http://informahealthcare.com/pgm ISSN: 0032-5481 (print), 1941-9260 (electronic) Postgrad Med, 2015; 127(5): 494–502 DOI: 10.1080/00325481.2015.1048181

CLINICAL FOCUS: DIABETES REVIEW

Obesity: Lifestyle management, bariatric surgery, drugs, and the therapeutic exploitation of gut hormones Preeshila Behary, Jaimini Cegla, Tricia M. Tan and Stephen R. Bloom Postgraduate Medicine Downloaded from informahealthcare.com by Nyu Medical Center on 06/05/15 For personal use only.

Division of Diabetes, Endocrinology and Metabolism, Hammersmith Hospital, Imperial College London, Du Cane Road, London, UK

Abstract

Keywords

Obesity is on the rise and the pursuit of efficient and safe treatment is ongoing. Available antiobesity medical therapies have so far proved to be disappointing, whereas bariatric surgery is leading the way and offers long-term health benefits. Part of the success of bariatric surgery is thought to be mediated by gut hormones. A better understanding of the role of gut hormones within the gut-brain signaling pathway in the control of hunger, satiety, and energy homeostasis, has led to their therapeutic exploitation as possible anti-obesity drugs. In this review, we provide a summary of currently available treatment options for obesity from simple lifestyle modifications and bariatric surgery to traditional and novel medical therapies.

Obesity, lifestyle management, bariatric surgery, drugs, gut hormones

Introduction Obesity is a growing global epidemic. The World Health Organization estimates that over 1 billion adults and over 42 million children under the age of 5 are overweight worldwide [1,2]. In the UK, a quarter of adults are obese [3] and 30% of children are classified as overweight or obese [4], whereas in the US, a third of the adult population and ~ 17% of children are obese [5,6]. Obesity is recognized as an important risk factor for the development of diabetes, cardiovascular disease, and certain types of cancer, all contributing to significant morbidity, mortality, and financial burden. This has driven considerable interest in understanding the pathophysiology of this condition, the ultimate aim being to devise innovative ways to tackle obesity [5,6]. Food intake is closely regulated to match energy intake to the metabolic demands of the body. This “homeostatic control” dictates our eating behavior and is believed to have played a critical role in human evolutionary self-preservation. The problem arises when this homeostatic system becomes overwhelmed, resulting in an imbalance between energy intake and energy expenditure, hence weight gain. There is no doubt that the food industry and modern technology promoting a sedentary lifestyle have been key factors in exacerbating this imbalance especially in the developed world. Accessibility, availability, and the appeal of convenient and energy dense foods have contributed to the disruption of the homeostatic control of eating behavior. Worryingly, obesity is no longer confined to the developed world but is also affecting many low and middle income

History Received 5 February 2015 Accepted 1 May 2015

countries [7,8]. Therefore, there is a drive to find solutions to this emerging problem. In this review, we will provide an overview of the treatments available for obesity, from conventional lifestyle management to emerging pharmaceutical and innovative surgical options. We will discuss the potential mechanisms being targeted in each therapy and their limitations.

Lifestyle management Reducing energy intake while maximizing energy expenditure appears to be the ideal solution to restore balance in body weight. For this reason, exercise and diet remain cornerstone treatments for obesity. Restricting calorie intake through dieting is an effective way to lose weight, with numerous weight loss diet plans to choose from. Some of the popular ones include the low and very low calorie diet, low glycemic, low carbohydrate, low fat, and high protein content diets. There is an ongoing debate in regard to the most effective weight loss diet and concern has been expressed over longterm safety, benefits, and sustainability of numerous diets [9-12]. Unsurprisingly, the prospective patient can get easily confused by the marketing of companies that claim the superiority of their weight loss diet plans. The available clinical evidence is not always helpful as robust long-term data directly comparing different weight loss dietary profiles are limited and contradictory. For example, a recent study advocates the superiority of low-carbohydrate content diets over low-fat diets on both weight loss and cardiovascular factor

Correspondence: Preeshila Behary, Division of Diabetes, Endocrinology and Metabolism, Hammersmith Hospital, Imperial College London, 6th Floor Commonwealth Building, Du Cane Road, London W12 0NN, UK. E-mail: [email protected]  2015 Informa UK Ltd.

Postgraduate Medicine Downloaded from informahealthcare.com by Nyu Medical Center on 06/05/15 For personal use only.

DOI: 10.1080/00325481.2015.1048181

Obesity: Lifestyle management, bariatric surgery, drugs, and gut hormones

risk reduction [13]. Others have shown that, irrespective of the macronutrient content of the designated weight loss diet, as long as they are consistent with a reduction in overall calorie content, people do lose clinically meaningful amounts of weight [14]. A recent meta-analysis showed that significant weight loss was observed with any low-carbohydrate or low-fat diet and weight loss differences between individual named diets were negligible [15]. Modest weight loss of 5% is associated with clinical improvement in cardiovascular risk factors including type 2 diabetes, with greater benefits derived from more substantial weight loss [16]. In fact, “reversal” of type 2 diabetes is possible following a short-term very low calorie diet of 600 kcal (2511 KJ) a day [17]. A very low calorie diet is a drastic short-term diet, potentially leading to weight loss of 15–25% over 3- to 4-month period when coupled with lifestyle modification [10]. Although effective for weight loss, there is a small increased risk of complications such as gallstones and micronutrient deficiency [9,18]. Irrespective of the diet plan, there is a high relapse rate of obesity. Weight regain of 50% by 1 year has been reported and net weight loss of 40 kg/m2) obesity (Table 1) [33,36,38]. However, there is now emerging evidence to validate its use in some patients with class I obesity (BMI = 30.0–34.9 kg/m2) as well [39,40]. The most commonly performed bariatric procedures worldwide, in order of popularity, are the Roux-en-Y gastric

496

P. Behary et al.

Postgrad Med, 2015; 127(5):494–502

Table 1. Classification of obesity by body mass index. 2

Body mass index (kg/m ) Overweight Class 1 obesity Class 2 obesity Class 3 obesity

25.0–29.9 30.0–34.9 35.0–39.9 ‡40.0

Postgraduate Medicine Downloaded from informahealthcare.com by Nyu Medical Center on 06/05/15 For personal use only.

