Review

Get Permission
Rev Diabet Stud, 2011, 8(3):339-347 DOI 10.1900/RDS.2011.8.339

Therapy for Obesity Based on Gastrointestinal Hormones

Jonatan I. Bagger, Mikkel Christensen, Filip K. Knop, Tina Vilsboll

Diabetes Research Division, Department of Internal Medicine F, Gentofte Hospital, University of Copenhagen, Denmark
Address correspondence to: Tina Vilsboll, e-mail: t.vilsboll@dadlnet.dk

Manuscript submitted September 24, 2011; resubmitted October 24, 2011; accepted October 28, 2011.

Keywords: DPP-4 inhibitors, GLP-1 receptor agonist, incretin hormone, obesity, type 2 diabetes

Abstract

It has long been known that peptide hormones from the gastrointestinal tract have significant impact on the regulation of nutrient metabolism. Among these hormones, incretins have been found to increase insulin secretion, and thus incretin-based therapies have emerged as new modalities for the treatment of type 2 diabetes. In contrast to other antidiabetic treatments, these agents have a positive outcome profile on body weight. Worldwide there are 500 million obese people, and 3 million are dying every year from obesity-related diseases. Recently, incretin-based therapy was proposed for the treatment of obesity. Currently two different incretin therapies are widely used in the treatment of type 2 diabetes: 1) the GLP-1 receptor agonists which cause significant and sustained weight loss in overweight patients, and 2) dipeptidyl peptidase 4 (DPP-4) inhibitors being weight neutral. These findings have led to a greater interest in the physiology of intestinal peptides with potential weight-reducing properties. This review discusses the effects of the incretin-based therapies in obesity, and provides an overview of intestinal peptides with promising effects as potential new treatments for obesity.

Abbreviations: BMI - body mass index; CCK - cholecystokinin; CI - confidence interval; CNS - central nervous system; DPP-4 - dipeptidyl peptidase 4; EMA - European Medicines Agency; GIP - glucose-dependent insulinotropic polypeptide; GLP-1 - glucagon-like peptide-1; GLP-1R - GLP-1 receptor; GOAT - ghrelin O-acyl tranferase; NPY - neuropeptide Y; PP - pancreatic polypeptide; PYY - peptide YY; WHO - World Health Organization; Y - tyrosine

Introduction

Worldwide, obesity has more than doubled since 1980 [1, 2]. In the USA, more than two thirds of the population is overweight (body mass index (BMI) 25.0-29.9 kg/m2), or obese (BMI ≥ 30 kg/m2). The proportion of the European population having weight problems is smaller, but the number continues to increase [1]. The most recent data from the World Health Organization (WHO) indicate that 1.5 billion adults worldwide are overweight, and 500 million are obese. It is particularly concerning that nearly 50 million children under the age of five are overweight [2]. The main problem seems to be the western lifestyle, combined with a genetic predisposition, which leads to obesity, type 2 diabetes, fatty liver disease, and eventually cardiovascular disease. Almost 3 million adults die each year as a result of being overweight, or obese. It is estimated that approximately 44% of the diabetes burden, 23% of the ischemic heart disease burden, and between 7% and 41% of certain cancer burdens are attributable to overweight and obesity [2]. By 2015, WHO projects that worldwide 2.3 billion adults will be overweight, with more than 700 million being obese.

Unfortunately, weight loss is not easily accomplished, or maintained. Meta-analyses of clinical trials on non-pharmacologic strategies for weight reduction report only modest results (i.e. weight reductions of 1 to 6 kg) that are short-lived [3-5]. Pharmacologic weight loss interventions have shown similar limited success [6-8]. Meta-analyses of sibutamine and orlistat trials, report average reductions of 3 to 5 kg; but attrition rates tend to be very high in the included trials, with almost 50% of the patients leaving the trials prematurely [6-8]. The high attrition rates, and the fact that many trials had an inadequate control bias, suggest that real-world use of these agents may be less successful [6-9]. Other meta-analyses show that bariatric surgery is associated with robust body weight-reducing effects, and suggest that certain bariatric procedures may reduce long-term mortality in obese patients [10, 11]. Thus, the combined evidence suggests that bariatric surgery may be a beneficial option for some obese patients. On the other hand, the safety and the cost of bariatric surgery limit the use of this intervention for large populations of patients.

The risk of developing diabetes escalates with the degree of excess weight, increasing 3-fold with a BMI of 25.0 to 29.9 kg/m2, and 20-fold with a BMI of ≥35 kg/m2 (compared with a BMI of 18.5 to 24.9 kg/m2) [12]. Thus, for each unit of increase in BMI, the risk for developing diabetes increases by approximately 12% [13]. On top of this, most of the currently available drugs for type 2 diabetes are associated with body weight increase (thiazolidinediones, sulphonylureas, and insulin) [14]. However, with the new incretin-based therapies for diabetes, positive outcomes on obesity-related parameters have been reported [15]. The incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are gut hormones secreted from endocrine cells in the intestinal mucosa acting as key regulators of the glucose-dependent alpha-cell and beta-cell responses in the pancreatic islets of Langerhans. Moreover, GLP-1 has body weight-reducing capabilities. In addition to GLP-1, the endocrine cells in the mucosal layer of the intestinal tract produce a wide range of substances known to influence appetite and food intake. This review summarizes the effects of incretin-based therapies in obesity, and also gives an overview of intestinal peptides with promising effects as potential new treatments for obesity.

Glucagon-like peptide-1 (GLP-1) physiology

GLP-1 is an incretin hormone released from the endocrine L-cells, situated primarily in the distal part of the ileum and in the colon, in response to ingestion of nutrients [16]. GLP-1 is processed from proglucagon and is rapidly degraded by the enzyme dipeptidyl peptidase 4 (DPP-4) [17]. DPP-4 cleaves off the two N-terminal amino acids, and leaves the molecule inactive, resulting in a half-life of less than 2 minutes [18]. Because of this rapid elimination, native GLP-1 is unsuitable for clinical use. GLP-1 potentiates glucose-induced insulin secretion and inhibits glucagon secretion, thereby improving glucose homeostasis. GLP-1 receptors (GLP-1R) are expressed in many regions of the brain and in particular in the arcuate nucleus and other hypothalamic regions involved in the regulation of food intake [19]. Correspondingly, animal studies using the GLP-1R antagonist, exendin(9-39), have demonstrated that GLP-1R activation is important in the regulation of appetite and food intake [20, 21].

