Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Apr 7.
Published in final edited form as: Cell Metab. 2020 Mar 17;31(4):679–698. doi: 10.1016/j.cmet.2020.02.014

Leveraging the gut to treat metabolic disease

Ruth E Gimeno 1, Daniel A Briere 1, Randy J Seeley 2
PMCID: PMC7184629  NIHMSID: NIHMS1577171  PMID: 32187525

Abstract

Twenty-five years ago, the future of treating obesity and diabetes focused on end organs known to be involved in energy balance and glucose regulation, including the brain, muscle, adipose tissue and pancreas. Today, the most effective therapies are focused around the gut. This includes surgical options, such as vertical sleeve gastrectomy and Roux-en-Y gastric bypass, that can produce sustained weight loss and diabetes remission, but also extends to pharmacological treatments that simulate or amplify various signals that come from the gut. The purpose of this review is to discuss the wealth of approaches currently under development that seek to further leverage the gut as a source of novel therapeutic opportunities with the hope that we can achieve the effects of surgical interventions with less invasive and more scalable solutions.

eToC

While traditional approaches to treating obesity and type 2 diabetes have focused on end-organs involved in energy metabolism, the gut has emerged as a source of our most effective therapies. From bariatric surgical procedures to pharmacological agents, harnessing the gut has and will continue to provide important therapeutic options for the treatment of these epidemic-level diseases.

Introduction

Type 2 Diabetes Mellitus (T2DM) is a large and growing public health problem that affects almost 10% of the US population (National Diabetes Statistics Report, 2017). Combined with rapidly rising obesity rates, currently estimated at 40% of the US adult population (National Health and Nutrition Examination Survey, 2015–2016), there is potential for a future crisis of T2DM. These statistics are more alarming when one considers that T2DM and obesity are associated with a broad spectrum of metabolic dysfunctions, including high blood pressure, dyslipidemia, heart failure, and nonalcoholic steatohepatitis. While there are a growing number of pharmacological treatments for T2DM and obesity, fewer than 50% of T2DM patients achieve treatment goals (Garcia-Perez, Alvarez et al. 2013) and none of the currently available therapies halt the progression of diabetes treatment towards insulinfocused therapy. Furthermore, rising rates of T2DM will result in escalating monetary and human costs.

Both existing and future therapies for T2DM and obesity have targeted several organ systems, with most of the research focused on the effector organs. In the case of T2DM, targeting pancreas, adipose tissue, muscle and liver are sensible approaches borne from decades of research on blood glucose regulation. While several strategies have been deployed to increase insulin secretion by acting directly or indirectly on the pancreas, an alternative strategy aims to increase the action of insulin (insulin sensitizers) by targeting effector organs such as adipose tissue, muscle and/or liver to lower glucose levels. In the case of obesity, most approaches target the appetite regulating circuits within the central nervous system. However, an alternative strategy aims to target muscle and/or adipose tissue to increase energy expenditure, capitalizing on decades of research that links these tissues directly to the regulation of energy intake or energy expenditure.

Today, the most effective strategies to treat T2DM and obesity involve the gastrointestinal (GI) tract, or gut, an outcome that would have been almost impossible to predict based on the available research literature 25 years ago. The purpose of this review is to outline current treatments options based on manipulations of the GI tract and to discuss next-generation, gut-based therapies that have been or are currently under investigation. We will also lay out potential strategies to identify novel therapeutic strategies based on targeting the GI tract or mimicking signals from the GI tract.

Current gut-based therapies

Surgical interventions

Among the available treatments for obesity and T2DM, various bariatric surgical interventions are clearly the most effective. The two most common are Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) (Figure 1). The clinical effects of both procedures are impressive. In RYGB, the average patient loses 70% of their excess body weight in the first 12 months after surgery, with VSG showing similar weight loss (Demssie, Jawaheer et al. 2012). However, what truly sets surgical interventions apart from other weight loss programs is durability. In the case of RYGB, the Swedish Outcome Study finds that patients still had a 28% reduction in body weight 14 years after the procedure (Sjostrom 2013). This stands in contrast to lifestyle interventions that have little overall benefit on weight even 5 years after the intervention (Look, Wing et al. 2013, Sjostrom 2013).

Figure 1:

Figure 1:

Traditionally, therapeutic approaches to obesity and type 2 diabetes have focused on altering the actions of key effector organs such as brain, liver, pancreas, adipose tissue and muscle. Surgical alterations of the GI tract by either RYGB or VSG result in sustained weight loss and potent improvements and even remission of type 2 diabetes. Targeting the GI tract for therapies, therefore, can leverage signals from the GI tract to regulate each of these effector organs.

Both RYGB and VSG also provide profound improvements in the regulation of glucose. In one randomized trial, RYGB or VSG surgically treated type 2 diabetic patients had a greater mean percentage reduction from baseline in percent glycated hemoglobin level than did patients who received medical therapy alone after 5 years (−2.1% vs. −0.3%, P = 0.003)(Schauer, Bhatt et al. 2017). This is a single study, but a wide variety of trial and observational data points towards surgical intervention as producing higher rates of remission for type 2 diabetes than can be achieved by current medical therapies (Tsilingiris, Koliaki et al. 2019). These clinical effects have resulted in referring to these procedures as “metabolic surgery” when used primarily to treat T2DM. This is encapsulated in a recent position statement that has urged metabolic surgeries being adopted as treatment options for type 2 diabetes (Rubino, Nathan et al. 2016). This leads to an important question: are these potent effects of T2DM a benefit that accrues primarily from the profound weight loss that occurs with these procedures? This is a complicated and controversial topic that falls outside the scope of this review. However, profound improvements in glucose regulation are observed in the first few days after surgical intervention and prior to the weight loss which typically peaks close to 1 year after surgery (Miras, Herring et al. 2017). Moreover, improved glucose regulation is not the only benefit, as a number of other obesity-related complications are reduced after surgical intervention, including cardiovascular risk factors, sleep apnea and even obesity-related cancers (Sjostrom 2013). Overall, the rearrangement of the GI tract from either RYGB or VSG produces profound physiological effects that result in successful weight loss and improved glucose regulation.

Mechanical restriction and caloric malabsorption are often used to explain the potent effects from metabolic surgery. Consequently, VSG is referred to as a “restrictive” procedure since it reduces the volume of the stomach by approximately 80%, and RYGB is considered “restrictive” since the functional volume of the stomach is drastically reduced. However, RYGB is also “malabsorptive” since rerouting of the intestine results in a bypass of the absorptive capacity of the upper portion of the small intestine. While mechanical explanations are likely playing a role, these do not provide a full account of post-surgical improvements in energy and glucose homeostasis. After surgery, patients clearly report being less hungry despite profound weight loss (Al-Najim, Docherty et al. 2018). If surgery exerted its effect by preventing ingestion and/or absorption of calories, patients would lose weight but would be hungrier (Seeley, Chambers et al. 2015). Hence, the notion that surgery works primarily by preventing the ingestion or absorption of calories simply does not square with available evidence.

Since mechanical explanations are inadequate to explain the benefits of metabolic surgery, an increased level of research has focused on other mechanisms, notably neuronal and hormonal signals from the gut. Though local innervation of the gut provides important signals back to the CNS that can alter both food intake and metabolism, direct evidence that such signals are the basis for the effects of surgery are relatively scant. Moreover, while the vagus nerve may play a role in alterations of food choice after bariatric surgery (Wilson-Perez, Chambers et al. 2013, Hankir, Seyfried et al. 2017), there is little evidence at present that such signals contribute to weight loss and improvements in glucose regulation. While neural signals from the gut seem less likely, it is very clear that both VSG and RYGB alter the secretion of numerous hormonal signals from the GI tract, including several validated therapeutic strategies for T2DM and obesity, and likely highlighting future therapeutics. These include traditional gut peptides like GLP-1 and less traditional hormones such as bile acids.

While specific gut peptides will be discussed in the next section, RYGB and VSG also result in weight-independent increases in levels and altered composition of bile acids in circulation. Bile acids can bind to and activate both a cell-surface receptor (TGR5) and a nuclear receptor (FXR). In turn, loss-of-function of FXR greatly reduces the ability of VSG to improve either body weight or glucose tolerance (Ryan, Tremaroli et al. 2014), and a number of other studies link bile acid signaling to the beneficial effects of surgery (McGavigan, Garibay et al. 2017, Albaugh, Banan et al. 2019). It is important to note that the source of these signals may not always be the host organism, as bariatric surgical procedures can greatly affect the composition of the bacteria in the gut. These gut bacteria are also capable of producing a wide number of both neural and hormonal signals that may also underlie the effects of surgery. In short, it is unlikely that a single factor or signal underlies the effect of metabolic surgery. Rather, it more likely that “polypharmacy” makes these surgical procedures more effective than currently available pharmacological treatments.

Despite the effectiveness of RYGB and VSG, such interventions for obesity and/or T2DM are limited to treating small populations. Only about 228,000 patients received bariatric surgery in 2017, representing 1% of the eligible population (Gasoyan, Tajeu et al. 2019). Put simply, there are not enough surgeons and operating rooms for surgery to be our main therapeutic option. Further, surgery is not without risk. As surgery involves permanent changes in the anatomy of the GI tract, there are no easy solutions for patients who experience adverse side effects, which can include malnutrition, frequent hypoglycemia and severe reflux in the case of VSG (Gletsu-Miller and Wright 2013, Kassem, Durda et al. 2017, Lim, Lee et al. 2019).

Gut peptide-based approaches

An emerging alternative to bariatric surgery is pharmacotherapy based on gut peptides (Figure 2). The gut is the source of numerous factors, primarily peptides, that act to regulate intestinal motility and nutrient absorption. But gut peptides also function to prime other organs for the availability of nutrients and to initiate counter regulatory responses such as satiety. Some of these peptides, particularly GLP-1, but also other glucagon-related peptides, such as GIP and oxyntomodulin, as well as PYY, have become important therapeutic targets for obesity and type 2 diabetes. The efficacies observed place them among the most attractive new therapeutic approaches in these areas. Understanding how these gut-derived hormones function and how they could act together to potentially mimic the effects of bariatric surgery is an exciting area of research that could potentially lead to additional therapies for diabetes and obesity.

Figure 2:

Figure 2:

Major gut hormones that regulate metabolic function

Here, we highlight some major gut peptides in terms of site of secretion, site and effort organ of action, and functions with emphasis on molecules whose therapeutic potential have been explored in clinical studies.

Incretins: GLP-1 and GIP

The term “incretin effect” was coined in the 1960s to describe the observation that oral administration of glucose leads to more pronounced insulin secretion compared to intravenous administration. In turn, this led to the hypothesis that gut-derived factors, so-called “incretins”, contribute to postprandial stimulation of insulin secretion. Two gut peptides, glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), are now recognized to be responsible for most, if not all, of the incretin effect (Nauck and Meier 2016). These peptides are released from enteroendocrine cells in the gut in response to nutrients, and act directly on pancreatic beta cells to activate insulin secretion in a glucose-dependent manner (Drucker, Habener et al. 2017). In healthy individuals, GLP-1 and GIP act in an additive manner, with GIP responsible for the majority of the incretin effect (Gasbjerg, Helsted et al. 2019). In contrast, in individuals with T2DM, GIP has little effect on insulin secretion, and the efficacy of GLP-1 appears to be greatly decreased, resulting in significant defects in the incretin effect (Nauck, Heimesaat et al. 1993, Nauck, Kleine et al. 1993). However, administration of exogenous GLP-1 partially restores the incretin effect in individuals with T2DM, albeit at doses that result in supraphysiological GLP-1 serum levels (approximately 5–10-fold above normal) (Nauck, Heimesaat et al. 1993, Nauck, Kleine et al. 1993). Consequently, much of the initial work focused on GLP-1 as a potential target for T2DM.

GLP-1 receptor agonists

Native GLP-1, while useful to characterize the physiology and pharmacology of this incretin, is not suitable for therapeutic use due to its very short half-life. Exendin-4, a GLP-1 related peptide isolated from the saliva of the Gila monster, is more stable, allowing for therapeutic development, and exenatide, a synthetic version of exendin-4 administered via twice daily injection, was the first GLP-1 – based therapy for T2DM to reach the market (Knop, Bronden et al. 2017, Aroda 2018)(Table 1). Subsequently, long-acting GLP-1 receptor agonists suitable for once daily or once weekly injection, including an extended release formulation of exenatide, have been approved for clinical use (Table 1)(Aroda 2018).

Table 1:

Select gut-peptide based therapies currently on the market or in clinical development

Class Key Molecules Administration Status Indication References
GLP-1 receptor agonists exenatide SC BID, SC QW Marketed T2DM (Knop, Bronden et al. 2017)
liraglutide SC QD Marketed T2DM, Obesity (Nauck, Meier et al. 2017, Chukir, Shukla et al. 2018)
dulaglutide SC QW Marketed T2DM (Nauck, Meier et al. 2017)
semaglutide SC QW Marketed T2DM (Nauck, Meier et al. 2017)
Oral QD Marketed T2DM (Hedrington and Davis 2019)
SC QW Phase 3 Obesity (Nauck, Meier et al. 2017, Christou, Katsiki et al. 2019)
efpeglenatide SC QW Phase 3 T2DM NCT03684642; NCT03353350
Dual GIP/GLP-1 receptor agonists Tirzepatide (LY3298176) SC QW Phase 3 T2DM (Frias, Nauck et al. 2018)
NNC0090–2746 (RG7697) SC QD Discontinued (Phase 1) T2DM (Frias, Bastyr et al. 2017, Schmitt, Portron et al. 2017)
Cpd 21 SC QW Discontinued (Phase 1) T2DM (Finan, Ma et al. 2013)
Dual Glucagon/ GLP-1 receptor agonists cotadutide (MEDI0382) SC QD Phase 2 Obesity, NASH (Ambery, Parker et al. 2018)
HM-12525A SC QW Phase 2 T2DM, Obesity NCT03235219
OPK88003 SC QW Phase 2 T2DM NCT03406377
SAR425899 SC QD Discontinued (Phase 2) T2DM (Tillner, Posch et al. 2019)
MK-8521 SC QD Discontinued (Phase 2) T2DM NCT02492763 (results available on CT.gov)
NN9277/NN6177 SC QW Phase 1 Obesity, NASH NCT02941042
LY3305677 SC QW Phase 1 T2DM NCT03325387
BI-456906 SC QW Phase 1 Obesity NCT03591718
Glucagon/GIP/ GLP-1 receptor triagonists NN9423 SC QD Phase 1 Obesity NCT03661879
HM15211 SC QW Phase 1 Obesity, NASH NCT03744182
LY3437943 SC QW Phase 1 T2DM NCT04143802
PYY analogs NN-9775 SC QW Phase 1 Obesity NCT03707990
NN-9747 SC QW Discontinued (Phase 1) Obesity NCT03574584

Molecules on the market or in active clinical development (global programs) with emphasis on molecules supported by preclinical or clinical publications. Select non-published molecules in active clinical development as well as discontinued molecules with public disclosures are also included.

Several important principles have emerged from clinical studies with GLP-1 receptor agonists. All molecules approved to date show potent glucose-lowering, driven partly by their direct effect on the beta cell to stimulate glucose-dependent insulin secretion, but also partly due to inhibition of glucagon secretion and, with some molecules, delayed gastric emptying (Nauck, Meier et al. 2017). These molecules cause weight loss, which has been primarily attributed to direct effects of GLP-1 on appetite-regulatory neurons in the hypothalamus (Secher, Jelsing et al. 2014); actions on the brainstem as well as vagal-mediated effects may also contribute under certain circumstances (Plamboeck, Veedfald et al. 2013, Holt, Richards et al. 2019). While short-acting GLP-1 receptor agonists show significant inhibition of gastric emptying, this effect can be attenuated in long-acting molecules (Knop, Bronden et al. 2017). Nausea and vomiting are the most common side effects and can limit dose and thus therapeutic efficacy. Molecules with lower peak-to-trough ratios appear to have fewer gastrointestinal side effects, and these side effects can be further managed through slow, step-wise dose escalation resulting in higher doses and better efficacy (Knop, Bronden et al. 2017, Aroda 2018, Nauck and Meier 2018). These insights have led to the development of the current generation of GLP-1 receptor agonists where molecules with optimized pharmacokinetic properties delivered via optimized dosing regimens achieve impressive glucose lowering and weight loss in patients with T2DM. Weight loss induced by these molecules is even more pronounced in non-diabetic individuals (Davies, Bergenstal et al. 2015, Pi-Sunyer, Astrup et al. 2015), leading to the development of some of these molecules as obesity therapies (Table 1).