Data taken from [38].

bypass (RYGB), the sleeve gastrectomy (SG), and laparoscopic gastric banding (LGB) (Figure 2). However, in the US, Canada, and countries of the Asia-Pacific region, more SG surgeries are being performed compared to RYGB, whereas LGB is declining in popularity globally [41]. The biliopancreatic diversion is another surgical procedure which is rarely performed nowadays due to its association with profound malabsorption and nutritional deficits [42,43]. LGB is a minimally invasive and technically easy procedure where a band is placed around the stomach just inferior to the gastroesophageal junction, to restrict gastric volume and regulate how much the patient can eat. One specific shortcoming of this procedure is that it requires repeated follow up for adjustment of the pressure in the band to achieve the desired level of restriction. SG involves a longitudinal resection of the stomach leading to the formation of a tube-like structure called the “sleeve”. This procedure is irreversible. RYGB involves the creation of a small gastric pouch of ~ 30 ml in volume, which is then anastomosed to a divided section of the distal small intestine. This is referred to as the Roux limb. The majority of the stomach and the proximal small intestine are bypassed. The bypassed bowel, known as the biliopancreatic limb, is then connected further down the Roux limb to allow the flow of pancreatic secretions and bile. This procedure combines restriction of food intake with some degree of malabsorption to induce weight loss. RYGB typically causes up to 30% body weight loss and a remission rate of diabetes of between 40 and 80%, depending on the criteria used to define remission [44-46]. Furthermore, long-term data suggest that bariatric surgery confers sustained weight loss of up to 20 years and reduction in cardiovascular death and all causes of mortality. This is despite a gradual weight regain observed after 2–3 years post surgery which subsequently appears to level off by 8–10 years [47]. These expert center trials can, however, be misleadingly optimistic with high attrition rates. SG performed in centers with the surgical expertise can also produce comparable weight loss and improvement in obesity related comorbidities, although long-term data to

challenge the RYGB procedure are not yet available [48-50]. Data available indicate that weight loss through SG is less than with RYGB; in addition, even if improvement in glycemic control in type 2 diabetes patients may not be significantly different between the two procedures, more antidiabetic medications need to be used in patients who undergo the SG [51]. However, compared to conventional nonsurgical management of obesity such as lifestyle modification, all three types of bariatric surgery are superior in achieving significant and sustained weight loss [47,51]. The beneficial metabolic effects of RYGB are thought to be mediated beyond the simplistic restrictive and malabsorption mechanisms. Compared to LGB, RYGB leads to additional weight loss and induce diabetes remission. The diabetes improves or “remits” within days of the RYGB surgery, before substantial weight loss has occurred, suggestive of weight-independent mechanisms [52,53]. The gut hormones GLP-1, OXM, and PYY are elevated early after RYGB and stay elevated for years, whereas ghrelin levels tends to be reduced [54-58]. It is thought that the rapid delivery of nutrients to the lower gut enhances the nutrient sensing ability of L cells which promote secretion of GLP-1, PYY, and OXM after eating [52]. These hormones have been shown to induce satiety, promote insulin release, and increase energy expenditure in humans [59-63]. Hence they have the potential to be the key mediators of weight loss and diabetes remission post-RYGB. In fact, Roux et al. showed that blocking the action of PYY in rats post-jejuno-intestinal bypass led to an increase in ad libitum food intake comparable to sham-operated animals; whereas exogenous injection of PYY in sham-operated rats led to a decrease in ad libitum food intake [57]. The same group also showed that inhibiting PYY and GLP-1 with octreotide post-RYGB resulted in an increase in appetite and food intake in both human and animal studies [56,64]. In other studies, satiety increases dramatically post-RYGB and has been associated with a rise in gut hormones [55,65]. In further support of the “gut hormone hypothesis”, it has been shown that patients who successfully lose weight post-RYGB “responders” have higher circulating GLP-1 and PYY compared to “non-responders” [66]”. In opposition to the “gut hormone hypothesis”, some studies observed significant weight loss and improvement in glycemia post-RYGB in both humans and animals, despite abolishing the effect of GLP-1 [67-69]. GLP-1 blockage was achieved by either an intravenous infusion of exendin-9 to -39), a potent GLP-1 antagonist, or by using a GLP-1R knocked-out mouse model. The authors therefore concluded

Figure 2. Illustrations showing common bariatric procedures: a) laparoscopic gastric banding; b) Roux-en-Y gastric bypass; c) sleeve gastrectomy.

Postgraduate Medicine Downloaded from informahealthcare.com by Nyu Medical Center on 06/05/15 For personal use only.

DOI: 10.1080/00325481.2015.1048181

Obesity: Lifestyle management, bariatric surgery, drugs, and gut hormones

that GLP-1 was unlikely to mediate the beneficial metabolic effects of RYGB and other factors are important. Besides gut hormones, other mechanisms have also been postulated to contribute toward the observed weight loss after RYGB. The vagus nerve represents an important relay system by which gut hormones exert their effect on the brain [70]. Therefore, it is feasible that disruption of vagal innervation to the gastrointestinal tract following surgery could modulate appetite and satiety toward weight loss [71-73]. Other implicated mechanisms involve a change in the composition and levels of bile acids, fibroblast growth factor 19 (FGF-19) and FGF-21 post-RYGB. These have been reported to be elevated post-RYGB and there is increasing evidence to suggest that these molecules act as regulators of glucose and energy metabolism [74-76]. Furthermore, a shift in gut microbiota composition has been observed after RYGB which favors an environment promoting glucose and energy metabolism possibly through short-chain free fatty acids production by colonic bacteria [77,78]. It is also believed there is an interplay between the microbiome post-RYGB and bile acids in regard to altering their absorption and deconjugation and enhancing overall enterohepatic recycling [79]. In comparison to the hormonal changes observed postRYGB, GLP-1, OXM, and PYY levels are unchanged or decreased following calorie restriction-induced weight loss and unchanged following LGB [80,81]. This may explain the superiority of RYGB over LGB and calorie restriction in inducing weight loss. The success of RYGB in inducing weight loss could also relate to the changes in food preferences post-surgery. There is some evidence to suggest that post-RYGB, healthier food options have a greater appeal to patients compared to high fat, high sugar food [82]. This is consistent with fMRI findings of decreased brain activation within the hedonic regions to high fat food but not low fat food after RYGB surgery [83]. Bariatric surgery is a life-changing procedure and, as with any surgery, carries risks and complications. The 30-day mortality of RYGB is estimated at 0.1% to 0.3%, comparable to a laparoscopic cholecystectomy, and hence considered a relatively “low risk” procedure [84]. Common short-term complications are bleeding, anastomotic leaks, wound infections, and thromboembolic events. Long-term complications may include nutritional deficiencies, weight regain, or hyperinsulinemic hypoglycemia. However, these are uncommon and the key factors impacting on surgical risk profile are experience of the surgeon, center’s volume, patient’s comorbidities, and complexity of surgery among others [85]. There is no doubt that bariatric surgery is currently the best ammunition we have to battle obesity but it does come with financial implications and resource limitations [86]. Furthermore, bariatric surgery is not always readily accessible to everyone, especially to those who are more likely to benefit such as individuals from lower socioeconomic background [86,87].

