to surgery may postoperatively develop other compulsive or addictive behaviors, such as binging, compulsive shopping, gambling and, more rarely, drug abuse [227]. The prevalence of this phenomena, and the factors associated with it, are not well understood at this time.

Statement 3.7.5

Patients with binge eating disorder (BED) are likely to achieve less postoperative weight loss than patients without BED. Therefore, BED should be evaluated by the a multidisciplinary team before and after surgery for obesity and weight-related diseases.

(Level of evidence 3, grade of recommendation B)

The presence of BED is common among patients undergoing surgery. Several studies as well as a recent meta-analysis [194] have suggested that individuals with BED prior to surgery experience less postoperative weight loss as well as greater psychosocial distress postoperatively. For these reasons, the presence of BED should be assessed by the multidisciplinary team both before and after surgery. The condition is only considered a contraindication to surgery for obesity and weight-related diseases when the behavior is severe, untreated, and the multidisciplinary team is confident that the behavior would negatively impact postoperative outcomes.

Statement 3.7.6

Bulimia nervosa is considered a contraindication for surgery for obesity and weight-related diseases.

(Level of evidence 2, grade of recommendation B)

Bulimia nervosa is relatively rare among individuals who present for surgery for obesity and weight-related diseases. However, it is believed to be a contraindication for surgery [228, 229]. Patients with the diagnosis are recommened for psychiatric treatment and a period of symptom remission prior to being offered surgery for obesity and weight-related diseases.

Statement 3.7.7

Patients with the night eating syndrome (NES) are likely to achieve less postoperative weight loss after surgery for obesity and weight-related diseases than patients without NES. Therefore, NES should be evaluated by the multidisciplinary team before and after surgery.

(Level of evidence 3, grade of recommendation C)

Relatively few studies investigated the relationship between the NES in patients undergoing surgery for obesity and weight-related diseases and postoperative outcomes [190, 194, 230232]. Those studies have suggested that the NES, like BED, can negatively impact postoperative outcomes. For these reasons, the presense of the NES should be assessed by the multidisciplinary team both before and after surgery for obesity and weight-related diseases. The condition is only considered a contraindication to surgery when the behavior is severe, untreated, and the multidisciplinary team believes that the behavior would negatively impact postoperative outcomes.

Sub-chapter 3.8 Endocrinopathies and fertility

Statement 3.8.1

Endocrinopathies that are responsible for secondary obesity or that require therapeutic intervention represent contraindications to surgery for obesity and weight-related diseases. Their diagnostic classification and treatment are in fact necessary to support the success of surgery and to reduce the perioperative morbidity and mortality.

(Level of evidence 4, grade of recommendation C)

The routine assessments of thyroid, adrenal, and other endocrine gland functions are advisable in the presence of medical history or clinical features that suggest pathologies of the same nature. [233236]. It is recommended that screening for Cushing’s syndrome be carried out by the dexamethasone suppression (1 mg) night test because patients with severe obesity may have clinical characteristics related to hypercortisolism which are quite often hardly recognizable [233, 237, 238]. Lastly, it is useful to exclude the presence of hypoparathyroidism in patients previously treated with total thyroidectomy [239, 240], as surgery for obesity and weight-related diseases might cause or aggravate hypocalcemia [241243].

Statement 3.8.2

Inadequate drug treatment of pre-existing endocrine medical conditions is a contraindication to surgery for obesity and weight-related diseases. In fact, the re-evaluation and optimization of the treatment of these conditions are necessary to reduce perioperative morbidity and mortality.

(Level of evidence 3, grade of recommendation C)

In patients with hypoparathyroidism, calcium and calcitriol treatment must be implemented prior to surgery for obesity and weight-related diseases [239, 240, 244]. Although surgery results in a significant reduction in serum levels of uric acid [245], prophylactic treatment should be considered in patients with a history of gout to prevent acute attacks. It is believed that these attacks occur more frequently after surgery for obesity and weight-related diseases than after other types of upper abdominal surgery [246]. It is recommended to discontinue estrogen therapy before surgery both in premenopausal and in postmenopausal women to reduce the risk of thromboembolic events in the postoperative period [247]. While the trans-dermal administration of progestogen preparations results in a lower risk of deep vein thrombosis than oral administration [248, 249], there is no data to demonstrate the safety of this treatment in patients who are being considered for surgery for obesity and weight-related diseases.

Statement 3.8.3

In women with obesity, weight loss should be considered the first line of treatment for infertility regardless of the presence or absence of polycystic ovary syndrome. Weight loss can be obtained by lifestyle therapy, pharmacologic therapy, or surgery for obesity and weight-related diseases.

(Level of evidence: 2, grade of recommendation B)

Obesity has a detrimental effect on female fertility [250]. The relative risk of infertility secondary to anovulatory cycles is 2.7 times higher in women over the age of 18 whose BMI is higher than 32 kg/m2 [251]. For women having ovulatory cycles, the chance of conception is decreased by 5 % for every BMI increase by one unit [252]. The etiologies of this decrease in fertility are multivariate and include higher leptin and lower adiponectin levels. Additionally, the expression of steroids in the ovaries is altered and, in conjunction with elevated insulin levels, cause an inhibition of the sex hormone-binding globulin production in the liver which ultimately results in hyperandrogenemia [253]. Reduced fertility is also due to the higher incidence of polycystic ovary syndrome in women with obesity [254]. Weight loss in women with poly-cystic ovary syndrome, obtained by lifestyle therapy, pharmacological therapy, and surgery for obesity and weight-related diseases, improves spontaneous ovulation and pregnancy rates [254]. Since weight loss prior to conception improves live birth rates in women suffering from obesity with or without polycystic ovary syndrome, it has to be considered the first line therapy for infertility in women with obesity [255].

Statement 3.8.4

In women with obesity the risk for a wide range of maternal and fetal complications in pregnancy is reduced by weight loss. If weight loss cannot be obtained by nonsurgical management, surgery for obesity and weight-related diseases is an option for these women.

(Level of evidence 2, grade of recommendation C)

In pregnant women, obesity increases the risk for suffering complications such as preeclampsia, hypertension, gestational diabetes, and the need for delivery by cesarean section [256258]. Not only is the pregnant woman herself at risk, the children of pregnant women suffering from obesity are at a higher risk for stillbirth, prematurity, congenital abnormalities, macrosomia, and obesity [259, 260]. With a BMI between 30 and 39.9 kg/m2, the relative risk for gestational diabetes is increased 4.0 times, the relative risk for gestational hypertension up to 3.2 times, and the risk for preeclampsia up to 3.3 times, compared to individuals with BMI under 30 kg/m2. Therefore, preconception assessment of and counseling on obesity are highly encouraged [261]. If a normal body weight cannot be obtained by conservative treatment, surgery for obesity and weight-related diseases maybe considered. After surgery for obesity and weight-related diseases, the complications of pregnancy, such as gestational diabetes, preeclampsia, gestational hypertension, and macrosomia are less likely to occur [262, 263].

Sub-chapter 3.9 Cancer and organ transplantation

Statement 3.9.1

Surgery for obesity and weight-related diseases reduces the incidence of some malignancies and the mortality related to them.

(Level of evidence 3, grade of recommendation C)

Obesity is associated with an increased risk of developing malignancies, especially gastrointestinal, genitourinary, reproductive, and hematopoietic cancers [264266]. In the USA, 14 % of cancer deaths are attributed to obesity; this percentage reaches 20 % if we consider only female individuals. Furthermore, it was estimated that the hypothetical correction of the excess weight would have been able to prevent about 900,000 cancer deaths in the US population [267]. Although there are no randomized clinical trials, several studies conducted on very large samples of patients showed a significant decrease in the incidence of cancer and cancer-related mortality in patients undergoing surgery for obesity and weight-related diseases compared to obese non-operated patients [268270]. This reduction appears to be more pronounced in women than in men [8].

Statement 3.9.2

Obesity is a risk factor for developing postmenopausal breast cancer and is also a predictor for a poor prognosis in breast cancer patients. The reduction of body weight by lifestyle changes, medications, or surgery for obesity and weight-related diseases reduces the risk for breast cancer and related mortality.

(Level of evidence: 3, grade of recommendation B)

Whereas obesity reduces the risk to develop a premenopausal breast cancer, it increases the risk to develop a postmenopausal breast cancer [271273]. About 25 % of breast cancer cases after menopause are in women with obesity [271, 272]. A weight gain of only 10 kg between the age of 18 and menopause increases the risk of postmenopausal breast cancer by 16 % [274, 275]. A BMI over 25 kg/m2 increases the breast cancer risk proportionally. Five BMI units increase mortality within the range of 12–20 % [271273]. However, obesity does not only increase the risk of developing breast cancer but it is also correlated with a shorter overall survival in patients with breast cancer [276]. A BMI higher than 27 kg/m2 triples the risk for a breast cancer patient to die from the cancer [277, 278]. The reduction of body weight leads to a decrease in the risk of developing breast cancer [279, 280].

Statement 3.9.3

Weight loss in women with obesity reduces the risks of endometrial cancer development and related mortality. Surgery for obesity and weight-related diseases is one of the options for achieving weight loss and for reducing endometrial cancer risk.

(Level of evidence 3, grade of recommendation C)

Obesity is a well-known risk factor for endometrial cancer, with a direct link between body weight and the risk of developing the disease [271]. Being obese increases the risk for endometrial cancer in women by a factor of 2 to 3 [281, 282]. Being overweight by 25 kg increases the risk by a factor 10 [283]. Additionally, whereas endometrial cancer is a curable disease, the mortality risk is also increased for women with obesity [284, 285]. The increased risk of endometrial cancer is thought to be due to the “unopposed estrogen hypothesis.” The increased estrogen and androgen levels along with hyperinsulinemia in women with obesity lead to mitogenous effects on the endometrium, especially after menopause, when these hormones are no longer opposed by progestin [286]. There are preliminary data showing that weight loss after surgery for obesity and weight-related diseases may reduce the risk for endometrial cancer by as much as 70 %, provided the women do not regain the lost weight [287289].

Statement 3.9.4

In patients with obesity who have a history of cancer, surgery for obesity and weight-related diseases is indicated only in selected cases with proven remission of tumors, provided the absence of signs that suggest a possible relapse and provided the history of neoplasia is associated with a reasonably long life expectancy.

(Level of evidence 3, grade of recommendation C)

Given the exponential increase in the number of surgery for obesity and weight-related diseases procedures performed worldwide each year [22], it is increasingly likely that a candidate for surgery for obesity and weight-related diseases has previously received medical and/or surgical treatment for a malignancy.

To date, there is only one published paper in the literature that retrospectively analyzes the relationship between surgery for obesity and weight-related diseases and cancer. A group of 58 patients was extrapolated from a cohort of 1566 patients with obesity who underwent surgery for obesity and weight-related diseases, in whom the neoplastic disease was first diagnosed, respectively, before (40 patients), during (2 patients), or after (16 patients) the surgical procedure. In light of these results, the authors conclude that a previous diagnosis of cancer is not a contraindication to perform a surgery for obesity and weight-related diseases if remission of the neoplastic disease is complete and there are no suspicions of possible recurrence or metastasis [290].

Statement 3.9.5

Surgical treatment of obesity and related diseases is an effective therapeutic option with an acceptable rate of complications and postoperative mortality for both patients with obesity awaiting organ transplant and patients who have received an organ transplant.

(Level of evidence 3, grade of recommendation C)

Given its exponential distribution, the diagnosis of obesity is increasingly common among patients undergoing organ transplantation [291298]. Within the therapeutic management of the patient suffering from obesity who is a candidate for organ transplant, the role of surgery for obesity and related diseases before, during, or after transplantation, has only been described in a few case reports or papers on small groups of patients. [106, 108, 299308]. Surgery for obesity and related diseases has been shown to increase eligibility for a transplant (in some cases, weight loss constitutes the indication for surgery) [108, 307, 308]. For patients with obesity who have already undergone transplant surgery, the weight loss after surgery for obesity and weight-related diseases is quite similar to that achieved in non-transplant candidates who are obese, but with an increase in peri-and postoperative complications and mortality [293296, 309]. This increased incidence of adverse events can be judged acceptable in view of the functional status of patients undergoing surgery for obesity and related diseases.

Sub-chapter 3.10: Pseudotumor cerebri

Statement 3.10

Weight reduction is recommended for patients with obesity suffering from pseudotumor cerebri or idiophatic intracranial hypertension (IIH).

(Level of evidence 3, grade of reccomendation C)

Pseudotumor cerebri, also called idiopathic IIH, is defined by an elevated intracranial pressure with normal cerebrospinal fluid (CSF) composition and no evidence of other findings on neuroimaging or other evaluations. Typical symptoms of IIH are headache, papilledema, and vision loss [310]. The overall annual incidence of IIH among the general population is approximately two per 100,000. However, it is especially high among young women suffering from obesity, which remains one of the most important risk factors [311313]. A theory by Sugerman et al. suggests that among patients with obesity, increased intra-abdominal pressure may result in an increase in pleural and cardiac filling pressures, which in turn increases central venous pressure, and ultimately decreases CSF absorption by reducing the pressure gradient between the dural venous sinuses and the subarachnoid space [314]. There are different medical (carbonic anhydrase inhibitors, topiramate, oral steroids) and surgical approaches (ventriculo-or lumboperitoneal shunting and stenting of the transverse venous sinus stenoses), but there are no randomized controlled trials prospectively assessing and comparing these treatments. Ventriculo- or lumboperitoneal shunts are widely used but they have a high frequency of failure [312, 315]. Weight reduction is recommended for all patients with obesity and with IIH. Sugerman demonstrated that surgery for obesity and weight-related diseases is the long-term procedure of choice for severely patients with obesity and IIH. It is shown to have a much higher rate of success than CSF-peritoneal shunting, as well as providing resolution of additional obesity comorbidity [316]. Cohort studies, case series and a retrospective review confirm reduction in intracranial pressure, symptom prevalence, and improvement of papilledema and visual fields can be obtained with a salt-restricted and low-calorie diet [316326].

Sub-Chapter 3.11 Chronic inflammation

Statement 3.11.1

Obesity leads to chronic inflammation resulting in multiple inflammation-triggered diseases. Surgery for obesity and weight-related diseases improves chronic inflammation status and appears to be an immune restorative procedure. This issue is considered as a supporting indication for surgical treatment of obesity and weight-related diseases.

(Level of evidence 3, grade of recommendation C)

New findings in the pathophysiology of adipose tissue support the intimate relationship between the adipose tissue and the hematopoietic system [327, 328]. Several studies report that chronic systemic inflammation in obesity originates from local immune responses in visceral adipose tissue. Expansion of adipocytes and a concomitant reduction in capillary density and blood flow lead to oxidative stress and endoplasmic reticulum stress in adipocytes, resulting in increased macrophage infiltration, abnormal cytokine production, and increased acute-phase reactants, thus causing chronic inflammation [329332]. Chronic inflammation directly promotes insulin resistance and type 2 diabetes mellitus, cardiovascular disease, and increased cancer risk [333336]. Surgically induced weight loss improves inflammatory mediators such as acute-phase proteins and cytokines. Abnormal levels are described to reverse rapidly after LAGB, vertical banded gastroplasty (VBG) and GBP [337340]. Thus, surgery for obesity and related diseases leads to an improvement of the chronic inflammation status and in a prevention of inflammation-triggered diseases.

Sub-chapter 3.12 Renal alterations

Statement 3.12.1

Surgery for obesity and weight-related diseases improves renal function in patients with obesity with or without type 2 diabetes.

(Level of evidence 2, grade of recommendation C)

In the SOS study with a median follow-up of 10 years, hyperalbuminuria (defined as urinary albumin excretion of >30 mg per 24 h) among the 1498 patients in the surgery group and 1610 controls without hyperalbuminuria at baseline, occurred in 246 participants in the control group and in 126 in the surgical group, corresponding to an incidence of 20.4 and 9.4 per 1000 person years [341]. No RCT have been performed comparing the effects of surgery for obesity and weight-related diseases with nonsurgical therapies on hard renal endpoints or markers of diabetes-related kidney disease [342]. Nonetheless, results of the studies published, thus far, are consistent in pointing towards improved outcomes [92, 343348]. A reduction in the median albumin/creatinine ratio (ACR) after 6 months from surgery has been shown. While the reduced ACR after GBP correlated with postoperative insulin resistance, it did not correlate with postoperative BMI. Retrospective analysis of prospectively collected data in a cohort of 52 patients undergoing GBP, SG, or LAGB and followed up for 5 years, showed that 58 % of patients achieved remission (an ACR <30 mg/g) with favorable outcomes correlating with postoperative weight loss and blood pressure [347]. In general terms, improvements are particularly pronounced in the subgroup with preoperative hyperalbuminuria and did not correlate with reductions in BMI.

Statement 3.12.2

Chronic renal failure requiring dialysis should not be considered a contraindication to surgery for obesity and weight-related diseases.

(Level of evidence 2, grade of recommendation B)

A retrospective review of a prospectively collected database was conducted for dialysis patients who underwent surgery for obesity and weight-related diseases. From the 3048 patients who had undergone surgery, only 0.7 % were dialysis patients. Chronic renal failure requiring dialysis should not be considered a contraindication to surgery for obesity and weight-related diseases since excellent medium-term weight loss and an acceptable (albeit increased) risk/benefit ratio are observed [349]. For subjects who are severely obese or unable to undertake an exercise program, surgery for obesity and weight-related diseases is recommended.

Statement 3.12.3

Surgery for obesity and weight-related diseases can be used as a bridge to renal transplantation in patients with renal failure who suffer from obesity, type 2 diabetes, or both.

(Level of evidence 3, grade of recommendation C)

Surgery for obesity and weight-related diseases has been proposed as a bridge to renal transplantation in patients with renal failure who suffer from obesity, type 2 diabetes, or both. Small case studies of patients with end-stage renal disease have shown that surgery reduces weight and its associated comorbidities, making renal transplantation safer and potentially more successful [350]. Outcome measures of more than 209,000 renal transplant recipients demonstrated significant differences in favor of factors such as lower BMI (<30 kg/m2); lower mortality; delayed graft function; acute rejection; 1-, 2-, and 3-year graft survival; 1-, 2-, and 3-year patient survival; wound infection and dehiscence; length of hospital stay; operation duration; hypertension; and incisional hernia [351]. Therefore, end-stage renal disease patients with a BMI >30 kg/m2 should preferably lose weight prior to renal transplantation. If this cannot be achieved with nonsurgical measures, surgery for obesity and weight-related diseases could be considered in renal transplant candidates who are morbidly obese.

Statement 3.12.4

Improvement in urinary incontinence may be an important benefit of surgery for obesity and weight-related diseases.

(Level of evidence 3, grade of recommendation C)

A study on 2458 participants of ten obesity centers and 3 years follow-up, showed that weight loss was related to urinary incontinence remission. Among women and men with severe obesity, surgery for obesity and weight-related diseases was associated with substantially reduced urinary incontinence over 3 years [352].

Sub-chapter 3.13 Functional status

Statement 3.13.1

Obesity is associated with poor physical functioning. Weight loss induced by surgery for obesity and weight-related diseases has been shown to improve objective measures of physical functioning. Initial data suggest that the physical functioning of bariatric patients might also be further improved by participation in physical exercise training programs.

(Level of evidence 2, grade of recommendation A)

Obese adult patients with obesity frequently have marked impairment in physical functioning, as measured by the ability to perform simple physical activities (walking, getting out of a chair, climbing stairs) [353]. Nine longitudinal observational studies reporting objective measures of physical functioning in adults with obesity before and after surgery for obesity and weight-related diseases were recently identified and reviewed [353]. The 6-min walk test (6MWT), which measures the maximal distance a participant can walk in 6 min, was reported in seven studies [354360], and found to be improved after surgery in all of them, with a relative increase after surgery ranging from 8.8 to 33.3 % [353]. The timed up-and-go (TUG) test, which measures the time taken to get up, walk 3 min, turn around, walk back, and sit down, was reported in three studies [349, 359361], and also found to be improved after surgery in all of them [353]. Finally, two studies [357, 362] demonstrated a significant improvement in global physical performance, as tested by the short physical performance battery (SPPB), a three-component test in which balance, walking time, and chair rise were assessed [353].

In the same review [353], six studies evaluated physical performance with exercise testing protocols [358, 363367]. Exercise capacity improved in all studies. Where reported, peak oxygen uptake markedly improved in relation to body weight, suggesting improved aerobic fitness. However, absolute values were either unchanged or decreased, possibly because of the unavoidable loss of muscle mass that accompanies rapid surgical weight loss. This pattern suggests that improvement in exercise capacity may be more related to a mechanical advantage attributable to weight loss, and not to absolute improvements in cardiorespiratory or muscle function [353].