Long-term cardiovascular outcome studies, a requirement for T2DM development programs, have revealed additional benefits of the GLP-1 class. The majority of GLP-1 receptor agonists shows a cardiovascular benefit, as evidenced by a decrease in the composite endpoint of myocardial infarction, stroke and cardiovascular death (Marso, Bain et al. 2016, Marso, Daniels et al. 2016, Nauck, Meier et al. 2017, Hernandez, Green et al. 2018, Gerstein, Colhoun et al. 2019). In addition, a decrease in albuminuria and a slower decline in glomerular filtration rate was observed in several studies, raising the possibility of renal benefits (Mann, Orsted et al. 2017, Luo, Lu et al. 2018, Tuttle, Lakshmanan et al. 2018, Gerstein, Colhoun et al. 2019). The mechanisms underlying these benefits are not clear. GLP-1 receptor agonists consistently show increases in heart rate and decreases in blood pressure, as well as small improvements in serum lipid profile, most notably decreased excursion of postprandial triglycerides and small improvements in LDL- and HDL-cholesterol, as well as anti-inflammatory effects, which could contribute to the observed cardiovascular and renal profiles (Nauck, Meier et al. 2017). Initial concerns about potential safety issues with GLP-1 receptor analogs, including medullary thyroid carcinoma and pancreatitis, have not manifested themselves as significant issues in the clinic (Nauck, Meier et al. 2017). Indeed, GLP-1 receptor agonists are now firmly established in the T2DM treatment paradigm and are the preferred therapy for patients with atherosclerotic cardiovascular disease, patients that would benefit from weight loss and individuals needing to achieve excellent glucose control without the risk of hypoglycemia (American Diabetes Association 2019). The next wave of innovation, now in progress, aims to develop orally bioavailable versions of GLP-1 receptor agonists to extend the benefits of GLP-1-based therapy to earlier stages of the diabetes treatment paradigm.

GIP and dual GIP/GLP-1 receptor agonists

Unlike GLP-1, very little work has been conducted to develop GIP as a stand-alone therapeutic (Finan, Muller et al. 2016, Holst 2019). In individuals with T2DM, GIP has little effect on insulin secretion, leading to the concept of diabetes as a GIP-resistant state (Nauck, Heimesaat et al. 1993). While the molecular basis for GIP resistance is not well understood, it was found to be closely linked to hyperglycemia (Vilsboll, Knop et al. 2003), and GIP-induced insulin secretion can be at least partially restored by treatments that normalize glucose levels such as insulin (Vilsboll, Knop et al. 2003, Hojberg, Vilsboll et al. 2009) or sulfonylureas (Meneilly, Bryer-Ash et al. 1993, Aaboe, Knop et al. 2009). Thus, combinations of GIP with other glucose-lowering agents could help unlock its therapeutic benefits. GIPinfusion studies have informed our understanding of GIP physiology and pharmacology. Most prominently, GIP is well known to increase circulating glucagon levels under conditions of hypoglycemia, likely reflecting direct effects of GIP on pancreatic alpha cells (Meier, Gallwitz et al. 2003, Christensen, Calanna et al. 2014). GIP activates adipose tissue blood flow and increases glucose and fatty acid uptake into adipose tissue in man, consistent with high level of GIP receptor expression in adipose tissue (Ahlqvist, Osmark et al. 2013, Asmar, Asmar et al. 2017). Unlike GLP-1, GIP does not affect gastric emptying, and no inhibition of food intake has been reported with peripherally administered GIP molecules either preclinically or in man (Finan, Muller et al. 2016, Holst 2019).

The ability of GIP to stimulate fatty acid uptake into adipose tissue, together with the observation of decreased weight gain in GIP or GIP receptor knock-out mice (Miyawaki, Yamada et al. 2002, Nasteska, Harada et al. 2014), has led to the notion that GIP may be an obesogenic molecule, prompting the development of GIP receptor antagonists as a therapeutic approach to obesity. Indeed, potent and selective GIP or GIP receptor antagonist antibodies decrease body weight in mice and non-human primates (Boylan, Glazebrook et al. 2015, Killion, Wang et al. 2018). On the other hand, long-acting GIP agonist peptides do not cause weight gain in preclinical models, and rather potentiate the weight loss induced by GLP-1, challenging the assumption of GIP as an obesogenic factor (Frias, Bastyr et al. 2017, Coskun, Sloop et al. 2018, Norregaard, Deryabina et al. 2018, Mroz, Finan et al. 2019).

The observation of synergistic weight loss with combined GIP and GLP-1 treatment in pre-clinical models prompted the development of dual GIP and GLP-1 receptor agonists (also termed “twincretins”) for the treatment of T2DM and obesity. The first molecules to be tested in man showed significant glucose lowering and weight loss compared to placebo, but did not differentiate itself significantly from the single GLP-1 receptor agonist liraglutide in early clinical studies (Finan, Ma et al. 2013, Frias, Bastyr et al. 2017). Recently, a novel dual GIP and GLP-1 receptor agonist, LY3298176 (now known as tirzepatide), demonstrated profound improvements in glucose control and body weight in a Phase 2b study when compared to dulaglutide, a single GLP-1 receptor agonist (Coskun, Sloop et al. 2018, Frias, Nauck et al. 2018). Tirzepatide achieved weight loss of >15% in roughly one fourth of patients, and normoglycemia (HbA1c of less than 5.7%) in roughly one third of patients, defining a new level of efficacy for T2DM therapy (Frias, Nauck et al. 2018). The mechanisms underlying the profound efficacy of tirzepatide is not fully understood. Compared to previous dual GIP/GLP-1 receptor agonists, tirzepatide has greater potency for the GIP receptor compared to the GLP-1 receptor, providing one possible explanation for its profound clinical benefit. Preclinical data demonstrate that both the GIP and the GLP-1 receptor contribute to the glucose lowering efficacy of tirzepatide (Coskun, Sloop et al. 2018). Clinical studies are ongoing to examine the mechanisms underlying the body weight and glucose lowering effects of tirzepatide in more detail. It will be interesting to see whether well-characterized GIP actions, such as effects on adipose tissue blood flow and nutrient uptake, contribute to the clinical efficacy of tirzepatide, and how the recently identified role of GIP in modulating appetite-regulatory circuits in mice (Adriaenssens, Biggs et al. 2019) could modulate body weight in the context of GLP-1 action. Overall, tirzepatide demonstrates the potential of dual incretin therapy and is sure to spark additional investigation into the actions of GIP. Tirzepatide recently advanced into Phase 3 clinical testing for T2DM with additional plans for testing in obesity and non-alcoholic steatohepatitis (NASH).

Oxyntomodulin, glucagon, and dual glucagon-GLP-1 receptor agonists

Oxyntomodulin (OXM), originally described as one of several gut-derived, glucagon-reactive peptides collective referred to as “enteroglucagon”, is a naturally occurring 37-amino acid product of the glucagon gene secreted by enteroendocrine L cells in response to a meal, with serum levels of < 2 pM and 10 pM in fasting versus postprandial state in humans (Pocai 2012, Cox, Berna et al. 2016, Lee, Chappell et al. 2016). Consistent with a common precursor and secretion from the same cell type, serum levels of OXM tightly correlate with circulating GLP-1 levels (Lee, Chappell et al. 2016). While no endogenous OXM receptor has been identified, OXM is a weak agonist to both the glucagon and the GLP-1 receptors with ~ 10- to 20-fold lower potency compared to glucagon or GLP-1 (Day, Ottaway et al. 2009, Kosinski, Hubert et al. 2012, Willard, Wootten et al. 2012). Due to its low serum levels, it is unlikely that OXM contributes significantly to GLP-1 or glucagon receptor signaling under normal conditions, and its physiological functions remain unknown. Interestingly, however, after bariatric surgery, postprandial OXM levels are increased up to 200 pM, reaching levels that might be expected to contribute to metabolic changes in this state (Wewer Albrechtsen, Hornburg et al. 2016).

Glucagon is well known as a hormone secreted by the alpha cells of the pancreas that acts on the liver to increase hepatic glucose output, thus raising glucose levels under conditions of hypoglycemia. Low levels of glucagon may also be secreted from the gut, as recently demonstrated in pancreatectomized individuals (Lund, Bagger et al. 2016), though the significance of this is unclear. Interestingly, glucagon has also been recognized as a weight regulatory hormone. Acute administration of glucagon inhibits food intake and increases energy expenditure in man (Penick and Hinkle 1961, Geary, Kissileff et al. 1992, Salem, Izzi-Engbeaya et al. 2016). In mice, administration of a long-acting glucagon analog decreases body weight primarily by increasing energy expenditure, an effect that appears to be mediated by the liver via both FGF21 and FXR (Habegger, Stemmer et al. 2013, Kim, Nason et al. 2018). In man, glucagon increases FGF21 serum levels and induces lipolysis, suggesting that some of the same pathways are engaged (Arafat, Kaczmarek et al. 2013, Habegger, Stemmer et al. 2013). Despite the promising effects of glucagon on body weight regulation, the potent hyperglycemic effects of glucagon in rodents and man (Habegger, Stemmer et al. 2013, Neylon, Moran et al. 2013) have prevented therapeutic development of glucagon analogs for obesity or T2DM.

Interestingly, coadministration of glucagon and GLP-1 in preclinical species results in weight loss beyond what can be achieved by GLP-1 alone, while maintaining the glucose-lowering effects of GLP-1 (Day, Ottaway et al. 2009, Pocai, Carrington et al. 2009, Kosinski, Hubert et al. 2012, Henderson, Konkar et al. 2016). In man, similar effects could be achieved by administering oxyntomodulin at doses expected to activate both the glucagon and the GLP-1 receptor (Cohen, Ellis et al. 2003, Wynne, Park et al. 2005, Wynne, Park et al. 2006, Tan, Behary et al. 2017, Shankar, Shankar et al. 2018). In both preclinical species and in man, glucagon increased energy expenditure, though modestly, even in the presence of GLP-1. Furthermore, in preclinical models, lipid profiles were profoundly improved compared to a selective GLP-1 receptor agonist, suggesting a potential benefit in the treatment of NASH. These findings sparked significant interest in exploring unimolecular combinations of glucagon and GLP-1 with several molecules progressing to clinical testing (Table 1). The most advanced of these molecules, SAR425899 and MEDI0382 (now known as cotadutide), show impressive weight loss in short term studies in obese or T2DM individuals as well as lowering of hepatic lipids (Ambery, Parker et al. 2018, Tillner, Posch et al. 2019). Glucose lowering was similar to what would be observed with single GLP-1 receptor agonists, suggesting that these molecules are able to balance GLP-1 and glucagon activity. It is important to note that no direct comparisons of dual glucagon/GLP-1 receptor agonists to single GLP-1 receptor agonists in man have been reported to date. While MEDI0382 has progressed into Phase 2b studies for T2DM, obesity and NASH, SAR425899 as well as another dual glucagon/GLP-1 receptor agonist, MRK-8521, have been discontinued, likely reflecting challenges in developing multifunctional glucagon/GLP-1 receptor agonists with a competitive profile, possibly due to suboptimal engagement of one of the two receptors (Table 1) or suboptimal pharmacokinetic profile.

Concurrent with the development of dual analogs, efforts are underway to combine the pharmacology of GLP-1, GIP and glucagon to achieve even more weight loss. In preclinical models, these combinations can normalize the body weight of very obese mice, and similar efficacy can be obtained with unimolecular GLP-1, GIP, and glucagon triagonists (Finan, Yang et al. 2015, Jall, Sachs et al. 2017). Several unimolecular triagonists are currently in development, with at least three molecules in early clinical testing (Table 1).

PYY

A gut-derived peptide that may play a complementary role to GLP-1 is peptide tyrosine tyrosine (PYY). PYY is a 36-amino acid peptide belonging to the pancreatic polypeptide family, which includes pancreatic polypeptide (PP) secreted from the pancreas and neuropeptide Y (NPY) produced by neurons in the central nervous system. While composed of similar hairpin-fold motif structures and sharing the same G-protein coupled NPY receptors (Y1, Y2, Y4, and Y5), PYY is predominantly secreted from L cells of the gut and functionally distinct from PP and NPY (Holzer, Reichmann et al. 2012).

PYY exists in two endogenous forms, PYY1–36 and PYY3–36, which differ in abundance in fasting versus postprandial states and in selectivity to NPY receptors. Similar to GLP-1, postprandial PYY concentrations increase and correlate with prolonged weight loss obtained by 8-week low calorie dieting followed by weight loss maintenance for 1 year (Iepsen, Lundgren et al. 2016). PYY3–36 is produced from PYY1–36 through the action of dipeptidyl peptidase IV (DPP4) and is the major postprandial circulating form of PYY (Adrian, Ferri et al. 1985, Adrian, Sagor et al. 1986, Medeiros and Turner 1994). Unlike GLP-1, the enzymatic cleavage of PYY1–36 by DPP4 does not reduce the affinity of PYY to a single receptor, rather it improves selectivity (Dumont, Fournier et al. 1995). Thus, while PYY1–36 binds all NPY receptors, PYY336 acts as a selective Y2 agonist whereby it exerts its unique action.

Initially discovered in the early 1980s, the first reports suggested a confined role of PYY in the gastrointestinal tract, including promoting gastric and pancreatic secretion and controlling gastrointestinal motility (Tatemoto and Mutt 1980, Lundberg, Tatemoto et al. 1982, Tatemoto 1982). However, the discovery that PYY3–36 plays a role in energy homeostasis via central action, demonstrated by experiments where exogenous administration of PYY3–36 reduces food intake in both rodents and humans and attenuates body weight gain in rodents, inspired interest in the Y2 receptor as a potential target in treating metabolic disease (Batterham, Cowley et al. 2002, Batterham, Cohen et al. 2003, Pittner, Moore et al. 2004). Moreover, further evidence suggests that PYY may play a role in insulin sensitivity and glucose uptake, likely via the Y2 receptor (Pittner, Moore et al. 2004, van den Hoek, Heijboer et al. 2004, Boey, Sainsbury et al. 2007). While these mechanisms are still being intensely investigated, the benefit of insulin sensitizing action for the treatment of metabolic diseases cannot be overstated.

PYY and GLP-1 have been demonstrated to be co-localized to the same L cells, same secretory vesicles, and shown to respond to the same stimuli, suggesting that PYY and GLP-1 play a complementary role (Habib, Richards et al. 2013). In support, co-infusions of PYY3–36 and GLP-1 additively / synergistically lower food intake in humans (Neary, Small et al. 2005, De Silva, Salem et al. 2011, Schmidt, Gregersen et al. 2014) and acute and chronic studies in rodents demonstrate additive / synergistic effects of PYY3–36 and GLP-1 on improvements in energy homeostasis (Neary, Small et al. 2005, Talsania, Anini et al. 2005, Kjaergaard, Salinas et al. 2019). While gastrointestinal side effects could limit use of PYY3–36 analogs / Y2 receptor agonists as a therapeutic target, especially when combined with GLP-1 analogs, a long-acting PYY analog reduced food intake in non-human primates with minimal emesis, even when coadministered with liraglutide which further reduced food intake (Rangwala, D’Aquino et al. 2019). Several long-acting PYY3–36 analogs / Y2 receptor agonists are being tested in early clinical development (Table 1), and it is anticipated that these molecules will ultimately be tested in combination with GLP-1 analogs. In addition, unimolecular approaches to PYY/GLP1 are being explored preclinically (Chepurny, Bonaccorso et al. 2018).