The mechanisms behind the anorectic actions of gut-derived GLP-1 seems to be mediated through both central and peripheral mechanisms [22-25]. Interestingly, in a study where a destruction of the arcuate nucleus was induced, the inhibitory effect of intra-cerebroventricular GLP-1 administration on food intake and appetite disappeared. Whereas, the appetite suppressive effects of peripherally administered GLP-1 was maintained [26]. The study indicates that the arcuate nucleus is essential for the central action of GLP-1, but not for the peripheral action.

GLP-1 is recognized as a neurotransmitter involved in the regulation of appetite and food intake [21, 24]. Furthermore, in rats GLP-1-producing neurons are activated by distension of the stomach and enteroceptive stress [27, 28]. This could indicate a neuronal transmission of satiety signals from the gastrointestinal tract into the brain through the "brain GLP-1 system". Theoretically, gut-derived GLP-1 could access the brain through leaks in the blood-brain barrier, such as the subfornical organ and the area postrema, as demonstrated in rats [29]. Thus, systemic infusions of GLP-1 have been shown to inhibit food intake in rats, independent of the vagal sensory afferents [30]. On the other hand, studies with large molecular size GLP-1R agonists (albumin-conjugated GLP-1R agonists) in both humans and mice have demonstrated that these large proteins inhibit feeding, even though they probably cannot cross the blood-brain barrier [31, 32]. These studies support that at least some effects of GLP-1 may not be exerted directly through the central nervous system (CNS), but possibly through the afferent neurons of the vagus nerve [33]. The nerve endings of these nerves are placed in the lamina propria of the intestine just beneath the mucosal surface in the space between the secreting cells and the capillary bed, with cell bodies in the nodose ganglion [33, 34]. Because of the massive and rapid degradation of the GLP-1 by DPP-4, the concentration of active GLP-1 'available' for these nerve endings, which are very close to the secretory origin of GLP-1, must be many times higher than for any other target cell.

Furthermore, inhibition of food intake by GLP-1 could be related to the well-described inhibition of gastric motility and emptying occurring after GLP-1 administration [35, 36]. This consideration is in line with the fact that gastric emptying has a central role in the regulation of food intake [37]. However, this mechanism does not stand alone, since GLP-1 also elicits appetite reduction in fasting humans [38]. On top of this, the effects of gastric emptying do not explain the weight-reducing effects of GLP-1R agonists, as the effect of GLP-1 on the gastrointestinal motility wanes with time in contrast to the continuing effect on appetite and food intake [39, 40].

GLP-1 receptor agonists

The GLP-1R agonists are resistant to degradation, and thereby inactivation by DPP-4, although a high affinity to the GLP-1R is maintained. Two different types of GLP-1R agonists have been developed for drug treatment:

1. Peptides based on the lizard venom-extracted peptide exendin-4 having ~50% amino acid sequence homology with native GLP-1 (e.g. exenatide (Byetta®); first market authorization in 2005 [41, 42]).
2. Peptides based on a back bone of the amino acid sequence of native GLP-1 having 97% sequence homology with the native peptide (e.g. liraglutide (Victoza®); first market authorization in 2009) [43]).

Exenatide has a half-life of 4-6 hours and must be administered twice daily, whereas liraglutide can be administered once-daily, because of its half-life of 12-14 hours. In October 2011, exenatide once-weekly was launched in Europe (Bydureon®). Other compounds with long half-lives (dulaglutide and albiglutide), are in late clinical development.

A recent meta-analysis investigated the therapeutic effects of GLP-1R agonists in the treatment of obese subjects with, and without, diabetes [15]. The analysis included twenty-five randomized controlled trials, including a total of more than 6,400 overweight participants treated with exenatide, exenatide once-weekly, or liraglutide for at least 20 weeks. It showed that patients randomized to GLP-1R agonists versus comparator (placebo, sulphonylurea, thiazolidinedione, DPP-4 inhibitor, or insulin) obtained a greater weight loss (weighted mean difference -2.9 kg; 95% confidence interval (CI) -3.6 kg to -2.2 kg). This treatment effect was seen in patients without diabetes (-3.2 kg, 95% CI: -4.3 kg to -2.1 kg) and in patients with type 2 diabetes (-2.8 kg, 95% CI: -3.2 kg to -2.1 kg). These results suggest that GLP-1R agonists might be beneficial as a new treatment of obesity, but long-term studies are needed to verify the effects and to answer safety issues.

In accordance with the meta-analysis mentioned above, a newly published study shows the superior effect of liraglutide compared to orlistat, the only anti-obesity-drug available in Europe [44]. After two years of treatment, a significant mean difference of 3 kg (95% CI: 1.3 kg to 4.7 kg) between the two drugs was seen in favor of liraglutide. In addition to the superior weight loss, liraglutide demonstrated beneficial effects on systolic and diastolic blood pressure, plasma levels of cholesterol and liver enzymes, and glycemic control. On the other hand, liraglutide was associated with nausea, diarrhea, and vomiting [44]. After the approval of exenatide and liraglutide, post-marketing reports of several incidents of acute pancreatitis in patients treated with GLP-1R agonists have been observed [45]. Current knowledge is insufficient to determine whether the incidence of acute pancreatitis is higher in those patients receiving exenatide or liraglutide compared with the background population of patients with type 2 diabetes. The latter group have an almost 3-fold increased risk of pancreatitis compared to a non-diabetic population [46]. It is recommended that GLP-1R agonists should not be used in subjects with a history of or increased risk of pancreatitis. In carcinogenicity studies with liraglutide, thyroid C-cell tumors were observed in mice and rats [47]. However, recent data has identified key differences between rodent models and humans regarding this. Up to the present date, no changes in thyroid function have been reported in clinical trials with GLP-1R agonists [48]. However, the long-term safety of sustained GLP-1R activation in human thyroid tissue requires continuing pharmaco-vigilance [49]. Lately, a debate has arisen on the plausible risk of pancreatic cancer in patients treated with exenatide, as compared to other anti-diabetic medications [50]. The European Medicines Agency (EMA) recently concluded that a causal relationship between GLP-1R agonists and pancreatic malignancies could not be confirmed nor excluded; but again this area requires careful pharmaco-vigilance [51]. Furthermore, large clinical studies are on the way, aiming to assess and confirm the cardiovascular safety of both exenatide and liraglutide.