Physical activity during weight loss is known to be beneficial and combined resistance and aerobic training has been shown to have the most favorable outcome on aerobic fitness [353]. Castello et al. prospectively randomized 32 eligible morbidly obese women treated with GBP to an aerobic exercise training program group or to a control group. The “walking distance at 6-min walk” test increased after surgery only in the group undergoing the physical training program [368]. Stegen et al. divided 15 morbidly obese patients treated with GBP in a mixed strength and endurance training program group and in a control group. Weight loss in the control group resulted in a decrease in muscle strength and no improvement in aerobic capacity. As a consequence, most components of functional capacity did not improve. In contrast, muscle strength, aerobic capacity, and functional capacity were all significantly improved in the training group [369].

Statement 3.13.2

Work absenteeism due to sick leave and disability-related early retirement is higher in workers with obesity. Weight loss induced by surgery for obesity and weight-related diseases results in a reduction of sick leave and may be associated with favorable effects on disability pension in men.

(Level of evidence 2, grade of recommendation A)

Obese workers generally have higher work absenteeism by sick leave than their normal-weight colleagues [370]. Having a BMI above 50 kg/m2, lower levels of education, and suffering from four or more comorbidities were found to be significant predictors of sick leave in a morbidly obese population [371]. Moreover, the productive life of patients with obesity is shorter than in non-obese subjects not only because of premature mortality but also because of earlier retirement due to disability. In Sweden, the use of disability pension was 2.3 times higher in men with morbid obesity and 2.7 times higher in women with morbid obesity than in the general population [372]. Most of these disability pensions were generated by cardiovascular diseases and orthopedic problems related to arthritis, both conditions that are known to be preventable with weight loss [372]. The loss of work productivity related to sick leave and disability retirement has a profound impact on the general well-being and quality of life of individuals who are obese and generates enormous economic costs for the society [370].

The effects of surgically induced weight loss on sick leave and disability pension have been analyzed in the SOS study [373, 374]. In the year prior to treatment, adjusted average number of days of sick leave plus disability pension was similar in surgical patients and controls [373]. Compared with controls, the surgical group had 35 % more days of sick leave during the first year after surgery, but 10–14 % fewer days during years 2–3. At year 4, the total number of days of sick leave tended to be lower in the surgical group [373]. The reduction in sick leave days observed in the surgical group during years 2–3 after surgery for obesity and weight-related diseases was more significant in patients aged 47–60 years than in younger patients [373]. In a more recent report covering an extended 10-year follow-up period, SOS investigators reported that when adjusting for baseline confounders, a reduced risk of disability pension was suggested in men in the surgery group (hazard ratio 0.79, 95 % confidence interval 0.62–1.00; P = 0.05). Moreover, the adjusted average number of disability pension days was lower in the surgical group [374]. In conclusion, results from the SOS study suggest that surgery for obesity and weight-related diseases results in a reduction of sick leave and may be associated with favorable effects on disability pension in men.

Chapter 4 General considerations for indications of surgery for obesity

Sub-chapter 4.1: Quality of life

Statement 4.1.1

Surgery for obesity and weight-related diseases has proven to be effective in determining the overall improvement of the quality of life of patients suffering from obesity.

(Level of evidence 1, grade of recommendation A)

Excess weight leads to an overall deterioration of quality of life (health-related quality of life—HRQL) that can affect both the psychosocial and the physical dimensions. The higher the degree of obesity, the greater the deterioration [375]. Other factors such as age (over 35 years), female gender, and the presence of comorbidities may aggravate the burden of obesity on the physical, mental, and social variables [376, 377]. In contrast, weight loss has a beneficial effect on HRQL that is directly proportional to the amount of weight lost [378, 379]. This effect is therefore “magnified” by surgery for obesity and weight-related diseases that results in significantly greater weight loss [380384]. In terms of quality of life, teenagers and individuals with a BMI between 30 and 35 kg m2 similarly benefit from surgery for obesity and weight-related diseases [385, 386]. Numerous published studies, including randomized clinical trials, compare the effects of surgery for obesity and weight-related diseases and the outcome of different methods aiming at the dietary-behavioral well-being of the individual. These studies unequivocally show the superiority of surgery for obesity and weight-related diseases for improving the psychosocial and mental components that characterize the individual HRQL [62, 115, 380, 387391].

Statement 4.1.2

The improvement in the quality of life of the patient with obesity treated by surgery for obesity and weight-related diseases is independent from the type of performed procedure.

(Level of evidence 1, grade of recommendation A)

Two randomized clinical trials investigated the correlation between a specific type of procedure and the extent of the benefits on HRQL [392, 393]. In a study, LAGB and GBP are compared [392]. In another study, GBP and BPD were compared [393]. In both studies, while registering statistically significant differences between the two procedures regarding weight loss, the degree of improvement of quality of life was very similar. These results are corroborated by similar data reported in a recent non-randomized trial that compared the four most frequently performed procedures (GBP, SG, LAGB, and BPD) [394].

Statement 4.1.3

In long-term follow-up after surgery for obesity and weight-related diseases, the quality of life follows the trend of body weight. The quality of life remains satisfactory provided the percentage of excess weight loss (%EWL) is maintained above 10 %.

(Level of evidence 1, grade of recommendation B)

At present, there are two published studies that analyze the changes of quality of life of the patient with obesity undergoing surgery for obesity and weight-related diseases. At a median follow-up of 6 [391] and 10 years, respectively [380], the results obtained appear similar. The different aspects of HRQL significantly and rapidly improve in the first 12–24 months after the surgical procedure, and then gradually worsen, likely due to weight regain. In the SOS, there is, between 6 and 10 years after surgery, a substantial stabilization of the quality of life of the patient that is still satisfactory compared to the preoperative condition [380].

Sub-chapter 4.2 Adolescents

Statement 4.2.1

Surgery for obesity and weight-related diseases is effective in patients with obesity who are under 18 years of age.

(Level of evidence 2, grade of recommendation B).

In the absence of changes of lifestyle, children with obesity may suffer a reduction of life expectancy between 10 and 20 years and are at risk to develop serious health problems between 40 and 60 years of age. Studies show that without proper treatment, children with extreme obesity may continue to suffer from obesity in adulthood, and that overweight adolescents have a 70 % chance of becoming adults with obesity or overweight [395].

In addition, two thirds of children with morbid obesity have two or more cardiovascular risk factors and a significant proportion of these children suffer comorbidities that are usually seen in adults. Obesity is a risk factor for cardiovascular diseases such as hypertension, dyslipidemia, left ventricular hypertrophy, and atherosclerosis. A study that included adolescents aged 5 to 17 years, demonstrated that 70 % of children with obesity had at least one cardiovascular risk factor and that 39 % of children suffering from obesity had at least two risk factors. The incidence of type 2 diabetes has increased dramatically among adolescents in this study, representing a fifth of newly diagnosed cases of type 2 diabetes. At this point diagnostic criteria for MSy are already present for adolescents aged more than years of age or greater who present at a greater than 90th percentile on their waist circumference [396].

Obesity in youngsters is also associated with liver disease, particularly steatohepatitis with fibrosis and eventually cirrhosis progression. In addition, the negative impact on the psychologic health, notoriously important to the emotional development, cannot be overstated. In this context, the incidence of depression, low self-esteem and poor interaction with peers, anxiety, suicide, and serious psychosocial problems increases with increased weight [397]. From a neurological point of view, obesity is associated with IIH or pseudotumor cerebri (see sub-chapter 3–10). Children suffering from obesity have a six times higher incidence of obstructive sleep apnea than children of normal BMI.

In general, children and adolescents with obesity should be evaluated, followed, and treated by endocrinologists and pediatricians. The lifestyle of these children, their living environments, the degrees of obesity in their family histories and their neonatal or psychomotor developments should be noted. A history for a possible medication use related to the development of obesity must also be investigated. The collection of these data is important to help rule out secondary, syndromic, and monogenic causes of obesity.

In 2007, the European interdisciplinary guidelines, in agreement with the views expressed by a document from US pediatricians, supported the use of surgery for patients with obesity who are younger than 18 years of age, with some limitations. These parameters included supporting surgery for obesity and weight-related diseases only for patients whose BMI >40 kg/m2 (or >99.5th percentile for age) with at least one comorbidity. Patients had to have been treated medically for at least 6 months in a specialized center and demonstrated skeletal maturity and completed development (Tanner Score >4). They had to also demonstrate an ability to adhere to multidisciplinary pre- and postoperative programs and have good access to a facility with a pediatric support specialist [398].

In the face of an increased incidence of adolescent obesity, the knowledge that a teenager with obesity has a high propensity to become a severely obese adult, the growing safety and efficacy of surgery for obesity and weight-related diseases, and the increasing experience of multidisciplinary teams has triggered greater use of surgery for obesity and weight-related diseases for the adolescent suffering from obesity [387, 395, 399].

One of the main concerns when dealing with adolescents with obesity is their degree of maturity. Previous recommendations stated that the earliest age for adolescents to be considered for surgery for obesity and weight-related diseases was 13 years of age for girls and 15 years for boys. Future recommendations should support surgery for patients who have reached the 95th percentile for parental height (skeletal maturation nearly complete). Age limits should, however, also take into account the advent of puberty.

The selection criteria published by ASMBS in 2012 [400] revised and simplified the criteria published by the interdisciplinary European group in 2007 [398] and Pratt in 2009 [397].

The criteria stated that the BMI values used in adults appear to be appropriate for the selection of adolescent patients but must include additional specific parameters such as BMI >35 kg/m2 with at least one significant comorbidity such as T2DM, moderate or severe OSAS (AHI >15 events/h), pseudotumor cerebri, severe steatohepatitis, or BMI >40 kg/m2 with the presence of other comorbidities such as mild OSAS (AHI 5–15 events/h), high blood pressure, impaired glucose tolerance, insulin resistance, dyslipidemia, impaired quality of life, and difficulties in daily activities. It is possible that in the future, like for adults, BMI as the major criteria for determining if an adolescent with obesity can undergo surgery for obesity and weight-related diseases, will play a more minor role in the decision-making. Other factors may become more prevalent such as the multidisciplinary evaluation, psychological factors, metabolic issues, functional comorbidities, quality of life, suicide risk for adolescents, and consideration of long-term health risks in the absence of treatment in a patient who has a long life expectancy [396].

Sub-chapter 4.3 Elderly patients

Statement 4.3.1

Surgery for obesity and weight-related diseases is effective in patients with obesity who are over the age of 60 years.

(Level of evidence 2, grade of recommendation A).

Surgery for obesity and weight-related diseases in the elderly must pay particular attention to the cost/benefit ratio. The comorbidities are usually more common and more severe, and the operative complications are more frequent resulting in an increased mortality risk. The expected weight loss is often lower and the effects on the quantity and quality of remaining life are variable and not really quantifiable. Additionally, there is less compliance to new dietary guidelines imposed by interventions [401]. While the literature demonstrates that surgery for obesity and weight-related diseases in the elderly results in a higher percentage of postoperative complications and lower weight loss related to younger patients, there are similar improvements/resolutions of the comorbidities and improvement of functional independence and quality of life [402404].

Taylor et al. conducted a prospective study in 40 patients with mean age 65.8 years (range 60–72) and preoperative mean BMI of 42.2 kg/m2 treated by LAGB [405]. Mean excess weight loss at 2 years was 54 %. Three complications (7.5 %) occurred (1 slippage and 2 access-port infections). There were no deaths. After a mean postoperative interval of 27 months, SF-36 scores improved significantly in four of the eight components and exceeded the age-matched population controls in three components. Comorbidity improvement was reported in 80 % of patients with diabetes, 79 % with dyslipidemia, 75 % with obstructive sleep apnea, 72 % with heartburn, 69 % with hypertension, 60 % with musculoskeletal pain, and 56 % with anxiety/depression. Medication requirements reduced or ceased in 66 % of individuals who required musculoskeletal analgesics, 43 % of diabetics, 33 % bronchodilators used, and 29 % with hypertension. Sleep improved in 48 %, self-esteem increased in 70 %, and 72 % had a better outlook on life. Eighty-two of patients were pleased that they had undergone LAGB, and 91 % would recommend LAGB to other older people.

Dunckle-Blatter et al. retrospectively analyzed the data of 1065 patients who underwent GBP [406]. Seventy-six (7.1 %) were aged >60 years and 989 patients (92.9 %) were <60 years old. In the older group, the mean number of comorbidities was 10; 70.5 % had diabetes, and 83.6 % had hypertension. In the younger group, the mean number of comorbidities was 4.7. The mean number of preoperative medications was 10 in the older group compared with 6.0 in the younger group. The mean length of stay was 2.9 days in both groups. Postoperatively, medications were reduced by nearly 50 % in both groups. Diabetes and hypertension resolved or improved significantly in both groups. The mean percentage of excess body weight loss was lower in the older patients (54.9 % versus 60.1 %; P = 0.09). The 90-day operative mortality rate was 1.64 % in the older group versus 0.53 % for the younger group (P = NS). The authors concluded that the mortality rate was acceptable in the older group, despite the greater number of comorbidities. Both diabetes and hypertension were more common in this population, with trends toward better improvement after GBP than in younger patients [406].

Most of the studies conclude that the overriding goal of surgery for obesity and weight-related diseases in patients older than 60 years is to improve the quality of life but not to expect a prolonged median survival [407, 408].

Sub-chapter 4.4 Body composition

Statement 4.4.1

Body composition and body fat distribution determine the individual risk for obesity-associated metabolic and cardiovascular disorders.

(Level of evidence 3, grade of recommendation B)

Differences in body composition and fat distribution are predictors of the individual risk to develop metabolic syndrome (e.g., T2DM, dyslipidemia, hepatic steatosis) and cardiovascular diseases [409]. Adipose tissue stored in visceral fat depots make individuals with obesity more prone to metabolic and cardiovascular complications than fat distributed subcutaneously [32]. Normal weight individuals with low subcutaneous but increased visceral fat mass have an increased cardio-metabolic risk [410]. At the same time insulin-sensitive healthy patients with obesity may be protected against obesity-associated metabolic diseases [411]. In addition, it is well-known that the extent of fat loss in patients with lipodystrophies determines the severity of associated metabolic complications such as diabetes mellitus, hypertriglyceridemia, and hepatic steatosis [412]. However, there is insufficient evidence-based data to suggest how to assign a patient to a specific surgical procedure on the basis of body composition and fat distribution.

Statement 4.4.2

Reduction of subcutaneous fat mass alone (e.g., by liposuction) does not improve circulating metabolic and inflammatory parameters.

(Level of evidence 3, grade of recommendation C).

It has been demonstrated that a significant reduction in subcutaneous fat mass by liposuction does not improve circulating metabolic and inflammatory parameters [413], whereas the reduction of visceral fat mass by omentectomy in addition to LAGB has significant beneficial and long-term effects on measures of glucose metabolism and insulin sensitivity in individuals with obesity [414]. These positive effects were not observed in patients with obesity undergoing omentectomy without the concomitant LAGB [415].

Statement 4.4.3

Substantial weight loss following surgery for obesity and weight-related diseases may lead to bone mass loss and, subsequently, to an increased risk of fractures.

(Level of evidence 3, grade of recommendation C)

Several studies, using dual-energy X-ray absorptiometry, have reported substantial bone loss after surgery for obesity and weight-related diseases [416, 417]. Bariatric patients have been found to be more prone to fractures when compared to the general population [418420]. A recent analysis of the SOS has shown that women who undergo surgery for obesity and weight-related diseases are at an increased risk of developing fractures and osteoporosis long-term. The findings do not seem to apply to men who have surgery for obesity and weight-related diseases. During a follow-up of as long as 25 years, women in the study who underwent one of three bariatric procedures (LAGB, VBG, GBP) were 1.5 times more likely to suffer a fracture compared with controls and significantly more likely to develop osteoporosis. GBP, compared to LAGB and VBG (no data about SG), presented higher risk of osteoporosis [421]. It is noteworthy, that profound weight loss may cause artifactual changes in DXA areal bone mineral density results. Assessment of volumetric bone mineral density by quantitative computed tomography may be less susceptible to such artifacts [422].

Statement 4.4.4

Loss of lean body mass after surgery for obesity and weight-related diseases is substantial and strategies to limit lean body mass loss should be emphasized.

(Level of evidence: 2, grade of recommendation: B)

Reduced skeletal muscle mass is a major predictor of impaired physical function and survival [423]. It has been suggested that loss of lean body mass may be substantial after surgery for obesity and weight-related diseases [424, 425]. In a recent systematic review of the literature, the majority of reports found positive effects of exercise before and following surgery for obesity and related diseases on anthropometric parameters, cardiovascular risk factors and physical fitness [426]. In the long-term, higher physical activity and lower sedentary time is associated with greater excess weight loss and maintenance of reduced body weight following surgery for obesity and weight-related diseases [427]. Chaston et al., in a systematic review, evaluated changes in fat free mass (FFM) following significant weight loss. The authors included in the review RCTs, clinical controlled trials, and observational studies and concluded that the percentage of weight lost as FFM in medical interventions was influenced by the level of calorie restriction, exercise, and rate of weight loss. BPD and GBP were associated with greater %FFML than LAGB [428].

Sub-chapter 4.5 Low BMI

Statement 4.5.1

Surgery for obesity and weight-related diseases is effective in patients with class I obesity (BMI 30–35 kg/m2) and comorbidity.

(Level of evidence 1, grade of recommendation A)

It has been demonstrated that patients with the same BMI can have health conditions and risk factors that fundamentally differ. On the other hand, patients with class I obesity may have similar or even worse comorbidities, compared to subjects with obesity of higher classes. Consequently, there is a need to identify parameters other than BMI to better characterize patients eligible for surgery [61].

The evidence for the good results of surgery for obesity and weight-related diseases on the metabolic control of patients with class I and class II obesity and type II diabetes have stimulated and encouraged the expansion of the indications for surgery even for patients with BMI 25–30 kg/m2. [45, 429, 430].

In 2004 and in 2005, the ASMBS and the European Association of Endoscopic Surgeons (EAES) recommended the extension of surgery for obesity and weight-related diseases to clinical trials involving patients with class I obesity in presence of a comorbidity [431, 432].

The interest in patients with class I obesity began to rise at the same time as data on the beneficial effects of surgery on diabetes control began to spread. In fact, in 2011, the International Diabetes Federation (IDF) recommended surgical treatment in patients with type 2 diabetes and class I obesity who have failed conventional therapies. The IDF also suggested that the surgeries be done within the limits of research protocols and not as the first therapeutic approach [11, 433].

In 2013, the ASMBS concluded that surgery for obesity and weight-related diseases is recommended in patients with BMI 30–35 kg/m2 if there is an inability to maintain weight loss or improvement/resolution of comorbidities obtained with nonsurgical therapies. In randomized controlled trials, LAGB, SG, and GBP have been shown to be safe, well tolerated, and effective in the short- and medium-term. Furthermore, it is only appropriate to proceed with surgery after an attempt at nonsurgical therapy [60].

In 2014, the IFSO created a position statement concerning surgery for patients with class I obesity. The IFSO stated that surgery for obesity and weight-related diseases is a highly effective weight loss strategy in patients with class I obesity at least in the medium-term. The adverse events rate in patients with class 1 obesity appears to be the same than in morbid obesity [61].

The overall analysis of randomized controlled trials, meta-analyses and prospective and retrospective studies have shown good results in terms of weight loss and reduction of comorbidity compared with patients who present a higher class obesity [10, 13, 45, 6265, 429, 430, 434437].

Conclusion

Obesity is a chronic disease that has already reached pandemic proportions and is becoming one of the leading causes of death and disability worldwide. A comprehensive, sustainable, and proactive strategy to deal with the challenges posed by the obesity epidemic is urgently needed. Weight loss induced by surgery has proven to be highly efficacious in treating obesity and its comorbidities.

Many international scientific societies and health organizations have proposed over the years position statements or guidelines related to the indications for surgical treatment for patients with obesity. These include scientific statements from the American Heart Association (AHA) in 2011, American Association of Clinical Endocrinologists (AACE) in 2011, American Diabetes Association (ADA) in 2011, International Diabetes Federation (IDF) in 2011, Bariatric Scientific Collaborative Group (IFSO, IFSO-EC, EASO, IOTF, ECOG) in 2007, U.S. Internal Revenue Service (IRS) in 2002, Centers for Medicare & Medicaid Services (CMS) in 2006, National Institutes of Health (NIH) in 1991.

This position statement takes account the major changes that have occured in recent years. On one hand, the increased interaction between the various scientific societies, and health organizations dealing with obesity, and on the other hand, the improvements in the surgical techniques that have led to a significant reduction in morbidity and operative mortality.