Pharmacotherapy There is an urgent need for noninvasive, simpler, and more accessible treatment options for obesity. The development of

497

anti-obesity drugs represents a major research area and has been the focus of reputable pharmaceutical companies for many years. The drug regulatory agencies, the FDA in the US, and the European Medicines Agency (EMA) in Europe, considers a mean weight loss of at least 5% from baseline to be a valid primary efficacy point for approving a weight loss drug [88,89] Rimonabant, a cannabinoid CB1 receptor antagonist, and sibutramine, a serotonin and noradrenaline reuptake inhibitor, initially showed promise when they were first launched. However, they were both withdrawn from the market because of the increased risk of depression and suicidal ideation in the case of rimonabant and cardiovascular disease with sibutramine in 2008 and 2010, respectively [90,91]. Orlistat, a potent selective pancreatic lipase inhibitor, was, until recently, the only licensed anti-obesity drug in Europe and is also approved by the FDA for long-term use. Orlistat can be purchased over the counter in some countries. An average weight loss of 3 kg is typically seen following a year’s treatment on orlistat [92,93]. However, its gastrointestinal side effects such as steatorrhea mean that orlistat is poorly tolerated and therefore limits its use. Two newer agents, lorcaserin and phentermine/topiramate have been approved by the FDA for the US market 2 years ago [94]. Lorcaserin is a selective serotonin 2C receptor agonist, which promotes satiety via stimulation of proopiomelanocortin (POMC) neurons. Clinical studies involving overweight and obese individuals with one obesity-related comorbidity have shown modest weight reduction of ~ 5% [95]. On the up side, the drug has shown a relatively good tolerability profile and the increased incidence of valvulopathy observed in rodents has not been borne out in humans thus far [94]. Phentermine, a central noradrenaline releasing drug and topiramate, is a licensed antiepileptic drug. The combination of both has been shown in clinical trials to induce average weight loss of 10% in around half of the study subjects when given the higher dose of the drug and most importantly, weight loss was sustained up to 56 weeks [96]. Despite phentermine’s long-established use as an appetite suppressant, the mechanism by which it suppresses appetite is unclear. Major safety concerns such as the risk of teratogenicity remain and therefore this drug may not be the ideal choice for women of childbearing age. As from September 2014, two anti-obesity agents have been approved by the FDA in the US. They are liraglutide 3mg (Saxenda), a GLP-1 agonist, and the combination drug naltrexone/bupropion [97,98]. In January 2015, the EMA also voted in favor of Saxenda to be brought to the European market [99]. This is discussed later in the review. Naltrexone is an opioid antagonist, which can be used for treating opioid and alcohol addiction, whereas bupropion is an antidepressant, which inhibits noradrenaline and dopamine re-uptake. It is believed that the combination drug works by promoting satiety via the activation of POMC neurons and modulation of the mesolimbic pathways [100]. In separate double-blinded, randomized clinical trials (RCTs) of 56 weeks duration, between 44% and 55% of study participants lost at least 5% of their body weight following treatment with

498

P. Behary et al.

naltrexone/bupropion. In subjects with type 2 diabetes, mean placebo-subtracted weight loss of 3.2% was seen [101-103]. The most common reported side effect from the RCTs was nausea with no difference observed in the incidence of psychiatric symptoms between the treatment group and placebo. In summary, there are limited pharmacotherapeutic options which are deemed safe and efficient in achieving clinically meaningful weight loss. Furthermore, long-term safety data is limited and therefore caution should be exercised with newer agents.

Postgraduate Medicine Downloaded from informahealthcare.com by Nyu Medical Center on 06/05/15 For personal use only.

Gut hormones Exploiting the gut–brain axis by taking advantage of the properties of gut hormones is the focus of intensive research looking for a new therapeutic approach for obesity. However, most gut hormones are rapidly broken down by peptidases in the circulation, and oral therapies are unable to withstand the harsh gastric environment. Therefore, injectable therapy remains the main form of administration. We have summarized below past and current manipulation of gut hormones in designing novel anti-obesity agents resistant to degradation. We chose to discuss these gut hormones specifically as they are the main ones implicated in mediating the beneficial metabolic effects of RYGB and are being investigated in current human clinical trials. GLP-1 GLP-1 has been the most therapeutically exploited gut hormone to date. It is secreted from the L cells of the ileum in response to oral nutrients. It is also a potent incretin, augmenting glucose-dependent insulin release from the pancreatic b cells. The biological active forms are GLP-1 (7–37) amide and GLP-1 (7–36) amide, although the latter is more prevalent in the circulation. Central and peripheral administration of GLP-1 has shown reduction in food intake and weight in rodents. Peripheral administration in both lean and overweight humans produces a reduction in ad libitum food intake [60,104]. It is believed that GLP-1 acts centrally on the hypothalamus and brainstem to inhibit appetite [105,106]. GLP-1 is broken down rapidly by the enzyme dipeptidyl peptidase-4 (DPP-4) and has a half-life of 2 minutes in the circulation. Manipulation of the peptide to extend its life is therefore necessary. Exenatide (Byetta) was the first GLP-1 analog to be marketed for the treatment of type 2 diabetes in 2005. This was originally based on exendin-4, which was isolated from the saliva of the Gila monster and is more robust to DPP-4 degradation. Further GLP-1 analogs then followed: liraglutide (Victoza), a GLP-1 analog engineered to bind to albumin and therefore to persist in circulation and exenatide LAR (Bydureon), a weekly preparation of exenatide in slowdissolving microspheres. Other GLP-1 mimetics currently on the market for the treatment of type 2 diabetes are albiglutide, dulaglutide, and lixisenatide [107]. Liraglutide is the analog that has seen most application in obesity. At a dose of 1.2 and 1.8 mg/day, liraglutide was initially approved for patients with type 2 diabetes and has been shown to cause a sustained weight loss of ~ 3 kg in clinical