DPP-4 inhibitors

The effects of GLP-1 can also be exploited by protecting endogenous GLP-1 from degradation by the enzyme DPP-4 [52]. Oral administration of inhibitors of this enzyme increase the circulating levels of active GLP-1 which is associated with anti-diabetic effects [53]. DPP-4 inhibitors are small molecules that are active upon oral administration. They have shown a clinically significant and sustained effect on glycemic control [54]. However, DPP-4 inhibitors have little effect on body weight, presumably because the plasma concentrations of active GLP-1 are not elevated sufficiently to exert this effect [55]. Several DPP-4 inhibitors are undergoing clinical development. Currently four of them (sitagliptin (Januvia®, Merck Sharp & Dohme), vildagliptin (Galvus®, Novartis), saxagliptin (Onglyza®, AstraZeneca/Bristol-Myers Squibb), and linagliptin (Trajenta®, Boehringer Ingelheim)) have been approved as medications for type 2 diabetes. Presumably, none of these agents will be approved for treatment of obesity, because of their overall weight-neutrality.

Perspectives on other intestinal hormones

GLP-1 is not the only intestinal hormone possessing weight-regulating properties. A handful of peptides secreted by the intestinal linings has proven impact on body weight in preclinical settings. Currently, several potential receptor agonists (and also receptor antagonists) of these peptides are being developed. Some of these are even dual receptor agonists with an affinity for two distinctive receptors.

Oxyntomodulin

Oxyntomodulin is a native peptide with affinity for both the glucagon receptor and the GLP-1R [56-58]. It is thus a glucagon-GLP-1 dual agonist. As for GLP-1, it originates from the cleavage of proglucagon, and is released from intestinal L-cells immediately after meal ingestion, with plasma concentrations being closely related to calorie intake [57]. The amino acid sequence of oxyntomodulin corresponds to the entire 29-amino acid sequence of the glucagon molecule, with a C-terminal extension of eight amino acids [59]. Therefore, it was formerly known as enteroglucagon.

The peptide is rapidly degraded by DPP-4 and neprylsin with a half-life of approximately 12 minutes [60, 61]. The effects of acute administration of oxyntomodulin in humans include inhibition of gastric emptying, gastric and pancreatic exocrine secretion, and food intake [60, 62]. Also, repeated subcutaneous administration causes marked weight loss in obese subjects [63]. Weight loss occurs as a result of reduced food intake, and possibly because of increased energy expenditure [64]. As mentioned above, oxyntomodulin acts as an agonist on the glucagon receptor, but with a 10 to 100-fold decreased affinity than glucagon [56, 57].

Nevertheless, an oxyntomodulin analog with an increased affinity for the glucagon receptor in mice, demonstrated increased potency regarding inhibition of food intake and body weight reduction compared to the native oxyntomodulin [65]. This suggests that this potentiated effect was mediated via the glucagon receptor. On the other hand, the central effect of native oxyntomodulin seems to be at least mediated primarily through the GLP-1R, since the effect of oxyntomodulin infused in the rat brain is blocked by the GLP-1R antagonist exendin(9-39) [62]. Also, the effect of oxyntomodulin is abolished in GLP-1R knock-out mice [66].

Peptide YY

Peptide YY (PYY) is a 36-amino acid peptide released by mucosal enteroendocrine L-cells (along with GLP-1 and oxyntomodulin) in response to intraluminal nutrients (postprandial plasma responses are strictly correlated to energy intake) [67]. Carbohydrates, proteins, and lipids all stimulate the secretion of PYY [68]. PYY was originally isolated from porcine intestine, and is characterized by tyrosine (Y) residues at both ends of the molecule, therefore the name [69]. PYY shares considerable homology with both pancreatic polypeptide (PP) and the neurotransmitter neuropeptide Y (NPY) in sequence and tertiary structure. When PYY(1-36) is secreted from the L-cell (or exogenous infused) it is readily cleaved by DPP-4 to form PYY(3-36), which is the major circulating form of PYY [70, 71]. PYY activates three of the mammalian NPY receptors (Y1, Y2, and Y5). PYY(1-36) has a relatively high affinity for all three receptors, whereas PYY(3-36) is a selective Y2 receptor agonist [72]. In humans, PYY(1-36) infusion delays gastric emptying [73], and inhibits gastric acid secretion and gallbladder contraction [74]. However, the anorectic effect of PYY seems to be mediated by by PYY(3-36), but not by PYY(1-36). This appears to be due to the specific affinity of the dominating peptide PYY(3-36) for the Y2 receptor, and due to the fact that the Y1 and Y5 receptors are associated with increased feeding behavior [75].

The fact that PYY(3-36) seems to be the active ’body weight-reducing‘ form might partially explain the failure of DPP-4 inhibitors to reduce weight to the same extent as the GLP-1R agonists, since an active DPP-4 enzyme is essential for the metamorphosis of PYY(1-36) to PYY(3-36) [76]. In humans, intravenous infusion of PYY(3-36) reduces food intake in both lean and obese subjects [77]. Unfortunately, adverse effects like nausea and vomiting are very common. In a recent study by Sloth et al. only 4 out of the first 9 participants succeeded in completing the high dose PYY(3-36) infusion (0.8 pmol/kg/min) due to the effects of nausea, vomiting, stomach pain, and flushing [77]. Thus, the dose of PYY(3-36) had to be decreased markedly. Similarly, a pharmacologic formulation of PYY(3-36) failed because of side-effects (nausea and vomiting) [78]. Recently, PYY(3-36) was suggested for use as anti-obesity treatment in combination with oxyntomodulin [79]. This would allow augmented action on appetite suppression through additive effects of PYY(3-36) and oxyntomodulin. Since both substances are L-cell derivatives and probably co-secreted this might correlate more with the native anorectic L-cell signal.

Cholecystokinin

Cholecystokinin (CCK) is a gut hormone secreted from the enteroendocrine I cells in the small intestine. It is also a neuropeptide localized at nerve terminals in the CNS and in the peripheral nervous system [80]. CCK is processed in different lengths (e.g. CCK-58, -33, -22, and -8), which predicts its activity. CCK shares its active site and receptors with another gut hormone, namely gastrin [81]. CCK has relative high affinity for the CCK receptor (CCK-A receptor) and a relative low affinity for the gastrin receptor (CCK-B receptor).

In the periphery, CCK primarily exerts its effects through the CCK receptor. It stimulates gallbladder contraction and pancreatic enzyme secretion, and retards gastric emptying [82, 83]. Furthermore, the release of PYY and GLP-1, after lipid ingestion, is dependent on signaling through the CCK receptor [84, 85]. In the CNS, the gastrin receptor seems to be the main receptor; and the effects of CCK-8 and CCK-5 are probably mediated through this receptor as only trace amounts of gastrin are found in the CNS [80, 86]. Intra-cranial infusion of CCK-8 seems to inhibit food intake in rats, but when it is administrated peripherally these effects are not persistent [87]. When CCK is administered to rats by intra-peritoneal infusion at the start of every meal, reduced meal size is rapidly compensated for by increased meal frequency [88]. Similar results were seen in humans where acute administration of CCK-8 decreased food intake [89].