Other gut peptide-based approaches

Many additional gut-derived peptides have been discussed as candidates for new therapeutics for obesity and/or T2DM (Sun, Martin et al. 2018). However, to date, only a few have progressed to rigorous clinical testing. The reason for this is multi-fold, but concerns about efficacy, safety and/or tolerability, as well as lack of a differentiation, particularly when compared to current incretin-based therapies, are all likely contributing factors. For example, cholecystokinin (CCK), a hormone secreted by enteroendocrine I-cells, received significant attention following studies that demonstrated CCK-mediated inhibition of food intake in man (Miller and Desai 2016). Several CCK1-receptor agonists were generated and at least one molecule was tested in man in a robust Phase 2 study. However, while inhibition of gastric emptying and stimulation of gall bladder contraction were observed, body weight was not changed even in the face of increased gastrointestinal side effects, suggesting that this approach would not result in a viable therapy for weight loss (Jordan, Greenway et al. 2008, Miller and Desai 2016, Hornigold, Roth et al. 2018).

Ghrelin is a stomach-derived hormone that has been termed the “hunger hormone” due to its ability to stimulate appetite and increase food intake in preclinical studies and in man (Kojima, Hosoda et al. 1999, Schellekens, Dinan et al. 2010). Levels of ghrelin are highest in the fasting state and attenuate during nutrient consumption; caloric restriction that occurs during dieting leads to an increase in levels of circulating ghrelin and ghrelin levels are decreased in certain types of bariatric surgery (Geloneze, Tambascia et al. 2003, Lin, Gletsu et al. 2004, Schellekens, Dinan et al. 2010, Hernandez, Green et al. 2018). Numerous efforts aimed at identifying antagonists to ghrelin, its receptor and the enzyme responsible for ghrelin acylation, ghrelin-O-acyl transferase (GOAT), were conducted; however, molecules tested to date failed in early-stage clinical testing due to a variety of factors, including lack of clinically significant weight loss, poor selectivity, and side effect profiles (Schalla and Stengel 2019).

New and existing gut peptides continue to be explored with an emphasis on potential new therapeutic avenues. Secretin, a gut-derived peptide best known for regulating stomach pH and pancreatic bicarbonate secretion, was recently found to mediate thermogenesis and satiation via its action on brown adipose tissue, demonstrating that even supposedly well understood peptides can have unexplored functions (Li, Schnabl et al. 2018). Acute infusion of secretin activates glucose uptake into brown adipose tissue in man, suggesting a potential role as a mediator of postprandial thermogenesis. Unfortunately, chronic infusion of a secretin analog to mice failed to result in sustained body weight reductions, making it unlikely that secretin will become a therapeutic for weight loss (Li, Schnabl et al. 2018). Another interesting gut-derived peptide is neurotensin, an anorexigenic hormone whose circulating levels are upregulated by bariatric surgery (Ratner, Skov et al. 2016). Recently, long-acting neurotensin analogs were found to inhibit food intake in rodents and synergize with GLP-1 analogs to reduce body weight (Ratner, He et al. 2019). Neurotensin has a broad range of activities, including effects on the cardiovascular system, glucose metabolism, thermal regulation and pain that may limit its therapeutic usefulness; to date, no neurotensin analogues have been tested in man.

The ability to rapidly evaluate pharmacology and physiology in acute settings in man using peptide infusion, and the potential for added efficacy through combination therapy make gut peptides uniquely suited for evaluation as novel therapeutic approaches. The short-half-life and rapid renal clearance that have limited the therapeutic evaluation of gut peptides in the past can now be overcome by peptide engineering, allowing assessment of pharmacology in a chronic setting as well. These advances, together with increasingly sensitive methods for peptide detection and quantitation, make it likely that additional gut-derived therapies will emerge in the future.

What is the role of gut peptides in bariatric surgery?

Can gut peptide-based therapy mimic the effects of bariatric surgery?

During bariatric surgery, multiple gut peptides are dramatically regulated. Within a few days after bariatric surgery, postprandial levels of GLP-1, PYY and oxyntomodulin increase, in line with improvements in fasting glucose and insulin concentrations (le Roux, Welbourn et al. 2007, Falkén, Hellström et al. 2011, Wewer Albrechtsen, Hornburg et al. 2016). Further increases in GLP-1 up to 6-fold above pre-surgery levels are observed in the months following surgery (Korner, Inabnet et al. 2009, Yousseif, Emmanuel et al. 2014). Interestingly, no consistent changes in GIP levels are reported following bariatric surgery (Rao and Kini 2011), suggesting little effect of bariatric surgery on gut peptide secretion from the proximal small intestine, and ghrelin levels are decreased in some, but not all types of bariatric surgery, generally correlating with effects of surgery on the stomach rather than overall weight loss (Kalinowski, Paluszkiewicz et al. 2017). Importantly, in observational studies, elevations in GLP-1 and PYY correlate with the durability of weight loss in patients at 1, 10, and even 20 years after surgery, suggesting alterations in gut hormone secretion contribute to sustained weight loss and glucose control (Naslund, Gryback et al. 1997, Falkén, Hellström et al. 2011, Dar, Chapman et al. 2012).

Given the potent effect of bariatric surgery to increase GLP-1 levels and the effects of GLP-1 receptor agonists to affect both food intake and glucose regulation, it is reasonable to hypothesize that increased GLP-1 secretion is a primary driver of the effects of surgery. Yet, this topic remains highly controversial. In humans, short-term blockade of the GLP-1 receptor leads to a clear worsening of glucose tolerance in patients who have had RYGB (Salehi and D’Alessio 2016, Holst, Madsbad et al. 2018, Salehi, Gastaldelli et al. 2019). However, the limitation of these experiments include the short duration of these blockades as well as the impairment of glucose tolerance by pharmacological blockade of the GLP-1 receptor in patients without surgery. Hence, it is not easy to show an interaction between surgery-induced increases in GLP-1 levels and sensitivity to the blockade of GLP-1 signaling. In rodents, loss-of-function of the GLP-1 receptor does not reduce the effectiveness of VSG or RYGB to cause either weight loss or improve glucose regulation (Wilson-Perez, Chambers et al. 2013, Mokadem, Zechner et al. 2014), suggesting that GLP-1 receptor signaling is not required for these effects. Recently, β-cell specific knockouts of the GLP-1 receptor have been tested in a mouse model of VSG. Unfortunately, two reports come to different conclusions with one showing that such tissue-specific knockdown impairs the ability of VSG to increase insulin secretion and improve glucose tolerance (Garibay, Lou et al. 2018), while another shows no effect (Douros, Lewis et al. 2018). While these results may suggest that GLP-1 is not solely responsible for the metabolic effects of surgery, GLP-1 likely plays an important part of the cocktail of hormone changes that occurs after surgical interventions. Interestingly, antagonizing GLP-1 and PYY secretion with the somatostatin analog octreotide can increase food appeal post bariatric-surgery-induced weight loss (Goldstone, Miras et al. 2016), suggesting an important role for gut peptide combinations. However, in mice where both GLP-1 and PYY receptors are knocked out, RYGB is equally effective to cause weight loss and improve glucose control (Boland, Mumphrey et al. 2019).

An alternate approach has been to attempt to mimic the results of bariatric surgery with infusions of native gut peptides at concentrations similar to the ones observed after bariatric surgery. Acute studies that co-infused GLP-1, PYY and oxyntomodulin into obese volunteers showed promising results on food intake (Tan, Behary et al. 2017), prompting the initiation of chronic studies designed to examine the effects of these combinations on body weight (Behary, Tharakan et al. 2019) and reflective of efforts to create dual and triple agonists of these peptides as described above.

It is important to note that mechanisms other than gut peptides likely contribute to the effects of bariatric surgery. As we have already discussed, a wide range of evidence points to alterations in bile acids coming from the liver to contribute to the effects of bariatric surgery and there remains the possibility that neural signals from the gut contribute. In addition, bariatric surgery is associated with alterations in the gut (Murphy, Tsai et al. 2017), and some evidence from fecal transplants indicate a role for the changes in the gut microbiome in the effectiveness of surgery (Liou, Paziuk et al. 2013, Tremaroli, Karlsson et al. 2015, Murphy, Tsai et al. 2017). However, in none of these experiments does the effect of the transplant entirely recapitulate the potent effects of surgery. Whether this is a limitation of the fecal transplant methodology or a reflection of a limited role of the microbiome remains controversial. While combining either pre- or probiotic approaches with gut-peptide based therapies may ultimately achieve efficacy similar to what is seen with bariatric surgery, we should not underestimate the difficulties associated with manipulating the microbiome for therapeutic benefit. The current approaches such as, probiotics, prebiotics and fecal transplants are all very blunt tools that do alter the gut bacterial communities in numerous ways. Fecal transplants focus on using the bacteria from the distal colon. While the number of bacteria is much lower in the small intestine, there is evidence that these bacteria play a larger role in influencing host metabolism (Arora, Seyfried et al. 2017). Given that the composition in the ileum is quite different, we simply may not be transplanting the most helpful bacteria or insuring they colonize the appropriate location within the gastrointestinal tract. Despite a potentially important role, there is no guarantee that we can manipulate the gut microbiome with the precision necessary for therapeutic benefit.

Is the gut really the primary source of gut peptides?

Endogenous GLP-1 is rapidly processed by DPP4 in the gastrointestinal tract, resulting in very low circulating levels of active GLP-1. This raises the question of how gut-derived GLP-1 can sufficiently activate the GLP-1 receptor in distant tissues to achieve its physiological functions (Hjollund, Deacon et al. 2011, Mulvihill, Varin et al. 2017, Mulvihill 2018). The prohormone encoding GLP-1, preproglucagon, is also expressed in tissues outside of the gut. Recent work using tissue-specific reactivation of preproglucagon has allowed for a more careful examination of this issue. In the gut, reactivation of preproglucagon had little impact on overall glucose tolerance, while in the pancreas it resulted in a reversal of the phenotype observed in preproglucagon null mice. More importantly, the ability of GLP-1 receptor blockade to impair glucose tolerance was absent in both whole-body null and gut reactivated mice. However, when preproglucagon is reactivated in the pancreas, the ability to respond to GLP-1 receptor blockade is restored (Chambers, Sorrell et al. 2017). Along with other observations (Ellingsgaard, Hauselmann et al. 2011), such data indicate that a key source of endogenous GLP-1 is from the pancreas rather than from the gut. Despite the fact that the majority of circulating GLP-1 does largely originate in the gut, it is not the critical actor in at least some tissues. Additionally, recent data indicates that pancreatic glucagon can activate GLP-1 receptors to increase insulin secretion (Capozzi, Svendsen et al. 2019). This has two important implications. First, it implies that the effects of GLP-1 agonists may not be truly mimicking the effect of GLP-1 as a gut hormone. Rather, such agonists probably engage the system in a way that is quite distinct from circulating native GLP-1. Second, as discussed below, one strategy towards novel therapies is to identify ways to activate intestinal cells that make GLP-1 and cause them to increase their secretion. If gut-derived GLP-1 is not a critical factor, this strategy may be less effective than what was originally thought.

Other Gut-based Approaches

Inhibition of Gut Peptide Degradation as a Therapeutic Strategy: DPP4 Inhibitors

Gut peptides are typically short-acting hormones whose duration and site of action is limited by rapid degradation, suggesting inhibition of gut peptide degradation as an alternative therapeutic approach. It is estimated that >90% of GLP-1 secreted by the L cell is degraded before it reaches the portal circulation (Hjollund, Deacon et al. 2011, Mulvihill, Varin et al. 2017, Mulvihill 2018). DPP4, an enzyme present both on the surface of vascular endothelial cells and in circulation, has been identified as the major GLP-1 degrading enzyme (Kieffer, McIntosh et al. 1995), resulting in the development of DPP4 inhibitors as an alternate way to harness the therapeutic benefits of GLP-1.

Ten DPP4 inhibitors are currently approved for the treatment of T2DM with several additional molecules under development (Table 2)(Deacon and Lebovitz 2016). DPP4 inhibitors have become an important component of diabetes therapy and are recommended as first-line therapy for a subset of individuals with T2DM (American Diabetes 2019). The pharmacological properties of these molecules, their clinical efficacy, safety, and long-term outcomes are remarkably consistent (Deacon and Lebovitz 2016, Nauck, Meier et al. 2017, Yamada, Shojima et al. 2018). DPP4 inhibitors as a class have excellent tolerability, show modest glucose lowering, and are weight neutral. Large outcome studies, conducted for five DPP4 inhibitors to date, confirmed safety, but interestingly showed no significant cardiovascular benefit (Nauck, Meier et al. 2017, McGuire, Alexander et al. 2019). Furthermore, no signals suggesting a renal benefit were detected in a meta-analysis of outcomes studies conducted to date (Luo, Lu et al. 2018).

Table 2:

Small molecule approaches to pathways related to gut peptides.

Class Key Molecules Administration Status Indication References
Gut peptide degradation inhibitors
DPP4 inhibitors sitagliptin oral QD Marketed T2DM (Deacon and Lebovitz 2016)
vildagliptin oral QD Marketed T2DM (Deacon and Lebovitz 2016)
linagliptin oral QD Marketed T2DM (Deacon and Lebovitz 2016)
saxagliptin oral QD Marketed T2DM (Deacon and Lebovitz 2016)
alogliptin oral QD Marketed T2DM (Deacon and Lebovitz 2016)
Putative gut peptide secretagogues tested in the clinic
GPR119 agonists DS-8500a oral QD Phase 2 T2DM (Matsumoto, Yoshitomi et al. 2018, Yamada, Terauchi et al. 2018)
GSK1292263 oral QD Phase 2 (discontinued) T2DM (Nunez, Bush et al. 2014)
BMS-903452 oral QD Phase 1 (discontinued) T2DM (Wacker, Wang et al. 2014)
JNJ-38431055 oral QD Phase 1 (discontinued) T2DM (Katz, Gambale et al. 2011, Semple, Ren et al. 2011)
GPR40 agonists (partial) SHR0534 oral QD Phase 1 T2DM (Li, Qiu et al. 2016)
TAK-875 (Fasiglifam) oral QD Phase 3 (discontinued) T2DM (Kaku, Enya et al. 2016, Menon, Lincoff et al. 2018)
LY2881835 LY2922083 LY2922470 oral QD Phase 1 (discontinued) T2DM (Chen, Li et al. 2016, Hamdouchi, Kahl et al. 2016)
MK-8666 oral QD Phase 1 (discontinued) T2DM (Krug, Vaddady et al. 2017, Lu, Byrne et al. 2017)
Putative gut peptide secretagogues with preclinical data only
GPR40 full agonists AMG-1638; AM-6226 NA Preclinical T2DM (Brown, Dransfield et al. 2012, Luo, Swaminath et al. 2012)
BMS-986118 NA Preclinical T2DM (Shi, Gu et al. 2018)
Cpd 12 NA Preclinical T2DM (Meegalla, Huang et al. 2018)
AP1; AP3 NA Preclinical T2DM (Pachanski, Kirkland et al. 2017)
GPR120 agonists TUG-891 NA Preclinical T2DM (Schilperoort, van Dam et al. 2018)
Cpd 4x NA Preclinical T2DM (Zhang, Cai et al. 2017)
Cpd 34 NA Preclinical T2DM (Azevedo, Watterson et al. 2016)
TGR5 agonists INT-777 NA Preclinical T2DM (Pellicciari, Gioiello et al. 2009, Li, Holmstrom et al. 2011)
Cpd 18 NA Preclinical T2DM (Briere, Ruan et al. 2015)
GPR142 agonists Cpd 33; Cpd 23 NA Preclinical T2DM (Toda, Hao et al. 2013, Yu, Lizarzaburu et al. 2013)
Cpd 40 NA Preclinical T2DM (Wilson, Kurukulasuriya et al. 2016)
GPR39 agonists Cpd 3 NA Preclinical T2DM (Peukert, Hughes et al. 2014)
SSTR5 antagonists S5A1 NA Preclinical T2DM (Farb, Adeva et al. 2017)
Cpd 10 NA Preclinical T2DM (Liu, Shao et al. 2018)
Cpd 25a NA Preclinical T2DM (Hirose, Yamasaki et al. 2017)

Molecules on the market or in active clinical development; discontinued molecules included if published information available. Select preclinical molecules included for mechanisms that have not progressed to the clinic to date.