Postgrad Med, 2015; 127(5):494–502

trials [108]. This is indeed modest but still useful for treating the obese patient with diabetes as several current glucoselowering therapies, including insulin as well as sulfonylureas, are associated with further weight gain. More exciting data from the Phase IIIa SCALE Diabetes trial were recently revealed which showed that liraglutide 3 mg/day achieved a placebo-subtracted weight loss difference of 5.9 kg over 56 weeks. Furthermore, it was also demonstrated that >80% of subjects treated with a 12-week run-in period of low calorie diet followed with liraglutide 3mg, maintained a weight loss of >5% for the study duration [109]. These results are in agreement with a previous study showing significant weight loss with liraglutide in a dose-dependent manner compared to both placebo and orlistat [110]. Weight loss was sustained for over 2 years and a good tolerability profile was observed with liraglutide given up to 3 mg/day [111]. Liraglutide 3 mg/day (Saxenda), as mentioned earlier, has now been approved both in the US and the UK as monotherapy for obesity in patients without diabetes. A common side effect of GLP-1 analogs is nausea. Nausea is likely to reflect the extreme end of the spectrum of GLP-1’s anorectic effects, with a gray zone in between where satiety is achieved without inducing nausea. However, nausea appears to subside with long-term therapy in the majority of patients [112]. Recently, concerns have been raised regarding the increased risks of pancreatitis, pancreatic and medullary thyroid cancer related to the long-term use of GLP-1 analogs [113]. This has fuelled much debate; however, the overall consensus is that the benefits far outweigh the risks and although caution needs to be exercised, there is insufficient evidence to firmly link GLP-1 analogs to the above mentioned adverse effects [114,115]. Pancreatic polypeptide PP is a 36 amino acid peptide secreted from the Islets of Langerhans in response to eating. It is believed to preferentially act on neuropeptide Y4 receptors within the hypothalamus and brain stem to promote satiety, although there is also some evidence for its anorectic action to be mediated via the vagus [116]. Intravenous infusion of PP has been shown to achieve food intake reduction of 25% over 24 hours in lean healthy volunteers [117]. Furthermore, a Phase Ia clinical trial of PP1420, a long-acting potent PP analog, showed promise in terms of safety and tolerability profile when given as a subcutaneous injection [118]. Phase Ib and Ic studies, which will focus on further safety and efficacy parameters such as food intake, at various ascending doses, are currently in progress (ClinicalTrials.gov identifier: NCT01052493). Peptide YY PYY is also a 36 amino acid peptide belonging to the same “PP fold” family as PP. It is secreted from the L cells in response to eating and exerts anorectic effects by preferentially acting on neuropeptide Y2 receptors, mostly situated in the arcuate nucleus of the hypothalamus. Batterham et al. demonstrated a reduction in food intake following PYY infusion in obese subjects and a reduction in weight following

DOI: 10.1080/00325481.2015.1048181

Obesity: Lifestyle management, bariatric surgery, drugs, and gut hormones

chronic PYY administration in rodents. They also showed significantly lower basal and postprandial rise in PYY levels in the obese compared to lean control which may suggest a role for PYY in the pathogenesis of obesity [62,119]. Currently, several novel long-acting injectable PYY analogs are under investigation in first-in-human studies (ClinicalTrials.gov identifier: NCT01515319).

Postgraduate Medicine Downloaded from informahealthcare.com by Nyu Medical Center on 06/05/15 For personal use only.

Oxyntomodulin OXM is a 37 amino acid peptide co-secreted with GLP-1 from L cells of the distal gut following a meal. OXM binds to both GLP-1 and glucagon receptors although with lower affinity compared to the individual peptides. Therefore, OXM represents an attractive possibility to exploit dual receptors and augment weight loss. This approach combines the appetite suppressive effects of GLP-1 and glucagon, with the energy expenditure increasing effects of glucagon [120,121]. In fact, peripheral infusion and chronic subcutaneous injection of OXM resulted in reduction of food intake, weight loss, and increase in activity-related energy expenditure in humans [61,63]. The concept of dual receptor agonism at both the GLP-1 and glucagon receptors is an exciting prospect and analogs such as ZP2929 by Zealand Pharma combining these properties are under development [122].

Conclusion There is no doubt that gut hormones have a key role in the regulation of appetite and weight and we have come a long way in both our understanding of this intricate neuroendocrine system as well as in using this knowledge to advance anti-obesity drug development. Bariatric surgery has set the benchmark and neither gut hormone analogs nor any of the current medical therapies can rival its dramatic effects on weight. In attempting to understand the mechanisms by which bariatric surgery exerts its effects, a novel therapeutic approach to the treatment of obesity has been uncovered. Perhaps the secret of its success lies in the changes of gut hormone profiles observed post-surgery. The elevations of at least three different anorexic gut hormones after RYGB may give rise to an additive or even synergistic effect on inducing and maintaining weight loss. It might also explain why single gut hormone agonists such as liraglutide do not achieve weight loss on the same scale as RYGB surgery. Our best chances of unraveling a “medical bypass” may be to mimic the changes seen in the post-RYGB hormonal milieu with combination drug therapy or designing dual or even triple receptor gut hormone agonists.

Declaration of interest The section is funded by grants from the MRC, BBSRC, NIHR, an Integrative Mammalian Biology Capacity Building Award, an FP7- HEALTH- 2009-241592 EuroCHIP grant and is supported by the NIHR Imperial Biomedical Research Centre Funding Scheme. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the

499

subject matter or materials discussed in the manuscript apart from those disclosed.