Nevertheless, the results from phase II trials using CCK receptor agonists have been disappointing to date. Thus, GI181771X (a CCK receptor agonist) did not elicit body weight loss when administered to more than 700 obese subjects (tested in a 24-week, cross-over, double-blinded setting) [90]. On the other hand, animal studies with CCK receptor agonists have demonstrated promising results [91, 92]. CCK has been found to enhance cerebral uptake of leptin in rats, wich results in a synergistic effect on weight loss [93]. Also, the synergistic effect in combination with cannabinoid receptor blockage [94] keeps CCK receptor signaling alive as a potential target for obesity treatment.

Ghrelin

Ghrelin is secreted from endocrine X/A-like cells in the gastric fundus and is activated upon acetylation by ghrelin O-acyl tranferase (GOAT) [95, 96]. It is a potent ligand for the growth hormone secretagogue receptor [97]. Also it seems to be an important regulator of appetite [98]. In humans, ghrelin peaks just before a meal intake, and decreases immediately after meal ingestion in lean subjects [99, 100]. On top of this, intravenous infusion of ghrelin in healthy lean subjects leads to heavily increased appetite and food intake [98]. Furthermore, patients with Prader-Willi syndrome (a rare genetic disease characterized by overeating) are characterized by elevated plasma levels of ghrelin [101].

Interestingly, in obese subjects, plasma ghrelin concentrations do not change after a test meal [102]. This suggests that blocking the ghrelin signal could be a tempting target for the treatment of obesity. Although ghrelin receptor knock-out mice are only modestly smaller than wild type mice, appetite and body composition are comparable to that of wild type littermates [103]. However, some data indicate an impaired growth hormone axis in the ghrelin receptor knock-out mice, a finding that raises some concern. Although, preclinical studies with agonists, and antagonists, to the growth hormone secretagogue receptor indicate that it is possible to distinguish between the actions of ghrelin on growth hormone secretion and food intake [104, 105]. These findings indicate that the effects on food intake might be facilitated through an alternative receptor to the well-known growth hormone secretagogue receptor, thus opening the opportunity to develop specific drugs.

Conclusions

The development of GLP-1R agonists for the treatment of type 2 diabetes with improved glycemic control combined with a sustained weight loss, is a major breakthrough, in the medical treatment of type 2 diabetes; and potentially, for the treatment of obesity. The accumulated data on GLP-1R agonists has led to a remarkable interest in other very important and potent gut-derived peptides. Most have been known for quite a while.

At present, the most promising data concern the L-cell derivatives, of which GLP-1 is by far the best described. However, both oxyntomodulin and PYY have gained some attention in recent years, and the idea of using the entire L cell response as template for the treatment of obesity is very attractive. As such, the results from well-conducted studies combining the effects of the three peptides in various ways are much anticipated.

In addition to the L-cell products, both CCK and especially ghrelin are potent peptides in the regulation of appetite, food intake, and body weight. Nonetheless, the road to the development of compounds specifically designed for the treatment of obesity seems to be a longer than for the L-cell derivatives. The development of all these gastrointestinal hormone derivatives into pharmacological agents will certainly provide novel insight and understanding of the role of the gastrointestinal tract in the regulation of appetite, hunger, food intake, and body weight.

Disclosures: No financial support was received for the preparation of this review. TV has received fees for giving lectures and for consultation from Novo Nordisk, Bristol-Myers Squibb, AstraZeneca, Novartis, MSD, Boehringer Ingelheim, and Eli Lilly & Co. TV is on the advisory boards of Novo Nordisk, MSD, and Bristol-Myers Squibb. FKK has received fees for giving lectures from Novo Nordisk, Bristol-Myers Squibb, AstraZeneca, Novartis, MSD, Gilead, Ono Pharmaceutical Co, and Eli Lilly & Co, and has consulted for AstraZeneca, BMS, and Gilead.