The therapeutic profile of DPP4 inhibitors differs significantly from GLP-1 receptor agonists. For one, glucose lowering mediated by DPP4 inhibitors is more modest than what is achievable with GLP-1 analogs. In addition, weight loss is not observed, gastrointestinal side effects, such as nausea and vomiting, are not found, and other on-target effects of GLP-1 receptor agonists, such as heart rate increases and inhibition of gastric emptying, are also not observed with DPP4 inhibitors (Nauck, Meier et al. 2017, Aroda 2018). A likely explanation is that the increase of GLP-1 by DPP4 inhibitors is limited by the total amount of GLP-1 secreted endogenously. In man, concentrations of DPP4 inhibitors that inhibit circulating DPP4 activity by >90% and achieve maximum therapeutic benefits, result in serum levels of intact, active GLP-1 of ~ 15–30 pM postprandially (Herman, Bergman et al. 2006, Addy, Tatosian et al. 2016). This is significantly lower than the ~200 pM of native GLP-1 required for maximal pharmacological effects in GLP-1 infusion studies (Nauck and Meier 2018) and the 50 pM amount of GLP-1 equivalents that has been estimated to be required to achieve pharmacological effects (Calara, Taylor et al. 2005, Kothare, Linnebjerg et al. 2008).

A large number of proteins and peptides have been identified as potential substrates for DPP4 (Nauck, Meier et al. 2017), raising the question of whether gut peptides other than GLP-1 may contribute to the efficacy of DPP4 inhibitors. Indeed, mice lacking GLP-1 (and other preproglucagon-derived peptides), are able to still respond to DPP4 inhibitors with glucose-lowering (Mulvihill 2018) and a study in T2DM subjects treated with a combination of a DPP4 inhibitor and the GLP-1 receptor antagonist exendin-9 suggested that as much as 50% of the glucose lowering observed with DPP4 inhibitors in man may be attributable to non-GLP-1 effects (Aulinger, Bedorf et al. 2014). Interestingly, DPP4 is also the major degrading enzyme for GIP (Kieffer, McIntosh et al. 1995), and DPP4 inhibitors increase the levels of circulating active GIP even more than circulating GLP-1 levels (up to 60 pM postprandially) (Yamada, Shojima et al. 2018). Additional peptides may contribute to the overall therapeutic profile of DPP4 inhibitors, although direct evidence is lacking. For example, processing of PYY1–36 to PYY3–36 by DPP4 changes the receptor selectivity profile, allowing it to activate the NPYR2 receptor to inhibit food intake (Mentlein, Dahms et al. 1993, Ostergaard, Kofoed et al. 2018). DPP4 inhibitors significantly reduce levels of circulating PYY3–36 (Aaboe, Knop et al. 2010), possibly limiting the effects of endogenous PYY on food intake. On the other hand, inhibition of intra-islet PYY processing via DPP4 has been linked to increased insulin secretion in vitro via the NPYR1 receptor, suggesting that PYY could contribute to glucose-lowering activities of DPP4 inhibitors (Guida, McCulloch et al. 2018). It is important to note, however, that definite proof for a role of PYY in the actions of DPP4 inhibitor is lacking. It is well established that DPP4 inhibitors decrease GLP-1 secretion through feedback inhibition on the L-cell, lowering the circulating concentration of total GLP-1 as well as other gut peptides. Overall, the mechanism of action of DPP4 inhibitors may be more complex than initially appreciated, reflecting its action on multiple peptides.

Activation of Nutrient Receptors in the Gut as a Therapeutic Strategy

Given that inhibition of gut peptide degradation resulted in DPP4 inhibitors as a new class of diabetes therapy, could increasing gut peptide secretion also open new therapeutic avenues? Furthermore, could the combination of both approaches allow even more powerful glucose lowering and weight loss? Numerous receptors – many of them highly druggable G protein coupled receptors – modulate gut peptide secretion in response to nutrients, such as fatty acids, amino acids, carbohydrates and other stimuli (Gribble and Reimann 2019). Despite a large amount of effort dedicated to these receptors, however, therapeutics have so far been elusive (see Table 2 for key clinical and preclinical efforts to date).

Receptors for bile acids

In addition to aiding the processing of dietary fats and fat-soluble nutrients, bile acids activate the nuclear hormone receptor farnesoid X receptor (FXR) and the G protein-coupled bile acid receptor 1, more commonly called TGR5. While FXR activation regulates bile acid synthesis and triglyceride metabolism (Ding, Yang et al. 2015), TGR5 activation results in the release of GLP-1 and PYY from L-cells and increases energy expenditure, at least in mice (Katsuma, Hirasawa et al. 2005, Watanabe, Houten et al. 2006). FXR agonists are currently under investigation for the treatment of NASH with promising results in clinical studies to date (Malhi and Camilleri 2017, Connolly, Ooka et al. 2018). While early studies with TGR5 agonists, such as the bile acid analog, INT-777, established strong effects on incretin secretion and glucose excursions in rodents (Pellicciari, Gioiello et al. 2009, Thomas, Gioiello et al. 2009), target-mediated side effects have so far prevented TGR5 activators from entering clinical evaluation. Specifically, activation of TGR5 leads to smooth muscle relaxation and stimulation of gallbladder filling, resulting in large increases in gall bladder size (Li, Holmstrom et al. 2011) that could not be separated from elevations in GLP-1 or glucose homeostasis (Briere, Ruan et al. 2015). Given the high expression of TGR5 in gallbladder (Keitel, Cupisti et al. 2009), current efforts are focused on reducing systemic exposure of a TGR5 agonist by creating gut-restricted compounds.

Receptors for long-chain fatty acids and related lipids

The G-protein coupled receptors GPR40, GPR119 and GPR120 share an affinity for long-chain fatty acids and related molecules. Interest in GPR119 increased significantly after its identification as a receptor that mediates the inhibitory effects of 2-monoacylglycerols, specifically oleoylethanolamide, on food intake (Overton, Babbs et al. 2006). In addition, GPR119 is highly expressed in enteroendocrine cells in the gut as well as in pancreatic islets. Numerous potent and selective GPR119 agonists have been identified and preclinical studies show a consistent picture: molecules induce glucose-dependent insulin secretion and modest increases in GLP-1 and GIP secretion, but have little or no effect on body weight (Ritter, Buning et al. 2016, Matsumoto, Yoshitomi et al. 2018). Clinical studies were conducted with more than 10 molecules. While glucose lowering was observed, the effect size was modest, impact on GLP-1 and GIP levels was minimal (<50%) and no significant weight loss was observed (Katz, Gambale et al. 2011, Semple, Ren et al. 2011, Katz, Gambale et al. 2012, Nunez, Bush et al. 2014, Wacker, Wang et al. 2014, Matsumoto, Yoshitomi et al. 2018, Yamada, Terauchi et al. 2018). These modest effects led to discontinuation of the vast majority of GPR119 agonist efforts.

GPR40 agonists were originally developed as glucose-dependent insulin secretagogues and later recognized to also have potential as GLP-1 secretagogues, reflecting expression of this receptor on pancreatic beta cells as well as enteroendocrine cells. First generation GPR40 agonists, such as fasiglifam (TAK-875), are partial agonists that showed significant glucose-lowering in the clinic (Araki, Hirayama et al. 2012), but did not increase circulating GLP-1 levels either preclinically or clinically (Luo, Swaminath et al. 2012, Chen, Li et al. 2016, Hamdouchi, Kahl et al. 2016); no effect on body weight was noted. While development of fasiglifam was discontinued due to concerns about drug-induced liver injury (Menon, Lincoff et al. 2018), several partial GPR40 agonists remain in clinical development. Recently, full agonists of the GPR40 receptor have been developed that bind to a distinct site and activate both Gαq and Gαs signaling (Brown, Dransfield et al. 2012, Lin, Guo et al. 2012, Luo, Swaminath et al. 2012). In preclinical studies, these molecules are among the strongest insulin and incretin secretagogues described, resulting in 3- to 5-fold increases in circulating GLP-1 levels as well as elevated levels of PYY and GIP, and strong increases of glucose-dependent insulin secretion. Despite significant efforts across the industry, to date, however, no full GPR40 agonists have entered clinical development.

GPR120 is a receptor for long-chain fatty acids expressed in multiple tissues, most prominently adipocytes, immune cells and enteroendocrine cells of the gut. Numerous companies as well as academic investigators have developed specific small molecule GPR120 agonists and demonstrated glucose lowering in rodents in acute and chronic settings (Oh, Walenta et al. 2014, Azevedo, Watterson et al. 2016, Cox, Chu et al. 2017, Sparks, Aquino et al. 2017, Sundstrom, Myhre et al. 2017, Zhang, Cai et al. 2017, Sheng, Yang et al. 2018, Winters, Sui et al. 2018). However, GLP-1 secretion has only been reported for a couple of these molecules with mixed results (Oh, Walenta et al. 2014, Sundstrom, Myhre et al. 2017), and development of potent and selective GPR120 molecules has been challenging. To date, no clinical studies with a GPR120 agonist have been reported.

Receptors for amino acids

A relatively recent nutrient receptor to be explored is GPR142, now recognized to be a receptor for aromatic amino acids that regulates secretion of incretins from the gut, as well as glucagon and insulin secretion from pancreatic islets (Rudenko, Shang et al. 2019). Potent and selective small molecule GPR142 agonists increase insulin and glucagon secretion from mouse and human islets, improve oral glucose tolerance upon acute administration in lean and obese mice, obese rats as well as in monkeys, and improve insulin sensitivity and basal glucose levels upon chronic administration to diet-induced obese mice (Toda, Hao et al. 2013, Yu, Lizarzaburu et al. 2013, Guo, Parker et al. 2016, Wilson, Kurukulasuriya et al. 2016, Rudenko, Shang et al. 2019). Circulating gut peptides, including GLP-1, GIP, and CCK are significantly increased in both rodents and non-human primates with GPR142 agonists with a particularly strong (~ 5-fold) effect on GIP (Lin, Efanov et al. 2016, Rudenko, Shang et al. 2019). Interestingly, GPR142 agonists can induce both beta-cell proliferation as well as protect beta-cells from stress in isolated islets in vitro, suggesting a potential for increased durability of therapy (Lin, Wang et al. 2018). To date, no clinical studies with a GPR142 agonist have been reported.

Other gut receptors with potential therapeutic relevance

Many additional receptors are highly expressed on intestinal L and/or K cells, raising the possibility of novel therapeutic targets that could potentially mediate GLP-1 secretion under physiological or pharmacological conditions. Among these, the short-chain fatty acid receptors GPR41 (FFAR2) and GPR43 (FFAR3), as well as GPR39, an orphan GPCR highly expressed in various metabolically important tissues including enteroendocrine cells, pancreatic beta cells and hepatocytes, have received significant attention (Tolhurst, Heffron et al. 2012, Peukert, Hughes et al. 2014, Christiansen, Gabe et al. 2018). However, pharmacological data to date are mixed and no molecules have entered clinical development (Peukert, Hughes et al. 2014, Fjellstrom, Larsson et al. 2015, Frimurer, Mende et al. 2017).

Inhibiting the brake: Can somatostatin antagonists be useful for the treatment of diabetes?

Somatostatin (SST) is a hormone produced by delta cells in the duodenum and pyloric regions of the gut, the pancreas, and the brain (Patel and Reichlin 1978, Hauge-Evans, King et al. 2009). SST has two active forms, SST-14, secreted from the hypothalamus, gut, and pancreas, and SST-28, secreted almost exclusively by intestinal cells (Shen, Pictet et al. 1982, O’Carroll, Lolait et al. 1992, Patel 1999). In humans, the major circulating form is SST-28 (Polonsky, Shoelson et al. 1983) and SST-28 serum levels appear nutritionally regulated, increasing by a small amount after a fat challenge (D’Alessio and Ensinck 1990). SST isoforms bind to a family of five receptors (SSTR1, SSTR2, SSTR3, SSTR4, and SSTR5) that are all Gαi-coupled receptors that signal via cAMP (Patel 1999). While SST-14 binds to all SSTRs similarly, SST-28 preferentially binds to SSTR5. Expression profiles of SSTRs differ from tissue to tissue; for a complete review see (Gunther, Tulipano et al. 2018).

Somatostatin is a potent inhibitor of intestinal gut hormone secretion, affecting GLP-1, GIP, PYY, ghrelin and other hormones (Vu, van Oostayen et al. 2001, Tan, Vanderpump et al. 2004, Rigamonti, Cella et al. 2011). In fact, a study in healthy volunteers showed complete suppression of the postprandial rise in GLP-1 and GIP serum levels upon treatment with the somatostatin analog pasireotide (Henry, Ciaraldi et al. 2013). Moreover, somatostatin analogs have been used clinically to reverse high levels of plasma GLP-1 and PYY observed after bariatric surgery (Goldstone, Miras et al. 2016). Of the five characterized somatostatin receptors, SSTR5 is highly expressed in enteroendocrine cells (Chisholm and Greenberg 2002) and the selectivity profile of the somatostatin analog pasireotide, as well as in vitro and in vivo studies in rodents, swine and sheep all point to activation of somatostatin receptor 5 (SSTR5) on intestinal L-cells as the most likely mechanism mediating its inhibitory effects on GLP-1 secretion (Martin and Faulkner 1996, Hansen, Hartmann et al. 2000, Farb, Adeva et al. 2017). An intriguing approach is to inhibit SSTR5 to relieve the inhibitory effect of endogenous somatostatin on gut peptide secretion. Studies in preclinical models suggest that this can lead to very large increases in GLP-1 while at the same time increasing insulin secretion due to relieving inhibition of SST-14 in human islets (Farb, Adeva et al. 2017). It will be interesting to see if somatostatin 5 receptor antagonists will show efficacy in the clinic.

Combination approaches

Can one combine different approaches to modulate gut peptide secretion and stability to achieve efficacy more similar to what is possible with combination gut peptide approaches? One proposed combination strategy aims to mimic the changes observed in bariatric surgery by simultaneously targeting three GLP-1 regulatory nodes: an agonist to a gut-localized GPCR (TGR5 or second generation GPR40 agonist) to promote GLP-1 secretion, a SSTR5 antagonist to blunt inhibition of GLP-1 release, and a DPP4 inhibitor to preserve active GLP-1 (Briere, Bueno et al. 2018). Preclinically, this approach results in levels of active GLP-1 (100–400 pM) that are higher than what is observed with bariatric surgery or with GLP-1 receptor agonists (Briere, Bueno et al. 2018, Sloop, Briere et al. 2018). Whether these increases translate into efficacy similar to what is observed with current combination peptide approaches in man remains to be determined

The future

There remain many further opportunities to reverse engineer the physiological effects of surgery to find less invasive and more scalable therapeutic strategies. Moreover, they may involve less invasive procedures, new pharmacotherapies or nutrition strategies that will make effective therapies available to more individuals. A wide range of new technologies will facilitate these activities. The ability to use TRAPseq and DROPseq in the GI tract will clearly provide for a much deeper understanding of gut function and provide clues as to how existing interventions alter critical signals coming from the gut. A deeper map of the entire gut will be available in the next several years that will make it possible to mine many more potential targets for intervention. In addition, the ability to visualize ongoing neural activity in vivo and the ability to manipulate specific populations of neurons using both DREADD and optogenetics are already revolutionizing our understanding of the vagus and its targets in the caudal brainstem (Williams, Chang et al. 2016). Such strategies can be applied to the enteric nervous system as well. It is hard to imagine that a better understanding of the major neural signals from the gut won’t lead to additional targets for intervention.