References [1] WHO. Report of the advisory committee on health research. Geneva: Genomics and World Health; 2002. [2] Tunstall- Pedoe H, Tunstall-Pedoe H. Preventing chronic diseases. A Vital investment: WHO global report. World Health Organization, Geneva; 2005. pp 200. Available from http:// www.who.int/chp/chronic_disease_report/en/. Int J Epidemiol 2006;35(4):1107. [3] Vasant H. Adult anthropometric measures, overweight and obesity. In Health Survey for England. Volume 1. Adult anthropometric measures, overweight and obesity In Health Survey for England. Health Social Care Heath Centre 2010. [4] Bridges ST. Children’s BMI, overweight and obesity. Health Survey Engl 2010. [5] Flegal KM, Carroll MD, Kit BK, Ogden CL. Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999-2010. JAMA 2012;307:491–7. [6] Ogden CL, Carroll MD, Kit BK, Flegal KM. Prevalence of obesity and trends in body mass index among US children and adolescents, 1999-2010. JAMA 2012;307:483–90. [7] Fried M, Yumuk V, Oppert JM, Scopinaro N, Torres A, Weiner R, et al. Interdisciplinary European guidelines on metabolic and bariatric surgery. Obes Surg 2014;24:42–55. [8] Swinburn BA, Sacks G, Hall KD, McPherson K, Finegood DT, Moodie ML, Gortmaker SL. The global obesity pandemic: shaped by global drivers and local environments. Lancet 2011;378:804–14. [9] Johansson K, Sundstro€m J, Marcus C, Hemmingsson E, Neovius M. Risk of symptomatic gallstones and cholecystectomy after a very-low-calorie diet or low-calorie diet in a commercial weight loss program: 1-year matched cohort study. Int J Obesity 2014;38:279–84. [10] Tsai AG, Wadden TA. The evolution of very-low-calorie diets: an update and meta-analysis. Obesity 2006;14:1283–93. [11] Avenell A, Brown TJ, McGee MA, Campbell MK, Grant AM, Broom J, et al. What are the long-term benefits of weight reducing diets in adults? A systematic review of randomized controlled trials. J Human Nutr Diet 2004;17:317–35. [12] Malik VS, Hu FB. Popular weight-loss diets: from evidence to practice. Nat Clin Pract Cardiov Med 2007;4:34–41. [13] Bazzano LA, Hu T, Reynolds K, Yao L, Bunol C, Liu Y, et al. Effects of Low-Carbohydrate and Low-Fat Diets. Ann Int Med 2014;161:309. [14] Sacks FM, Bray GA, Carey VJ, Smith SR, Ryan DH, Anton SD, et al. Comparison of weight-loss diets with different compositions of fat, protein, and carbohydrates. N Eng J Med 2009;360:859–73. [15] Johnston BC, Kanters S, Bandayrel K, Wu P, Naji F, Siemieniuk RA, et al. Comparison of Weight Loss Among Named Diet Programs in Overweight and Obese Adults. JAMA 2014;312:923. [16] Wing RR, Lang W, Wadden TA, Safford M, Knowler WC, Bertoni AG, et al. Benefits of modest weight loss in improving cardiovascular risk factors in overweight and obese individuals with type 2 diabetes. Diabetes Care 2011;34:1481–6. [17] Lim EL, Hollingsworth KG, Aribisala BS, Chen MJ, Mathers JC, Taylor R. Reversal of type 2 diabetes: normalisation of beta cell function in association with decreased pancreas and liver triacylglycerol. Diabetologia 2011;54:2506–14. [18] Damms-Machado A, Weser G, Bischoff SC. Micronutrient deficiency in obese subjects undergoing low calorie diet. Nutr J 2012;11:34. [19] Curioni CC, Lourenc¸o PM. Long-term weight loss after diet and exercise: a systematic review. Int J Obesity 2005;29:1168–74. [20] Douketis JD, Macie C, Thabane L, Williamson DF. Systematic review of long-term weight loss studies in obese adults: clinical significance and applicability to clinical practice. Int J Obesity 2005;29:1153–67. [21] Dansinger ML, Gleason JA, Griffith JL, Selker HP, Schaefer EJ. Comparison of the Atkins, Ornish, Weight Watchers, and Zone

500

[22] [23] [24]

[25]

[26]

Postgraduate Medicine Downloaded from informahealthcare.com by Nyu Medical Center on 06/05/15 For personal use only.

[27] [28] [29]

[30]

[31] [32]

[33]

[34] [35] [36]

[37] [38]

[39] [40]

[41] [42]

P. Behary et al. diets for weight loss and heart disease risk reduction: a randomized trial. JAMA 2005;293:43–53. Keys A. Human starvation and its consequences. J Am Diet Assoc 1946;22:582–7. Lomer M. Advanced Nutrition and dietetics in Gastroenterology. Wiley Blackwell, West Sussex, UK; 2014. Sumithran P, Prendergast LA, Delbridge E, Purcell K, Shulkes A, Kriketos A, Proietto J. Long-term persistence of hormonal adaptations to weight loss. N Eng J Med 2011;365: 1597–604. Ebbeling CB, Swain JF, Feldman HA, Wong WW, Hachey DL, Garcia-Lago E, Ludwig DS. Effects of dietary composition on energy expenditure during weight-loss maintenance. JAMA 2012;307:2627–34. Leibel RL, Rosenbaum M, Hirsch J. Changes in energy expenditure resulting from altered body weight. N Eng J Med 1995;332:621–8. Camps SG, Verhoef SP, Westerterp KR. Weight loss, weight maintenance, and adaptive thermogenesis 1-3. Am J Clin Nutr 2013;97:990–5. Ochner CN, Barrios DM, Lee CD, Pi-Sunyer FX. Biological mechanisms that promote weight regain following weight loss in obese humans. Physiol Behav 2013;120:106–13. Rosenbaum M, Sy M, Pavlovich K, Leibel RL, Hirsch J. Leptin reverses weight loss-induced changes in regional neural activity responses to visual food stimuli. J Clin Investig 2008; 118:2583–91. Hinkle W, Cordell M, Leibel R, Rosenbaum M, Hirsch J. Effects of reduced weight maintenance and leptin repletion on functional connectivity of the hypothalamus in obese humans. PLoS One 2013;8:e59114. Catenacci VA, Wyatt HR. The role of physical activity in producing and maintaining weight loss. Nat Clin Pract Endocrinol Metabol 2007;3:518–29. Tate DF, Jeffery RW, Sherwood NE, Wing RR. Long-term weight losses associated with prescription of higher physical activity goals. Are higher levels of physical activity protective against weight regain? Am J Clin Nutr 2007;85:954–9. NICE guidelines [CG189]. Obesity: identification, assessment and management of overweight and obesity in children, young people and adults 1-recommendations Guidance and guidelines. 2014. Physical Activity Guidelines for Americans. 2008. Available from: http://www.health.gov/paguidelines/guidelines/chapter4. aspx. Trost SG, Owen N, Bauman AE, Sallis JF, Brown W. Correlates of adults’ participation in physical activity: review and update. Med Sci Sports Exercise 2002;34:1996–2001. Jensen MD, Ryan DH, Apovian CM, Ard JD, Comuzzie AG, Donato KA, et al. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: A report of the American college of cardiology/American heart association task force on practice guidelines and the obesity society. J Am Coll Cardiol 2014;63:2985–3023. Kayman S, Bruvold W, Stern JS. Maintenance and relapse after weight loss in women: behavioral aspects. Am J Clin Nutr 1990;52:800–7. WHO. Obesity: preventing and managing the global epidemic. Report of WHO Consultation on Obesity. WHO Technical. World Health Organization, Geneva; 2000. Available from http://www.who.int/nutrition/publications/obesity/ WHO_TRS_894/en/. NIH conference. Gastrointestinal surgery for severe obesity. consensus development conference panel. Ann Int Med 1991;115:956–61. Busetto L, Dixon J, De Luca M, Shikora S, Pories W, Angrisani L. 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;24:487–519. Angrisani L, Santonicola A, Iovino P, Formisano G, Buchwald H, Scopinaro N. Bariatric Surgery Worldwide 2013. Obesity Surgery 2015. [Epub ahead of print]. Slater GH, Ren CJ, Siegel N, Williams T, Barr D, Wolfe B, et al. Serum fat-soluble vitamin deficiency and abnormal