The interaction with other scientific societies and health organizations has led to the conclusion that the disease of obesity can no longer be identified only by weight/height ratio (BMI) or anthropometric measures but must also take into account the different clinical manifestations of functional type, metabolic type, psychological/psychiatric type and, not less important, social type. These considerations, along with the development of less aggressive surgery, have created the need to draw up the position statement with upgraded indications. This statement was created by a working group formed by prominent members of the International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO) and then discussed and approved by the Executive Board of (IFSO).

Consequently, after a careful review of the available data concerning the safety and efficacy of surgery for obesity and weight-related diseases and its effectiveness in treating obesity and its comorbidities, IFSO’s panel has endorsed new recommendations that consider the most modern functional, metabolic, psychological/psychiatric, and social aspect of obesity.

Footnotes

Compliance with Ethical Standards

Ethical Approval All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and national research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards.

Informed Consent Informed consent was obtained from all individual participants included in the study.

References

  • 1.Buchwald H, Varco RL, editors. Metabolic surgery. New York: Grune and Stratton; 1978. [Google Scholar]
  • 2.Buchwald H, Varco RL, Matts JP, et al. Effect of partial ileal bypass surgery on mortality and morbidity from coronary heart disease in patients with hypercholesterolemia. Report of the Program on the Surgical Control of the Hyperlipidemias (POSCH) N Engl J Med. 1990;323(14):946–55. doi: 10.1056/NEJM199010043231404. [DOI] [PubMed] [Google Scholar]
  • 3.Kremen AJ, Linner LH, Nelson CH. An experimental evaluation of the nutritional importance of proximal and distal small intestine. Ann Surg. 1954;140(3):439–48. doi: 10.1097/00000658-195409000-00018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Buchwald H. The evolution of metabolic/bariatric surgery. Obes Surg. 2014;24(8):1126–35. doi: 10.1007/s11695-014-1354-3. [DOI] [PubMed] [Google Scholar]
  • 5.Kuk JL, Ardern CI. Are metabolically normal but obese individuals at lower risk for all-cause mortality? Diabetes Care. 2009;32(12):2297–9. doi: 10.2337/dc09-0574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.NIH conference. Gastrointestinal surgery for severe obesity. consensus development conference panel. Ann Intern Med. 1991;115(12):956–61. [PubMed] [Google Scholar]
  • 7.Sjöström L, Lindroos AK, Peltonen M, et al. Swedish Obese Subjects Study Scientific Group. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med. 2004;351(26):2683–93. doi: 10.1056/NEJMoa035622. [DOI] [PubMed] [Google Scholar]
  • 8.Sjöström L, Gummesson A, Sjöström CD, et al. Swedish Obese Subjects Study. Effects of bariatric surgery on cancer incidence in obese patients in Sweden (Swedish Obese Subjects Study): a prospective, controlled intervention trial. Lancet Oncol. 2009;10(7):653–62. doi: 10.1016/S1470-2045(09)70159-7. [DOI] [PubMed] [Google Scholar]
  • 9.Sjöström L, Peltonen M, Jacobson P, et al. Bariatric surgery and long-term cardiovascular events. JAMA. 2012;307(1):56–65. doi: 10.1001/jama.2011.1914. [DOI] [PubMed] [Google Scholar]
  • 10.Schauer PR, Kashyap SR, Wolski K, et al. Bariatric surgery versus intensive medical therapy in obese patients with diabetes. N Engl J Med. 2012;366(17):1567–76. doi: 10.1056/NEJMoa1200225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Dixon JB, Zimmet P, Alberti KG, et al. International Diabetes Federation Taskforce on Epidemiology and Prevention. Bariatric surgery: an IDF statement for obese type 2 diabetes. Diabet Med. 2011;28(6):628–42. doi: 10.1111/j.1464-5491.2011.03306.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Sjöström L. Review of the key results from the Swedish Obese Subjects (SOS) trial—a prospective controlled intervention study of bariatric surgery. J Intern Med. 2013;273(3):219–34. doi: 10.1111/joim.12012. [DOI] [PubMed] [Google Scholar]
  • 13.Ikramuddin S, Korner J, Lee WJ, et al. Roux-en-Y gastric bypass vs intensive medical management for the control of type 2 diabetes, hypertension, and hyperlipidemia: the diabetes surgery study randomized clinical trial. JAMA. 2013;309(21):2240–9. doi: 10.1001/jama.2013.5835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Buchwald H, Avidor Y, Braunwald E, et al. Bariatric surgery. a systemic review and meta-analysis. JAMA. 2004;292(14):1724–37. doi: 10.1001/jama.292.14.1724. [DOI] [PubMed] [Google Scholar]
  • 15.Christou NV, Lieberman M, Sampalis F, et al. Bariatric surgery reduces cancer risk in morbidly obese patients. Surg Obes Relat Dis. 2008;4(6):691–5. doi: 10.1016/j.soard.2008.08.025. [DOI] [PubMed] [Google Scholar]
  • 16.Christou NV, Sampalis JS, Liberman M, et al. Surgery decreases long-term mortality, morbidity, and health care use in morbidly obese patients. Ann Surg. 2004;240(3):416–23. doi: 10.1097/01.sla.0000137343.63376.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Busetto L, Mirabelli D, Petroni ML, et al. Comparative long-term mortality after laparoscopic adjustable gastric banding versus nonsurgical controls. Surg Obes Relat Dis. 2007;3(5):496–502. doi: 10.1016/j.soard.2007.06.003. [DOI] [PubMed] [Google Scholar]
  • 18.Aguiar IC, Freitas WR, Jr, Santos IR, et al. Obstructive sleep apnea and pulmonary function in patients with severe obesity before and after bariatric surgery: a randomized clinical trial. Multidiscip Respir Med. 2014;9(1):43. doi: 10.1186/2049-6958-9-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Greenburg DL, Lettieri CJ, Eliasson AH. Effects of surgical weight loss on measures of obstructive sleep apnea: a meta-analysis. Am J Med. 2009;122(6):535–42. doi: 10.1016/j.amjmed.2008.10.037. [DOI] [PubMed] [Google Scholar]
  • 20.Poirier P, Cornier MA, Mazzone T, et al. Bariatric surgery and cardiovascular risk factors: a scientific statement from the American Heart Association. Circulation. 2011;123(15):1683–701. doi: 10.1161/CIR.0b013e3182149099. [DOI] [PubMed] [Google Scholar]
  • 21.Buchwald H, Estok R, Fahrbach K, et al. Weight and type 2 diabetes after bariatric surgery: systematic review and meta-analysis. Am J Med. 2009;122(3):248–56.e5. doi: 10.1016/j.amjmed.2008.09.041. [DOI] [PubMed] [Google Scholar]
  • 22.Buchwald H, Oien DM. Metabolic/bariatric surgery worldwide 2011. Obes Surg. 2013;23(4):427–36. doi: 10.1007/s11695-012-0864-0. [DOI] [PubMed] [Google Scholar]
  • 23.Physical status: the use and interpretation of anthropometry. Report of a WHO Expert Committee. World Health Organ Tech Rep Ser. 1995;854:1–452. [PubMed] [Google Scholar]
  • 24.Wang J, Thornton JC, Russell M, et al. Asians have lower body mass index (BMI) but higher percent body fat than do Whites: comparisons of anthropometric measurements. Am J Clin Nutr. 1994;60(1):23–8. doi: 10.1093/ajcn/60.1.23. [DOI] [PubMed] [Google Scholar]
  • 25.Yajnik CS. Obesity epidemic in India: intrauterine origins? Proc Nutr Soc. 2004;63(3):387–96. doi: 10.1079/pns2004365. [DOI] [PubMed] [Google Scholar]
  • 26.Ko GT, Tang JS. Waist circumference and BMI cut-off based on 10-year cardiovascular risk: evidence for “central pre-obesity”. Obesity (Silver Spring) 2007;15(11):2832–9. doi: 10.1038/oby.2007.336. [DOI] [PubMed] [Google Scholar]
  • 27.Neel JV. Diabetes mellitus: a “thrifty” genotype rendered detrimental by “progress”? Am J Hum Genet. 1962;14:353–62. [PMC free article] [PubMed] [Google Scholar]
  • 28.Saydah SH, Fradkin J, Cowie CC. Poor control of risk factors for vascular disease among adults with previously diagnosed diabetes. JAMA. 2004;291(3):335–42. doi: 10.1001/jama.291.3.335. [DOI] [PubMed] [Google Scholar]
  • 29.Summary of revisions for the 2009 clinical practice recommendations. Diabetes Care. 2009;32(Suppl 1):S3–5. doi: 10.2337/dc09-S003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Dixon JB, Zimmet P, Alberti KG, Rubino F. International Diabetes Federation Taskforce on Epidemiology and Prevention. Bariatric surgery: an IDF statement for obese type 2 diabetes. Surg Obes Relat Dis. 2011;7(4):433–47. doi: 10.1016/j.soard.2011.05.013. [DOI] [PubMed] [Google Scholar]
  • 31.Lakdawala M, Bhasker A. Asian Consensus Meeting on Metabolic Surgery (ACMOMS). Report: Asian consensus meeting on metabolic surgery. Recommendations for the use of bariat-ric and gastrointestinal metabolic surgery for treatment of obesity and type II diabetes mellitus in the Asian population: August 9th and 10th, 2008, Trivandrum, India. Obes Surg. 2010;20(7):929–36. doi: 10.1007/s11695-010-0162-7. [DOI] [PubMed] [Google Scholar]
  • 32.Müller MJ, Lagerpusch M, Enderle J, et al. Beyond the body mass index: tracking body composition in the pathogenesis of obesity and the metabolic syndrome. Obes Rev. 2012;13(Suppl 2):6–13. doi: 10.1111/j.1467-789X.2012.01033.x. [DOI] [PubMed] [Google Scholar]
  • 33.Unger RH. Minireview: weapons of lean body mass destruction: the role of ectopic lipids in the metabolic syndrome. Endocrinology. 2003;144(12):5159–65. doi: 10.1210/en.2003-0870. [DOI] [PubMed] [Google Scholar]
  • 34.Kragelund C, Omland T. A farewell to body-mass index? Lancet. 2005;366(9497):1589–91. doi: 10.1016/S0140-6736(05)67642-8. [DOI] [PubMed] [Google Scholar]
  • 35.Okorodudu DO, Jumean MF, Montori VM, et al. Diagnostic performance of body mass index to identify obesity as defined by body adiposity: a systematic review and meta-analysis. Int J Obes (Lond) 2010;34(5):791–9. doi: 10.1038/ijo.2010.5. [DOI] [PubMed] [Google Scholar]
  • 36.Expert Consultation WHO. Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies. Lancet. 2004;363(9403):157–63. doi: 10.1016/S0140-6736(03)15268-3. [DOI] [PubMed] [Google Scholar]
  • 37.Sharma AM, Kushner RF. A proposed clinical staging system for obesity. Int J Obes (Lond) 2009;33(3):289–95. doi: 10.1038/ijo.2009.2. [DOI] [PubMed] [Google Scholar]
  • 38.Padwal RS, Pajewski NM, Allison DB, et al. Using the Edmonton obesity staging system to predict mortality in a population-representative cohort of people with overweight and obesity. CMAJ. 2011;183(14):E1059–66. doi: 10.1503/cmaj.110387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kuk JL, Ardern CI, Church TS, et al. Edmonton obesity staging system: association with weight history and mortality risk. Appl Physiol Nutr Metab. 2011;36(4):570–6. doi: 10.1139/h11-058. [DOI] [PubMed] [Google Scholar]
  • 40.Gill RS, Karmali S, Sharma AM. The potential role of the Edmonton obesity staging system in determining indications for bariatric surgery. Obes Surg. 2011;21(12):1947–9. doi: 10.1007/s11695-011-0533-8. [DOI] [PubMed] [Google Scholar]
  • 41.Longitudinal Assessment of Bariatric Surgery (LABS) Consortium. Flum DR, Belle SH, et al. Perioperative safety in the longitudinal assessment of bariatric surgery. N Engl J Med. 2009;361(5):445–54. doi: 10.1056/NEJMoa0901836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.De Maria EJ, Portenier D, Wolfe L. Obesity surgery mortality risk score: proposal for a clinically useful score to predict mortality risk in patients undergoing gastric bypass. Surg Obes Relat Dis. 2007;3(2):134–40. doi: 10.1016/j.soard.2007.01.005. [DOI] [PubMed] [Google Scholar]
  • 43.Blackstone RP, Cortes MC. Metabolic acuity score: effect on major complications after bariatric surgery. Surg Obes Relat Dis. 2010;6(3):267–73. doi: 10.1016/j.soard.2009.09.010. [DOI] [PubMed] [Google Scholar]
  • 44.Guo X, Liu X, Wang M, et al. The effects of bariatric procedures versus medical therapy for obese patients with type 2 diabetes: meta-analysis of randomized controlled trials. Biomed Res Int. 2013;2013:410609. doi: 10.1155/2013/410609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Dixon JB, O’Brien PE, Playfair J, et al. Adjustable gastric banding and conventional therapy for type 2 diabetes: a randomized controlled trial. JAMA. 2008;299(3):316–23. doi: 10.1001/jama.299.3.316. [DOI] [PubMed] [Google Scholar]
  • 46.Mingrone G, Panunzi S, De Gaetano A, et al. Bariatric surgery versus conventional medical therapy for type 2 diabetes. N Engl J Med. 2012;366(17):1577–85. doi: 10.1056/NEJMoa1200111. [DOI] [PubMed] [Google Scholar]
  • 47.Courcoulas AP, Belle SH, Neiberg RH, Pierson SK, Eagleton JK, Kalarchian MA, et al. Three-year outcomes of bariatric surgery vs lifestyle intervention for type 2 diabetes mellitus treatment: a randomized clinical trial. JAMA Surg. 2015 doi: 10.1001/jamasurg.2015.1534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zhuo X, Zhang P, Barker L, et al. The lifetime cost of diabetes and its implications for diabetes prevention. Diabetes Care. 2014;37(9):2557–64. doi: 10.2337/dc13-2484. [DOI] [PubMed] [Google Scholar]
  • 49.Picot J, Jones J, Colquitt JL, et al. The clinical effectiveness and cost-effectiveness of bariatric (weight loss) surgery for obesity: a systematic review and economic evaluation. Health Technol Assess. 2009;13(41):1–190. 215–357, iii–iv. doi: 10.3310/hta13410. [DOI] [PubMed] [Google Scholar]
  • 50.Keating CL, Dixon JB, Moodie ML, et al. Cost-effectiveness of surgically induced weight loss for the management of type 2 diabetes: modeled lifetime analysis. Diabetes Care. 2009;32(4):567–74. doi: 10.2337/dc08-1749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Hoerger TJ, Zhang P, Segel JE, et al. Cost-effectiveness of bariatric surgery for severely obese adults with diabetes. Diabetes Care. 2010;33(9):1933–9. doi: 10.2337/dc10-0554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Cremieux PY, Buchwald H, Shikora SA, et al. A study on the economic impact of bariatric surgery. Am J Manag Care. 2008;14(9):589–96. [PubMed] [Google Scholar]
  • 53.Warren JA, Ewing JA, Hale AL, et al. Cost-effectiveness of bar-iatric Surgery: increasing the economic viability of the most effective treatment for type II diabetes mellitus. Am Surg. 2015;81(8):807–11. [PubMed] [Google Scholar]
  • 54.Li JF, Lai DD, Ni B, et al. Comparison of laparoscopic Roux-en-Y gastric bypass with laparoscopic sleeve gastrectomy for morbid obesity or type 2 diabetes mellitus: a meta-analysis of randomized controlled trials. Can J Surg. 2013;56(6):E158–64. doi: 10.1503/cjs.026912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yip S, Plank LD, Murphy R. Gastric bypass and sleeve gastrectomy for type 2 diabetes: a systematic review and meta-analysis of outcomes. Obes Surg. 2013;23(12):1994–2003. doi: 10.1007/s11695-013-1030-z. [DOI] [PubMed] [Google Scholar]
  • 56.Zhang C, Yuan Y, Qiu C, et al. A meta-analysis of 2-year effect after surgery: laparoscopic Roux-en-Y gastric bypass versus laparoscopic sleeve gastrectomy for morbid obesity and diabetes mellitus. Obes Surg. 2014;24(9):1528–35. doi: 10.1007/s11695-014-1303-1. [DOI] [PubMed] [Google Scholar]
  • 57.Yang X, Yang G, Wang W, et al. A meta-analysis: to compare the clinical results between gastric bypass and sleeve gastrectomy for the obese patients. Obes Surg. 2013;23(7):1001–10. doi: 10.1007/s11695-013-0938-7. [DOI] [PubMed] [Google Scholar]
  • 58.Cho JM, Kim HJ, Menzo EL, et al. Effect of sleeve gastrectomy on type 2 diabetes as an alternative treatment modality to Roux-en-Y gastric bypass: systemic review and meta-analysis. Surg Obes Relat Dis. 2015 doi: 10.1016/j.soard.2015.03.001. [DOI] [PubMed] [Google Scholar]
  • 59.Wang MC, Guo XH, Zhang YW, et al. Laparoscopic Roux-en-Y gastric bypass versus sleeve gastrectomy for obese patients with Type 2 diabetes: a meta-analysis of randomized controlled trials. Am Surg. 2015;81(2):166–71. [PubMed] [Google Scholar]
  • 60.ASMBS Clinical Issues Committee. Bariatric surgery in class I obesity (body mass index 30–35 kg/m2) Surg Obes Relat Dis. 2013;9(1):e1–10. doi: 10.1016/j.soard.2012.09.002. [DOI] [PubMed] [Google Scholar]
  • 61.Busetto L, Dixon J, De Luca M, et al. Bariatric surgery in class I obesity: a position statement from the International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO) Obes Surg. 2014;24(4):487–519. doi: 10.1007/s11695-014-1214-1. [DOI] [PubMed] [Google Scholar]
  • 62.O’Brien PE, Dixon JB, Laurie C, et al. Treatment of mild to moderate obesity with laparoscopic adjustable gastric banding or an intensive medical program: a randomized trial. Ann Intern Med. 2006;144(9):625–33. doi: 10.7326/0003-4819-144-9-200605020-00005. [DOI] [PubMed] [Google Scholar]
  • 63.Lee WJ, Chong K, Ser KH, et al. Gastric bypass vs sleeve gastrectomy for type 2 diabetes mellitus: a randomized controlled trial. Arch Surg. 2011;146(2):143–8. doi: 10.1001/archsurg.2010.326. [DOI] [PubMed] [Google Scholar]
  • 64.Li Q, Chen L, Yang Z, et al. Metabolic effects of bariatric surgery in type 2 diabetic patients with body mass index <35 kg/m2. Diabetes Obes Metab. 2012;14(3):262–70. doi: 10.1111/j.1463-1326.2011.01524.x. [DOI] [PubMed] [Google Scholar]
  • 65.Reis CE, Alvarez-Leite JI, Bressan J, et al. Role of bariatric-metabolic surgery in the treatment of obese type 2 diabetes with body mass index <35 kg/m2: a literature review. Diabetes Technol Ther. 2012;14(4):365–72. doi: 10.1089/dia.2011.0127. [DOI] [PubMed] [Google Scholar]
  • 66.Chikunguwo SM, Wolfe LG, Dodson P, et al. Analysis of factors associated with durable remission of diabetes after Roux-en-Y gastric bypass. Surg Obes Relat Dis. 2010;6(3):254–9. doi: 10.1016/j.soard.2009.11.003. [DOI] [PubMed] [Google Scholar]
  • 67.DiGiorgi M, Rosen DJ, Choi JJ, et al. Re-emergence of diabetes after gastric bypass in patients with mid- to long-term follow-up. Surg Obes Relat Dis. 2010;6(3):249–53. doi: 10.1016/j.soard.2009.09.019. [DOI] [PubMed] [Google Scholar]
  • 68.Hall TC, Pellen MG, Sedman PC, et al. Preoperative factors predicting remission of type 2 diabetes mellitus after Roux-en-Y gastric bypass surgery for obesity. Obes Surg. 2010;20(9):1245–50. doi: 10.1007/s11695-010-0198-8. [DOI] [PubMed] [Google Scholar]