Finally, we would be remiss to not mention the possibility of combining gut-peptide based approaches with therapies based on circulating hormones derived from tissues other than the gut. For example, analogs of FGF21, a liver-derived circulating hormone, have been demonstrated to cause weight-loss, lipid lowering and insulin sensitizing effects in man, and have emerged as an attractive therapeutic approach for NASH (Sumida and Yoneda 2018, Kliewer and Mangelsdorf 2019). Preclinically, FGF21 analogs show synergistic effects on body weight when combined with GLP-1. Similarly, the beta-cell-derived hormone amylin has well characterized effects on food intake and glucose control, and long-acting amylin analogs as well as dual amylin-calcitonin receptor agonists are being evaluated for obesity and T2DM both alone and in combination with GLP-1 analogs (Srivastava and Apovian 2018). It is likely that in the future gut- and non-gut-based therapies will be combined with the goal of normalizing body weight and glucose control to a degree not possible with current therapies.

Perspective

We are amid a revolution that will rapidly increase our understanding of the complexities of gut function and its impact on a wide range of physiology. The challenge will be to turn these insights into safe and efficacious therapies to more effectively deal with the twin epidemics of obesity and type 2 diabetes. Gut function and the signals generated by the gut have already led to important advances in the treatments for obesity and diabetes. However, there is an urgent need for next generation therapies for obesity and T2DM that not only address weight loss and glucose control in the short term, but also provide durability of these effects. Ideally, some of these treatments could provide sustained benefits, resulting in diabetes remission similar to what is observed after bariatric surgery. It will be interesting to see whether the gut can live up to the challenge and help identify this next generation of therapies.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Aaboe K, Knop FK, Vilsboll T, Deacon CF, Holst JJ, Madsbad S and Krarup T (2010). “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 12(4): 323–333. [DOI] [PubMed] [Google Scholar]
  2. Aaboe K, Knop FK, Vilsboll T, Volund A, Simonsen U, Deacon CF, Madsbad S, Holst JJ and Krarup T (2009). “KATP channel closure ameliorates the impaired insulinotropic effect of glucose-dependent insulinotropic polypeptide in patients with type 2 diabetes.” J Clin Endocrinol Metab 94(2): 603–608. [DOI] [PubMed] [Google Scholar]
  3. Addy C, Tatosian D, Glasgow XS, Gendrano IN 3rd, Kauh E, Martucci A, Johnson-Levonas AO, Selverian D, Matthews CZ, Gutierrez M, Wagner JA and Aubrey Stoch S (2016). “Pharmacokinetic and Pharmacodynamic Effects of Multiple-dose Administration of Omarigliptin, a Once-weekly Dipeptidyl Peptidase-4 Inhibitor, in Obese Participants With and Without Type 2 Diabetes Mellitus.” Clin Ther 38(3): 516–530. [DOI] [PubMed] [Google Scholar]
  4. Adriaenssens AE, Biggs EK, Darwish T, Tadross J, Sukthankar T, Girish M, Polex-Wolf J, Lam BY, Zvetkova I, Pan W, Chiarugi D, Yeo GSH, Blouet C, Gribble FM and Reimann F (2019). “Glucose-Dependent Insulinotropic Polypeptide Receptor-Expressing Cells in the Hypothalamus Regulate Food Intake.” Cell Metab 30(5): 987–996 e986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Adrian TE, Ferri GL, Bacarese-Hamilton AJ, Fuessl HS, Polak JM and Bloom SR (1985). “Human distribution and release of a putative new gut hormone, peptide YY.” Gastroenterology 89(5): 1070–1077. [DOI] [PubMed] [Google Scholar]
  6. Adrian TE, Sagor GR, Savage AP, Bacarese-Hamilton AJ, Hall GM and Bloom SR (1986). “Peptide YY kinetics and effects on blood pressure and circulating pancreatic and gastrointestinal hormones and metabolites in man.” J Clin Endocrinol Metab 63(4): 803–807. [DOI] [PubMed] [Google Scholar]
  7. Ahlqvist E, Osmark P, Kuulasmaa T, Pilgaard K, Omar B, Brons C, Kotova O, Zetterqvist AV, Stancakova A, Jonsson A, Hansson O, Kuusisto J, Kieffer TJ, Tuomi T, Isomaa B, Madsbad S, Gomez MF, Poulsen P, Laakso M, Degerman E, Pihlajamaki J, Wierup N, Vaag A, Groop L and Lyssenko V (2013). “Link between GIP and osteopontin in adipose tissue and insulin resistance.” Diabetes 62(6): 2088–2094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Al-Najim W, Docherty NG and le Roux CW (2018). “Food Intake and Eating Behavior After Bariatric Surgery.” Physiol Rev 98(3): 1113–1141. [DOI] [PubMed] [Google Scholar]
  9. Albaugh VL, Banan B, Antoun J, Xiong Y, Guo Y, Ping J, Alikhan M, Clements BA, Abumrad NN and Flynn CR (2019). “Role of Bile Acids and GLP-1 in Mediating the Metabolic Improvements of Bariatric Surgery.” Gastroenterology 156(4): 1041–1051 e1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Ambery P, Parker VE, Stumvoll M, Posch MG, Heise T, Plum-Moerschel L, Tsai LF, Robertson D, Jain M, Petrone M, Rondinone C, Hirshberg B and Jermutus L (2018). “MEDI0382, a GLP-1 and glucagon receptor dual agonist, in obese or overweight patients with type 2 diabetes: a randomised, controlled, double-blind, ascending dose and phase 2a study.” Lancet 391(10140): 2607–2618. [DOI] [PubMed] [Google Scholar]
  11. American Diabetes Association (2019). “9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes-2019.” Diabetes Care 42(Suppl 1): S90–S102. [DOI] [PubMed] [Google Scholar]
  12. Arafat AM, Kaczmarek P, Skrzypski M, Pruszynska-Oszmalek E, Kolodziejski P, Szczepankiewicz D, Sassek M, Wojciechowicz T, Wiedenmann B, Pfeiffer AF, Nowak KW and Strowski MZ (2013). “Glucagon increases circulating fibroblast growth factor 21 independently of endogenous insulin levels: a novel mechanism of glucagon-stimulated lipolysis?” Diabetologia 56(3): 588–597. [DOI] [PubMed] [Google Scholar]
  13. Araki T, Hirayama M, Hiroi S and Kaku K (2012). “GPR40-induced insulin secretion by the novel agonist TAK-875: first clinical findings in patients with type 2 diabetes.” Diabetes Obes Metab 14(3): 271–278. [DOI] [PubMed] [Google Scholar]
  14. Aroda VR (2018). “A review of GLP-1 receptor agonists: Evolution and advancement, through the lens of randomised controlled trials.” Diabetes Obes Metab 20 Suppl 1: 22–33. [DOI] [PubMed] [Google Scholar]
  15. Arora T, Seyfried F, Docherty NG, Tremaroli V, le Roux CW, Perkins R and Backhed F (2017). “Diabetes-associated microbiota in fa/fa rats is modified by Roux-en-Y gastric bypass.” ISME J 11(9): 2035–2046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Asmar M, Asmar A, Simonsen L, Gasbjerg LS, Sparre-Ulrich AH, Rosenkilde MM, Hartmann B, Dela F, Holst JJ and Bulow J (2017). “The Gluco- and Liporegulatory and Vasodilatory Effects of Glucose-Dependent Insulinotropic Polypeptide (GIP) Are Abolished by an Antagonist of the Human GIP Receptor.” Diabetes 66(9): 2363–2371. [DOI] [PubMed] [Google Scholar]
  17. Aulinger BA, Bedorf A, Kutscherauer G, de Heer J, Holst JJ, Goke B and Schirra J (2014). “Defining the role of GLP-1 in the enteroinsulinar axis in type 2 diabetes using DPP-4 inhibition and GLP-1 receptor blockade.” Diabetes 63(3): 1079–1092. [DOI] [PubMed] [Google Scholar]
  18. Azevedo CM, Watterson KR, Wargent ET, Hansen SV, Hudson BD, Kepczynska MA, Dunlop J, Shimpukade B, Christiansen E, Milligan G, Stocker CJ and Ulven T (2016). “Non-Acidic Free Fatty Acid Receptor 4 Agonists with Antidiabetic Activity.” J Med Chem 59(19): 8868–8878. [DOI] [PubMed] [Google Scholar]
  19. Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, Frost GS, Ghatei MA and Bloom SR (2003). “Inhibition of food intake in obese subjects by peptide YY3–36.” N Engl J Med 349(10): 941–948. [DOI] [PubMed] [Google Scholar]
  20. Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, Wren AM, Brynes AE, Low MJ, Ghatei MA, Cone RD and Bloom SR (2002). “Gut hormone PYY(3–36) physiologically inhibits food intake.” Nature 418(6898): 650–654. [DOI] [PubMed] [Google Scholar]
  21. Behary P, Tharakan G, Alexiadou K, Johnson N, Wewer Albrechtsen NJ, Kenkre J, Cuenco J, Hope D, Anyiam O, Choudhury S, Alessimii H, Poddar A, Minnion J, Doyle C, Frost G, Le Roux C, Purkayastha S, Moorthy K, Dhillo W, Holst JJ, Ahmed AR, Prevost AT, Bloom SR and Tan TM (2019). “Combined GLP-1, Oxyntomodulin, and Peptide YY Improves Body Weight and Glycemia in Obesity and Prediabetes/Type 2 Diabetes: A Randomized, Single-Blinded, Placebo-Controlled Study.” Diabetes Care 42(8): 1446–1453. [DOI] [PubMed] [Google Scholar]
  22. Boey D, Sainsbury A and Herzog H (2007). “The role of peptide YY in regulating glucose homeostasis.” Peptides 28(2): 390–395. [DOI] [PubMed] [Google Scholar]
  23. Boland BB, Mumphrey MB, Hao Z, Townsend RL, Gill B, Oldham S, Will S, Morrison CD, Yu S, Munzberg H, Rhodes CJ, Trevaskis JL and Berthoud HR (2019). “Combined loss of GLP-1R and Y2R does not alter progression of high-fat diet-induced obesity or response to RYGB surgery in mice.” Mol Metab 25: 64–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Boylan MO, Glazebrook PA, Tatalovic M and Wolfe MM (2015). “Gastric inhibitory polypeptide immunoneutralization attenuates development of obesity in mice.” Am J Physiol Endocrinol Metab 309(12): E1008–1018. [DOI] [PubMed] [Google Scholar]
  25. Briere DA, Bueno AB, Gunn EJ, Michael MD and Sloop KW (2018). “Mechanisms to Elevate Endogenous GLP-1 Beyond Injectable GLP-1 Analogs and Metabolic Surgery.” Diabetes 67(2): 309–320. [DOI] [PubMed] [Google Scholar]
  26. Briere DA, Ruan X, Cheng CC, Siesky AM, Fitch TE, Dominguez C, Sanfeliciano SG, Montero C, Suen CS, Xu Y, Coskun T and Michael MD (2015). “Novel Small Molecule Agonist of TGR5 Possesses Anti-Diabetic Effects but Causes Gallbladder Filling in Mice.” PLoS One 10(8): e0136873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Brown SP, Dransfield PJ, Vimolratana M, Jiao X, Zhu L, Pattaropong V, Sun Y, Liu J, Luo J, Zhang J, Wong S, Zhuang R, Guo Q, Li F, Medina JC, Swaminath G, Lin DC and Houze JB (2012). “Discovery of AM-1638: A Potent and Orally Bioavailable GPR40/FFA1 Full Agonist.” ACS Med Chem Lett 3(9): 726–730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Calara F, Taylor K, Han J, Zabala E, Carr EM, Wintle M and Fineman M (2005). “A randomized, open-label, crossover study examining the effect of injection site on bioavailability of exenatide (synthetic exendin-4).” Clin Ther 27(2): 210–215. [DOI] [PubMed] [Google Scholar]
  29. Capozzi ME, Svendsen B, Encisco SE, Lewandowski SL, Martin MD, Lin H, Jaffe JL, Coch RW, Haldeman JM, MacDonald PE, Merrins MJ, D’Alessio DA and Campbell JE (2019). “beta Cell tone is defined by proglucagon peptides through cAMP signaling.” JCI Insight 4(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Chambers AP, Sorrell JE, Haller A, Roelofs K, Hutch CR, Kim KS, Gutierrez-Aguilar R, Li B, Drucker DJ, D’Alessio DA, Seeley RJ and Sandoval DA (2017). “The Role of Pancreatic Preproglucagon in Glucose Homeostasis in Mice.” Cell Metab 25(4): 927–934 e923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Chen C, Li H and Long YQ (2016). “GPR40 agonists for the treatment of type 2 diabetes mellitus: The biological characteristics and the chemical space.” Bioorg Med Chem Lett 26(23): 5603–5612. [DOI] [PubMed] [Google Scholar]
  32. Chepurny OG, Bonaccorso RL, Leech CA, Wöllert T, Langford GM, Schwede F, Roth CL, Doyle RP and Holz GG (2018). “Chimeric peptide EP45 as a dual agonist at GLP-1 and NPY2R receptors.” Scientific Reports 8(1): 3749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Chisholm C and Greenberg GR (2002). “Somatostatin-28 regulates GLP-1 secretion via somatostatin receptor subtype 5 in rat intestinal cultures.” Am J Physiol Endocrinol Metab 283(2): E311–317. [DOI] [PubMed] [Google Scholar]
  34. Christensen MB, Calanna S, Holst JJ, Vilsboll T and Knop FK (2014). “Glucose-dependent insulinotropic polypeptide: blood glucose stabilizing effects in patients with type 2 diabetes.” J Clin Endocrinol Metab 99(3): E418–426. [DOI] [PubMed] [Google Scholar]
  35. Christiansen CB, Gabe MBN, Svendsen B, Dragsted LO, Rosenkilde MM and Holst JJ (2018). “The impact of short-chain fatty acids on GLP-1 and PYY secretion from the isolated perfused rat colon.” Am J Physiol Gastrointest Liver Physiol 315(1): G53–G65. [DOI] [PubMed] [Google Scholar]
  36. Christou GA, Katsiki N, Blundell J, Fruhbeck G and Kiortsis DN (2019). “Semaglutide as a promising antiobesity drug.” Obes Rev. [DOI] [PubMed] [Google Scholar]
  37. Chukir T, Shukla AP, Saunders KH and Aronne LJ (2018). “Pharmacotherapy for obesity in individuals with type 2 diabetes.” Expert Opin Pharmacother 19(3): 223–231. [DOI] [PubMed] [Google Scholar]
  38. Cohen MA, Ellis SM, Le Roux CW, Batterham RL, Park A, Patterson M, Frost GS, Ghatei MA and Bloom SR (2003). “Oxyntomodulin Suppresses Appetite and Reduces Food Intake in Humans.” The Journal of Clinical Endocrinology & Metabolism 88(10): 4696–4701. [DOI] [PubMed] [Google Scholar]
  39. Connolly JJ, Ooka K and Lim JK (2018). “Future Pharmacotherapy for Non-alcoholic Steatohepatitis (NASH): Review of Phase 2 and 3 Trials.” J Clin Transl Hepatol 6(3): 264–275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Coskun T, Sloop KW, Loghin C, Alsina-Fernandez J, Urva S, Bokvist KB, Cui X, Briere DA, Cabrera O, Roell WC, Kuchibhotla U, Moyers JS, Benson CT, Gimeno RE, D’Alessio DA and Haupt A (2018). “LY3298176, a novel dual GIP and GLP-1 receptor agonist for the treatment of type 2 diabetes mellitus: From discovery to clinical proof of concept.” Mol Metab 18: 3–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Cox JM, Berna MJ, Jin Z, Cox AL, Sloop KW, Gutierrez JA and Ackermann BL (2016). “Characterization and quantification of oxyntomodulin in human and rat plasma using high-resolution accurate mass LC-MS.” Bioanalysis 8(15): 1579–1595. [DOI] [PubMed] [Google Scholar]