References

  1. Finucane MM, Stevens GA, Cowan MJ, Danaei G, Lin JK, Paciorek CJ, Singh GM, Gutierrez HR, Lu Y, Bahalim AN, et al. National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9.1 million participants. Lancet 2011. 377:557-567. [DOD] [CrossRef]
  2. WHO. Obesity and overweight. http://www.who.int/mediacentre/factsheets/fs311/en/index.html, 2011. [DOD] 
  3. Norris SL, Zhang X, Avenell A, Gregg E, Brown TJ, Schmid CH, Lau J. Long-term non-pharmacologic weight loss interventions for adults with type 2 diabetes. Cochrane Database Syst Rev 2005. 2:CD004095. [DOD] 
  4. Franz MJ, VanWormer JJ, Crain AL, Boucher JL, Histon T, Caplan W, Bowman JD, Pronk NP. Weight-loss outcomes: a systematic review and meta-analysis of weight-loss clinical trials with a minimum 1-year follow-up. J Am Diet Assoc 2007. 107:1755-1767. [DOD] [CrossRef]
  5. Gourlan MJ, Trouilloud DO, Sarrazin PG. Interventions promoting physical activity among obese populations: a meta-analysis considering global effect, long-term maintenance, physical activity indicators and dose characteristics. Obes Rev 2011. 12:E633-E645. [DOD] [CrossRef]
  6. Padwal R, Li SK, Lau DC. Long-term pharmacotherapy for obesity and overweight. Cochrane Database Syst Rev 2004. 3:CD004094. [DOD] 
  7. Rucker D, Padwal R, Li SK, Curioni C, Lau DC. Long term pharmacotherapy for obesity and overweight: updated meta-analysis. BMJ 2007. 335:1194-1199. [DOD] [CrossRef]
  8. Li M, Cheung BM. Pharmacotherapy for obesity. Br J Clin Pharmacol 2009. 68:804-810. [DOD] [CrossRef]
  9. Thomas O, Thabane L, Douketis J, Chu R, Westfall AO, Allison DB. Industry funding and the reporting quality of large long-term weight loss trials. Int J Obes (Lond) 2008. 32:1531-1536. [DOD] [CrossRef]
  10. Padwal R, Klarenbach S, Wiebe N, Birch D, Karmali S, Manns B, Hazel M, Sharma AM, Tonelli M. Bariatric surgery: a systematic review and network meta-analysis of randomized trials. Obes Rev 2011. 12(8):602-621. [DOD] [CrossRef]
  11. Pontiroli AE, Morabito A. Long-term prevention of mortality in morbid obesity through bariatric surgery. a systematic review and meta-analysis of trials performed with gastric banding and gastric bypass. Ann Surg 2011. 253:484-487. [DOD] [CrossRef]
  12. Field AE, Coakley EH, Must A, Spadano JL, Laird N, Dietz WH, Rimm E, Colditz GA. Impact of overweight on the risk of developing common chronic diseases during a 10-year period. Arch Intern Med 2001. 161:1581-1586. [DOD] [CrossRef]
  13. Ford ES, Williamson DF, Liu S. Weight change and diabetes incidence: findings from a national cohort of US adults. Am J Epidemiol 1997. 146:214-222. [DOD] 
  14. Bolen S, Feldman L, Vassy J, Wilson L, Yeh HC, Marinopoulos S, Wiley C, Selvin E, Wilson R, Bass EB, et al. Systematic review: comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Ann Intern Med 2007. 147:386-399. [DOD] 
  15. Vilsboll T, Christensen M, Junker A, Knop FK, Gluud LL. Effects of glucagon-like peptide-1 receptor agonists on weight loss: systematic review and meta-analyses of randomised controlled trials. BMJ 2011. In press. [DOD] 
  16. Elliott RM, Morgan LM, Tredger JA, Deacon S, Wright J, Marks V. Glucagon-like peptide-1 (7-36)amide and glucose-dependent insulinotropic polypeptide secretion in response to nutrient ingestion in man: acute post-prandial and 24-h secretion patterns. J Endocrinol 1993. 138:159-166. [DOD] [CrossRef]
  17. Deacon CF, Johnsen AH, Holst JJ. Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo. J Clin Endocrinol Metab 1995. 80:952-957. [DOD] [CrossRef]
  18. Vilsboll T, Agerso H, Krarup T, Holst JJ. Similar elimination rates of glucagon-like peptide-1 in obese type 2 diabetic patients and healthy subjects. J Clin Endocrinol Metab 2003. 88:220-224. [DOD] [CrossRef]
  19. Goke R, Larsen PJ, Mikkelsen JD, Sheikh SP. Distribution of GLP-1 binding sites in the rat brain: evidence that exendin-4 is a ligand of brain GLP-1 binding sites. Eur J Neurosci 1995. 7:2294-2300. [DOD] [CrossRef]
  20. Williams DL, Baskin DG, Schwartz MW. Evidence that intestinal glucagon-like peptide-1 plays a physiological role in satiety. Endocrinology 2009. 150:1680-1687. [DOD] [CrossRef]
  21. Turton MD, O'Shea D, Gunn I, Beak SA, Edwards CM, Meeran K, Choi SJ, Taylor GM, Heath MM, Lambert PD, et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 1996. 379:69-72. [DOD] [CrossRef]
  22. Holst JJ. On the physiology of GIP and GLP-1. Horm Metab Res 2004. 36:747-754. [DOD] [CrossRef]
  23. Raun K, von Voss P, Knudsen LB. Liraglutide, a once-daily human glucagon-like peptide-1 analog, minimizes food intake in severely obese minipigs. Obesity (Silver Spring) 2007. 15:1710-1716. [DOD] [CrossRef]
  24. Tang-Christensen M, Larsen PJ, Göke R, Fink-Jensen A, Jessop DS, Moller M, Sheikh SP. Central administration of GLP-1-(7-36) amide inhibits food and water intake in rats. Am J Physiol 1996. 271:R848-R856. [DOD] 
  25. Vrang N, Hansen M, Larsen PJ, Tang-Christensen M. Characterization of brainstem preproglucagon projections to the paraventricular and dorsomedial hypothalamic nuclei. Brain Res 2007. 1149:118-126. [DOD] [CrossRef]
  26. Tang-Christensen M, Vrang N, Larsen PJ. Glucagon-like peptide 1(7-36) amide's central inhibition of feeding and peripheral inhibition of drinking are abolished by neonatal monosodium glutamate treatment. Diabetes 1998. 47:530-537. [DOD] [CrossRef]
  27. Vrang N, Phifer CB, Corkern MM, Berthoud HR. Gastric distension induces c-Fos in medullary GLP-1/2-containing neurons. Am J Physiol Regul Integr Comp Physiol 2003. 285:R470-R478. [DOD] 
  28. Rinaman L. Interoceptive stress activates glucagon-like peptide-1 neurons that project to the hypothalamus. Am J Physiol 1999. 277:R582-R590. [DOD] 