  • 69.Hamza N, Abbas MH, Darwish A, et al. Predictors of remission of type 2 diabetes mellitus after laparoscopic gastric banding and bypass. Surg Obes Relat Dis. 2011;7(6):691–6. doi: 10.1016/j.soard.2010.03.292. [DOI] [PubMed] [Google Scholar]
  • 70.Hawa MI, Kolb H, Schloot N, et al. Action LADA consortium. Adult-onset autoimmune diabetes in Europe is prevalent with a broad clinical phenotype: Action LADA 7. Diabetes Care. 2013;36(4):908–13. doi: 10.2337/dc12-0931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Manning SB, Pucci A, Batterham RL, et al. Latent autoimmune diabetes in adults presenting as diabetes “recurrence” after bariatric surgery: a case report. Diabetes Care. 2013;36(8):e120. doi: 10.2337/dc13-0810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kashyap SR, Schauer P. Clinical considerations for the management of residual diabetes following bariatric surgery. Diabetes Obes Metab. 2012;14(9):773–79. doi: 10.1111/j.1463-1326.2012.01577.x. [DOI] [PubMed] [Google Scholar]
  • 73.Deitel M. Update: why diabetes does not resolve in some patients after bariatric surgery. Obes Surg. 2011;21(6):794–6. doi: 10.1007/s11695-010-0329-2. [DOI] [PubMed] [Google Scholar]
  • 74.Lee WJ, Ser KH, Chong K, et al. Laparoscopic sleeve gastrectomy for diabetes treatment in nonmorbidly obese patients: efficacy and change of insulin secretion. Surgery. 2010;147(5):664–9. doi: 10.1016/j.surg.2009.10.059. [DOI] [PubMed] [Google Scholar]
  • 75.Czupryniak L, Wiszniewski M, Szymański D, et al. Long-term results of gastric bypass surgery in morbidly obese type 1 diabetes patients. Obes Surg. 2010;20(4):506–8. doi: 10.1007/s11695-010-0074-6. [DOI] [PubMed] [Google Scholar]
  • 76.Mendez CE, Tanenberg RJ, Pories W. Outcomes of Roux-en-Y gastric bypass surgery for severely obese patients with type 1 diabetes: a case series report. Diabetes Metab Syndr Obes. 2010;3:281–3. [PMC free article] [PubMed] [Google Scholar]
  • 77.Reyes Garcia R, Romero Muñoz M, Galbis VH. Bariatric surgery in type 1 diabetes. Endocrinol Nutr. 2013;60(1):46–7. doi: 10.1016/j.endonu.2012.01.021. [DOI] [PubMed] [Google Scholar]
  • 78.Breen DM, Rasmussen BA, Kokorovic A, et al. Jejunal nutrient sensing is required for duodenal-jejunal bypass surgery to rapidly lower glucose concentrations in uncontrolled diabetes. Nat Med. 2012;18(6):950–5. doi: 10.1038/nm.2745. [DOI] [PubMed] [Google Scholar]
  • 79.Reaven GM. Role of insulin resistance in human disease (syndrome X): an expanded definition. Annu Rev Med. 1993;44:121–31. doi: 10.1146/annurev.me.44.020193.001005. [DOI] [PubMed] [Google Scholar]
  • 80.Alberti KG, Zimmet P, Shaw J, et al. The metabolic syndrome—a new worldwide definition. Lancet. 2005;366(9491):1059–62. doi: 10.1016/S0140-6736(05)67402-8. [DOI] [PubMed] [Google Scholar]
  • 81.Sattar N, McConnachie A, Shaper AG, et al. Can metabolic syndrome usefully predict cardiovascular disease and diabetes? Outcome data from two prospective studies. Lancet. 2008;371(9628):1927–35. doi: 10.1016/S0140-6736(08)60602-9. [DOI] [PubMed] [Google Scholar]
  • 82.Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey. JAMA. 2002;287(3):356–9. doi: 10.1001/jama.287.3.356. [DOI] [PubMed] [Google Scholar]
  • 83.Maggard-Gibbons M, Maglione M, Livhits M, et al. Bariatric surgery for weight loss and glycemic control in nonmorbidly obese adults with diabetes: a systematic review. JAMA. 2013;309(21):2250–61. doi: 10.1001/jama.2013.4851. [DOI] [PubMed] [Google Scholar]
  • 84.Schauer PR, Bhatt DL, Kirwan JP, et al. STAMPEDE Investigators. Bariatric surgery versus intensive medical therapy for diabetes—3-year outcomes. N Engl J Med. 2014;370(21):2002–13. doi: 10.1056/NEJMoa1401329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kahn R. The metabolic syndrome (Emperor) wears no clothes. Diabetes Care. 2006;29(7):1693–6. doi: 10.2337/dc06-1453. [DOI] [PubMed] [Google Scholar]
  • 86.Prospective Studies Collaboration. Whitlock G, Lewington S, et al. Body-mass index and cause-specific mortality in 900,000 adults: collaborative analyses of 57 prospective studies. Lancet. 2009;373(9669):1083–96. doi: 10.1016/S0140-6736(09)60318-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Yusuf S, Hawken S, Ounpuu S, et al. Obesity and the risk of myocardial infarction in 27,000 participants from 52 countries: a case-control study. Lancet. 2005;366(9497):1640–9. doi: 10.1016/S0140-6736(05)67663-5. [DOI] [PubMed] [Google Scholar]
  • 88.Batsis JA, Sarr MG, Collazo-Clavell ML, et al. Cardiovascular risk after bariatric surgery for obesity. Am J Cardiol. 2008;102(7):930–7. doi: 10.1016/j.amjcard.2008.05.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Vest AR, Heneghan HM, Agarwal S, et al. Bariatric surgery and cardiovascular outcomes: a systematic review. Heart. 2012;98(24):1763–77. doi: 10.1136/heartjnl-2012-301778. [DOI] [PubMed] [Google Scholar]
  • 90.Busetto L, De Stefano F, Pigozzo S, et al. Long-term cardiovascular risk and coronary events in morbidly obese patients treated with laparoscopic gastric banding. Surg Obes Relat Dis. 2014;10(1):112–20. doi: 10.1016/j.soard.2013.03.010. [DOI] [PubMed] [Google Scholar]
  • 91.Kwok CS, Pradhan A, Khan MA, et al. Bariatric surgery and its impact on cardiovascular disease and mortality: a systematic review and meta-analysis. Int J Cardiol. 2014;173(1):20–8. doi: 10.1016/j.ijcard.2014.02.026. [DOI] [PubMed] [Google Scholar]
  • 92.Johnson BL, Blackhurst DW, Latham BB, et al. Bariatric surgery is associated with a reduction in major macrovascular and micro-vascular complications in moderately to severely obese patients with type 2 diabetes mellitus. J Am Coll Surg. 2013;216(4):545–56. doi: 10.1016/j.jamcollsurg.2012.12.019. [DOI] [PubMed] [Google Scholar]
  • 93.Sturm W, Tschoner A, Engl J, et al. Effect of bariatric surgery on both functional and structural measures of premature atherosclerosis. Eur Heart J. 2009;30(16):2038–43. doi: 10.1093/eurheartj/ehp211. [DOI] [PubMed] [Google Scholar]
  • 94.Tschoner A, Sturm W, Gelsinger C, et al. Long-term effects of weight loss after bariatric surgery on functional and structural markers of atherosclerosis. Obesity (Silver Spring) 2013;21(10):1960–5. doi: 10.1002/oby.20357. [DOI] [PubMed] [Google Scholar]
  • 95.Habib P, Scocco JD, Terek M, et al. Effects of bariatric surgery on inflammatory, functional and structural markers of coronary atherosclerosis. Am J Cardiol. 2009;104(9):1251–5. doi: 10.1016/j.amjcard.2009.06.042. [DOI] [PubMed] [Google Scholar]
  • 96.Saleh MH, Bertolami MC, Assef JE, et al. Improvement of atherosclerotic markers in non-diabetic patients after bariatric surgery. Obes Surg. 2012;22(11):1701–7. doi: 10.1007/s11695-012-0706-0. [DOI] [PubMed] [Google Scholar]
  • 97.Sarmento PLFA, Plavnik FL, Zanella MT, et al. Association of carotid intima-media thickness and cardiovascular risk factors in women pre- and post-bariatric surgery. Obes Surg. 2009;19(3):339–44. doi: 10.1007/s11695-008-9783-5. [DOI] [PubMed] [Google Scholar]
  • 98.Nerla R, Tarzia P, Sestito A, et al. Effect of bariatric surgery on peripheral flow mediated dilation and coronary microvascular function. Nutr Metab Cardiovasc Dis. 2012;22(8):626–34. doi: 10.1016/j.numecd.2010.10.004. [DOI] [PubMed] [Google Scholar]
  • 99.Priester T, Ault TG, Davidson L, et al. Coronary calcium scores 6 years after bariatric surgery. Obes Surg. 2015;25(1):90–6. doi: 10.1007/s11695-014-1327-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Lopez-Jimenez F, Bhatia S, Collazo-Clavell ML, et al. Safety and efficacy of bariatric surgery in patients with coronary artery disease. Mayo Clin Proc. 2005;80(9):1157–62. doi: 10.4065/80.9.1157. [DOI] [PubMed] [Google Scholar]
  • 101.Afolabi BA, Novaro GM, Szomstein S, et al. Cardiovascular complications of obesity surgery in patients with increased preoperative cardiac risk. Surg Obes Relat Dis. 2009;5(6):653–6. doi: 10.1016/j.soard.2009.06.009. [DOI] [PubMed] [Google Scholar]
  • 102.Delling L, Karason K, Olbers T, Sjöström D, Wahlstrand B, Carlsson B, et al. Feasibility of bariatric surgery as a strategy for secondary prevention in cardiovascular disease: a report from the Swedish Obese Subjects trial. J Obes. 2010;2010 doi: 10.1155/2010/102341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Ramani GV, McCloskey C, Ramanathan RC, et al. Safety and efficacy of bariatric surgery in morbidly obese patients with severe systolic heart failure. Clin Cardiol. 2008;31(11):516–20. doi: 10.1002/clc.20315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Miranda WR, Batsis JA, Sarr MG, et al. Impact of bariatric surgery on quality of life, functional capacity, and symptoms in patients with heart failure. Obes Surg. 2013;23(7):1011–5. doi: 10.1007/s11695-013-0953-8. [DOI] [PubMed] [Google Scholar]
  • 105.Oreopoulos A, Padwal R, Kalantar-Zadeh K, et al. Body mass index and mortality in heart failure: a meta-analysis. Am Heart J. 2008;156(1):13–22. doi: 10.1016/j.ahj.2008.02.014. [DOI] [PubMed] [Google Scholar]
  • 106.Caceres M, Czer LS, Esmailian F, et al. Bariatric surgery in severe obesity and end-stage heart failure with mechanical circulatory support as a bridge to successful heart transplantation: a case report. Transplant Proc. 2013;45(2):798–9. doi: 10.1016/j.transproceed.2012.10.036. [DOI] [PubMed] [Google Scholar]
  • 107.Chaudhry UI, Kanji A, Sai-Sudhakar CB, et al. Laparoscopic sleeve gastrectomy in morbidly obese patients with end-stage heart failure and left ventricular assist device: medium-term results. Surg Obes Relat Dis. 2015;11(1):88–93. doi: 10.1016/j.soard.2014.04.003. [DOI] [PubMed] [Google Scholar]
  • 108.Wikiel KJ, McCloskey CA, Ramanathan RC. Bariatric surgery: a safe and effective conduit to cardiac transplantation. Surg Obes Relat Dis. 2014;10(3):479–84. doi: 10.1016/j.soard.2013.11.002. [DOI] [PubMed] [Google Scholar]
  • 109.Sarkhosh K, Switzer NJ, El-Hadi M, et al. The impact of bariatric surgery on obstructive sleep apnea: a systematic review. Obes Surg. 2013;23(3):414–23. doi: 10.1007/s11695-012-0862-2. [DOI] [PubMed] [Google Scholar]
  • 110.Tishler PV, Larkin EK, Schluchter MD, et al. Incidence of sleep-disordered breathing in an urban adult population: the relative importance of risk factors in the development of sleep-disordered breathing. JAMA. 2003;289(17):2230–7. doi: 10.1001/jama.289.17.2230. [DOI] [PubMed] [Google Scholar]
  • 111.Pannain S, Mokhlesi B. Bariatric surgery and its impact on sleep architecture, sleep-disordered breathing, and metabolism. Best Pract Res Clin Endocrinol Metab. 2010;24(5):745–61. doi: 10.1016/j.beem.2010.07.007. [DOI] [PubMed] [Google Scholar]
  • 112.Drager LF, Togeiro SM, Polotsky VY, et al. Obstructive sleep apnea: a cardiometabolic risk in obesity and the metabolic syndrome. J Am Coll Cardiol. 2013;62(7):569–76. doi: 10.1016/j.jacc.2013.05.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Dorkova Z, Petrasova D, Molcanyiova A, et al. Effects of continuous positive airway pressure on cardiovascular risk profile in patients with severe obstructive sleep apnea and metabolic syndrome. Chest. 2008;134(4):686–92. doi: 10.1378/chest.08-0556. [DOI] [PubMed] [Google Scholar]
  • 114.Sharma SK, Agrawal S, Damodaran D, et al. CPAP for the metabolic syndrome in patients with obstructive sleep apnea. N Engl J Med. 2011;365(24):2277–86. doi: 10.1056/NEJMoa1103944. [DOI] [PubMed] [Google Scholar]
  • 115.Dixon JB, Schachter LM, O’Brien PE, et al. Surgical vs conventional therapy for weight loss treatment of obstructive sleep apnea: a randomized controlled trial. JAMA. 2012;308(11):1142–9. doi: 10.1001/2012.jama.11580. [DOI] [PubMed] [Google Scholar]
  • 116.Gupta RM, Parvizi J, Hanssen AD, et al. Postoperative complications in patients with obstructive sleep apnea syndrome undergoing hip or knee replacement: a case-control study. Mayo Clin Proc. 2001;76(9):897–905. doi: 10.4065/76.9.897. [DOI] [PubMed] [Google Scholar]
  • 117.Hwang D, Shakir N, Limann B, et al. Association of sleep-disordered breathing with postoperative complications. Chest. 2008;133(5):1128–34. doi: 10.1378/chest.07-1488. [DOI] [PubMed] [Google Scholar]
  • 118.Ferreyra GP, Baussano I, Squadrone V, et al. Continuous positive airway pressure for treatment of respiratory complications after abdominal surgery: a systematic review and meta-analysis. Ann Surg. 2008;247(4):617–26. doi: 10.1097/SLA.0b013e3181675829. [DOI] [PubMed] [Google Scholar]
  • 119.Khan A, King WC, Patterson EJ, et al. Assessment of obstructive sleep apnea in adults undergoing bariatric surgery in the longitudinal assessment of bariatric surgery-2 (LABS-2) study. J Clin Sleep Med. 2013;9(1):21–9. doi: 10.5664/jcsm.2332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.O’Keeffe T, Patterson EJ. Evidence supporting routine polysomnography before bariatric surgery. Obes Surg. 2004;14(1):23–6. doi: 10.1381/096089204772787248. [DOI] [PubMed] [Google Scholar]
  • 121.Sharkey KM, Orff HJ, Tosi C, et al. Subjective sleepiness and daytime functioning in bariatric patients with obstructive sleep apnea. Sleep Breath. 2013;17(1):267–74. doi: 10.1007/s11325-012-0685-3. [DOI] [PubMed] [Google Scholar]
  • 122.Gasa M, Salord N, Fortuna AM, et al. Optimizing screening of severe obstructive sleep apnea in patients undergoing bariatric surgery. Surg Obes Relat Dis. 2013;9(4):539–46. doi: 10.1016/j.soard.2012.01.020. [DOI] [PubMed] [Google Scholar]
  • 123.Dixon JB, Schachter LM, O’Brien PE. Predicting sleep apnea and excessive day sleepiness in the severely obese: indicators for polysomnography. Chest. 2003;123(4):1134–41. doi: 10.1378/chest.123.4.1134. [DOI] [PubMed] [Google Scholar]
  • 124.Sareli AE, Cantor CR, Williams NN, et al. Obstructive sleep apnea in patients undergoing bariatric surgery—a tertiary center experience. Obes Surg. 2011;21(3):316–27. doi: 10.1007/s11695-009-9928-1. [DOI] [PubMed] [Google Scholar]
  • 125.Beuther DA, Sutherland ER. Overweight, obesity, and incident asthma: a meta-analysis of prospective epidemiologic studies. Am J Respir Crit Care Med. 2007;175(7):661–6. doi: 10.1164/rccm.200611-1717OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Juel CT, Ali Z, Nilas L, et al. Asthma and obesity: does weight loss improve asthma control? a systematic review. J Asthma Allergy. 2012;5:21–6. doi: 10.2147/JAA.S32232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Macgregor AM, Greenberg RA. Effect of surgically induced weight loss on asthma in the morbidly obese. Obes Surg. 1993;3(1):15–21. doi: 10.1381/096089293765559700. [DOI] [PubMed] [Google Scholar]
  • 128.Dixon JB, Chapman L, O’Brien P. Marked improvement in asthma after Lap-Band surgery for morbid obesity. Obes Surg. 1999;9(4):385–9. doi: 10.1381/096089299765552981. [DOI] [PubMed] [Google Scholar]
  • 129.Dávila-Cervantes A, Domínguez-Cherit G, Borunda D, et al. Impact of surgically-induced weight loss on respiratory function: a prospective analysis. Obes Surg. 2004;14(10):1389–92. doi: 10.1381/0960892042583996. [DOI] [PubMed] [Google Scholar]
  • 130.Spivak H, Hewitt MF, Onn A, et al. Weight loss and improvement of obesity-related illness in 500 U.S. patients following laparoscopic adjustable gastric banding procedure. Am J Surg. 2005;189(1):27–32. doi: 10.1016/j.amjsurg.2004.06.038. [DOI] [PubMed] [Google Scholar]
  • 131.Dhabuwala A, Cannan RJ, Stubbs RS. Improvement in co-morbidities following weight loss from gastric bypass surgery. Obes Surg. 2000;10(5):428–35. doi: 10.1381/096089200321594291. [DOI] [PubMed] [Google Scholar]
  • 132.Dixon AE, Pratley RE, Forgione PM, et al. Effects of obesity and bariatric surgery on airway hyperresponsiveness, asthma control, and inflammation. J Allergy Clin Immunol. 2011;128(3):508–15. doi: 10.1016/j.jaci.2011.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.van Huisstede A, Rudolphus A, Castro Cabezas M, et al. Effect of bariatric surgery on asthma control, lung function and bronchial and systemic inflammation in morbidly obese subjects with asthma. Thorax. 2015;70(7):659–67. doi: 10.1136/thoraxjnl-2014-206712. [DOI] [PubMed] [Google Scholar]
  • 134.Dandona P, Ghanim H, Monte SV, et al. Increase in the mediators of asthma in obesity and obesity with type 2 diabetes: reduction with weight loss. Obesity (Silver Spring) 2014;22(2):356–62. doi: 10.1002/oby.20524. [DOI] [PubMed] [Google Scholar]
  • 135.Blagojevic M, Jinks C, Jeffery A, et al. Risk factors for onset of osteoarthritis of the knee in older adults: a systematic review and meta-analysis. Osteoarthritis Cartilage. 2010;18(1):24–33. doi: 10.1016/j.joca.2009.08.010. [DOI] [PubMed] [Google Scholar]
  • 136.Grotle M, Hagen KB, Natvig B, et al. Obesity and osteoarthritis in knee, hip and/or hand: an epidemiological study in the general population with 10 years follow-up. BMC Musculoskelet Disord. 2008;9:132. doi: 10.1186/1471-2474-9-132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Cooper C, Inskip H, Croft P, et al. Individual risk factors for hip osteoarthritis: obesity, hip injury, and physical activity. Am J Epidemiol. 1998;147(6):516–22. doi: 10.1093/oxfordjournals.aje.a009482. [DOI] [PubMed] [Google Scholar]
  • 138.Ackerman IN, Osborne RH. Obesity and increased burden of hip and knee joint disease in Australia: results from a national survey. BMC Musculoskelet Disord. 2012;13:254. doi: 10.1186/1471-2474-13-254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Christensen R, Bartels EM, Astrup A, et al. Effect of weight reduction in obese patients diagnosed with knee osteoarthritis: a systematic review and meta-analysis. Ann Rheum Dis. 2007;66(4):433–9. doi: 10.1136/ard.2006.065904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Bordini B, Stea S, Cremonini S, et al. Relationship between obesity and early failure of total knee prostheses. BMC Musculoskelet Disord. 2009;10:29. doi: 10.1186/1471-2474-10-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kerkhoffs GM, Servien E, Dunn W, et al. The influence of obesity on the complication rate and outcome of total knee arthroplasty: a meta-analysis and systematic literature review. J Bone Joint Surg Am. 2012;94(20):1839–44. doi: 10.2106/JBJS.K.00820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Severson EP, Singh JA, Browne JA, et al. Total knee arthroplasty in morbidly obese patients treated with bariatric surgery: a comparative study. J Arthroplasty. 2012;27(9):1696–700. doi: 10.1016/j.arth.2012.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Parvizi J, Trousdale RT, Sarr MG. Total joint arthroplasty in patients surgically treated for morbid obesity. J Arthroplasty. 2000;15(8):1003–8. doi: 10.1054/arth.2000.9054. [DOI] [PubMed] [Google Scholar]