  42. Cox JM, Chu HD, Chelliah MV, Debenham JS, Eagen K, Lan P, Lombardo M, London C, Plotkin MA, Shah U, Sun Z, Vaccaro HM, Venkatraman S, Suzuki T, Wang N, Ashley ER, Crespo A, Madeira M, Leung DH, Alleyne C, Ogawa AM, Souza S, Thomas-Fowlkes B, Di Salvo J, Weinglass A, Kirkland M, Pachanski M, Powles MA, Tozzo E, Akiyama TE, Ujjainwalla F, Tata JR and Sinz CJ (2017). “Design, Synthesis, and Evaluation of Novel and Selective G-protein Coupled Receptor 120 (GPR120) Spirocyclic Agonists.” ACS Med Chem Lett 8(1): 49–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. D’Alessio DA and Ensinck JW (1990). “Fasting and Postprandial Concentrations of Somatostatin-28 and Somatostatin-14 in Type II Diabetes in Men.” Diabetes 39(10): 1198–1202. [DOI] [PubMed] [Google Scholar]
  44. Dar MS, Chapman WH 3rd, Pender JR, Drake AJ 3rd, O’Brien K, Tanenberg RJ, Dohm GL and Pories WJ (2012). “GLP-1 response to a mixed meal: what happens 10 years after Roux-en-Y gastric bypass (RYGB)?” Obes Surg 22(7): 1077–1083. [DOI] [PubMed] [Google Scholar]
  45. Davies MJ, Bergenstal R, Bode B, Kushner RF, Lewin A, Skjoth TV, Andreasen AH, Jensen CB, DeFronzo RA and Group NNS (2015). “Efficacy of Liraglutide for Weight Loss Among Patients With Type 2 Diabetes: The SCALE Diabetes Randomized Clinical Trial.” JAMA 314(7): 687–699. [DOI] [PubMed] [Google Scholar]
  46. Day JW, Ottaway N, Patterson JT, Gelfanov V, Smiley D, Gidda J, Findeisen H, Bruemmer D, Drucker DJ, Chaudhary N, Holland J, Hembree J, Abplanalp W, Grant E, Ruehl J, Wilson H, Kirchner H, Lockie SH, Hofmann S, Woods SC, Nogueiras R, Pfluger PT, Perez-Tilve D, DiMarchi R and Tschop MH (2009). “A new glucagon and GLP-1 co-agonist eliminates obesity in rodents.” Nat Chem Biol 5(10): 749–757. [DOI] [PubMed] [Google Scholar]
  47. De Silva A and Bloom SR (2012). “Gut Hormones and Appetite Control: A Focus on PYY and GLP-1 as Therapeutic Targets in Obesity.” Gut Liver 6(1): 10–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. De Silva A, Salem V, Long CJ, Makwana A, Newbould RD, Rabiner EA, Ghatei MA, Bloom SR, Matthews PM, Beaver JD and Dhillo WS (2011). “The gut hormones PYY 3–36 and GLP-1 7–36 amide reduce food intake and modulate brain activity in appetite centers in humans.” Cell Metab 14(5): 700–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Deacon CF and Lebovitz HE (2016). “Comparative review of dipeptidyl peptidase-4 inhibitors and sulphonylureas.” Diabetes Obes Metab 18(4): 333–347. [DOI] [PubMed] [Google Scholar]
  50. Demssie YN, Jawaheer J, Farook S, New JP and Syed AA (2012). “Metabolic outcomes 1 year after gastric bypass surgery in obese people with type 2 diabetes.” Med Princ Pract 21(2): 125–128. [DOI] [PubMed] [Google Scholar]
  51. Ding L, Yang L, Wang Z and Huang W (2015). “Bile acid nuclear receptor FXR and digestive system diseases.” Acta Pharm Sin B 5(2): 135–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Douros JD, Lewis AG, Smith EP, Niu J, Capozzi M, Wittmann A, Campbell J, Tong J, Wagner C, Mahbod P, Seeley R and D’Alessio DA (2018). “Enhanced Glucose Control Following Vertical Sleeve Gastrectomy Does Not Require a beta-Cell Glucagon-Like Peptide 1 Receptor.” Diabetes 67(8): 1504–1511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Drucker DJ, Habener JF and Holst JJ (2017). “Discovery, characterization, and clinical development of the glucagon-like peptides.” J Clin Invest 127(12): 4217–4227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Dumont Y, Fournier A, St-Pierre S and Quirion R (1995). Characterization of neuropeptide Y binding sites in rat brain membrane preparations using [125I][Leu31,Pro34]peptide YY and [125I]peptide YY3–36 as selective Y1 and Y2 radioligands. [PubMed]
  55. Ellingsgaard H, Hauselmann I, Schuler B, Habib AM, Baggio LL, Meier DT, Eppler E, Bouzakri K, Wueest S, Muller YD, Hansen AM, Reinecke M, Konrad D, Gassmann M, Reimann F, Halban PA, Gromada J, Drucker DJ, Gribble FM, Ehses JA and Donath MY (2011). “Interleukin-6 enhances insulin secretion by increasing glucagon-like peptide-1 secretion from L cells and alpha cells.” Nat Med 17(11): 1481–1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Falkén Y, Hellström PM, Holst JJ and Näslund E (2011). “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.” The Journal of Clinical Endocrinology & Metabolism 96(7): 2227–2235. [DOI] [PubMed] [Google Scholar]
  57. Farb TB, Adeva M, Beauchamp TJ, Cabrera O, Coates DA, Meredith TD, Droz BA, Efanov A, Ficorilli JV, Gackenheimer SL, Martinez-Grau MA, Molero V, Ruano G, Statnick MA, Suter TM, Syed SK, Toledo MA, Willard FS, Zhou X, Bokvist KB and Barrett DG (2017). “Regulation of Endogenous (Male) Rodent GLP-1 Secretion and Human Islet Insulin Secretion by Antagonism of Somatostatin Receptor 5.” Endocrinology 158(11): 3859–3873. [DOI] [PubMed] [Google Scholar]
  58. Finan B, Ma T, Ottaway N, Muller TD, Habegger KM, Heppner KM, Kirchner H, Holland J, Hembree J, Raver C, Lockie SH, Smiley DL, Gelfanov V, Yang B, Hofmann S, Bruemmer D, Drucker DJ, Pfluger PT, Perez-Tilve D, Gidda J, Vignati L, Zhang L, Hauptman JB, Lau M, Brecheisen M, Uhles S, Riboulet W, Hainaut E, Sebokova E, Conde-Knape K, Konkar A, DiMarchi RD and Tschop MH (2013). “Unimolecular dual incretins maximize metabolic benefits in rodents, monkeys, and humans.” Sci Transl Med 5(209): 209ra151. [DOI] [PubMed] [Google Scholar]
  59. Finan B, Muller TD, Clemmensen C, Perez-Tilve D, DiMarchi RD and Tschop MH (2016). “Reappraisal of GIP Pharmacology for Metabolic Diseases.” Trends Mol Med 22(5): 359–376. [DOI] [PubMed] [Google Scholar]
  60. Finan B, Yang B, Ottaway N, Smiley DL, Ma T, Clemmensen C, Chabenne J, Zhang L, Habegger KM, Fischer K, Campbell JE, Sandoval D, Seeley RJ, Bleicher K, Uhles S, Riboulet W, Funk J, Hertel C, Belli S, Sebokova E, Conde-Knape K, Konkar A, Drucker DJ, Gelfanov V, Pfluger PT, Muller TD, Perez-Tilve D, DiMarchi RD and Tschop MH (2015). “A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents.” Nat Med 21(1): 27–36. [DOI] [PubMed] [Google Scholar]
  61. Fjellstrom O, Larsson N, Yasuda S, Tsuchida T, Oguma T, Marley A, Wennberg-Huldt C, Hovdal D, Fukuda H, Yoneyama Y, Sasaki K, Johansson A, Lundqvist S, Brengdahl J, Isaacs RJ, Brown D, Geschwindner S, Benthem L, Priest C and Turnbull A (2015). “Novel Zn2+ Modulated GPR39 Receptor Agonists Do Not Drive Acute Insulin Secretion in Rodents.” PLoS One 10(12): e0145849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Frias JP, Bastyr EJ 3rd, Vignati L, Tschop MH, Schmitt C, Owen K, Christensen RH and DiMarchi RD (2017). “The Sustained Effects of a Dual GIP/GLP-1 Receptor Agonist, NNC0090–2746, in Patients with Type 2 Diabetes.” Cell Metab 26(2): 343–352 e342. [DOI] [PubMed] [Google Scholar]
  63. Frias JP, Nauck MA, Van J, Kutner ME, Cui X, Benson C, Urva S, Gimeno RE, Milicevic Z, Robins D and Haupt A (2018). “Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a randomised, placebo-controlled and active comparator-controlled phase 2 trial.” Lancet 392(10160): 2180–2193. [DOI] [PubMed] [Google Scholar]
  64. Frimurer TM, Mende F, Graae AS, Engelstoft MS, Egerod KL, Nygaard R, Gerlach LO, Hansen JB, Schwartz TW and Holst B (2017). “Model-Based Discovery of Synthetic Agonists for the Zn(2+)-Sensing G-Protein-Coupled Receptor 39 (GPR39) Reveals Novel Biological Functions.” J Med Chem 60(3): 886–898. [DOI] [PubMed] [Google Scholar]
  65. Garcia-Perez LE, Alvarez M, Dilla T, Gil-Guillen V and Orozco-Beltran D (2013). “Adherence to therapies in patients with type 2 diabetes.” Diabetes Ther 4(2): 175–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Garibay D, Lou J, Lee SA, Zaborska KE, Weissman MH, Sloma E, Donahue L, Miller AD, White AC, Michael MD, Sloop KW and Cummings BP (2018). “beta Cell GLP-1R Signaling Alters alpha Cell Proglucagon Processing after Vertical Sleeve Gastrectomy in Mice.” Cell Rep 23(4): 967–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Gasbjerg LS, Helsted MM, Hartmann B, Jensen MH, Gabe MBN, Sparre-Ulrich AH, Veedfald S, Stensen S, Lanng AR, Bergmann NC, Christensen MB, Vilsboll T, Holst JJ, Rosenkilde MM and Knop FK (2019). “Separate and Combined Glucometabolic Effects of Endogenous Glucose-Dependent Insulinotropic Polypeptide and Glucagon-like Peptide 1 in Healthy Individuals.” Diabetes 68(5): 906–917. [DOI] [PubMed] [Google Scholar]
  68. Gasoyan H, Tajeu G, Halpern MT and Sarwer DB (2019). “Reasons for underutilization of bariatric surgery: The role of insurance benefit design.” Surg Obes Relat Dis 15(1): 146–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Geary N, Kissileff HR, Pi-Sunyer FX and Hinton V (1992). “Individual, but not simultaneous, glucagon and cholecystokinin infusions inhibit feeding in men.” Am J Physiol 262(6 Pt 2): R975–980. [DOI] [PubMed] [Google Scholar]
  70. Geloneze B, Tambascia MA, Pilla VF, Geloneze SR, Repetto EM and Pareja JC (2003). “Ghrelin: a gut-brain hormone: effect of gastric bypass surgery.” Obes Surg 13(1): 17–22. [DOI] [PubMed] [Google Scholar]
  71. Gerstein HC, Colhoun HM, Dagenais GR, Diaz R, Lakshmanan M, Pais P, Probstfield J, Botros FT, Riddle MC, Ryden L, Xavier D, Atisso CM, Dyal L, Hall S, Rao-Melacini P, Wong G, Avezum A, Basile J, Chung N, Conget I, Cushman WC, Franek E, Hancu N, Hanefeld M, Holt S, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Cardona Munoz EG, Pirags V, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T and Investigators R (2019). “Dulaglutide and renal outcomes in type 2 diabetes: an exploratory analysis of the REWIND randomised, placebo-controlled trial.” Lancet 394(10193): 131–138. [DOI] [PubMed] [Google Scholar]
  72. Gerstein HC, Colhoun HM, Dagenais GR, Diaz R, Lakshmanan M, Pais P, Probstfield J, Riesmeyer JS, Riddle MC, Ryden L, Xavier D, Atisso CM, Dyal L, Hall S, Rao-Melacini P, Wong G, Avezum A, Basile J, Chung N, Conget I, Cushman WC, Franek E, Hancu N, Hanefeld M, Holt S, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Cardona Munoz EG, Pirags V, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T and Investigators R (2019). “Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial.” Lancet 394(10193): 121–130. [DOI] [PubMed] [Google Scholar]
  73. Gletsu-Miller N and Wright BN (2013). “Mineral malnutrition following bariatric surgery.” Adv Nutr 4(5): 506–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Goldstone AP, Miras AD, Scholtz S, Jackson S, Neff KJ, Penicaud L, Geoghegan J, Chhina N, Durighel G, Bell JD, Meillon S and le Roux CW (2016). “Link Between Increased Satiety Gut Hormones and Reduced Food Reward After Gastric Bypass Surgery for Obesity.” J Clin Endocrinol Metab 101(2): 599–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Gribble FM and Reimann F (2019). “Function and mechanisms of enteroendocrine cells and gut hormones in metabolism.” Nat Rev Endocrinol 15(4): 226–237. [DOI] [PubMed] [Google Scholar]
  76. Guida C, McCulloch LJ, Godazgar M, Stephen SD, Baker C, Basco D, Dong J, Chen D, Clark A and Ramracheya RD (2018). “Sitagliptin and Roux-en-Y gastric bypass modulate insulin secretion via regulation of intra-islet PYY.” Diabetes Obes Metab 20(3): 571–581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Gunther T, Tulipano G, Dournaud P, Bousquet C, Csaba Z, Kreienkamp HJ, Lupp A, Korbonits M, Castano JP, Wester HJ, Culler M, Melmed S and Schulz S (2018). “International Union of Basic and Clinical Pharmacology. CV. Somatostatin Receptors: Structure, Function, Ligands, and New Nomenclature.” Pharmacol Rev 70(4): 763–835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Guo L, Parker DL, Zang Y, Sweis RF, Liu W, Sherer EC, Buist N, Terebetski J, Kelly T, Bugianesi R, Priest BT, Dingley KH, Li X, Mitelman S, Salituro G, Trujillo ME, Pachanski M, Kirkland M, Powles MA, Eiermann GJ, Feng Y, Shang J, Howard AD, Ujjainwalla F, Sinz CJ, Debenham JS, Edmondson SD, Nargund RP, Hagmann WK and Li D (2016). “Discovery and Optimization of a Novel Triazole Series of GPR142 Agonists for the Treatment of Type 2 Diabetes.” ACS Med Chem Lett 7(12): 1107–1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Habegger KM, Stemmer K, Cheng C, Muller TD, Heppner KM, Ottaway N, Holland J, Hembree JL, Smiley D, Gelfanov V, Krishna R, Arafat AM, Konkar A, Belli S, Kapps M, Woods SC, Hofmann SM, D’Alessio D, Pfluger PT, Perez-Tilve D, Seeley RJ, Konishi M, Itoh N, Kharitonenkov A, Spranger J, DiMarchi RD and Tschop MH (2013). “Fibroblast growth factor 21 mediates specific glucagon actions.” Diabetes 62(5): 1453–1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Habib AM, Richards P, Rogers GJ, Reimann F and Gribble FM (2013). “Co-localisation and secretion of glucagon-like peptide 1 and peptide YY from primary cultured human L cells.” Diabetologia 56(6): 1413–1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Hamdouchi C, Kahl SD, Patel Lewis A, Cardona GR, Zink RW, Chen K, Eessalu TE, Ficorilli JV, Marcelo MC, Otto KA, Wilbur KL, Lineswala JP, Piper JL, Coffey DS, Sweetana SA, Haas JV, Brooks DA, Pratt EJ, Belin RM, Deeg MA, Ma X, Cannady EA, Johnson JT, Yumibe NP, Chen Q, Maiti P, Montrose-Rafizadeh C, Chen Y and Reifel Miller A (2016). “The Discovery, Preclinical, and Early Clinical Development of Potent and Selective GPR40 Agonists for the Treatment of Type 2 Diabetes Mellitus (LY2881835, LY2922083, and LY2922470).” J Med Chem 59(24): 10891–10916. [DOI] [PubMed] [Google Scholar]