Postgrad Med, 2015; 127(5):494–502

[43]

[44]

[45]

[46] [47] [48]

[49]

[50]

[51]

[52]

[53] [54]

[55]

[56]

[57]

[58]

[59]

[60]

[61]

calcium metabolism after malabsorptive bariatric surgery. J Gastroint Surg 2004;8:48–55; discussion 54–5. Aasheim ET, Bjo S, Hanvold SE, Engstro€m M, Hanvold SE, Mala T, et al. Vitamin status after bariatric surgery: a randomized study of gastric bypass and duodenal switch 1 – 3. Am J Clin Nutr 2009;90:15–22. Buchwald H, Estok R, Fahrbach K, Banel D, Jensen MD, Pories WJ, et al. Weight and type 2 diabetes after bariatric surgery: systematic review and meta-analysis. Am J Med 2009;122:248–56. Pournaras DJ, Aasheim ET, Søvik TT, Andrews R, Mahon D, Welbourn R, et al. Effect of the definition of type II diabetes remission in the evaluation of bariatric surgery for metabolic disorders. Br J Surg 2012;99:100–3. Mingrone G, Panunzi S, De Gaetano A, Guidone C, Iaconelli A, Leccesi L, et al. Bariatric surgery versus conventional medical therapy for type 2 diabetes. N Eng J Med 2012;366:1577–85. Sjo€stro€m L. Review of the key results from the Swedish Obese Subjects (SOS) trial - a prospective controlled intervention study of bariatric surgery. J Int Med 2013;273:219–34. Trastulli S, Desiderio J, Guarino S, Frampton GK. Laparoscopic sleeve gastrectomy compared with other bariatric surgical procedures: a systematic review of randomized trials. Surgery Obesity Related Dis 2013;9:816–29. Wang MC, Guo XH, Zhang YW, Zhang YL, Zhang HH, Zhang YC. 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;81:166–71. Diamantis T, Apostolou KG, Alexandrou A, Frampton GK. Review of long-term weight loss results after laparoscopic sleeve gastrectomy. Surgery Obesity Related Dis 2014;10:177– 83. Schauer PR, Bhatt DL, Kirwan JP, Wolski K, Brethauer SA, Navaneethan SD, et al. Bariatric surgery versus intensive medical therapy for diabetes–3-year outcomes. N Eng J Med 2014;370:2002–13. Dirksen C, Jørgensen NB, Bojsen-Møller KN, Jacobsen SH, Hansen DL, Worm D, et al. Mechanisms of improved glycaemic control after Roux-en-Y gastric bypass. Diabetologia 2012;55:1890–901. Cummings DE, Overduin J, Shannon MH, Foster-Schubert KE. Hormonal mechanisms of weight loss and diabetes resolution after bariatric surgery. Sur Obesity Relat Dis 2005;1:358–68. Cummings DE, Weigle DS, Frayo RS, Breen PA, Ma MK, Dellinger EP, Purnell JQ. Plasma ghrelin levels after dietinduced weight loss or gastric bypass surgery. N Eng J Med 2002;346:1623–30. Borg CM, le Roux CW, Ghatei MA, Bloom SR, Patel AG, Aylwin SJ. Progressive rise in gut hormone levels after Roux-en-Y gastric bypass suggests gut adaptation and explains altered satiety. Br J Surg 2006;93:210–15. Le Roux CW, Welbourn R, Werling M, Osborne A, Kokkinos A, Laurenius A, et al. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann Surg 2007;246:780–5. Roux CW, Aylwin SJ, Batterham RL, Borg CM, Coyle F, Prasad V, et al. Gut hormone profiles following bariatric surgery favor an anorectic state, facilitate weight loss, and improve metabolic parameters. Ann Surg 2006;243:108–14. Näslund E, Backman L, Holst JJ, Theodorsson E, Hellstro€m PM. Importance of small bowel peptides for the improved glucose metabolism 20 years after jejunoileal bypass for obesity. Obesity Surg 1998;8:253–60. MacDonald PE, El-kholy W, Riedel MJ, Salapatek AM, Light PE, Wheeler MB. The multiple actions of GLP-1 on the process of glucose-stimulated insulin secretion. Diabetes 2002;51:S434–42. Verdich C, Flint A, Gutzwiller JP, Näslund E, Beglinger C, Hellstro€m PM, et al. A meta-analysis of the effect of glucagonlike peptide-1 (7-36) amide on ad libitum energy intake in humans. J Clin Endocrinol Metab 2001;86:4382–9. Cohen MA, Ellis SM, Le Roux CW, Batterham RL, Park A, Patterson M, et al. Oxyntomodulin suppresses appetite and

DOI: 10.1080/00325481.2015.1048181

[62] [63]

[64]

[65]

Postgraduate Medicine Downloaded from informahealthcare.com by Nyu Medical Center on 06/05/15 For personal use only.

[66]

[67]

[68]

[69]

[70] [71]

[72] [73]

[74]

[75]

[76]

[77] [78]

[79]