  • 144.Inacio MC, Paxton EW, Fisher D, et al. Bariatric surgery prior to total joint arthroplasty may not provide dramatic improvements in post-arthroplasty surgical outcomes. J Arthroplasty. 2014;29(7):1359–64. doi: 10.1016/j.arth.2014.02.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Corley DA, Kubo A. Body mass index and gastroesophageal reflux disease: a systematic review and meta-analysis. Am J Gastroenterol. 2006;101(11):2619–28. doi: 10.1111/j.1572-0241.2006.00849.x. [DOI] [PubMed] [Google Scholar]
  • 146.Eslick GD. Gastrointestinal symptoms and obesity: a meta-analysis. Obes Rev. 2012;13(5):469–79. doi: 10.1111/j.1467-789X.2011.00969.x. [DOI] [PubMed] [Google Scholar]
  • 147.El-Serag H. The association between obesity and GERD: a review of the epidemiological evidence. Dig Dis Sci. 2008;53(9):2307–12. doi: 10.1007/s10620-008-0413-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Cai N, Ji GZ, Fan ZN, et al. Association between body mass index and erosive esophagitis: a meta-analysis. World J Gastroenterol. 2012;18(20):2545–53. doi: 10.3748/wjg.v18.i20.2545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hampel H, Abraham NS, El-Serag HB. Meta-analysis: obesity and the risk for gastroesophageal reflux disease and its complications. Ann Intern Med. 2005;143(3):199–211. doi: 10.7326/0003-4819-143-3-200508020-00006. [DOI] [PubMed] [Google Scholar]
  • 150.Kubo A, Corley DA. Body mass index and adenocarcinomas of the esophagus or gastric cardia: a systematic review and meta-analysis. Cancer Epidemiol Biomarkers Prev. 2006;15(5):872–8. doi: 10.1158/1055-9965.EPI-05-0860. [DOI] [PubMed] [Google Scholar]
  • 151.Singh S, Sharma AN, Murad MH, et al. Central adiposity is associated with increased risk of esophageal inflammation, metaplasia, and adenocarcinoma: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2013;11(11):1399–412.e7. doi: 10.1016/j.cgh.2013.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Friedenberg FK, Xanthopoulos M, Foster GD, et al. The association between gastroesophageal reflux disease and obesity. Am J Gastroenterol. 2008;103(8):2111–22. doi: 10.1111/j.1572-0241.2008.01946.x. [DOI] [PubMed] [Google Scholar]
  • 153.Ness-Jensen E, Lindam A, Lagergren J, et al. Weight loss and reduction in gastroesophageal reflux. A prospective population-based cohort study: the HUNT study. Am J Gastroenterol. 2013;108(3):376–82. doi: 10.1038/ajg.2012.466. [DOI] [PubMed] [Google Scholar]
  • 154.Singh M, Lee J, Gupta N, et al. Weight loss can lead to resolution of gastroesophageal reflux disease symptoms: a prospective intervention trial. Obesity (Silver Spring) 2013;21(2):284–90. doi: 10.1002/oby.20279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Patterson EJ, Davis DG, Khajanchee Y, et al. Comparison of objective outcomes following laparoscopic Nissen fundoplication versus laparoscopic gastric bypass in the morbidly obese with heartburn. Surg Endosc. 2003;17(10):1561–5. doi: 10.1007/s00464-002-8955-y. [DOI] [PubMed] [Google Scholar]
  • 156.Varela JE, Hinojosa MW, Nguyen NT. Laparoscopic fundoplication compared with laparoscopic gastric bypass in morbidly obese patients with gastroesophageal reflux disease. Surg Obes Relat Dis. 2009;5(2):139–43. doi: 10.1016/j.soard.2008.08.021. [DOI] [PubMed] [Google Scholar]
  • 157.Prachand VN, Alverdy JC. Gastroesophageal reflux disease and severe obesity: fundoplication or bariatric surgery? World J Gastroenterol. 2010;16(30):3757–61. doi: 10.3748/wjg.v16.i30.3757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Ikramuddin S. Surgical management of gastroesophageal reflux disease in obesity. Dig Dis Sci. 2008;53(9):2318–29. doi: 10.1007/s10620-008-0415-7. [DOI] [PubMed] [Google Scholar]
  • 159.Pallati PK, Shaligram A, Shostrom VK, et al. Improvement in gastroesophageal reflux disease symptoms after various bariatric procedures: review of the Bariatric Outcomes Longitudinal Database. Surg Obes Relat Dis. 2014;10(3):502–7. doi: 10.1016/j.soard.2013.07.018. [DOI] [PubMed] [Google Scholar]
  • 160.Nelson LG, Gonzalez R, Haines K, et al. Amelioration of gastroesophageal reflux symptoms following Roux-en-Y gastric bypass for clinically significant obesity. Am Surg. 2005;71(11):950–3. [PubMed] [Google Scholar]
  • 161.Tai CM, Lee YC, Wu MS, et al. The effect of Roux-en-Y gastric bypass on gastroesophageal reflux disease in morbidly obese Chinese patients. Obes Surg. 2009;19(5):565–70. doi: 10.1007/s11695-008-9731-4. [DOI] [PubMed] [Google Scholar]
  • 162.Madalosso CA, Gurski RR, Callegari-Jacques SM, et al. The impact of gastric bypass on gastroesophageal reflux disease in patients with morbid obesity: a prospective study based on the Montreal Consensus. Ann Surg. 2010;251(2):244–8. doi: 10.1097/SLA.0b013e3181bdff20. [DOI] [PubMed] [Google Scholar]
  • 163.Houghton SG, Romero Y, Sarr MG. Effect of Roux-en-Y gastric bypass in obese patients with Barrett’s esophagus: attempts to eliminate duodenogastric reflux. Surg Obes Relat Dis. 2008;4(1):1–4. doi: 10.1016/j.soard.2007.10.003. [DOI] [PubMed] [Google Scholar]
  • 164.Csendes A, Burgos AM, Smok G, et al. Effect of gastric bypass on Barrett’s esophagus and intestinal metaplasia of the cardia in patients with morbid obesity. J Gastrointest Surg. 2006;10(2):259–64. doi: 10.1016/j.gassur.2005.06.006. [DOI] [PubMed] [Google Scholar]
  • 165.Peterli R, Borbély Y, Kern B, et al. Early results of the Swiss Multicentre Bypass or Sleeve Study (SM-BOSS): a prospective randomized trial comparing laparoscopic sleeve gastrectomy and Roux-en-Y gastric bypass. Ann Surg. 2013;258(5):690–4. doi: 10.1097/SLA.0b013e3182a67426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Stefanidis D, Navarro F, Augenstein VA, et al. Laparoscopic fundoplication takedown with conversion to Roux-en-Y gastric bypass leads to excellent reflux control and quality of life after fundoplication failure. Surg Endosc. 2012;26(12):3521–7. doi: 10.1007/s00464-012-2380-7. [DOI] [PubMed] [Google Scholar]
  • 167.Kim M, Navarro F, Eruchalu CN, et al. Minimally invasive Roux-en-Y gastric bypass for fundoplication failure offers excellent gastroesophageal reflux control. Am Surg. 2014;80(7):696–703. [PubMed] [Google Scholar]
  • 168.de Jong JR, Besselink MG, van Ramshorst B, et al. Effects of adjustable gastric banding on gastroesophageal reflux and esophageal motility: a systematic review. Obes Rev. 2010;11(4):297–305. doi: 10.1111/j.1467-789X.2009.00622.x. [DOI] [PubMed] [Google Scholar]
  • 169.Klaus A, Gruber I, Wetscher G, et al. Prevalent esophageal body motility disorders underlie aggravation of GERD symptoms in morbidly obese patients following adjustable gastric banding. Arch Surg. 2006;141(3):247–51. doi: 10.1001/archsurg.141.3.247. [DOI] [PubMed] [Google Scholar]
  • 170.Gagner M, Deitel M, Erickson AL, et al. Survey on laparoscopic sleeve gastrectomy (LSG) at the Fourth International Consensus Summit on Sleeve Gastrectomy. Obes Surg. 2013;23(12):2013–7. doi: 10.1007/s11695-013-1040-x. [DOI] [PubMed] [Google Scholar]
  • 171.Laffin M, Chau J, Gill RS, et al. Sleeve gastrectomy and gastro-esophageal reflux disease. J Obes. 2013;2013:741097. doi: 10.1155/2013/741097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Chiu S, Birch DW, Shi X, et al. Effect of sleeve gastrectomy on gastroesophageal reflux disease: a systematic review. Surg Obes Relat Dis. 2011;7(4):510–5. doi: 10.1016/j.soard.2010.09.011. [DOI] [PubMed] [Google Scholar]
  • 173.Gulkarov I, Wetterau M, Ren CJ, et al. Hiatal hernia repair at the initial laparoscopic adjustable gastric band operation reduces the need for reoperation. Surg Endosc. 2008;22(4):1035–41. doi: 10.1007/s00464-007-9684-z. [DOI] [PubMed] [Google Scholar]
  • 174.Parikh MS, Fielding GA, Ren CJ. U.S. experience with 749 laparoscopic adjustable gastric bands: intermediate outcomes. Surg Endosc. 2005;19(12):1631–5. doi: 10.1007/s00464-005-0302-7. [DOI] [PubMed] [Google Scholar]
  • 175.Vernon G, Baranova A, Younossi ZM. Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther. 2011;34(3):274–85. doi: 10.1111/j.1365-2036.2011.04724.x. [DOI] [PubMed] [Google Scholar]
  • 176.Chavez-Tapia NC, Tellez-Avila FI, Barrientos-Gutierrez T, et al. Bariatric surgery for non-alcoholic steatohepatitis in obese patients. Cochrane Database Syst Rev. 2010(1):CD007340. doi: 10.1002/14651858.CD007340.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Bower G, Toma T, Harling L, Jiao LR, Efthimiou E, Darzi A, et al. Bariatric Surgery and non-alcoholic fatty liver disease: a systematic review of liver biochemistry and histology. Obes Surg. 2015 doi: 10.1007/s11695-015-1691-x. [DOI] [PubMed] [Google Scholar]
  • 178.Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of non-alcoholic fatty liver disease: practice guideline by the American Gastroenterological Association, American Association for the Study of Liver Diseases, and American College of Gastroenterology. Gastroenterology. 2012;142(7):1592–609. doi: 10.1053/j.gastro.2012.04.001. [DOI] [PubMed] [Google Scholar]
  • 179.Rabl C, Campos GM. The impact of bariatric surgery on nonalcoholic steatohepatitis. Semin Liver Dis. 2012;32(1):80–91. doi: 10.1055/s-0032-1306428. [DOI] [PubMed] [Google Scholar]
  • 180.Nagem RG, Lázaro-da-Silva A, de Oliveira RM, et al. Gallstone-related complications after Roux-en-Y gastric bypass: a prospective study. Hepatobiliary Pancreat Dis Int. 2012;11(6):630–5. doi: 10.1016/s1499-3872(12)60236-7. [DOI] [PubMed] [Google Scholar]
  • 181.Scopinaro N, Gianetta E, Civalleri D, et al. Two years of clinical experience with biliopancreatic bypass for obesity. Am J Clin Nutr. 1980;33(2 Suppl):506–14. doi: 10.1093/ajcn/33.2.506. [DOI] [PubMed] [Google Scholar]
  • 182.Tsirline VB, Keilani ZM, El Djouzi S, et al. How frequently and when do patients undergo cholecystectomy after bariatric surgery? Surg Obes Relat Dis. 2014;10(2):313–21. doi: 10.1016/j.soard.2013.10.011. [DOI] [PubMed] [Google Scholar]
  • 183.Sakorafas GH, Milingos D, Peros G. Asymptomatic cholelithiasis: is cholecystectomy really needed? A critical reappraisal 15 years after the introduction of laparoscopic cholecystectomy. Dig Dis Sci. 2007;52(5):1313–25. doi: 10.1007/s10620-006-9107-3. [DOI] [PubMed] [Google Scholar]
  • 184.Worobetz LJ, Inglis FG, Shaffer EA. The effect of ursodeoxycholic acid therapy on gallstone formation in the morbidly obese during rapid weight loss. Am J Gastroenterol. 1993;88(10):1705–10. [PubMed] [Google Scholar]
  • 185.Bardaro SJ, Gagner M, Consten E, et al. Routine cholecystectomy during laparoscopic biliopancreatic diversion with duodenal switch is not necessary. Surg Obes Relat Dis. 2007;3(5):549–53. doi: 10.1016/j.soard.2007.05.007. [DOI] [PubMed] [Google Scholar]
  • 186.Malik S, Mitchell JE, Engel S, et al. Psychopathology in bariatric surgery candidates: a review of studies using structured diagnostic interviews. Compr Psychiatry. 2014;55(2):248–59. doi: 10.1016/j.comppsych.2013.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Lin HY, Huang CK, Tai CM, et al. Psychiatric disorders of patients seeking obesity treatment. BMC Psychiatr. 2013;13:1. doi: 10.1186/1471-244X-13-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Mühlhans B, Horbach T, de Zwaan M. Psychiatric disorders in bariatric surgery candidates: a review of the literature and results of a German prebariatric surgery sample. Gen Hosp Psychiatry. 2009;31(5):414–21. doi: 10.1016/j.genhosppsych.2009.05.004. [DOI] [PubMed] [Google Scholar]
  • 189.Kalarchian MA, Marcus MD, Levine MD, et al. Psychiatric disorders among bariatric surgery candidates: relationship to obesity and functional health status. Am J Psychiatry. 2007;164(2):328–34. doi: 10.1176/ajp.2007.164.2.328. [DOI] [PubMed] [Google Scholar]
  • 190.Mechanick JI, Youdim A, Jones DB, et al. 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. Surg Obes Relat Dis. 2013;9(2):159–91. doi: 10.1016/j.soard.2012.12.010. [DOI] [PubMed] [Google Scholar]
  • 191.Eldar S, Heneghan HM, Brethauer S, et al. A focus on surgical preoperative evaluation of the bariatric patient—the Cleveland Clinic protocol and review of the literature. Surgeon. 2011;9(5):273–7. doi: 10.1016/j.surge.2011.04.002. [DOI] [PubMed] [Google Scholar]
  • 192.Pickering RP, Grant BF, Chou SP, et al. Are overweight, obesity, and extreme obesity associated with psychopathology? Results from the national epidemiologic survey on alcohol and related conditions. J Clin Psychiatry. 2007;68(7):998–1009. doi: 10.4088/jcp.v68n0704. [DOI] [PubMed] [Google Scholar]
  • 193.Mitchell JE, Selzer F, Kalarchian MA, et al. Psychopathology before surgery in the longitudinal assessment of bariatric surgery-3 (LABS-3) psychosocial study. Surg Obes Relat Dis. 2012;8(5):533–41. doi: 10.1016/j.soard.2012.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Livhits M, Mercado C, Yermilov I, et al. Preoperative predictors of weight loss following bariatric surgery: systematic review. Obes Surg. 2012;22(1):70–89. doi: 10.1007/s11695-011-0472-4. [DOI] [PubMed] [Google Scholar]
  • 195.de Zwaan M, Enderle J, Wagner S, et al. Anxiety and depression in bariatric surgery patients: a prospective, follow-up study using structured clinical interviews. J Affect Disord. 2011;133(1-2):61–8. doi: 10.1016/j.jad.2011.03.025. [DOI] [PubMed] [Google Scholar]
  • 196.Legenbauer T, De Zwaan M, Benecke A, et al. Depression and anxiety: their predictive function for weight loss in obese individuals. Obes Facts. 2009;2(4):227–34. doi: 10.1159/000226278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Brunault P, Jacobi D, Miknius V, et al. High preoperative depression, phobic anxiety, and binge eating scores and low medium-term weight loss in sleeve gastrectomy obese patients: a preliminary cohort study. Psychosomatics. 2012;53(4):363–70. doi: 10.1016/j.psym.2011.12.008. [DOI] [PubMed] [Google Scholar]
  • 198.Edwards-Hampton SA, Madan A, Wedin S, et al. A closer look at the nature of anxiety in weight loss surgery candidates. Int J Psychiatry Med. 2014;47(2):105–13. doi: 10.2190/PM.47.2.b. [DOI] [PubMed] [Google Scholar]
  • 199.Singh M. Mood, food, and obesity. Front Psychol. 2014;5:925. doi: 10.3389/fpsyg.2014.00925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Meye FJ, Adan RA. Feelings about food: the ventral tegmental area in food reward and emotional eating. Trends Pharmacol Sci. 2014;35(1):31–40. doi: 10.1016/j.tips.2013.11.003. [DOI] [PubMed] [Google Scholar]
  • 201.Akubuiro A, Bridget Zimmerman M, Boles Ponto LL, et al. Hyperactive hypothalamus, motivated and non-distractible chronic overeating in ADAR2 transgenic mice. Genes Brain Behav. 2013;12(3):311–22. doi: 10.1111/gbb.12020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Hryhorczuk C, Sharma S, Fulton SE. Metabolic disturbances connecting obesity and depression. Front Neurosci. 2013;7:177. doi: 10.3389/fnins.2013.00177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Sharma S, Fulton S. Diet-induced obesity promotes depressive-like behaviour that is associated with neural adaptations in brain reward circuitry. Int J Obes (Lond) 2013;37(3):382–9. doi: 10.1038/ijo.2012.48. [DOI] [PubMed] [Google Scholar]
  • 204.Kok P, Roelfsema F, Frölich M, et al. Activation of dopamine D2 receptors simultaneously ameliorates various metabolic features of obese women. Am J Physiol Endocrinol Metab. 2006;291(5):E1038–43. doi: 10.1152/ajpendo.00567.2005. [DOI] [PubMed] [Google Scholar]
  • 205.Ahmed AT, Warton EM, Schaefer CA, et al. The effect of bariatric surgery on psychiatric course among patients with bipolar disorder. Bipolar Disord. 2013;15(7):753–63. doi: 10.1111/bdi.12109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Yska JP, van der Linde S, Tapper VV, et al. Influence of bariatric surgery on the use and pharmacokinetics of some major drug classes. Obes Surg. 2013;23(6):819–25. doi: 10.1007/s11695-013-0882-6. [DOI] [PubMed] [Google Scholar]
  • 207.Cunningham JL, Merrell CC, Sarr M, et al. Investigation of antidepressant medication usage after bariatric surgery. Obes Surg. 2012;22(4):530–5. doi: 10.1007/s11695-011-0517-8. [DOI] [PubMed] [Google Scholar]
  • 208.Smith A, Henriksen B, Cohen A. Pharmacokinetic considerations in Roux-en-Y gastric bypass patients. Am J Health Syst Pharm. 2011;68(23):2241–7. doi: 10.2146/ajhp100630. [DOI] [PubMed] [Google Scholar]
  • 209.Brietzke E, Lafer B. Long-acting injectable risperidone in a bipolar patient submitted to bariatric surgery and intolerant to conventional mood stabilizers. Psychiatry Clin Neurosci. 2011;65(2):205. doi: 10.1111/j.1440-1819.2010.02175.x. [DOI] [PubMed] [Google Scholar]
  • 210.Tripp AC. Lithium toxicity after Roux-en-Y gastric bypass surgery. J Clin Psychopharmacol. 2011;31(2):261–2. doi: 10.1097/JCP.0b013e318210b203. [DOI] [PubMed] [Google Scholar]
  • 211.Pramyothin P, Khaodhiar L. Metabolic syndrome with the atypical antipsychotics. Curr Opin Endocrinol Diabetes Obes. 2010;17(5):460–6. doi: 10.1097/MED.0b013e32833de61c. [DOI] [PubMed] [Google Scholar]
  • 212.Deeks ED. Risperidone long-acting injection: in bipolar I disorder. Drugs. 2010;70(8):1001–12. doi: 10.2165/11204480-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 213.Bobbioni-Harsch E, Guillermin ML, Habicht F. Rev Med Suisse. 442. Vol. 10. French: 2014. Reciprocal interactions between bariatric surgery and psychopathology; pp. 1721–6. [PubMed] [Google Scholar]
  • 214.Svensson PA, Anveden Å, Romeo S, et al. Alcohol consumption and alcohol problems after bariatric surgery in the Swedish Obese Subjects study. Obesity (Silver Spring) 2013;21(12):2444–51. doi: 10.1002/oby.20397. [DOI] [PubMed] [Google Scholar]