  82. Hankir MK, Seyfried F, Hintschich CA, Diep TA, Kleberg K, Kranz M, Deuther-Conrad W, Tellez LA, Rullmann M, Patt M, Teichert J, Hesse S, Sabri O, Brust P, Hansen HS, de Araujo IE, Krugel U and Fenske WK (2017). “Gastric Bypass Surgery Recruits a Gut PPAR-alpha-Striatal D1R Pathway to Reduce Fat Appetite in Obese Rats.” Cell Metab 25(2): 335–344. [DOI] [PubMed] [Google Scholar]
  83. Hansen L, Hartmann B, Bisgaard T, Mineo H, Jorgensen PN and Holst JJ (2000). “Somatostatin restrains the secretion of glucagon-like peptide-1 and −2 from isolated perfused porcine ileum.” Am J Physiol Endocrinol Metab 278(6): E1010–1018. [DOI] [PubMed] [Google Scholar]
  84. Hauge-Evans AC, King AJ, Carmignac D, Richardson CC, Robinson IC, Low MJ, Christie MR, Persaud SJ and Jones PM (2009). “Somatostatin secreted by islet delta-cells fulfills multiple roles as a paracrine regulator of islet function.” Diabetes 58(2): 403–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Hedrington MS and Davis SN (2019). “Oral semaglutide for the treatment of type 2 diabetes.” Expert Opin Pharmacother 20(2): 133–141. [DOI] [PubMed] [Google Scholar]
  86. Henderson SJ, Konkar A, Hornigold DC, Trevaskis JL, Jackson R, Fritsch Fredin M, Jansson-Lofmark R, Naylor J, Rossi A, Bednarek MA, Bhagroo N, Salari H, Will S, Oldham S, Hansen G, Feigh M, Klein T, Grimsby J, Maguire S, Jermutus L, Rondinone CM and Coghlan MP (2016). “Robust anti-obesity and metabolic effects of a dual GLP-1/glucagon receptor peptide agonist in rodents and non-human primates.” Diabetes Obes Metab 18(12): 1176–1190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Henry RR, Ciaraldi TP, Armstrong D, Burke P, Ligueros-Saylan M and Mudaliar S (2013). “Hyperglycemia associated with pasireotide: results from a mechanistic study in healthy volunteers.” J Clin Endocrinol Metab 98(8): 3446–3453. [DOI] [PubMed] [Google Scholar]
  88. Herman GA, Bergman A, Stevens C, Kotey P, Yi B, Zhao P, Dietrich B, Golor G, Schrodter A, Keymeulen B, Lasseter KC, Kipnes MS, Snyder K, Hilliard D, Tanen M, Cilissen C, De Smet M, de Lepeleire I, Van Dyck K, Wang AQ, Zeng W, Davies MJ, Tanaka W, Holst JJ, Deacon CF, Gottesdiener KM and Wagner JA (2006). “Effect of single oral doses of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on incretin and plasma glucose levels after an oral glucose tolerance test in patients with type 2 diabetes.” J Clin Endocrinol Metab 91(11): 4612–4619. [DOI] [PubMed] [Google Scholar]
  89. Hernandez AF, Green JB, Janmohamed S, D’Agostino RB Sr., Granger CB, Jones NP, Leiter LA, Rosenberg AE, Sigmon KN, Somerville MC, Thorpe KM, McMurray JJV, Del Prato S, c. Harmony Outcomes and investigators (2018). “Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (Harmony Outcomes): a double-blind, randomised placebo-controlled trial.” Lancet 392(10157): 1519–1529. [DOI] [PubMed] [Google Scholar]
  90. Hirose H, Yamasaki T, Ogino M, Mizojiri R, Tamura-Okano Y, Yashiro H, Muraki Y, Nakano Y, Sugama J, Hata A, Iwasaki S, Watanabe M, Maekawa T and Kasai S (2017). “Discovery of novel 5-oxa-2,6-diazaspiro[3.4]oct-6-ene derivatives as potent, selective, and orally available somatostatin receptor subtype 5 (SSTR5) antagonists for treatment of type 2 diabetes mellitus.” Bioorg Med Chem 25(15): 4175–4193. [DOI] [PubMed] [Google Scholar]
  91. Hjollund KR, Deacon CF and Holst JJ (2011). “Dipeptidyl peptidase-4 inhibition increases portal concentrations of intact glucagon-like peptide-1 (GLP-1) to a greater extent than peripheral concentrations in anaesthetised pigs.” Diabetologia 54(8): 2206–2208. [DOI] [PubMed] [Google Scholar]
  92. Hojberg PV, Vilsboll T, Rabol R, Knop FK, Bache M, Krarup T, Holst JJ and Madsbad S (2009). “Four weeks of near-normalisation of blood glucose improves the insulin response to glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide in patients with type 2 diabetes.” Diabetologia 52(2): 199–207. [DOI] [PubMed] [Google Scholar]
  93. Holst JJ (2019). “The incretin system in healthy humans: the role of GIP and GLP-1.” Metabolism. [DOI] [PubMed] [Google Scholar]
  94. Holst JJ, Madsbad S, Bojsen-Moller KN, Svane MS, Jorgensen NB, Dirksen C and Martinussen C (2018). “Mechanisms in bariatric surgery: Gut hormones, diabetes resolution, and weight loss.” Surg Obes Relat Dis 14(5): 708–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Holt MK, Richards JE, Cook DR, Brierley DI, Williams DL, Reimann F, Gribble FM and Trapp S (2019). “Preproglucagon Neurons in the Nucleus of the Solitary Tract Are the Main Source of Brain GLP-1, Mediate Stress-Induced Hypophagia, and Limit Unusually Large Intakes of Food.” Diabetes 68(1): 21–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Holzer P, Reichmann F and Farzi A (2012). “Neuropeptide Y, peptide YY and pancreatic polypeptide in the gut-brain axis.” Neuropeptides 46(6): 261–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Hornigold DC, Roth E, Howard V, Will S, Oldham S, Coghlan MP, Blouet C and Trevaskis JL (2018). “A GLP-1:CCK fusion peptide harnesses the synergistic effects on metabolism of CCK-1 and GLP-1 receptor agonism in mice.” Appetite 127: 334–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Iepsen EW, Lundgren J, Holst JJ, Madsbad S and Torekov SS (2016). “Successful weight loss maintenance includes long-term increased meal responses of GLP-1 and PYY3–36.” Eur J Endocrinol 174(6): 775–784. [DOI] [PubMed] [Google Scholar]
  99. Jall S, Sachs S, Clemmensen C, Finan B, Neff F, DiMarchi RD, Tschop MH, Muller TD and Hofmann SM (2017). “Monomeric GLP-1/GIP/glucagon triagonism corrects obesity, hepatosteatosis, and dyslipidemia in female mice.” Mol Metab 6(5): 440–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Jordan J, Greenway FL, Leiter LA, Li Z, Jacobson P, Murphy K, Hill J, Kler L and Aftring RP (2008). “Stimulation of cholecystokinin-A receptors with GI181771X does not cause weight loss in overweight or obese patients.” Clin Pharmacol Ther 83(2): 281–287. [DOI] [PubMed] [Google Scholar]
  101. Kaku K, Enya K, Nakaya R, Ohira T and Matsuno R (2016). “Long-term safety and efficacy of fasiglifam (TAK-875), a G-protein-coupled receptor 40 agonist, as monotherapy and combination therapy in Japanese patients with type 2 diabetes: a 52-week open-label phase III study.” Diabetes Obes Metab 18(9): 925–929. [DOI] [PubMed] [Google Scholar]
  102. Kalinowski P, Paluszkiewicz R, Wroblewski T, Remiszewski P, Grodzicki M, Bartoszewicz Z and Krawczyk M (2017). “Ghrelin, leptin, and glycemic control after sleeve gastrectomy versus Roux-en-Y gastric bypass-results of a randomized clinical trial.” Surg Obes Relat Dis 13(2): 181–188. [DOI] [PubMed] [Google Scholar]
  103. Kassem MA, Durda MA, Stoicea N, Cavus O, Sahin L and Rogers B (2017). “The Impact of Bariatric Surgery on Type 2 Diabetes Mellitus and the Management of Hypoglycemic Events.” Front Endocrinol (Lausanne) 8: 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Katsuma S, Hirasawa A and Tsujimoto G (2005). “Bile acids promote glucagon-like peptide-1 secretion through TGR5 in a murine enteroendocrine cell line STC-1.” Biochem Biophys Res Commun 329(1): 386–390. [DOI] [PubMed] [Google Scholar]
  105. Katz LB, Gambale JJ, Rothenberg PL, Vanapalli SR, Vaccaro N, Xi L, Polidori DC, Vets E, Sarich TC and Stein PP (2011). “Pharmacokinetics, pharmacodynamics, safety, and tolerability of JNJ-38431055, a novel GPR119 receptor agonist and potential antidiabetes agent, in healthy male subjects.” Clin Pharmacol Ther 90(5): 685–692. [DOI] [PubMed] [Google Scholar]
  106. Katz LB, Gambale JJ, Rothenberg PL, Vanapalli SR, Vaccaro N, Xi L, Sarich TC and Stein PP (2012). “Effects of JNJ-38431055, a novel GPR119 receptor agonist, in randomized, double-blind, placebo-controlled studies in subjects with type 2 diabetes.” Diabetes Obes Metab 14(8): 709–716. [DOI] [PubMed] [Google Scholar]
  107. Keitel V, Cupisti K, Ullmer C, Knoefel WT, Kubitz R and Haussinger D (2009). “The membrane-bound bile acid receptor TGR5 is localized in the epithelium of human gallbladders.” Hepatology 50(3): 861–870. [DOI] [PubMed] [Google Scholar]
  108. Kieffer TJ, McIntosh CH and Pederson RA (1995). “Degradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IV.” Endocrinology 136(8): 3585–3596. [DOI] [PubMed] [Google Scholar]
  109. Killion EA, Wang J, Yie J, Shi SD, Bates D, Min X, Komorowski R, Hager T, Deng L, Atangan L, Lu SC, Kurzeja RJM, Sivits G, Lin J, Chen Q, Wang Z, Thibault SA, Abbott CM, Meng T, Clavette B, Murawsky CM, Foltz IN, Rottman JB, Hale C, Veniant MM and Lloyd DJ (2018). “Anti-obesity effects of GIPR antagonists alone and in combination with GLP-1R agonists in preclinical models.” Sci Transl Med 10(472). [DOI] [PubMed] [Google Scholar]
  110. Kim T, Nason S, Holleman C, Pepin M, Wilson L, Berryhill TF, Wende AR, Steele C, Young ME, Barnes S, Drucker DJ, Finan B, DiMarchi R, Perez-Tilve D, Tschop M and Habegger KM (2018). “Glucagon Receptor Signaling Regulates Energy Metabolism via Hepatic Farnesoid X Receptor and Fibroblast Growth Factor 21.” Diabetes 67(9): 1773–1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Kjaergaard M, Salinas CBG, Rehfeld JF, Secher A, Raun K and Wulff BS (2019). “PYY(3–36) and exendin-4 reduce food intake and activate neuronal circuits in a synergistic manner in mice.” Neuropeptides 73: 89–95. [DOI] [PubMed] [Google Scholar]
  112. Kliewer SA and Mangelsdorf DJ (2019). “A Dozen Years of Discovery: Insights into the Physiology and Pharmacology of FGF21.” Cell Metab 29(2): 246–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Knop FK, Bronden A and Vilsboll T (2017). “Exenatide: pharmacokinetics, clinical use, and future directions.” Expert Opin Pharmacother 18(6): 555–571. [DOI] [PubMed] [Google Scholar]
  114. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H and Kangawa K (1999). “Ghrelin is a growth-hormone-releasing acylated peptide from stomach.” Nature 402(6762): 656–660. [DOI] [PubMed] [Google Scholar]
  115. Korner J, Inabnet W, Febres G, Conwell IM, McMahon DJ, Salas R, Taveras C, Schrope B and Bessler M (2009). “Prospective study of gut hormone and metabolic changes after adjustable gastric banding and Roux-en-Y gastric bypass.” Int J Obes (Lond) 33(7): 786–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Kosinski JR, Hubert J, Carrington PE, Chicchi GG, Mu J, Miller C, Cao J, Bianchi E, Pessi A, Sinharoy R, Marsh DJ and Pocai A (2012). “The glucagon receptor is involved in mediating the body weight-lowering effects of oxyntomodulin.” Obesity (Silver Spring) 20(8): 1566–1571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Kothare PA, Linnebjerg H, Isaka Y, Uenaka K, Yamamura A, Yeo KP, de la Pena A, Teng CH, Mace K, Fineman M, Shigeta H, Sakata Y and Irie S (2008). “Pharmacokinetics, pharmacodynamics, tolerability, and safety of exenatide in Japanese patients with type 2 diabetes mellitus.” J Clin Pharmacol 48(12): 1389–1399. [DOI] [PubMed] [Google Scholar]
  118. Krug AW, Vaddady P, Railkar RA, Musser BJ, Cote J, Ederveen A, Krefetz DG, DeNoia E, Free AL, Morrow L, Chakravarthy MV, Kauh E, Tatosian DA and Kothare PA (2017). “Leveraging a Clinical Phase Ib Proof-of-Concept Study for the GPR40 Agonist MK-8666 in Patients With Type 2 Diabetes for Model-Informed Phase II Dose Selection.” Clin Transl Sci 10(5): 404–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. le Roux CW, Welbourn R, Werling M, Osborne A, Kokkinos A, Laurenius A, Lonroth H, Fandriks L, Ghatei MA, Bloom SR and Olbers T (2007). “Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass.” Ann Surg 246(5): 780–785. [DOI] [PubMed] [Google Scholar]
  120. Lee AY, Chappell DL, Bak MJ, Judo M, Liang L, Churakova T, Ayanoglu G, Castro-Perez J, Zhou H, Previs S, Souza SC, Lassman ME and Laterza OF (2016). “Multiplexed Quantification of Proglucagon-Derived Peptides by Immunoaffinity Enrichment and Tandem Mass Spectrometry after a Meal Tolerance Test.” Clin Chem 62(1): 227–235. [DOI] [PubMed] [Google Scholar]
  121. Li T, Holmstrom SR, Kir S, Umetani M, Schmidt DR, Kliewer SA and Mangelsdorf DJ (2011). “The G protein-coupled bile acid receptor, TGR5, stimulates gallbladder filling.” Mol Endocrinol 25(6): 1066–1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Li Y, Schnabl K, Gabler SM, Willershauser M, Reber J, Karlas A, Laurila S, Lahesmaa M, M UD, Bast-Habersbrunner A, Virtanen KA, Fromme T, Bolze F, O’Farrell LS, Alsina-Fernandez J, Coskun T, Ntziachristos V, Nuutila P and Klingenspor M (2018). “Secretin-Activated Brown Fat Mediates Prandial Thermogenesis to Induce Satiation.” Cell 175(6): 1561–1574 e1512. [DOI] [PubMed] [Google Scholar]
  123. Li Z, Qiu Q, Geng X, Yang J, Huang W and Qian H (2016). “Free fatty acid receptor agonists for the treatment of type 2 diabetes: drugs in preclinical to phase II clinical development.” Expert Opin Investig Drugs 25(8): 871–890. [DOI] [PubMed] [Google Scholar]
  124. Lim CH, Lee PC, Lim E, Tan J, Chan WH, Tan HC, Ganguly S, Tham KW and Eng A (2019). “Correlation Between Symptomatic Gastro-Esophageal Reflux Disease (GERD) and Erosive Esophagitis (EE) Post-vertical Sleeve Gastrectomy (VSG).” Obes Surg 29(1): 207–214. [DOI] [PubMed] [Google Scholar]