  29. Orskov C, Poulsen SS, Møller M, Holst JJ. Glucagon-like peptide I receptors in the subfornical organ and the area postrema are accessible to circulating glucagon-like peptide I. Diabetes 1996. 45:832-835. [DOD] [CrossRef]
  30. Ruttimann EB, Arnold M, Hillebrand JJ, Geary N, Langhans W. Intrameal hepatic portal and intraperitoneal infusions of glucagon-like peptide-1 reduce spontaneous meal size in the rat via different mechanisms. Endocrinology 2009. 150:1174-1181. [DOD] [CrossRef]
  31. Baggio LL, Huang Q, Cao X, Drucker DJ. An albumin-exendin-4 conjugate engages central and peripheral circuits regulating murine energy and glucose homeostasis. Gastroenterology 2008. 134:1137-1147. [DOD] [CrossRef]
  32. Rosenstock J, Reusch J, Bush M, Yang F, Stewart M. Potential of albiglutide, a long-acting GLP-1 receptor agonist, in type 2 diabetes: a randomized controlled trial exploring weekly, biweekly, and monthly dosing. Diabetes Care 2009. 32:1880-1886. [DOD] [CrossRef]
  33. Kakei M, Yada T, Nakagawa A, Nakabayashi H. Glucagon-like peptide-1 evokes action potentials and increases cytosolic Ca2+ in rat nodose ganglion neurons. Auton Neurosci 2002. 102:39-44. [DOD] [CrossRef]
  34. Holst JJ. The physiology of glucagon-like peptide 1. Physiol Rev 2007. 87:1409-1439. [DOD] [CrossRef]
  35. Geliebter A. Gastric distension and gastric capacity in relation to food intake in humans. Physiol Behav 1988. 44:665-668. [DOD] [CrossRef]
  36. Nauck MA, Niedereichholz U, Ettler R, Holst JJ, Orskov C, Ritzel R, Schmiegel WH. Glucagon-like peptide 1 inhibition of gastric emptying outweighs its insulinotropic effects in healthy humans. Am J Physiol 1997. 273:E981-E988. [DOD] 
  37. Delzenne N, Blundell J, Brouns F, Cunningham K, De Graaf K, Erkner A, Lluch A, Mars M, Peters HPF, Westerterp-Plantenga M. Gastrointestinal targets of appetite regulation in humans. Obes Rev 2010. 11:234-250. [DOD] [CrossRef]
  38. Gutzwiller JP, Göke B, Drewe J, Hildebrand P, Ketterer S, Handschin D, Winterhalder R, Conen D, Beglinger C. Glucagon-like peptide-1: a potent regulator of food intake in humans. Gut 1999. 44:81-86. [DOD] [CrossRef]
  39. Knudsen LB, Tang-Christensen M, Jelsing J, Vrang N, Raun K. Liraglutide: short-lived effect on gastric emptying - long-lasting effects on body-weight. Diabetologia 2010. 53:860. [DOD] 
  40. Nauck MA, Kemmeries G, Holst JJ, Meier JJ. Rapid tachyphylaxis of the glucagon-like peptide 1-induced deceleration of gastric emptying in humans. Diabetes 2011. 60:1561-1565. [DOD] [CrossRef]
  41. Goke R, Fehmann HC, Linn T, Schmidt H, Krause M, Eng J, Göke B. Exendin-4 is a high potency agonist and truncated exendin-(9-39)-amide an antagonist at the glucagon-like peptide 1-(7-36)-amide receptor of insulin-secreting beta-cells. J Biol Chem 1993. 268:19650-19655. [DOD] 
  42. Eng J, Kleinman WA, Singh L, Singh G, Raufman JP. Isolation and characterization of exendin-4, an exendin-3 analogue, from Heloderma suspectum venom. Further evidence for an exendin receptor on dispersed acini from guinea pig pancreas. J Biol Chem 1992. 267:7402-7405. [DOD] 
  43. Knudsen LB. Liraglutide: the therapeutic promise from animal models. Int J Clin Pract Suppl 2010. 64:4-11. [DOD] [CrossRef]
  44. Astrup A, Carraro R, Finer N, Harper A, Kunesova M, Lean ME, Niskanen L, Rasmussen MF, Rissanen A, Rössner S, et al. Safety, tolerability and sustained weight loss over 2 years with the once-daily human GLP-1 analog, liraglutide. Int J Obes (Lond) 2011. In press. [DOD] 
  45. Anderson SL, Trujillo JM. Association of pancreatitis with glucagon-like peptide-1 agonist use. Ann Pharmacother 2010. 44:904-909. [DOD] [CrossRef]
  46. Noel RA, Braun DK, Patterson RE, Bloomgren GL. Increased risk of acute pancreatitis and biliary disease observed in patients with type 2 diabetes: a retrospective cohort study. Diabetes Care 2009. 32:834-838. [DOD] [CrossRef]
  47. FDA briefing materials. Table of contents liraglutide. April 2, 2009. www.fda.gov/.../Drugs/EndocrinologicandMetabolicDrugsAdvisoryCommittee/UCM148645.pdf, 2011. [DOD] 
  48. Waser B, Beetschen K, Pellegata NS, Reubi JC. Incretin receptors in non-neoplastic and neoplastic thyroid C cells in rodents and humans: relevance for incretin-based diabetes therapy. Neuroendocrinology 2011. In press. [DOD] 
  49. Bjerre Knudsen L, Madsen LW, Andersen S, Almholt K, de Boer AS, Drucker DJ, Gotfredsen C, Egerod FL, Hegelund AC, Jacobsen H, et al. Glucagon-like Peptide-1 receptor agonists activate rodent thyroid C-cells causing calcitonin release and C-cell proliferation. Endocrinology 2010. 151:1473-1486. [DOD] [CrossRef]
  50. Elashoff M, Matveyenko AV, Gier B, Elashoff R, Butler PC. Pancreatitis, pancreatic, and thyroid cancer with glucagon-like peptide-1-based therapies. Gastroenterology 2011. 141:150-156. [DOD] [CrossRef]
  51. Anti-diabetic drugs: cardio/cerebrovascular adverse effect and pancreatitis/pancreatic cancer. European Medicines Agency, 2011, Priorities for Drug Safety Research. [DOD] 
  52. Holst JJ, Deacon CF. Inhibition of the activity of dipeptidyl-peptidase IV as a treatment for type 2 diabetes. Diabetes 1998. 47:1663-1670. [DOD] [CrossRef]
  53. Deacon CF, Holst JJ. Dipeptidyl peptidase IV inhibitors: a promising new therapeutic approach for the management of type 2 diabetes. Int J Biochem Cell Biol 2006. 38:831-844. [DOD] [CrossRef]
  54. Ahren B, Gomis R, Standl E, Mills D, Schweizer A. Twelve- and 52-week efficacy of the dipeptidyl peptidase IV inhibitor LAF237 in metformin-treated patients with type 2 diabetes. Diabetes Care 2004. 27:2874-2880. [DOD] [CrossRef]
  55. Holst JJ, Deacon CF, Vilsboll T, Krarup T, Madsbad S. Glucagon-like peptide-1, glucose homeostasis and diabetes. Trends Mol Med 2008. 14:161-168. [DOD] [CrossRef]
  56. Gros L, Thorens B, Bataille D, Kervran A. Glucagon-like peptide-1-(7-36) amide, oxyntomodulin, and glucagon interact with a common receptor in a somatostatin-secreting cell line. Endocrinology 1993. 133:631-638. [DOD] [CrossRef]