  • 215.Kudsi OY, Huskey K, Grove S, et al. Prevalence of preoperative alcohol abuse among patients seeking weight-loss surgery. Surg Endosc. 2013;27(4):1093–7. doi: 10.1007/s00464-012-2568-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Heinberg LJ, Ashton K, Coughlin J. Alcohol and bariatric surgery: review and suggested recommendations for assessment and management. Surg Obes Relat Dis. 2012;8(3):357–63. doi: 10.1016/j.soard.2012.01.016. [DOI] [PubMed] [Google Scholar]
  • 217.Suzuki J, Haimovici F, Chang G. Alcohol use disorders after bariatric surgery. Obes Surg. 2012;22(2):201–7. doi: 10.1007/s11695-010-0346-1. [DOI] [PubMed] [Google Scholar]
  • 218.Ertelt TW, Mitchell JE, Lancaster K, et al. Alcohol abuse and dependence before and after bariatric surgery: a review of the literature and report of a new data set. Surg Obes Relat Dis. 2008;4(5):647–50. doi: 10.1016/j.soard.2008.01.004. [DOI] [PubMed] [Google Scholar]
  • 219.Saules KK, Wiedemann A, Ivezaj V, et al. Bariatric surgery history among substance abuse treatment patients: prevalence and associated features. Surg Obes Relat Dis. 2010;6(6):615–21. doi: 10.1016/j.soard.2009.12.008. [DOI] [PubMed] [Google Scholar]
  • 220.King WC, Chen JY, Mitchell JE, et al. Prevalence of alcohol use disorders before and after bariatric surgery. JAMA. 2012;307(23):2516–25. doi: 10.1001/jama.2012.6147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Ostlund MP, Backman O, Marsk R, et al. Increased admission for alcohol dependence after gastric bypass surgery compared with restrictive bariatric surgery. JAMA Surg. 2013;148(4):374–7. doi: 10.1001/jamasurg.2013.700. [DOI] [PubMed] [Google Scholar]
  • 222.Polston JE, Pritchett CE, Tomasko JM, et al. Roux-en-Y gastric bypass increases intravenous ethanol self-administration in dietary obese rats. PLoS One. 2013;8(12):e83741. doi: 10.1371/journal.pone.0083741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Maluenda F, Csendes A, De Aretxabala X, et al. Alcohol absorption modification after a laparoscopic sleeve gastrectomy due to obesity. Obes Surg. 2010;20(6):744–8. doi: 10.1007/s11695-010-0136-9. [DOI] [PubMed] [Google Scholar]
  • 224.Thanos PK, Subrize M, Delis F, et al. Gastric bypass increases ethanol and water consumption in diet-induced obese rats. Obes Surg. 2012;22(12):1884–92. doi: 10.1007/s11695-012-0749-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.McCormick LM, Buchanan JR, Onwuameze OE, et al. Beyond alcoholism: Wernicke-Korsakoff syndrome in patients with psychiatric disorders. Cogn Behav Neurol. 2011;24(4):209–16. doi: 10.1097/WNN.0b013e31823f90c4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Ashton K, Heinberg L, Merrell J, et al. Pilot evaluation of a substance abuse prevention group intervention for at-risk bariatric surgery candidates. Surg Obes Relat Dis. 2013;9(3):462–7. doi: 10.1016/j.soard.2013.01.016. [DOI] [PubMed] [Google Scholar]
  • 227.Blum K, Bailey J, Gonzalez AM, Oscar-Berman M, Liu Y, Giordano J, et al. Neurogenetics of reward deficiency syndrome (RDS) as the root cause of “addiction transfer”: a new phenomenon common after bariatric surgery. J Genet Syndr Gene Ther. 2011;2012(1) doi: 10.4172/2157-7412.S2-001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Snyder AG. Psychological assessment of the patient undergoing bariatric surgery. Ochsner J. 2009;9(3):144–8. [PMC free article] [PubMed] [Google Scholar]
  • 229.Friedman KE, Applegate KL, Grant J. Who is adherent with preoperative psychological treatment recommendations among weight loss surgery candidates? Surg Obes Relat Dis. 2007;3(3):376–82. doi: 10.1016/j.soard.2007.01.008. [DOI] [PubMed] [Google Scholar]
  • 230.Parker K, O’Brien P, Brennan L. Measurement of disordered eating following bariatric surgery: a systematic review of the literature. Obes Surg. 2014;24(6):945–53. doi: 10.1007/s11695-014-1248-4. [DOI] [PubMed] [Google Scholar]
  • 231.Morrow J, Gluck M, Lorence M, et al. Night eating status and influence on body weight, body image, hunger, and cortisol pre-and post- Roux-en-Y Gastric Bypass (RYGB) surgery. Eat Weight Disord. 2008;13(4):e96–9. doi: 10.1007/BF03327512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Colles SL, Dixon JB. Night eating syndrome: impact on bariatric surgery. Obes Surg. 2006;16(7):811–20. doi: 10.1381/096089206777822160. [DOI] [PubMed] [Google Scholar]
  • 233.Fierabracci P, Pinchera A, Martinelli S, et al. Prevalence of endocrine diseases in morbidly obese patients scheduled for bariatric surgery: beyond diabetes. Obes Surg. 2011;21(1):54–60. doi: 10.1007/s11695-010-0297-6. [DOI] [PubMed] [Google Scholar]
  • 234.Poirier P, Alpert MA, Fleisher LA, et al. Cardiovascular evaluation and management of severely obese patients undergoing surgery: a science advisory from the American Heart Association. Circulation. 2009;120(1):86–95. doi: 10.1161/CIRCULATIONAHA.109.192575. [DOI] [PubMed] [Google Scholar]
  • 235.Stocker DJ. Management of the bariatric surgery patient. Endocrinol Metab Clin North Am. 2003;32(2):437–57. doi: 10.1016/s0889-8529(03)00002-1. [DOI] [PubMed] [Google Scholar]
  • 236.DeMaria EJ. Bariatric surgery for morbid obesity. N Engl J Med. 2007;356(21):2176–83. doi: 10.1056/NEJMct067019. [DOI] [PubMed] [Google Scholar]
  • 237.Ness-Abramof R, Nabriski D, Apovian CM, et al. Overnight dexamethasone suppression test: a reliable screen for Cushing’s syndrome in the obese. Obes Res. 2002;10(12):1217–21. doi: 10.1038/oby.2002.166. [DOI] [PubMed] [Google Scholar]
  • 238.Collazo-Clavell ML, Clark MM, McAlpine DE, et al. Assessment and preparation of patients for bariatric surgery. Mayo Clin Proc. 2006;81(10 Suppl):S11–7. doi: 10.1016/s0025-6196(11)61176-2. [DOI] [PubMed] [Google Scholar]
  • 239.Manco M, Nanni G, Tondolo V, et al. Hypocalcemia complicating near-total thyroidectomy in patients with coexisting lipid malab-sorption due to biliopancreatic diversion. Obes Surg. 2004;14(10):1429–34. doi: 10.1381/0960892042583752. [DOI] [PubMed] [Google Scholar]
  • 240.Pietras SM, Holick MF. Refractory hypocalcemia following near-total thyroidectomy in a patient with a prior Roux-en-Y gastric bypass. Obes Surg. 2009;19(4):524–6. doi: 10.1007/s11695-008-9614-8. [DOI] [PubMed] [Google Scholar]
  • 241.Shah M, Simha V, Garg A. Review: long-term impact of bariatric surgery on body weight, comorbidities, and nutritional status. J Clin Endocrinol Metab. 2006;91(11):4223–31. doi: 10.1210/jc.2006-0557. [DOI] [PubMed] [Google Scholar]
  • 242.Heber D, Greenway FL, Kaplan LM, et al. Endocrine and nutritional management of the post-bariatric surgery patient: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2010;95(11):4823–43. doi: 10.1210/jc.2009-2128. [DOI] [PubMed] [Google Scholar]
  • 243.Slater GH, Ren CJ, Siegel N, et al. Serum fat-soluble vitamin deficiency and abnormal calcium metabolism after malabsorptive bariatric surgery. J Gastrointest Surg. 2004;8(1):48–55. doi: 10.1016/j.gassur.2003.09.020. [DOI] [PubMed] [Google Scholar]
  • 244.Rojas-Marcos PM, Rubio MA, Kreskshi WI, et al. Severe hypo-calcemia following total thyroidectomy after biliopancreatic diversion. Obes Surg. 2005;15(3):431–4. doi: 10.1381/0960892053576712. [DOI] [PubMed] [Google Scholar]
  • 245.Dalbeth N, Chen P, White M, et al. Impact of bariatric surgery on serum urate targets in people with morbid obesity and diabetes: a prospective longitudinal study. Ann Rheum Dis. 2014;73(5):797–802. doi: 10.1136/annrheumdis-2013-203970. [DOI] [PubMed] [Google Scholar]
  • 246.Romero-Talamás H, Daigle CR, Aminian A, et al. The effect of bariatric surgery on gout: a comparative study. Surg Obes Relat Dis. 2014;10(6):1161–5. doi: 10.1016/j.soard.2014.02.025. [DOI] [PubMed] [Google Scholar]
  • 247.Bergendal A, Bremme K, Hedenmalm K, et al. Risk factors for venous thromboembolism in pre-and postmenopausal women. Thromb Res. 2012;130(4):596–601. doi: 10.1016/j.thromres.2012.05.024. [DOI] [PubMed] [Google Scholar]
  • 248.Laliberté F, Dea K, Duh MS, et al. Does the route of administration for estrogen hormone therapy impact the risk of venous thromboembolism? Estradiol transdermal system versus oral estrogen-only hormone therapy. Menopause. 2011;18(10):1052–9. doi: 10.1097/gme.0b013e3182175e5c. [DOI] [PubMed] [Google Scholar]
  • 249.Canonico M, Oger E, Conard J, et al. Obesity and risk of venous thromboembolism among postmenopausal women: differential impact of hormone therapy by route of estrogen administration. ESTHER Study J Thromb Haemost. 2006;4(6):1259–65. doi: 10.1111/j.1538-7836.2006.01933.x. [DOI] [PubMed] [Google Scholar]
  • 250.Pandey S, Pandey S, Maheshwari A, et al. The impact of female obesity on the outcome of fertility treatment. J Hum Reprod Sci. 2010;3(2):62–7. doi: 10.4103/0974-1208.69332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Rich-Edwards JW, Goldman MB, Willett WC, et al. Adolescent body mass index and infertility caused by ovulatory disorder. Am J Obstet Gynecol. 1994;171(1):171–7. doi: 10.1016/0002-9378(94)90465-0. [DOI] [PubMed] [Google Scholar]
  • 252.van der Steeg JW, Steures P, Eijkemans MJ, et al. Obesity affects spontaneous pregnancy chances in subfertile, ovulatory women. Hum Reprod. 2008;23(2):324–8. doi: 10.1093/humrep/dem371. [DOI] [PubMed] [Google Scholar]
  • 253.Gil-Campos M, Cañete RR, Gil A. Adiponectin, the missing link in insulin resistance and obesity. Clin Nutr. 2004;23(5):963–74. doi: 10.1016/j.clnu.2004.04.010. [DOI] [PubMed] [Google Scholar]
  • 254.Motta AB. The role of obesity in the development of polycystic ovary syndrome. Curr Pharm Des. 2012;18(17):2482–91. doi: 10.2174/13816128112092482. [DOI] [PubMed] [Google Scholar]
  • 255.Baeten JM, Bukusi EA, Lambe M. Pregnancy complications and outcomes among overweight and obese nulliparous women. Am J Public Health. 2001;91(3):436–40. doi: 10.2105/ajph.91.3.436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Cedergren MI. Maternal morbid obesity and the risk of adverse pregnancy outcome. Obstet Gynecol. 2004;103(2):219–24. doi: 10.1097/01.AOG.0000107291.46159.00. [DOI] [PubMed] [Google Scholar]
  • 257.Sebire NJ, Jolly M, Harris JP, et al. Maternal obesity and pregnancy outcome: a study of 287,213 pregnancies in London. Int J Obes Relat Metab Disord. 2001;25(8):1175–82. doi: 10.1038/sj.ijo.0801670. [DOI] [PubMed] [Google Scholar]
  • 258.Weiss JL, Malone FD, Emig D, et al. Obesity, obstetric complications and cesarean delivery rate—a population-based screening study. Am J Obstet Gynecol. 2004;190(4):1091–7. doi: 10.1016/j.ajog.2003.09.058. [DOI] [PubMed] [Google Scholar]
  • 259.Stothard KJ, Tennant PW, Bell R, et al. Maternal overweight and obesity and the risk of congenital anomalies: a systematic review and meta-analysis. JAMA. 2009;301(6):636–50. doi: 10.1001/jama.2009.113. [DOI] [PubMed] [Google Scholar]
  • 260.Oken E, Taveras EM, Kleinman KP, et al. Gestational weight gain and child adiposity at age 3 years. Am J Obstet Gynecol. 2007;196(4):322.e1–8. doi: 10.1016/j.ajog.2006.11.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.American College of Obstetricians and Gynecologists. ACOG committee opinion no. 549: obesity in pregnancy. Obstet Gynecol. 2013;121(1):213–7. doi: 10.1097/01.aog.0000425667.10377.60. [DOI] [PubMed] [Google Scholar]
  • 262.Sheiner E, Levy A, Silverberg D, et al. Pregnancy after bariatric surgery is not associated with adverse perinatal outcome. Am J Obstet Gynecol. 2004;190(5):1335–40. doi: 10.1016/j.ajog.2003.11.004. [DOI] [PubMed] [Google Scholar]
  • 263.Martin LF, Finigan KM, Nolan TE. Pregnancy after adjustable gastric banding. Obstet Gynecol. 2000;95(6 Pt 1):927–30. doi: 10.1016/s0029-7844(00)00819-x. [DOI] [PubMed] [Google Scholar]
  • 264.McTiernan A. Obesity and cancer: the risks, science, and potential management strategies. Oncology (Williston Park) 2005;19(7):871–81. [PubMed] [Google Scholar]
  • 265.Anderson AS, Caswell S. Obesity management—an opportunity for cancer prevention. Surgeon. 2009;7(5):282–5. doi: 10.1016/s1479-666x(09)80005-x. [DOI] [PubMed] [Google Scholar]
  • 266.Bianchini F, Kaaks R, Vainio H. Overweight, obesity, and cancer risk. Lancet Oncol. 2002;3(9):565–74. doi: 10.1016/s1470-2045(02)00849-5. [DOI] [PubMed] [Google Scholar]
  • 267.Calle EE, Rodriguez C, Walker-Thurmond K, et al. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med. 2003;348(17):1625–38. doi: 10.1056/NEJMoa021423. [DOI] [PubMed] [Google Scholar]
  • 268.Tee MC, Cao Y, Warnock GL, et al. Effect of bariatric surgery on oncologic outcomes: a systematic review and meta-analysis. Surg Endosc. 2013;27(12):4449–56. doi: 10.1007/s00464-013-3127-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Casagrande DS, Rosa DD, Umpierre D, et al. Incidence of cancer following bariatric surgery: systematic review and meta-analysis. Obes Surg. 2014;24(9):1499–509. doi: 10.1007/s11695-014-1276-0. [DOI] [PubMed] [Google Scholar]
  • 270.Afshar S, Kelly SB, Seymour K, et al. The effects of bariatric surgery on colorectal cancer risk: systematic review and meta-analysis. Obes Surg. 2014;24(10):1793–9. doi: 10.1007/s11695-014-1359-y. [DOI] [PubMed] [Google Scholar]
  • 271.Reeves GK, Pirie K, Beral V, et al. Cancer incidence and mortality in relation to body mass index in the Million Women Study: cohort study. BMJ. 2007;335(7630):1134. doi: 10.1136/bmj.39367.495995.AE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Renehan AG, Tyson M, Egger M, et al. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet. 2008;371(9612):569–78. doi: 10.1016/S0140-6736(08)60269-X. [DOI] [PubMed] [Google Scholar]
  • 273.Renehan AG, Soerjomataram I, Tyson M, et al. Incident cancer burden attributable to excess body mass index in 30 European countries. Int J Cancer. 2010;126(3):692–702. doi: 10.1002/ijc.24803. [DOI] [PubMed] [Google Scholar]
  • 274.Eliassen AH, Colditz GA, Rosner B, et al. Adult weight change and risk of postmenopausal breast cancer. JAMA. 2006;296(2):193–201. doi: 10.1001/jama.296.2.193. [DOI] [PubMed] [Google Scholar]
  • 275.Lahmann PH, Lissner L, Gullberg B, et al. A prospective study of adiposity and postmenopausal breast cancer risk: the Malmö Diet and Cancer Study. Int J Cancer. 2003;103(2):246–52. doi: 10.1002/ijc.10799. [DOI] [PubMed] [Google Scholar]
  • 276.Chang SH, Pollack LM, Colditz GA. Obesity, mortality, and life years lost associated with breast cancer in nonsmoking US Women, National Health Interview Survey, 1997–2000. Prev Chronic Dis. 2013;10:E186. doi: 10.5888/pcd10.130112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Minicozzi P, Berrino F, Sebastiani F, et al. High fasting blood glucose and obesity significantly and independently increase risk of breast cancer death in hormone receptor-positive disease. Eur J Cancer. 2013;49(18):3881–8. doi: 10.1016/j.ejca.2013.08.004. [DOI] [PubMed] [Google Scholar]
  • 278.Petrelli JM, Calle EE, Rodriguez C, et al. Body mass index, height, and postmenopausal breast cancer mortality in a prospective cohort of US women. Cancer Causes Control. 2002;13(4):325–32. doi: 10.1023/a:1015288615472. [DOI] [PubMed] [Google Scholar]
  • 279.Howell A, Sims AH, Ong KR, et al. Mechanisms of disease: prediction and prevention of breast cancer—cellular and molecular interactions. Nat Clin Pract Oncol. 2005;2(12):635–46. doi: 10.1038/ncponc0361. [DOI] [PubMed] [Google Scholar]
  • 280.Harvie M, Howell A, Vierkant RA, et al. Association of gain and loss of weight before and after menopause with risk of postmenopausal breast cancer in the Iowa women’s health study. Cancer Epidemiol Biomarkers Prev. 2005;14(3):656–61. doi: 10.1158/1055-9965.EPI-04-0001. [DOI] [PubMed] [Google Scholar]
  • 281.Havrilesky LJ, Maxwell GL, Myers ER. Cost-effectiveness analysis of annual screening strategies for endometrial cancer. Am J Obstet Gynecol. 2009;200(6):640.e1–8. doi: 10.1016/j.ajog.2009.02.022. [DOI] [PubMed] [Google Scholar]
  • 282.Kwon JS, Lu KH. Cost-effectiveness analysis of endometrial cancer prevention strategies for obese women. Obstet Gynecol. 2008;112(1):56–63. doi: 10.1097/AOG.0b013e31817d53a4. [DOI] [PubMed] [Google Scholar]
  • 283.Rose PG. Endometrial carcinoma. N Engl J Med. 1996;335(9):640–9. doi: 10.1056/NEJM199608293350907. [DOI] [PubMed] [Google Scholar]
  • 284.von Gruenigen VE, Tian C, Frasure H, et al. Treatment effects, disease recurrence, and survival in obese women with early endometrial carcinoma: a Gynecologic Oncology Group study. Cancer. 2006;107(12):2786–91. doi: 10.1002/cncr.22351. [DOI] [PubMed] [Google Scholar]
  • 285.Arem H, Park Y, Pelser C, et al. Prediagnosis body mass index, physical activity, and mortality in endometrial cancer patients. J Natl Cancer Inst. 2013;105(5):342–9. doi: 10.1093/jnci/djs530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Kaaks R, Lukanova A, Kurzer MS. Obesity, endogenous hormones, and endometrial cancer risk: a synthetic review. Cancer Epidemiol Biomarkers Prev. 2002;11(12):1531–43. [PubMed] [Google Scholar]
  • 287.McCawley GM, Ferriss JS, Geffel D, et al. Cancer in obese women: potential protective impact of bariatric surgery. J Am Coll Surg. 2009;208(6):1093–8. doi: 10.1016/j.jamcollsurg.2009.01.045. [DOI] [PubMed] [Google Scholar]
  • 288.Adams TD, Stroup AM, Gress RE, et al. Cancer incidence and mortality after gastric bypass surgery. Obesity (Silver Spring) 2009;17(4):796–802. doi: 10.1038/oby.2008.610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Ward KK, Roncancio AM, Shah NR, et al. Bariatric surgery decreases the risk of uterine malignancy. Gynecol Oncol. 2014;133(1):63–6. doi: 10.1016/j.ygyno.2013.11.012. [DOI] [PubMed] [Google Scholar]
  • 290.Gagné DJ, Papasavas PK, Maalouf M, et al. Obesity surgery and malignancy: our experience after 1500 cases. Surg Obes Relat Dis. 2009;5(2):160–4. doi: 10.1016/j.soard.2008.07.013. [DOI] [PubMed] [Google Scholar]
  • 291.Potluri K, Hou S. Obesity in kidney transplant recipients and candidates. Am J Kidney Dis. 2010;56(1):143–56. doi: 10.1053/j.ajkd.2010.01.017. [DOI] [PubMed] [Google Scholar]