Obesity: Lifestyle management, bariatric surgery, drugs, and gut hormones

reduces food intake in humans. J Clin Endocrinol Metab 2003;88:4696–701. Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, Frost GS, et al. Inhibition of food intake in obese subjects by peptide YY3-36. N Eng J Med 2003;349:941–8. Wynne K, Park AJ, Small CJ, Meeran K, Ghatei MA, Frost GS, Bloom SR. Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomised controlled trial. Int J Obesity 2006;30:1729–36. Fenske WK, Bueter M, Miras a D, Ghatei MA, Bloom SR, le Roux CW. Exogenous peptide YY3-36 and Exendin-4 further decrease food intake, whereas octreotide increases food intake in rats after Roux-en-Y gastric bypass. Int J Obesity 2012;36: 379–84. Falken Y, Hellstro€m PM, Holst JJ, Näslund E. Changes in glucose homeostasis after Roux-en-Y gastric bypass surgery for obesity at day three, two months, and one year after surgery: Role of gut peptides. J Clin Endoc Metab 2011;96:2227–35. Dirksen C, Jørgensen NB, Bojsen-Møller KN, Kielgast U, Jacobsen SH, Clausen TR, et al. Gut hormones, early dumping and resting energy expenditure in patients with good and poor weight loss response after Roux-en-Y gastric bypass. Int J Obesity 2013;37:1452–9. Jimenez A, Casamitjana R, Viaplana-Masclans J, Lacy A, Vidal J. GLP-1 action and glucose tolerance in subjects with remission of type 2 diabetes after gastric bypass surgery. Diabetes Care 2013;36:2062–9. Mokadem M, Zechner JF, Margolskee RF, Drucker DJ, Aguirre V. Effects of Roux-en-Y gastric bypass on energy and glucose homeostasis are preserved in two mouse models of functional glucagon-like peptide-1 deficiency. Mol Metab 2014;3:191–201. Jørgensen NB, Dirksen C, Bojsen-møller KN, Jacobsen SH, Worm D, Hansen DL, et al. Exaggerated glucagon-like peptide 1 response is important for improved b-cell function and glucose tolerance after roux-en-y gastric bypass in patients with type 2 diabetes. 2014;62:3044–52. Gribble FM. The gut endocrine system as a coordinator of postprandial nutrient homoeostasis. Proc Nutr Soc 2012;71:456–62. Hao Z, Townsend RL, Mumphrey MB, Patterson LM, Ye J, Berthoud HR. Vagal innervation of intestine contributes to weight loss After Roux-en-Y gastric bypass surgery in rats. Obesity Surg 2014;24:2145–51. Gautron L, Zechner JF, Aguirre V. Vagal innervation patterns following Roux-en-Y gastric bypass in the mouse. Int J Obesity 2013;37:1603–7. Le Roux CW, Neary NM, Halsey TJ, Small CJ, Martinez-Isla AM, Ghatei MA, et al. Ghrelin does not stimulate food intake in patients with surgical procedures involving vagotomy. J Clin Endocrinol Metab 2005;90:4521–4. Werling M, Vincent RP, Cross GF, Marschall HU, Fändriks L, Lo €nroth H, et al. Enhanced fasting and post-prandial plasma bile acid responses after Roux-en-Y gastric bypass surgery. Scand J Gastroenterol 2013;48:1257–64. Peterli R, Steinert RE, Woelnerhanssen B, Peters T, Christoffel-Courtin C, Gass M, et al. Metabolic and hormonal changes after laparoscopic Roux-en-Y gastric bypass and sleeve gastrectomy: a randomized, prospective trial. Obesity Surg 2012;22:740–8. Pournaras DJ, Glicksman C, Vincent RP, Kuganolipava S, Alaghband-Zadeh J, Mahon D, et al. The role of bile after Roux-en-Y gastric bypass in promoting weight loss and improving glycaemic control. Endocrinology 2012;153:3613–19. Li JV, Ashrafian H, Bueter M, Kinross J, Sands C, le Roux CW, et al. Metabolic surgery profoundly influences gut microbialhost metabolic cross-talk. Gut 2011;60:1214–23. Liou AP, Paziuk M, Luevano JM, Machineni S, Turnbaugh PJ, Kaplan LM. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med 2013;5:178ra41. Sweeney TE, Morton JM. Metabolic surgery: action via hormonal milieu changes, changes in bile acids or gut microbiota? A summary of the literature. Best Pract Res Clin Gastroenterol 2014;28:727–40.

501

[80] Laferrère B, Teixeira J, McGinty J, Tran H, Egger JR, Colarusso A, et al. Effect of weight loss by gastric bypass surgery versus hypocaloric diet on glucose and incretin levels in patients with type 2 diabetes. J Clin Endocrinol Metab 2008;93:2479–85. [81] Korner J, Inabnet W, Febres G, Conwell IM, McMahon DJ, Salas R, et al. Prospective study of gut hormone and metabolic changes after adjustable gastric banding and Roux-en-Y gastric bypass. Int J Obesity 2009;33:786–95. [82] Olbers T, Bjo€rkman S, Lindroos A, Maleckas A, Lo €nn L, Sjo€stro€m L, Lo €nroth H. Body composition, dietary intake, and energy expenditure after laparoscopic Roux-en-Y gastric bypass and laparoscopic vertical banded gastroplasty: a randomized clinical trial. Ann Surg 2006;244:715–22. [83] Scholtz S, Miras AD, Chhina N, Prechtl CG, Sleeth ML, Daud NM, et al. Obese patients after gastric bypass surgery have lower brain-hedonic responses to food than after gastric banding. Gut 2014;63:891–902. [84] Buchwald H, Estok R, Fahrbach K, Banel D, Sledge I. Trends in mortality in bariatric surgery: a systematic review and metaanalysis. Surgery 2007;142:621–32. [85] Dixon JB, Zimmet P, Alberti KG, Rubino F. Bariatric surgery: an IDF statement for obese Type 2 diabetes. Diabetic Med 2011;28:628–42. [86] Jackson TD, Zhang R, Glockler D, Pennington J, Reddigan JI, Rotstein OD, et al. Health inequity in access to bariatric surgery: a protocol for a systematic review. System Rev 2014;3:15. [87] Ahmad A, Laverty AA, Aasheim E, Majeed A, Millett C, Saxena S. Eligibility for bariatric surgery among adults in England: analysis of a national cross-sectional survey. JRSM open 2014;5:2042533313512479. [88] European Medicines Agency. Draft Guideline on clinical evaluation of medicinal products used in weight control. 2014. Available from http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2014/07/WC500170278.pdf. [89] Colman E. Food and drug administration’s obesity drug guidance document: a short history. Circulation 2012;125:2156–64. [90] James WP, Caterson ID, Coutinho W, Finer N, Van Gaal LF, Maggioni AP, et al. Effect of sibutramine on cardiovascular outcomes in overweight and obese subjects. N Eng J Med 2010;363:905–17. [91] Samat A, Tomlinson B, Taheri S, Thomas GN. Rimonabant for the treatment of obesity. Recent Patents Cardiov Drug Discov 2008;3:187–93. [92] Li Z, Maglione M, Tu W, Mojica W, Arterburn D, Shugarman LR, et al. Meta-analysis: pharmacologic treatment of obesity. Ann Int Med 2005;142:532–46. [93] Torgerson JS, Hauptman J, Boldrin MN, Sjo€stro€m L. XENical in the prevention of diabetes in obese subjects (XENDOS) study: a randomized study of orlistat as an adjunct to lifestyle changes for the prevention of type 2 diabetes in obese patients. Diabetes Care 2004;27:155–61. [94] Colman E, Golden J, Roberts M, Egan A, Weaver J, Rosebraugh C. The FDA’s assessment of two drugs for chronic weight management. N Eng J Med 2012;367:1577–9. [95] Smith SR, Weissman NJ, Anderson CM, Sanchez M, Chuang E, Stubbe S, et al. Multicenter, placebo-controlled trial of lorcaserin for weight management. N Eng J Med 2010;363:245–56. [96] Gadde KM, Allison DB, Ryan DH, Peterson CA, Troupin B, Schwiers ML, Day WW. Effects of low-dose, controlled-release, phentermine plus topiramate combination on weight and associated comorbidities in overweight and obese adults (CONQUER): a randomised, placebo-controlled, phase 3 trial. Lancet 2011;377:1341–52. [97] Press Announcements. FDA approves weight-management drug Saxenda [Internet]. 2015. Available from http://www.fda. gov/NewsEvents/Newsroom/PressAnnouncements/ucm427913. htm. [98] Press Announcements. FDA approves weight-management drug Contrave [Internet]. 2014. Available from http://www.fda.gov/ NewsEvents/Newsroom/PressAnnouncements/ucm413896.htm. [99] European Medicines Agency. News and Events - Saxenda recommended for approval in weight management in adults [Internet]. 2015. Available from http://www.ema.europa.eu/ema/