  57. Baldissera FG, Holst JJ, Knuhtsen S, Hilsted L, Nielsen OV. Oxyntomodulin (glicentin-(33-69)): pharmacokinetics, binding to liver cell membranes, effects on isolated perfused pig pancreas, and secretion from isolated perfused lower small intestine of pigs. Regul Pept 1988. 21:151-166. [DOD] [CrossRef]
  58. Holst JJ. Enteroglucagon. Annu Rev Physiol 1997. 59:257-271. [DOD] [CrossRef]
  59. Bataille D, Tatemoto K, Gespach C, Jörnvall H, Rosselin G, Mutt V. Isolation of glucagon-37 (bioactive enteroglucagon/oxyntomodulin) from porcine jejuno-ileum. Characterization of the peptide. FEBS Lett 1982. 146:79-86. [DOD] [CrossRef]
  60. Schjoldager BT, Baldissera FG, Mortensen PE, Holst JJ, Christiansen J. Oxyntomodulin: a potential hormone from the distal gut. Pharmacokinetics and effects on gastric acid and insulin secretion in man. Eur J Clin Invest 1988. 18:499-503. [DOD] [CrossRef]
  61. Druce MR, Minnion JS, Field BC, Patel SR, Shillito JC, Tilby M, Beale KE, Murphy KG, Ghatei MA, Bloom SR. Investigation of structure-activity relationships of Oxyntomodulin (Oxm) using Oxm analogs. Endocrinology 2009. 150:1712-1722. [DOD] [CrossRef]
  62. Dakin CL, Gunn I, Small CJ, Edwards CM, Hay DL, Smith DM, Ghatei MA, Bloom SR. Oxyntomodulin inhibits food intake in the rat. Endocrinology 2001. 142:4244-4250. [DOD] [CrossRef]
  63. Wynne K, Park AJ, Small CJ, Patterson M, Ellis SM, Murphy KG, Wren AM, Frost GS, Meeran K, Ghatei MA, et al. Subcutaneous oxyntomodulin reduces body weight in overweight and obese subjects: a double-blind, randomized, controlled trial. Diabetes 2005. 54:2390-2395. [DOD] [CrossRef]
  64. 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 Obes (Lond) 2006. 30:1729-1736. [DOD] [CrossRef]
  65. Pocai A, Carrington PE, Adams JR, Wright M, Eiermann G, Zhu L, Du X, Petrov A, Lassman ME, Jiang G, et al. Glucagon-like peptide 1/glucagon receptor dual agonism reverses obesity in mice. Diabetes 2009. 58:2258-2266. [DOD] [CrossRef]
  66. Baggio LL, Huang Q, Brown TJ, Drucker DJ. Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure. Gastroenterology 2004. 127:546-558. [DOD] [CrossRef]
  67. Adrian TE, Ferri GL, Bacarese-Hamilton AJ, Fuessl HS, Polak JM, Bloom SR. Human distribution and release of a putative new gut hormone, peptide YY. Gastroenterology 1985. 89:1070-1077. [DOD] 
  68. Pedersen-Bjergaard U, Host U, Kelbaek H, Schifter S, Rehfeld JF, Faber J, Christensen NJ. Influence of meal composition on postprandial peripheral plasma concentrations of vasoactive peptides in man. Scand J Clin Lab Invest 1996. 56:497-503. [DOD] [CrossRef]
  69. Tatemoto K, Mutt V. Isolation of two novel candidate hormones using a chemical method for finding naturally occurring polypeptides. Nature 1980. 285:417-418. [DOD] [CrossRef]
  70. Mentlein R, Dahms P, Grandt D, Krüger R. Proteolytic processing of neuropeptide Y and peptide YY by dipeptidyl peptidase IV. Regul Pept 1993. 49:133-144. [DOD] [CrossRef]
  71. Grandt D, Schimiczek M, Beglinger C, Layer P, Goebell H, Eysselein VE, Reeve JR Jr. Two molecular forms of peptide YY (PYY) are abundant in human blood: characterization of a radioimmunoassay recognizing PYY 1-36 and PYY 3-36. Regul Pept 1994. 51:151-159. [DOD] [CrossRef]
  72. Keire DA, Bowers CW, Solomon TE, Reeve JR Jr. Structure and receptor binding of PYY analogs. Peptides 2002. 23:305-321. [DOD] [CrossRef]
  73. Savage AP, Adrian TE, Carolan G, Chatterjee VK, Bloom SR. Effects of peptide YY (PYY) on mouth to caecum intestinal transit time and on the rate of gastric emptying in healthy volunteers. Gut 1987. 28:166-170. [DOD] [CrossRef]
  74. Adrian TE, Savage AP, Sagor GR, Allen JM, Bacarese-Hamilton AJ, Tatemoto K, Polak JM, Bloom SR. Effect of peptide YY on gastric, pancreatic, and biliary function in humans. Gastroenterology 1985. 89:494-499. [DOD] 
  75. Lecklin A, Lundell I, Paananen L, Wikberg JE, Männistö PT, Larhammar D. Receptor subtypes Y1 and Y5 mediate neuropeptide Y induced feeding in the guinea-pig. Br J Pharmacol 2002. 135:2029-2037. [DOD] [CrossRef]
  76. Aaboe K, Knop FK, Vilsboll T, Deacon CF, Holst JJ, Madsbad S, Krarup T. Twelve weeks treatment with the DPP-4 inhibitor, sitagliptin, prevents degradation of peptide YY and improves glucose and non-glucose induced insulin secretion in patients with type 2 diabetes mellitus. Diabetes Obes Metab 2010. 12:323-333. [DOD] [CrossRef]
  77. Sloth B, Holst JJ, Flint A, Gregersen NT, Astrup A. Effects of PYY1-36 and PYY3-36 on appetite, energy intake, energy expenditure, glucose and fat metabolism in obese and lean subjects. Am J Physiol Endocrinol Metab 2007. 292:E1062-E1068. [DOD] [CrossRef]
  78. Gantz I, Erondu N, Mallick M, Musser B, Krishna R, Tanaka WK, Snyder K, Stevens C, Stroh MA, Zhu H, et al. Efficacy and safety of intranasal peptide YY3-36 for weight reduction in obese adults. J Clin Endocrinol Metab 2007. 92:1754-1757. [DOD] [CrossRef]
  79. Field BC, Wren AM, Peters V, Baynes KC, Martin NM, Patterson M, Alsaraf S, Amber V, Wynne K, Ghatei MA, et al. PYY3-36 and oxyntomodulin can be additive in their effect on food intake in overweight and obese humans. Diabetes 2010. 59:1635-1639. [DOD] [CrossRef]
  80. Rehfeld JF. Immunochemical studies on cholecystokinin. II. Distribution and molecular heterogeneity in the central nervous system and small intestine of man and hog. J Biol Chem 1978. 253:4022-4030. [DOD] 
  81. Rehfeld JF. The endoproteolytic maturation of progastrin and procholecystokinin. J Mol Med 2006. 84:544-550. [DOD] [CrossRef]
  82. Fried M, Erlacher U, Schwizer W, Löchner C, Koerfer J, Beglinger C, Jansen JB, Lamers CB, Harder F, Bischof-Delaloye A. Role of cholecystokinin in the regulation of gastric emptying and pancreatic enzyme secretion in humans. Studies with the cholecystokinin-receptor antagonist loxiglumide. Gastroenterology 1991. 101:503-511. [DOD] 