  • 292.Charlton M. Nonalcoholic fatty liver disease: a review of current understanding and future impact. Clin Gastroenterol Hepatol. 2004;2(12):1048–58. doi: 10.1016/s1542-3565(04)00440-9. [DOI] [PubMed] [Google Scholar]
  • 293.Kemmer N, Neff GW, Franco E, et al. Nonalcoholic fatty liver disease epidemic and its implications for liver transplantation. Transplantation. 2013;96(10):860–2. doi: 10.1097/01.TP.0000436723.59879.01. [DOI] [PubMed] [Google Scholar]
  • 294.Leonard J, Heimbach JK, Malinchoc M, et al. The impact of obesity on long-term outcomes in liver transplant recipients—results of the NIDDK liver transplant database. Am J Transplant. 2008;8(3):667–72. doi: 10.1111/j.1600-6143.2007.02100.x. [DOI] [PubMed] [Google Scholar]
  • 295.Conzen KD, Vachharajani N, Collins KM, et al. Morbid obesity in liver transplant recipients adversely affects long-term graft and patient survival in a single-institution analysis. HPB (Oxford) 2015;17(3):251–7. doi: 10.1111/hpb.12340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Hakeem AR, Cockbain AJ, Raza SS, et al. Increased morbidity in overweight and obese liver transplant recipients: a single-center experience of 1325 patients from the United Kingdom. Liver Transpl. 2013;19(5):551–62. doi: 10.1002/lt.23618. [DOI] [PubMed] [Google Scholar]
  • 297.Pieloch D, Dombrovskiy V, Osband AJ, et al. Morbid obesity is not an independent predictor of graft failure or patient mortality after kidney transplantation. J Ren Nutr. 2014;24(1):50–7. doi: 10.1053/j.jrn.2013.07.001. [DOI] [PubMed] [Google Scholar]
  • 298.Curran SP, Famure O, Li Y, Kim SJ. Increased recipient body mass index is associated with acute rejection and other adverse outcomes after kidney transplantation. Transplantation. 2014;97(1):64–70. doi: 10.1097/TP.0b013e3182a688a4. [DOI] [PubMed] [Google Scholar]
  • 299.McCloskey CA, Ramani GV, Mathier MA, et al. Bariatric surgery improves cardiac function in morbidly obese patients with severe cardiomyopathy. Surg Obes Relat Dis. 2007;3(5):503–7. doi: 10.1016/j.soard.2007.05.006. [DOI] [PubMed] [Google Scholar]
  • 300.Newcombe V, Blanch A, Slater GH, et al. Laparoscopic adjustable gastric banding prior to renal transplantation. Obes Surg. 2005;15(4):567–70. doi: 10.1381/0960892053723349. [DOI] [PubMed] [Google Scholar]
  • 301.Campsen J, Zimmerman M, Shoen J, et al. Adjustable gastric banding in a morbidly obese patient during liver transplantation. Obes Surg. 2008;18(12):1625–7. doi: 10.1007/s11695-008-9633-5. [DOI] [PubMed] [Google Scholar]
  • 302.Porubsky M, Powelson JA, Selzer DJ, et al. Pancreas transplantation after bariatric surgery. Clin Transplant. 2012;26(1):E1–6. doi: 10.1111/j.1399-0012.2011.01559.x. [DOI] [PubMed] [Google Scholar]
  • 303.Al-Sabah S, Christou NV. Laparoscopic gastric bypass after cardiac transplantation. Surg Obes Relat Dis. 2008;4(5):668–70. doi: 10.1016/j.soard.2008.03.247. [DOI] [PubMed] [Google Scholar]
  • 304.Lin MY, Tavakol MM, Sarin A, et al. Safety and feasibility of sleeve gastrectomy in morbidly obese patients following liver transplantation. Surg Endosc. 2013;27(1):81–5. doi: 10.1007/s00464-012-2410-5. [DOI] [PubMed] [Google Scholar]
  • 305.Al-Nowaylati AR, Al-Haddad BJ, Dorman RB, et al. Gastric bypass after liver transplantation. Liver Transpl. 2013;19(12):1324–9. doi: 10.1002/lt.23734. [DOI] [PubMed] [Google Scholar]
  • 306.Duchini A, Brunson ME. Roux-en-Y gastric bypass for recurrent nonalcoholic steatohepatitis in liver transplant recipients with morbid obesity. Transplantation. 2001;72(1):156–9. doi: 10.1097/00007890-200107150-00029. [DOI] [PubMed] [Google Scholar]
  • 307.Takata MC, Campos GM, Ciovica R, et al. Laparoscopic bariatric surgery improves candidacy in morbidly obese patients awaiting transplantation. Surg Obes Relat Dis. 2008;4(2):159–64. doi: 10.1016/j.soard.2007.12.009. [DOI] [PubMed] [Google Scholar]
  • 308.Lin MY, Tavakol MM, Sarin A, et al. Laparoscopic sleeve gastrectomy is safe and efficacious for pretransplant candidates. Surg Obes Relat Dis. 2013;9(5):653–8. doi: 10.1016/j.soard.2013.02.013. [DOI] [PubMed] [Google Scholar]
  • 309.Lazzati A, Iannelli A, Schneck AS, et al. Bariatric surgery and liver transplantation: a systematic review a new frontier for bariatric surgery. Obes Surg. 2015;25(1):134–42. doi: 10.1007/s11695-014-1430-8. [DOI] [PubMed] [Google Scholar]
  • 310.Friedman DI, Jacobson DM. Diagnostic criteria for idiopathic intracranial hypertension. Neurology. 2002;59(10):1492–5. doi: 10.1212/01.wnl.0000029570.69134.1b. [DOI] [PubMed] [Google Scholar]
  • 311.Wakerley BR, Tan MH, Ting EY. Idiopathic intracranial hypertension. Cephalalgia. 2015;35(3):248–61. doi: 10.1177/0333102414534329. [DOI] [PubMed] [Google Scholar]
  • 312.Biousse V, Bruce BB, Newman NJ. Update on the pathophysiology and management of idiopathic intracranial hypertension. J Neurol Neurosurg Psychiatry. 2012;83(5):488–94. doi: 10.1136/jnnp-2011-302029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Curry WT, Jr, Butler WE, Barker FG., 2nd Rapidly rising incidence of cerebrospinal fluid shunting procedures for idiopathic intracranial hypertension in the United States, 1988–2002. Neurosurgery. 2005;57(1):97–108. doi: 10.1227/01.neu.0000163094.23923.e5. [DOI] [PubMed] [Google Scholar]
  • 314.Sugerman HJ, DeMaria EJ, Felton WL, III, et al. Increased intra-abdominal pressure and cardiac filling pressures in obesity-associated pseudotumor cerebri. Neurology. 1997;49(2):507–11. doi: 10.1212/wnl.49.2.507. [DOI] [PubMed] [Google Scholar]
  • 315.Menger RP, Connor DE, Jr, Thakur JD, et al. A comparison of lumboperitoneal and ventriculoperitoneal shunting for idiopathic intracranial hypertension: an analysis of economic impact and complications using the Nationwide Inpatient Sample. Neurosurg Focus. 2014;37(5):E4. doi: 10.3171/2014.8.FOCUS14436. [DOI] [PubMed] [Google Scholar]
  • 316.Sugerman HJ, Felton WL, 3rd, Sismanis A, et al. Gastric surgery for pseudotumor cerebri associated with severe obesity. Ann Surg. 1999;229(5):634–40. doi: 10.1097/00000658-199905000-00005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Sinclair AJ, Burdon MA, Nightingale PG, et al. Low energy diet and intracranial pressure in women with idiopathic intracranial hypertension: prospective cohort study. BMJ. 2010;341:c2701. doi: 10.1136/bmj.c2701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Newborg B. Pseudotumor cerebri treated by rice reduction diet. Arch Intern Med. 1974;133(5):802–7. [PubMed] [Google Scholar]
  • 319.Kupersmith MJ, Gamell L, Turbin R, et al. Effects of weight loss on the course of idiopathic intracranial hypertension in women. Neurology. 1998;50(4):1094–8. doi: 10.1212/wnl.50.4.1094. [DOI] [PubMed] [Google Scholar]
  • 320.Johnson LN, Krohel GB, Madsen RW, et al. The role of weight loss and acetazolamide in the treatment of idiopathic intracranial hypertension (pseudotumor cerebri) Ophthalmology. 1998;105(12):2313–7. doi: 10.1016/S0161-6420(98)91234-9. [DOI] [PubMed] [Google Scholar]
  • 321.Glueck CJ, Golnik KC, Aregawi D, et al. Changes in weight, papilledema, headache, visual field, and life status in response to diet and metformin in women with idiopathic intracranial hypertension with and without concurrent polycystic ovary syndrome or hyperinsulinemia. Transl Res. 2006;148(5):215–22. doi: 10.1016/j.trsl.2006.05.003. [DOI] [PubMed] [Google Scholar]
  • 322.Sugerman HJ, Felton WL, 3rd, Salvant JB, Jr, et al. Effects of surgically induced weight loss on idiopathic intracranial hypertension in morbid obesity. Neurology. 1995;45(9):1655–9. doi: 10.1212/wnl.45.9.1655. [DOI] [PubMed] [Google Scholar]
  • 323.Chandra V, Dutta S, Albanese CT, et al. Clinical resolution of severely symptomatic pseudotumor cerebri after gastric bypass in an adolescent. Surg Obes Relat Dis. 2007;3(2):198–200. doi: 10.1016/j.soard.2006.11.015. [DOI] [PubMed] [Google Scholar]
  • 324.Nadkarni T, Rekate HL, Wallace D. Resolution of pseudotumor cerebri after bariatric surgery for related obesity. Case report J Neurosurg. 2004;101(5):878–80. doi: 10.3171/jns.2004.101.5.0878. [DOI] [PubMed] [Google Scholar]
  • 325.Levin AA, Hess D, Hohler AD. Treatment of idiopathic intracranial hypertension with gastric bypass surgery. Int J Neurosci. 2015;125(1):78–80. doi: 10.3109/00207454.2014.898144. [DOI] [PubMed] [Google Scholar]
  • 326.Fridley J, Foroozan R, Sherman V, et al. Bariatric surgery for the treatment of idiopathic intracranial hypertension. J Neurosurg. 2011;114(1):34–9. doi: 10.3171/2009.12.JNS09953. [DOI] [PubMed] [Google Scholar]
  • 327.Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–7. doi: 10.1038/nature05485. [DOI] [PubMed] [Google Scholar]
  • 328.Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest. 2005;115(5):1111–9. doi: 10.1172/JCI25102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329.Furukawa S, Fujita T, Shimabukuro M, et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2004;114(12):1752–61. doi: 10.1172/JCI21625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Netzer N, Gatterer H, Faulhaber M, et al. Hypoxia, oxidative stress and fat. Biomolecules. 2015;5(2):1143–50. doi: 10.3390/biom5021143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331.Schmidt FM, Weschenfelder J, Sander C, et al. Inflammatory cytokines in general and central obesity and modulating effects of physical activity. PLoS One. 2015;10(3):e0121971. doi: 10.1371/journal.pone.0121971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Nijhuis J, Rensen SS, Slaats Y, et al. Neutrophil activation in morbid obesity, chronic activation of acute inflammation. Obesity (Silver Spring) 2009;17(11):2014–8. doi: 10.1038/oby.2009.113. [DOI] [PubMed] [Google Scholar]
  • 333.Hu T, Li LF, Shen J, et al. Chronic inflammation and colorectal cancer: the role of vascular endothelial growth factor. Curr Pharm Des. 2015;21(21):2960–7. doi: 10.2174/1381612821666150514104244. [DOI] [PubMed] [Google Scholar]
  • 334.Tilg H, Moschen AR. Mechanisms behind the link between obesity and gastrointestinal cancers. Best Pract Res Clin Gastroenterol. 2014;28(4):599–610. doi: 10.1016/j.bpg.2014.07.006. [DOI] [PubMed] [Google Scholar]
  • 335.Long E, Beales IL. The role of obesity in oesophageal cancer development. Therap Adv Gastroenterol. 2014;7(6):247–68. doi: 10.1177/1756283X14538689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336.Cottam D, Fisher B, Ziemba A, et al. Tumor growth factor expression in obesity and changes in expression with weight loss: another cause of increased virulence and incidence of cancer in obesity. Surg Obes Relat Dis. 2010;6(5):538–41. doi: 10.1016/j.soard.2010.04.011. [DOI] [PubMed] [Google Scholar]
  • 337.Cottam DR, Schaefer PA, Shaftan GW, et al. Effect of surgically-induced weight loss on leukocyte indicators of chronic inflammation in morbid obesity. Obes Surg. 2002;12(3):335–42. doi: 10.1381/096089202321088101. [DOI] [PubMed] [Google Scholar]
  • 338.van Dielen FM, Buurman WA, Hadfoune M, et al. Macrophage inhibitory factor, plasminogen activator inhibitor-1, other acute phase proteins, and inflammatory mediators normalize as a result of weight loss in morbidly obese subjects treated with gastric restrictive surgery. J Clin Endocrinol Metab. 2004;89(8):4062–8. doi: 10.1210/jc.2003-032125. [DOI] [PubMed] [Google Scholar]
  • 339.Hanusch-Enserer U, Cauza E, Spak M, et al. Acute-phase response and immunological markers in morbid obese patients and patients following adjustable gastric banding. Int J Obes Relat Metab Disord. 2003;27(3):355–61. doi: 10.1038/sj.ijo.0802240. [DOI] [PubMed] [Google Scholar]
  • 340.Santos J, Salgado P, Santos C, et al. Effect of bariatric surgery on weight loss, inflammation, iron metabolism, and lipid profile. Scand J Surg. 2014;103(1):21–5. doi: 10.1177/1457496913490467. [DOI] [PubMed] [Google Scholar]
  • 341.Carlsson LM, Romeo S, Jacobson P, et al. The incidence of albuminuria after bariatric surgery and usual care in Swedish Obese Subjects (SOS): a prospective controlled intervention trial. Int J Obes (Lond) 2015;39(1):169–75. doi: 10.1038/ijo.2014.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342.Miras AD, le Roux CW. Metabolic surgery: shifting the focus from glycaemia and weight to end-organ health. Lancet Diabetes Endocrinol. 2014;2(2):141–51. doi: 10.1016/S2213-8587(13)70158-X. [DOI] [PubMed] [Google Scholar]
  • 343.Navaneethan SD, Yehnert H, Moustarah F, et al. Weight loss interventions in chronic kidney disease: a systematic review and meta-analysis. Clin J Am Soc Nephrol. 2009;4(10):1565–74. doi: 10.2215/CJN.02250409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344.Iaconelli A, Panunzi S, De Gaetano A, et al. Effects of biliopancreatic diversion on diabetic complications: a 10-year followup. Diabetes Care. 2011;34(3):561–7. doi: 10.2337/dc10-1761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345.Miras AD, Chuah LL, Lascaratos G, et al. Bariatric surgery does not exacerbate and may be beneficial for the microvascular complications of type 2 diabetes. Diabetes Care. 2012;35(12):e81. doi: 10.2337/dc11-2353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Fenske WK, Dubb S, Bueter M, et al. Effect of bariatric surgery-induced weight loss on renal and systemic inflammation and blood pressure: a 12-month prospective study. Surg Obes Relat Dis. 2013;9(4):559–68. doi: 10.1016/j.soard.2012.03.009. [DOI] [PubMed] [Google Scholar]
  • 347.Heneghan HM, Cetin D, Navaneethan SD, et al. Effects of bariatric surgery on diabetic nephropathy after 5 years of follow-up. Surg Obes Relat Dis. 2013;9(1):7–14. doi: 10.1016/j.soard.2012.08.016. [DOI] [PubMed] [Google Scholar]
  • 348.Navaneethan SD, Malin SK, Arrigain S, et al. Bariatric surgery, kidney function, insulin resistance, and adipokines in patients with decreased GFR: a cohort study. Am J Kidney Dis. 2015;65(2):345–7. doi: 10.1053/j.ajkd.2014.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Jamal MH, Corcelles R, Daigle CR, et al. Safety and effectiveness of bariatric surgery in dialysis patients and kidney transplantation candidates. Surg Obes Relat Dis. 2015;11(2):419–23. doi: 10.1016/j.soard.2014.09.022. [DOI] [PubMed] [Google Scholar]
  • 350.Modanlou KA, Muthyala U, Xiao H, et al. Bariatric surgery among kidney transplant candidates and recipients: analysis of the United States renal data system and literature review. Transplantation. 2009;87(8):1167–73. doi: 10.1097/TP.0b013e31819e3f14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351.Lafranca JA, JN IJ, Betjes MG, Dor FJ. Body mass index and outcome in renal transplant recipients: a systematic review and meta-analysis. BMC Med. 2015;13(1):111. Erratum in BMC Med. 2015;13:141. doi: 10.1186/s12916-015-0340-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Subak LL, King WC, Belle SH, et al. Urinary incontinence before and after bariatric surgery. JAMA Intern Med. 2015;175(8):1378–87. doi: 10.1001/jamainternmed.2015.2609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Steele T, Cuthbertson DJ, Wilding JPH. Impact of bariatric surgery on physical functioning in obese adults. Obes Rev. 2015;16(3):248–58. doi: 10.1111/obr.12247. [DOI] [PubMed] [Google Scholar]
  • 354.Maniscalco M, Zedda A, Giardiello C, et al. Effect of bariatric surgery on the six-minute walk test in severe uncomplicated obesity. Obes Surg. 2006;16(7):836–41. doi: 10.1381/096089206777822331. [DOI] [PubMed] [Google Scholar]
  • 355.Tompkins J, Bosch PR, Chenowith R, et al. Changes in functional walking distance and health-related quality of life after gastric bypass surgery. Phys Ther. 2008;88(8):928–35. doi: 10.2522/ptj.20070296. [DOI] [PubMed] [Google Scholar]
  • 356.de Souza SA, Faintuch J, Fabris SM, et al. Six-minute walk test: functional capacity of severely obese before and after bariatric surgery. Surg Obes Relat Dis. 2009;5(5):540–3. doi: 10.1016/j.soard.2009.05.003. [DOI] [PubMed] [Google Scholar]
  • 357.Josbeno DA, Jakicic JM, Hergenroeder A, et al. Physical activity and physical function changes in obese individuals after gastric bypass surgery. Surg Obes Relat Dis. 2010;6(4):361–6. doi: 10.1016/j.soard.2008.08.003. [DOI] [PubMed] [Google Scholar]
  • 358.De Souza SA, Faintuch J, Sant’anna AF. Effect of weight loss on aerobic capacity in patients with severe obesity before and after bariatric surgery. Obes Surg. 2010;20(7):871–5. doi: 10.1007/s11695-010-0109-z. [DOI] [PubMed] [Google Scholar]
  • 359.Lyytinen T, Liikavainio T, Pääkkönen M, et al. Physical function and properties of quadriceps femoris muscle after bariatric surgery and subsequent weight loss. J Musculoskelet Neuronal Interact. 2013;13(3):329–38. [PubMed] [Google Scholar]
  • 360.Vargas CB, Picolli F, Dani C, et al. Functioning of obese individuals in pre- and postoperative periods of bariatric surgery. Obes Surg. 2013;23(10):1590–5. doi: 10.1007/s11695-013-0924-0. [DOI] [PubMed] [Google Scholar]
  • 361.Iossi MF, Konstantakos EK, Teel DD, 2nd, et al. Musculoskeletal function following bariatric surgery. Obesity (Silver Spring) 2013;21(6):1104–10. doi: 10.1002/oby.20155. [DOI] [PubMed] [Google Scholar]
  • 362.Miller GD, Nicklas BJ, You T, et al. Physical function improvements after laparoscopic Roux-en-Y gastric bypass surgery. Surg Obes Relat Dis. 2009;5(5):530–7. doi: 10.1016/j.soard.2008.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363.Kanoupakis E, Michaloudis D, Fraidakis O, et al. Left ventricular function and cardiopulmonary performance following surgical treatment of morbid obesity. Obes Surg. 2001;11(5):552–8. doi: 10.1381/09608920160556715. [DOI] [PubMed] [Google Scholar]
  • 364.Serés L, Lopez-Ayerbe J, Coll R, et al. Increased exercise capacity after surgically induced weight loss in morbid obesity. Obesity (Silver Spring) 2006;14(2):273–9. doi: 10.1038/oby.2006.35. [DOI] [PubMed] [Google Scholar]
  • 365.Valezi AC, Machado VH. Morphofunctional evaluation of the heart of obese patients before and after bariatric surgery. Obes Surg. 2011;21(11):1693–7. doi: 10.1007/s11695-011-0431-0. [DOI] [PubMed] [Google Scholar]