502

[100] [101]

[102]

Postgraduate Medicine Downloaded from informahealthcare.com by Nyu Medical Center on 06/05/15 For personal use only.

[103]

[104]

[105] [106]

[107] [108]

[109]

[110]

P. Behary et al. index.jsp?curl=pages/news_and_events/news/2015/01/news_detail_002255.jsp&mid=WC0b01ac058004d5c1. Ornellas T, Chavez B. Naltrexone SR/Bupropion SR (Contrave): A new approach to weight loss in obese adults. P T 2011;36:255–62. Greenway FL, Fujioka K, Plodkowski RA, Mudaliar S, Guttadauria M, Erickson J, et al. Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (CORI): A multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2010;376:595–605. Apovian CM, Aronne L, Rubino D, Still C, Wyatt H, Burns C, et al. A randomized, phase 3 trial of naltrexone SR/bupropion SR on weight and obesity-related risk factors (COR-II). Obesity 2013;21:935–43. Hollander P, Gupta AK, Plodkowski R, Greenway F, Bays H, Burns C, et al. Effects of naltrexone sustained-release/bupropion sustained-release combination therapy on body weight and glycemic parameters in overweight and obese patients with type2 diabetes. Diabetes Care 2013;36:4022–9. Meeran K, O’Shea D, Edwards CM, Turton MD, Heath MM, Gunn I, et al. Repeated intracerebroventricular administration of glucagon-like peptide-1-(7-36) amide or exendin-(9-39) alters body weight in the rat. Endocrinology 1999;140:244–50. Turton MD, O’Shea D, Gunn I, Beak SA, Edwards CM, Meeran K, et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 1996;379:69–72. Parker JA, McCullough KA, Field BC, Minnion JS, Martin NM, Ghatei MA, Bloom SR. Glucagon and GLP-1 inhibit food intake and increase c-fos expression in similar appetite regulating centres in the brainstem and amygdala. Int J Obesity 2013;37:1391–8. Trujillo JM, Nuffer W, Ellis SL. GLP-1 receptor agonists: a review of head-to-head clinical studies. Ther Adv Endocrinol Metab 2015;6:19–28. Buse JB, Rosenstock J, Sesti G, Schmidt WE, Montanya E, Brett JH, et al. Liraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6). Lancet 2009;374:39–47. Wadden TA, Hollander P, Klein S, Niswender K, Woo V, Hale PM, et al. Weight maintenance and additional weight loss with liraglutide after low-calorie-diet-induced weight loss: the SCALE Maintenance randomized study. Int J Obesity 2013;37:1443–51. Astrup A, Ro €ssner S, Van Gaal L, Rissanen A, Niskanen L, Al Hakim M, et al. Effects of liraglutide in the treatment of

Postgrad Med, 2015; 127(5):494–502

[111]

[112]

[113]

[114] [115] [116]

[117]

[118]

[119] [120]

[121] [122]

obesity: a randomised, double-blind, placebo-controlled study. Lancet 2009;374:1606–16. Astrup A, Carraro R, Finer N, Harper A, Kunesova M, Lean ME, et al. Safety, tolerability and sustained weight loss over 2 years with the once-daily human GLP-1 analog, liraglutide. Int J Obesity 2012;36:843–54. Ratner RE, Maggs D, Nielsen LL, Stonehouse AH, Poon T, Zhang B, et al. Long-term effects of exenatide therapy over 82 weeks on glycaemic control and weight in over-weight metformin-treated patients with type 2 diabetes mellitus. Diabetes 2006;8:419–28. Elashoff M, Matveyenko A V, Gier B, Elashoff R, Butler PC. Pancreatitis, pancreatic, and thyroid cancer with glucagon-like peptide-1-based therapies. Gastroenterology 2011;141:150–6. Nauck MA. A critical analysis of the clinical use of incretinbased therapies: The benefits by far outweigh the potential risks. Diabetes Care 2013;36:2126–32. Nauck MA, Friedrich N. Do GLP-1-based therapies increase cancer risk? Diabetes Care 2013;36:S245–52. Asakawa A, Inui A, Yuzuriha H, Ueno N, Katsuura G, Fujimiya M, et al. Characterization of the effects of pancreatic polypeptide in the regulation of energy balance. Gastroenterology 2003;124:1325–36. Batterham RL, Le Roux CW, Cohen MA, Park AJ, Ellis SM, Patterson M, et al. Pancreatic polypeptide reduces appetite and food intake in humans. J Clin Endocrinol Metab 2003;88:3989–92. Tan TM, Field BCT, Minnion JS, Cuenco-Shillito J, Chambers ES, Zac-Varghese S, et al. Pharmacokinetics, adverse effects and tolerability of a novel analogue of human pancreatic polypeptide, PP 1420. Br J Clin Pharmacol 2012;73:232–9. Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, et al. Gut hormone PYY(3-36) physiologically inhibits food intake. Nature 2002;418:650–4. Tan TM, Field BCT, McCullough KA, Troke RC, Chambers ES, Salem V, et al. Coadministration of glucagon-like peptide-1 during glucagon infusion in humans results in increased energy expenditure and amelioration of hyperglycemia. Diabetes 2013;62:1131–8. Schulman JL, Carleton JL, Whitney G, Whitehorn JC. Effect of glucagon on food intake and body weight in man. J Appl Physiol 1957;11:419–21. Fosgerau K, Hoffmann T. Peptide therapeutics: Current status and future directions. Drug Discov Today 2015;20:122–8.

Obesity: Lifestyle management, bariatric surgery, drugs, and the therapeutic exploitation of gut hormones.

Obesity is on the rise and the pursuit of efficient and safe treatment is ongoing. Available anti-obesity medical therapies have so far proved to be d...
473KB Sizes 0 Downloads 11 Views