  83. Takahashi T, May D, Owyang C. Cholinergic dependence of gallbladder response to cholecystokinin in the guinea pig in vivo. Am J Physiol 1991. 261:G565-G569. [DOD] 
  84. Beglinger S, Drewe J, Schirra J, Göke B, D'Amato M, Beglinger C. Role of fat hydrolysis in regulating glucagon-like Peptide-1 secretion. J Clin Endocrinol Metab 2010. 95:879-886. [DOD] [CrossRef]
  85. Degen L, Drewe J, Piccoli F, Gräni K, Oesch S, Bunea R, D'Amato M, Beglinger C. Effect of CCK-1 receptor blockade on ghrelin and PYY secretion in men. Am J Physiol Regul Integr Comp Physiol 2007. 292:R1391-R1399. [DOD] [CrossRef]
  86. Rehfeld JF. Localisation of gastrins to neuro- and adenohypophysis. Nature 1978. 271:771-773. [DOD] [CrossRef]
  87. Mori T, Nagai K, Nakagawa H, Yanaihara N. Intracranial infusion of CCK-8 derivatives suppresses food intake in rats. Am J Physiol 1986. 251:R718-R723. [DOD] 
  88. West DB, Fey D, Woods SC. Cholecystokinin persistently suppresses meal size but not food intake in free-feeding rats. Am J Physiol 1984. 246:R776-R787. [DOD] 
  89. Kissileff HR, Pi-Sunyer FX, Thornton J, Smith GP. C-terminal octapeptide of cholecystokinin decreases food intake in man. Am J Clin Nutr 1981. 34:154-160. [DOD] 
  90. Jordan J, Greenway FL, Leiter LA, Li Z, Jacobson P, Murphy K, Hill J, Kler L, Aftring RP. Stimulation of cholecystokinin-A receptors with GI181771X does not cause weight loss in overweight or obese patients. Clin Pharmacol Ther 2008. 83:281-287. [DOD] [CrossRef]
  91. Simmons RD, Kaiser FC, Hudzik TJ. Behavioral effects of AR-R 15849, a highly selective CCK-A agonist. Pharmacol Biochem Behav 1999. 62:549-557. [DOD] [CrossRef]
  92. Asin KE, Bednarz L, Nikkel AL, Gore PA Jr, Montana WE, Cullen MJ, Shiosaki K, Craig R, Nadzan AM. Behavioral effects of A71623, a highly selective CCK-A agonist tetrapeptide. Am J Physiol 1992. 263:R125-R135. [DOD] 
  93. Merino B, Cano V, Guzman R, Somoza B, Ruiz-Gayo M. Leptin-mediated hypothalamic pathway of cholecystokinin (CCK-8) to regulate body weight in free-feeding rats. Endocrinology 2008. 149:1994-2000. [DOD] [CrossRef]
  94. Orio L, Crespo I, Lopez-Moreno JA, Reyes-Cabello C, Rodriguez de Fonseca F, Gomez de Heras R. Additive effects of cannabinoid CB1 receptors blockade and cholecystokinin on feeding inhibition. Pharmacol Biochem Behav 2011. 98:220-226. [DOD] [CrossRef]
  95. Yang J, Brown MS, Liang G, Grishin NV, Goldstein JL. Identification of the acyltransferase that octanoylates ghrelin, an appetite-stimulating peptide hormone. Cell 2008. 132:387-396. [DOD] [CrossRef]
  96. van der Lely AJ, Tschöp M, Heiman ML, Ghigo E. Biological, physiological, pathophysiological, and pharmacological aspects of ghrelin. Endocr Rev 2004. 25:426 -457. [DOD] [CrossRef]
  97. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 1999. 402:656-660. [DOD] [CrossRef]
  98. Wren AM, Seal LJ, Cohen MA, Brynes AE, Frost GS, Murphy KG, Dhillo WS, Ghatei MA, Bloom SR. Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab 2001. 86:5992. [DOD] [CrossRef]
  99. Cummings DE, Purnell JQ, Frayo RS, Schmidova K, Wisse BE, Weigle DS. A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 2001. 50:1714-1719. [DOD] [CrossRef]
  100. Cummings DE, Frayo RS, Marmonier C, Aubert R, Chapelot D. Plasma ghrelin levels and hunger scores in humans initiating meals voluntarily without time- and food-related cues. Am J Physiol Endocrinol Metab 2004. 287:E297-E304. [DOD] [CrossRef]
  101. Tauber M, Conte Auriol F, Moulin P, Molinas C, Delagnes V, Salles JP. Hyperghrelinemia is a common feature of Prader-Willi syndrome and pituitary stalk interruption: a pathophysiological hypothesis. Horm Res 2004. 62:49-54. [DOD] [CrossRef]
  102. English PJ, Ghatei MA, Malik IA, Bloom SR, Wilding JP. Food fails to suppress ghrelin levels in obese humans. J Clin Endocrinol Metab 2002. 87:2984. [DOD] [CrossRef]
  103. Sun Y, Wang P, Zheng H, Smith RG. Ghrelin stimulation of growth hormone release and appetite is mediated through the growth hormone secretagogue receptor. Proc Natl Acad Sci USA 2004. 101:4679-4684. [DOD] [CrossRef]
  104. Halem HA, Taylor JE, Dong JZ, Shen Y, Datta R, Abizaid A, Diano S, Horvath TL, Culler MD. A novel growth hormone secretagogue-1a receptor antagonist that blocks ghrelin-induced growth hormone secretion but induces increased body weight gain. Neuroendocrinology 2005. 81:339-349. [DOD] [CrossRef]
  105. Demange L, Boeglin D, Moulin A, Mousseaux D, Ryan J, Berge G, Gagne D, Heitz A, Perrissoud D, Locatelli V, et al. Synthesis and pharmacological in vitro and in vivo evaluations of novel triazole derivatives as ligands of the ghrelin receptor. J Med Chem 2007. 50:1939-1957. [DOD] [CrossRef]

This article has been cited by other articles:

Short-term effects of glucagon-like peptide 1 (GLP-1) receptor agonists on fat distribution in patients with type 2 diabetes mellitus: an ultrasonography study.

Morano S, Romagnoli E, Filardi T, Nieddu L, Mandosi E, Fallarino M, Turinese I, Dagostino MP, Lenzi A, Carnevale V

Acta Diabetol 2015. In press

Does a GLP-1 receptor agonist change glucose tolerance in patients treated with antipsychotic medications? Design of a randomised, double-blinded, placebo-controlled clinical trial

Larsen JR, Vedtofte L, Holst JJ, Oturai P, Kjaer A, Corell CU, Vilsboll T, Fink-Jensen A

Bmj Open 2014. 4(3):e004227

The glucagon-like peptide-1 analog liraglutide suppresses ghrelin and controls diabetes in a patient with Prader-Willi syndrome

Senda M, Ogawa S, Nako K, Okamura M, Sakamoto T, Ito S

Endocr J 2012. 59(10):889-894

The continuing need for drug development and clinical trials in type 2 diabetes and its complications: introduction to the RDS Special Issue

Raz I, Gallwitz B

Rev Diabet Stud 2011. 8(3):288-292