  • 366.Wasmund SL, Owan T, Yanowitz FG, et al. Improved heart rate recovery after marked weight loss induced by gastric bypass surgery: two-year follow up in the Utah Obesity Study. Heart Rhythm. 2011;8(1):84–90. doi: 10.1016/j.hrthm.2010.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367.Wilms B, Ernst B, Thurnheer M, et al. Differential changes in exercise performance after massive weight loss induced by bariatric surgery. Obes Surg. 2013;23(3):365–71. doi: 10.1007/s11695-012-0795-9. [DOI] [PubMed] [Google Scholar]
  • 368.Castello V, Simões RP, Bassi D, et al. Impact of aerobic exercise training on heart rate variability and functional capacity in obese women after gastric bypass surgery. Obes Surg. 2011;21(11):1739–49. doi: 10.1007/s11695-010-0319-4. [DOI] [PubMed] [Google Scholar]
  • 369.Stegen S, Derave W, Calders P, et al. Physical fitness in morbidly obese patients: effect of gastric bypass surgery and exercise training. Obes Surg. 2011;21(1):61–70. doi: 10.1007/s11695-009-0045-y. [DOI] [PubMed] [Google Scholar]
  • 370.Terranova L, Busetto L, Vestri A, et al. Bariatric surgery: cost-effectiveness and budget impact. Obes Surg. 2012;22(4):646–53. doi: 10.1007/s11695-012-0608-1. [DOI] [PubMed] [Google Scholar]
  • 371.Hernæs UJ, Andersen JR, Norheim OF, et al. Work participation among the morbidly obese seeking bariatric surgery: an exploratory study from Norway. Obes Surg. 2015;25(2):271–8. doi: 10.1007/s11695-014-1333-8. [DOI] [PubMed] [Google Scholar]
  • 372.Sjöström L, Larsson B, Backman L, et al. Swedish Obese Subjects (SOS). Recruitment for an intervention study and a selected description of the obese state. Int J Obes Relat Metab Disord. 1992;16(6):465–79. [PubMed] [Google Scholar]
  • 373.Narbro K, Agren G, Jonsson E, et al. Sick leave and disability pension before and after treatment for obesity: a report from the Swedish Obese Subjects (SOS) study. Int J Obes Relat Metab Disord. 1999;23(6):619–24. doi: 10.1038/sj.ijo.0800890. [DOI] [PubMed] [Google Scholar]
  • 374.Gripeteg L, Lindroos AK, Peltonen M, et al. Effects of bariatric surgery on disability pension in Swedish obese subjects. Int J Obes (Lond) 2012;36(3):356–62. doi: 10.1038/ijo.2011.15. [DOI] [PubMed] [Google Scholar]
  • 375.Kolotkin RL, Meter K, Williams GR. Quality of life and obesity. Obes Rev. 2001;2(4):219–29. doi: 10.1046/j.1467-789x.2001.00040.x. [DOI] [PubMed] [Google Scholar]
  • 376.Larsson U, Karlsson J, Sullivan M. Impact of overweight and obesity on health-related quality of life—a Swedish population study. Int J Obes Relat Metab Disord. 2002;26(3):417–24. doi: 10.1038/sj.ijo.0801919. [DOI] [PubMed] [Google Scholar]
  • 377.Doll HA, Petersen SE, Stewart-Brown SL. Obesity and physical and emotional well-being: associations between body mass index, chronic illness, and the physical and mental components of the SF-36 questionnaire. Obes Res. 2000;8(2):160–70. doi: 10.1038/oby.2000.17. [DOI] [PubMed] [Google Scholar]
  • 378.Kolotkin RL, Crosby RD, Williams GR, et al. The relationship between health-related quality of life and weight loss. Obes Res. 2001;9(9):564–71. doi: 10.1038/oby.2001.73. [DOI] [PubMed] [Google Scholar]
  • 379.Karlsson J, Taft C, Sjöström L, et al. Psychosocial functioning in the obese before and after weight reduction: construct validity and responsiveness of the Obesity-related Problems scale. Int J Obes Relat Metab Disord. 2003;27(5):617–30. doi: 10.1038/sj.ijo.0802272. [DOI] [PubMed] [Google Scholar]
  • 380.Karlsson J, Taft C, Rydén A, et al. 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) 2007;31(8):1248–61. doi: 10.1038/sj.ijo.0803573. [DOI] [PubMed] [Google Scholar]
  • 381.Magallares A, Schomerus G. Mental and physical health-related quality of life in obese patients before and after bariatric surgery: a meta-analysis. Psychol Health Med. 2015;20(2):165–76. doi: 10.1080/13548506.2014.963627. [DOI] [PubMed] [Google Scholar]
  • 382.Batsis JA, Lopez-Jimenez F, Collazo-Clavell ML, et al. Quality of life after bariatric surgery: a population-based cohort study. Am J Med. 2009;122(11):1055.e1–1055.e10. doi: 10.1016/j.amjmed.2009.05.024. [DOI] [PubMed] [Google Scholar]
  • 383.Suter M, Donadini A, Romy S, et al. Laparoscopic Roux-en-Y gastric bypass: significant long-term weight loss, improvement of obesity-related comorbidities and quality of life. Ann Surg. 2011;254(2):267–73. doi: 10.1097/SLA.0b013e3182263b66. [DOI] [PubMed] [Google Scholar]
  • 384.Burgmer R, Legenbauer T, Müller A, et al. Psychological outcome 4 years after restrictive bariatric surgery. Obes Surg. 2014;24(10):1670–8. doi: 10.1007/s11695-014-1226-x. [DOI] [PubMed] [Google Scholar]
  • 385.Black JA, White B, Viner RM, et al. Bariatric surgery for obese children and adolescents: a systematic review and meta-analysis. Obes Rev. 2013;14(8):634–44. doi: 10.1111/obr.12037. [DOI] [PubMed] [Google Scholar]
  • 386.Kakoulidis TP, Karringer A, Gloaguen T, et al. Initial results with sleeve gastrectomy for patients with class I obesity (BMI 30-35 kg/m2) Surg Obes Relat Dis. 2009;5(4):425–8. doi: 10.1016/j.soard.2008.09.009. [DOI] [PubMed] [Google Scholar]
  • 387.O’Brien PE, Sawyer SM, Laurie C, et al. Laparoscopic adjustable gastric banding in severely obese adolescents; a randomized trial. JAMA. 2010;303(6):519–26. doi: 10.1001/jama.2010.81. [DOI] [PubMed] [Google Scholar]
  • 388.Colquitt JL, Pickett K, Loveman E, et al. Surgery for weight loss in adults. Cochrane Database Syst Rev. 2014;8:CD003641. doi: 10.1002/14651858.CD003641.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 389.Faulconbridge LF, Wadden TA, Thomas JG, et al. Changes in depression and quality of life in obese individuals with binge eating disorder: bariatric surgery versus lifestyle modification. Surg Obes Relat Dis. 2013;9:790–6. doi: 10.1016/j.soard.2012.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 390.Gloy VL, Briel M, Bhatt DL, et al. Bariatric surgery versus nonsurgical treatment for obesity: a systematic review and meta-analysis of randomised controlled trials. BMJ. 2013;347:f5934. doi: 10.1136/bmj.f5934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 391.Kolotkin RL, Davidson LE, Crosby RD, et al. Six-year changes in health-related quality of life in gastric bypass patients versus obese comparison groups. Surg Obes Relat Dis. 2012;8(5):625–33. doi: 10.1016/j.soard.2012.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 392.Nguyen NT, Slone JA, Nguyen XM, et al. A prospective randomized trial of laparoscopic gastric bypass versus laparoscopic adjustable gastric banding for the treatment of morbid obesity: outcomes, quality of life, and costs. Ann Surg. 2009;250(4):631–41. doi: 10.1097/SLA.0b013e3181b92480. [DOI] [PubMed] [Google Scholar]
  • 393.Søvik TT, Aasheim ET, Taha O, et al. Weight loss, cardiovascular risk factors, and quality of life after gastric bypass and duodenal switch: a randomized trial. Ann Intern Med. 2011;155(5):281–91. doi: 10.7326/0003-4819-155-5-201109060-00005. [DOI] [PubMed] [Google Scholar]
  • 394.Strain GW, Kolotkin RL, Dakin GF, et al. The effects of weight loss after bariatric surgery on health-related quality of life and depression. Nutr Diabetes. 2014;4:e132. doi: 10.1038/nutd.2014.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 395.Inge TH, Krebs NF, Garcia VF, et al. Bariatric surgery for severely overweight adolescents: concerns and recommendations. Pediatrics. 2004;114(1):217–23. doi: 10.1542/peds.114.1.217. [DOI] [PubMed] [Google Scholar]
  • 396.Freedman DS, Mei Z, Srinivasan SR, et al. Cardiovascular risk factors and excess adiposity among overweight children and adolescents: the Bogalusa Heart Study. J Pediatr. 2007;150(1):12–7.e2. doi: 10.1016/j.jpeds.2006.08.042. [DOI] [PubMed] [Google Scholar]
  • 397.Pratt JS, Lenders CM, Dionne EA, et al. Best practice updates for pediatric/adolescent weight loss surgery. Obesity (Silver Spring) 2009;17(5):901–10. doi: 10.1038/oby.2008.577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 398.Fried M, Hainer V, Basdevant A, et al. Inter-disciplinary European guidelines on surgery of severe obesity. Int J Obes (Lond) 2007;31(4):569–77. doi: 10.1038/sj.ijo.0803560. [DOI] [PubMed] [Google Scholar]
  • 399.Treadwell JR, Sun F, Schoelles K. Systematic review and meta-analysis of bariatric surgery for pediatric obesity. Ann Surg. 2008;248(5):763–76. doi: 10.1097/SLA.0b013e31818702f4. [DOI] [PubMed] [Google Scholar]
  • 400.Michalsky M, Reichard K, Inge T, et al. American Society for Metabolic and Bariatric Surgery. ASMBS pediatric committee best practice guidelines. Surg Obes Relat Dis. 2012;8(1):1–7. doi: 10.1016/j.soard.2011.09.009. [DOI] [PubMed] [Google Scholar]
  • 401.Società Italiana di Chirurgia dell’Obesità e delle malattie metaboliche (S.I.C.OB.) Linee guida e stato dell’arte della chirurgia bariatrica e metabolica in Italia. Napoli: EdiSES; 2008. [Google Scholar]
  • 402.Sugerman HJ, DeMaria EJ, Kellum JM, et al. Effects of bariatric surgery in older patients. Ann Surg. 2004;240(2):243–7. doi: 10.1097/01.sla.0000133361.68436.da. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 403.Quebbemann B, Engstrom D, Siegfried T, et al. Bariatric surgery in patients older than 65 years is safe and effective. Surg Obes Relat Dis. 2005;1(4):389–92. doi: 10.1016/j.soard.2005.05.003. [DOI] [PubMed] [Google Scholar]
  • 404.Hazzan D, Chin EH, Steinhagen E, et al. Laparoscopic bariatric surgery can be safe for treatment of morbid obesity in patients older than 60 years. Surg Obes Relat Dis. 2006;2(6):613–6. doi: 10.1016/j.soard.2006.09.009. [DOI] [PubMed] [Google Scholar]
  • 405.Taylor CJ, Layani L. Laparoscopic adjustable gastric banding in patients > or =60 years old: is it worthwhile? Obes Surg. 2006;16(12):1579–83. doi: 10.1381/096089206779319310. [DOI] [PubMed] [Google Scholar]
  • 406.Dunkle-Blatter SE, St Jean MR, Whitehead C, et al. Outcomes among elderly bariatric patients at a high-volume center. Surg Obes Relat Dis. 2007;3(2):163–9. doi: 10.1016/j.soard.2006.12.004. [DOI] [PubMed] [Google Scholar]
  • 407.Busetto L, Angrisani L, Basso N, et al. Safety and efficacy of laparoscopic adjustable gastric banding in the elderly. Obesity (Silver Spring) 2008;16(2):334–8. doi: 10.1038/oby.2007.85. [DOI] [PubMed] [Google Scholar]
  • 408.Villareal DT, Apovian CM, Kushner RF, et al. Obesity in older adults: technical review and position statement of the American Society for Nutrition and NAASO, the Obesity Society. Am J Clin Nutr. 2005;82(5):923–34. doi: 10.1093/ajcn/82.5.923. [DOI] [PubMed] [Google Scholar]
  • 409.InterAct Consortium. Langenberg C, Sharp SJ, et al. Long-term risk of incident type 2 diabetes and measures of overall and regional obesity: the EPIC-InterAct case-cohort study. PLoS Med. 2012;9(6):e1001230. doi: 10.1371/journal.pmed.1001230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 410.Thomas EL, Parkinson JR, Frost GS, et al. The missing risk: MRI and MRS phenotyping of abdominal adiposity and ectopic fat. Obesity (Silver Spring) 2012;20(1):76–87. doi: 10.1038/oby.2011.142. [DOI] [PubMed] [Google Scholar]
  • 411.Klöting N, Fasshauer M, Dietrich A, et al. Insulin-sensitive obesity. Am J Physiol Endocrinol Metab. 2010;299(3):E506–15. doi: 10.1152/ajpendo.00586.2009. [DOI] [PubMed] [Google Scholar]
  • 412.Stefan N, Kantartzis K, Machann J, et al. Identification and characterization of metabolically benign obesity in humans. Arch Intern Med. 2008;168(15):1609–16. doi: 10.1001/archinte.168.15.1609. [DOI] [PubMed] [Google Scholar]
  • 413.Klein S, Fontana L, Young VL, et al. Absence of an effect of liposuction on insulin action and risk factors for coronary heart disease. N Engl J Med. 2004;350(25):2549–57. doi: 10.1056/NEJMoa033179. [DOI] [PubMed] [Google Scholar]
  • 414.Thorne A, Lonnqvist F, Apelman J, et al. A pilot study of long-term effects of a novel obesity treatment: omentectomy in connection with adjustable gastric banding. Int J Obes Relat Metab Disord. 2002;26(2):193–9. doi: 10.1038/sj.ijo.0801871. [DOI] [PubMed] [Google Scholar]
  • 415.Fabbrini E, Tamboli RA, Magkos F, et al. Surgical removal of omental fat does not improve insulin sensitivity and cardiovascular risk factors in obese adults. Gastroenterology. 2010;139(2):448–55. doi: 10.1053/j.gastro.2010.04.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 416.Vilarrasa N, de Gordejuela AG, Gómez-Vaquero C, et al. Effect of bariatric surgery on bone mineral density: comparison of gastric bypass and sleeve gastrectomy. Obes Surg. 2013;23(12):2086–91. doi: 10.1007/s11695-013-1016-x. [DOI] [PubMed] [Google Scholar]
  • 417.Wucher H, Ciangura C, Poitou C, et al. Effects of weight loss on bone status after bariatric surgery: association between adipokines and bone markers. Obes Surg. 2008;18(1):58–65. doi: 10.1007/s11695-007-9258-0. [DOI] [PubMed] [Google Scholar]
  • 418.Nakamura KM, Haglind EG, Clowes JA, et al. Fracture risk following bariatric surgery: a population-based study. Osteoporos Int. 2014;25(1):151–8. doi: 10.1007/s00198-013-2463-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 419.Lalmohamed A, de Vries F, Bazelier MT, et al. Risk of fracture after bariatric surgery in the United Kingdom: population based, retrospective cohort study. BMJ. 2012;345:e5085. doi: 10.1136/bmj.e5085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 420.Berarducci A, Haines K, Murr MM. Incidence of bone loss, falls, and fractures after Roux-en-Y gastric bypass for morbid obesity. Appl Nurs Res. 2009;22(1):35–41. doi: 10.1016/j.apnr.2007.03.004. [DOI] [PubMed] [Google Scholar]
  • 421.Ahlin S, Peltonen M, Anveden Ĺ, Jacobson P, Sjöholm K, Svensson PA, et al. Bariatric surgery increases the risk of osteoporosis and fractures in women in the Swedish Obese subjects study. Obes Facts. 2015;8(suppl 1):T8:OS3.3. [Google Scholar]
  • 422.Yu EW, Bouxsein ML, Roy AE, et al. Bone loss after bariatric surgery: discordant results between DXA and QCT bone density. J Bone Miner Res. 2014;29(3):542–50. doi: 10.1002/jbmr.2063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 423.Bosy-Westphal A, Müller MJ. Identification of skeletal muscle mass depletion across age and BMI groups in health and disease—there is need for a unified definition. Int J Obes (Lond) 2015;39(3):379–86. doi: 10.1038/ijo.2014.161. [DOI] [PubMed] [Google Scholar]
  • 424.Tamboli RA, Hossain HA, Marks PA, et al. Body composition and energy metabolism following Roux-en-Y gastric bypass surgery. Obesity (Silver Spring) 2010;18(9):1718–24. doi: 10.1038/oby.2010.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 425.Carey DG, Pliego GJ, Raymond RL. Body composition and metabolic changes following bariatric surgery: effects on fat mass, lean mass and basal metabolic rate: six months to one-year follow-up. Obes Surg. 2006;16(12):1602–8. doi: 10.1381/096089206779319347. [DOI] [PubMed] [Google Scholar]
  • 426.Pouwels S, Wit M, Teijink JA, et al. Aspects of exercise before or after bariatric surgery: a systematic review. Obes Facts. 2015;8(2):132–46. doi: 10.1159/000381201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 427.Herman KM, Carver TE, Christou NV, et al. Physical activity and sitting time in bariatric surgery patients 1–16 years post-surgery. Clin Obes. 2014;4(5):267–76. doi: 10.1111/cob.12069. [DOI] [PubMed] [Google Scholar]
  • 428.Chaston TB, Dixon JB, O’Brien PE. Changes in fat-free mass during significant weight loss: a systematic review. Int J Obes (Lond) 2007;31(5):743–50. doi: 10.1038/sj.ijo.0803483. [DOI] [PubMed] [Google Scholar]
  • 429.Fried M, Ribaric G, Buchwald JN, et al. Metabolic surgery for the treatment of type 2 diabetes in patients with BMI <35 Kg/m2: an integrative review of early studies. Obes Surg. 2010;20(6):776–90. doi: 10.1007/s11695-010-0113-3. [DOI] [PubMed] [Google Scholar]
  • 430.Scopinaro N, Adami GF, Papadia FS, et al. The effects of biliopancreatic diversion on type 2 diabetes mellitus in patients with mild obesity (BMI 30–35 kg/m2) and simple overweight (BMI 25–30 kg/m2): a prospective controlled study. Obes Surg. 2011;21(7):880–8. doi: 10.1007/s11695-011-0407-0. [DOI] [PubMed] [Google Scholar]
  • 431.2004 ASBS Consensus Conference on Surgery for Severe Obesity. Surg Obes Relat Dis. 2005;1(3):297–381. doi: 10.1016/j.soard.2005.03.003. [DOI] [PubMed] [Google Scholar]
  • 432.Sauerland S, Angrisani L, Belachew M, et al. Obesity surgery: evidence-based guidelines of the European Association for Endoscopic Surgery (EAES) Surg Endosc. 2005;19(2):200–21. doi: 10.1007/s00464-004-9194-1. [DOI] [PubMed] [Google Scholar]
  • 433.Rubino F, Kaplan LM, Schauer PR, et al. The Diabetes Surgery Summit consensus conference: recommendations for the evaluation and use of gastrointestinal surgery to treat type 2 diabetes mellitus. Ann Surg. 2010;251(3):399–405. doi: 10.1097/SLA.0b013e3181be34e7. [DOI] [PubMed] [Google Scholar]
  • 434.Maglione MA, Gibbons MM, Livhits M, Ewing B, Hu J, Ruelaz Maher A, et al. Bariatric surgery and nonsurgical therapy in adults with metabolic conditions and a body mass index of 30.0 to 34.9 kg/m2. Rockville (MD): Agency for Healthcare Research and Quality (US); 2013. Report No.: 12(13)-EHC139-EF. [PubMed] [Google Scholar]
  • 435.Parikh M, Issa R, Vieira D, et al. Role of bariatric surgery as treatment for type 2 diabetes in patients who do not meet current NIH criteria: a systematic review and meta-analysis. J Am Coll Surg. 2013;217(3):527–32. doi: 10.1016/j.jamcollsurg.2013.04.023. [DOI] [PubMed] [Google Scholar]
  • 436.Demaria EJ, Winegar DA, Pate VW, et al. Early postoperative outcomes of metabolic surgery to treat diabetes from sites participating in the ASMBS bariatric surgery center of excellence program as reported in the bariatric outcomes longitudinal database. Ann Surg. 2010;252(3):559–66. doi: 10.1097/SLA.0b013e3181f2aed0. [DOI] [PubMed] [Google Scholar]
  • 437.Angrisani L, Favretti F, Furbetta F, et al. Italian Group for Lap-Band System: results of multicenter study on patients with BMI < or =35 kg/m2. Obes Surg. 2004;14(3):415–8. doi: 10.1381/096089204322917963. [DOI] [PubMed] [Google Scholar]

RESOURCES