  125. Lin DC, Guo Q, Luo J, Zhang J, Nguyen K, Chen M, Tran T, Dransfield PJ, Brown SP, Houze J, Vimolratana M, Jiao XY, Wang Y, Birdsall NJ and Swaminath G (2012). “Identification and pharmacological characterization of multiple allosteric binding sites on the free fatty acid 1 receptor.” Mol Pharmacol 82(5): 843–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Lin E, Gletsu N, Fugate K, McClusky D, Gu LH, Zhu JL, Ramshaw BJ, Papanicolaou DA, Ziegler TR and Smith CD (2004). “The effects of gastric surgery on systemic ghrelin levels in the morbidly obese.” Arch Surg 139(7): 780–784. [DOI] [PubMed] [Google Scholar]
  127. Lin HV, Efanov AM, Fang X, Beavers LS, Wang X, Wang J, Gonzalez Valcarcel IC and Ma T (2016). “GPR142 Controls Tryptophan-Induced Insulin and Incretin Hormone Secretion to Improve Glucose Metabolism.” PLoS One 11(6): e0157298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Lin HV, Wang J, Wang J, Li W, Wang X, Alston JT, Thomas MK, Briere DA, Syed SK and Efanov AM (2018). “GPR142 prompts glucagon-like Peptide-1 release from islets to improve beta cell function.” Mol Metab 11: 205–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Liou AP, Paziuk M, Luevano JM Jr., Machineni S, Turnbaugh PJ and Kaplan LM (2013). “Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity.” Sci Transl Med 5(178): 178ra141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Liu W, Shao PP, Liang GB, Bawiec J, He J, Aster SD, Wu M, Chicchi G, Wang J, Tsao KL, Shang J, Salituro G, Zhou YP, Li C, Akiyama TE, Metzger DE, Murphy BA, Howard AD, Weber AE and Duffy JL (2018). “Discovery and Pharmacology of a Novel Somatostatin Subtype 5 (SSTR5) Antagonist: Synergy with DPP-4 Inhibition.” ACS Med Chem Lett 9(11): 1082–1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Look ARG, Wing RR, Bolin P, Brancati FL, Bray GA, Clark JM, Coday M, Crow RS, Curtis JM, Egan CM, Espeland MA, Evans M, Foreyt JP, Ghazarian S, Gregg EW, Harrison B, Hazuda HP, Hill JO, Horton ES, Hubbard VS, Jakicic JM, Jeffery RW, Johnson KC, Kahn SE, Kitabchi AE, Knowler WC, Lewis CE, Maschak-Carey BJ, Montez MG, Murillo A, Nathan DM, Patricio J, Peters A, Pi-Sunyer X, Pownall H, Reboussin D, Regensteiner JG, Rickman AD, Ryan DH, Safford M, Wadden TA, Wagenknecht LE, West DS, Williamson DF and Yanovski SZ (2013). “Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes.” N Engl J Med 369(2): 145–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Lu J, Byrne N, Wang J, Bricogne G, Brown FK, Chobanian HR, Colletti SL, Di Salvo J, Thomas-Fowlkes B, Guo Y, Hall DL, Hadix J, Hastings NB, Hermes JD, Ho T, Howard AD, Josien H, Kornienko M, Lumb KJ, Miller MW, Patel SB, Pio B, Plummer CW, Sherborne BS, Sheth P, Souza S, Tummala S, Vonrhein C, Webb M, Allen SJ, Johnston JM, Weinglass AB, Sharma S and Soisson SM (2017). “Structural basis for the cooperative allosteric activation of the free fatty acid receptor GPR40.” Nat Struct Mol Biol 24(7): 570–577. [DOI] [PubMed] [Google Scholar]
  133. Lund A, Bagger JI, Wewer Albrechtsen NJ, Christensen M, Grondahl M, Hartmann B, Mathiesen ER, Hansen CP, Storkholm JH, van Hall G, Rehfeld JF, Hornburg D, Meissner F, Mann M, Larsen S, Holst JJ, Vilsboll T and Knop FK (2016). “Evidence of Extrapancreatic Glucagon Secretion in Man.” Diabetes 65(3): 585–597. [DOI] [PubMed] [Google Scholar]
  134. Lundberg JM, Tatemoto K, Terenius L, Hellstrom PM, Mutt V, Hokfelt T and Hamberger B (1982). “Localization of peptide YY (PYY) in gastrointestinal endocrine cells and effects on intestinal blood flow and motility.” Proc Natl Acad Sci U S A 79(14): 4471–4475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Luo J, Swaminath G, Brown SP, Zhang J, Guo Q, Chen M, Nguyen K, Tran T, Miao L, Dransfield PJ, Vimolratana M, Houze JB, Wong S, Toteva M, Shan B, Li F, Zhuang R and Lin DC (2012). “A potent class of GPR40 full agonists engages the enteroinsular axis to promote glucose control in rodents.” PLoS One 7(10): e46300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Luo Y, Lu K, Liu G, Wang J, Laurent I and Zhou X (2018). “The Effects of Novel Antidiabetic Drugs on Albuminuria in Type 2 Diabetes Mellitus: A Systematic Review and Meta-analysis of Randomized Controlled Trials.” Clin Drug Investig 38(12): 1089–1108. [DOI] [PubMed] [Google Scholar]
  137. Malhi H and Camilleri M (2017). “Modulating bile acid pathways and TGR5 receptors for treating liver and GI diseases.” Curr Opin Pharmacol 37: 80–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Mann JFE, Orsted DD, Brown-Frandsen K, Marso SP, Poulter NR, Rasmussen S, Tornoe K, Zinman B, Buse JB, L. S. Committee and Investigators (2017). “Liraglutide and Renal Outcomes in Type 2 Diabetes.” N Engl J Med 377(9): 839–848. [DOI] [PubMed] [Google Scholar]
  139. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsboll T and S.−. Investigators (2016). “Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes.” N Engl J Med 375(19): 1834–1844. [DOI] [PubMed] [Google Scholar]
  140. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner M, Zinman B, Bergenstal RM, Buse JB, Committee LS and Investigators LT (2016). “Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes.” N Engl J Med 375(4): 311–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Martin PA and Faulkner A (1996). “Effects of somatostatin-28 on circulating concentrations of insulin and gut hormones in sheep.” J Endocrinol 151(1): 107–112. [DOI] [PubMed] [Google Scholar]
  142. Matsumoto K, Yoshitomi T, Ishimoto Y, Tanaka N, Takahashi K, Watanabe A and Chiba K (2018). “DS-8500a, an Orally Available G Protein-Coupled Receptor 119 Agonist, Upregulates Glucagon-Like Peptide-1 and Enhances Glucose-Dependent Insulin Secretion and Improves Glucose Homeostasis in Type 2 Diabetic Rats.” J Pharmacol Exp Ther 367(3): 509–517. [DOI] [PubMed] [Google Scholar]
  143. McGavigan AK, Garibay D, Henseler ZM, Chen J, Bettaieb A, Haj FG, Ley RE, Chouinard ML and Cummings BP (2017). “TGR5 contributes to glucoregulatory improvements after vertical sleeve gastrectomy in mice.” Gut 66(2): 226–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. McGuire DK, Alexander JH, Johansen OE, Perkovic V, Rosenstock J, Cooper ME, Wanner C, Kahn SE, Toto RD, Zinman B, Baanstra D, Pfarr E, Schnaidt S, Meinicke T, George JT, von Eynatten M, Marx N and Investigators C (2019). “Linagliptin Effects on Heart Failure and Related Outcomes in Individuals With Type 2 Diabetes Mellitus at High Cardiovascular and Renal Risk in CARMELINA.” Circulation 139(3): 351–361. [DOI] [PubMed] [Google Scholar]
  145. Medeiros MD and Turner AJ (1994). “Processing and metabolism of peptide-YY: pivotal roles of dipeptidylpeptidase-IV, aminopeptidase-P, and endopeptidase-24.11.” Endocrinology 134(5): 2088–2094. [DOI] [PubMed] [Google Scholar]
  146. Meegalla SK, Huang H, Martin T, Xu J, Zhao S, Liu J, Hall M, Gunnet J, Wang Y, Rady B, Silva J, Otieno M, Arnoult E, Paul Lee S, Pocai A and Player MR (2018). “Discovery of a novel potent GPR40 full agonist.” Bioorg Med Chem Lett 28(4): 720–726. [DOI] [PubMed] [Google Scholar]
  147. Meier JJ, Gallwitz B, Siepmann N, Holst JJ, Deacon CF, Schmidt WE and Nauck MA (2003). “Gastric inhibitory polypeptide (GIP) dose-dependently stimulates glucagon secretion in healthy human subjects at euglycaemia.” Diabetologia 46(6): 798–801. [DOI] [PubMed] [Google Scholar]
  148. Meneilly GS, Bryer-Ash M and Elahi D (1993). “The effect of glyburide on beta-cell sensitivity to glucose-dependent insulinotropic polypeptide.” Diabetes Care 16(1): 110–114. [DOI] [PubMed] [Google Scholar]
  149. Menon V, Lincoff AM, Nicholls SJ, Jasper S, Wolski K, McGuire DK, Mehta CR, Rosenstock J, Lopez C, Marcinak J, Cao C, Nissen SE and Investigators G (2018). “Fasiglifam-Induced Liver Injury in Patients With Type 2 Diabetes: Results of a Randomized Controlled Cardiovascular Outcomes Safety Trial.” Diabetes Care 41(12): 2603–2609. [DOI] [PubMed] [Google Scholar]
  150. Mentlein R, Dahms P, Grandt D and Kruger R (1993). “Proteolytic processing of neuropeptide Y and peptide YY by dipeptidyl peptidase IV.” Regul Pept 49(2): 133–144. [DOI] [PubMed] [Google Scholar]
  151. Miller LJ and Desai AJ (2016). “Metabolic Actions of the Type 1 Cholecystokinin Receptor: Its Potential as a Therapeutic Target.” Trends Endocrinol Metab 27(9): 609–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Miras AD, Herring R, Vusirikala A, Shojaee-Moradi F, Jackson NC, Chandaria S, Jackson SN, Goldstone AP, Hakim N, Patel AG, Umpleby AM and Le Roux CW (2017). “Measurement of hepatic insulin sensitivity early after the bypass of the proximal small bowel in humans.” Obes Sci Pract 3(1): 95–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Miyawaki K, Yamada Y, Ban N, Ihara Y, Tsukiyama K, Zhou H, Fujimoto S, Oku A, Tsuda K, Toyokuni S, Hiai H, Mizunoya W, Fushiki T, Holst JJ, Makino M, Tashita A, Kobara Y, Tsubamoto Y, Jinnouchi T, Jomori T and Seino Y (2002). “Inhibition of gastric inhibitory polypeptide signaling prevents obesity.” Nat Med 8(7): 738–742. [DOI] [PubMed] [Google Scholar]
  154. Mokadem M, Zechner JF, Margolskee RF, Drucker DJ and Aguirre V (2014). “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 3(2): 191–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Mroz PA, Finan B, Gelfanov V, Yang B, Tschop MH, DiMarchi RD and Perez-Tilve D (2019). “Optimized GIP analogs promote body weight lowering in mice through GIPR agonism not antagonism.” Mol Metab 20: 51–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Mulvihill EE (2018). “Dipeptidyl peptidase inhibitor therapy in type 2 diabetes: Control of the incretin axis and regulation of postprandial glucose and lipid metabolism.” Peptides 100: 158–164. [DOI] [PubMed] [Google Scholar]
  157. Mulvihill EE, Varin EM, Gladanac B, Campbell JE, Ussher JR, Baggio LL, Yusta B, Ayala J, Burmeister MA, Matthews D, Bang KWA, Ayala JE and Drucker DJ (2017). “Cellular Sites and Mechanisms Linking Reduction of Dipeptidyl Peptidase-4 Activity to Control of Incretin Hormone Action and Glucose Homeostasis.” Cell Metab 25(1): 152–165. [DOI] [PubMed] [Google Scholar]
  158. Murphy R, Tsai P, Jullig M, Liu A, Plank L and Booth M (2017). “Differential Changes in Gut Microbiota After Gastric Bypass and Sleeve Gastrectomy Bariatric Surgery Vary According to Diabetes Remission.” Obes Surg 27(4): 917–925. [DOI] [PubMed] [Google Scholar]
  159. Naslund E, Gryback P, Hellstrom PM, Jacobsson H, Holst JJ, Theodorsson E and Backman L (1997). “Gastrointestinal hormones and gastric emptying 20 years after jejunoileal bypass for massive obesity.” Int J Obes Relat Metab Disord 21(5): 387–392. [DOI] [PubMed] [Google Scholar]
  160. Nasteska D, Harada N, Suzuki K, Yamane S, Hamasaki A, Joo E, Iwasaki K, Shibue K, Harada T and Inagaki N (2014). “Chronic reduction of GIP secretion alleviates obesity and insulin resistance under high-fat diet conditions.” Diabetes 63(7): 2332–2343. [DOI] [PubMed] [Google Scholar]
  161. Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R and Creutzfeldt W (1993). “Preserved incretin activity of glucagon-like peptide 1 [7–36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus.” J Clin Invest 91(1): 301–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Nauck MA, Kleine N, Orskov C, Holst JJ, Willms B and Creutzfeldt W (1993). “Normalization of fasting hyperglycaemia by exogenous glucagon-like peptide 1 (7–36 amide) in type 2 (non-insulin-dependent) diabetic patients.” Diabetologia 36(8): 741–744. [DOI] [PubMed] [Google Scholar]
  163. Nauck MA and Meier JJ (2016). “The incretin effect in healthy individuals and those with type 2 diabetes: physiology, pathophysiology, and response to therapeutic interventions.” Lancet Diabetes Endocrinol 4(6): 525–536. [DOI] [PubMed] [Google Scholar]
  164. Nauck MA and Meier JJ (2018). “Incretin hormones: Their role in health and disease.” Diabetes Obes Metab 20 Suppl 1: 5–21. [DOI] [PubMed] [Google Scholar]
  165. Nauck MA, Meier JJ, Cavender MA, Abd El Aziz M and Drucker DJ (2017). “Cardiovascular Actions and Clinical Outcomes With Glucagon-Like Peptide-1 Receptor Agonists and Dipeptidyl Peptidase-4 Inhibitors.” Circulation 136(9): 849–870. [DOI] [PubMed] [Google Scholar]
  166. Neary NM, Small CJ, Druce MR, Park AJ, Ellis SM, Semjonous NM, Dakin CL, Filipsson K, Wang F, Kent AS, Frost GS, Ghatei MA and Bloom SR (2005). “Peptide YY3–36 and glucagon-like peptide-17–36 inhibit food intake additively.” Endocrinology 146(12): 5120–5127. [DOI] [PubMed] [Google Scholar]
  167. Neylon OM, Moran MM, Pellicano A, Nightingale M and O’Connell MA (2013). “Successful subcutaneous glucagon use for persistent hypoglycaemia in congenital hyperinsulinism.” J Pediatr Endocrinol Metab 26(11–12): 1157–1161. [DOI] [PubMed] [Google Scholar]
  168. Norregaard PK, Deryabina MA, Tofteng Shelton P, Fog JU, Daugaard JR, Eriksson PO, Larsen LF and Jessen L (2018). “A novel GIP analogue, ZP4165, enhances glucagon-like peptide-1-induced body weight loss and improves glycaemic control in rodents.” Diabetes Obes Metab 20(1): 60–68. [DOI] [PubMed] [Google Scholar]
  169. Nunez DJ, Bush MA, Collins DA, McMullen SL, Gillmor D, Apseloff G, Atiee G, Corsino L, Morrow L and Feldman PL (2014). “Gut hormone pharmacology of a novel GPR119 agonist (GSK1292263), metformin, and sitagliptin in type 2 diabetes mellitus: results from two randomized studies.” PLoS One 9(4): e92494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. O’Carroll AM, Lolait SJ, Konig M and Mahan LC (1992). “Molecular cloning and expression of a pituitary somatostatin receptor with preferential affinity for somatostatin-28.” Mol Pharmacol 42(6): 939–946. [PubMed] [Google Scholar]
  171. Oh DY, Walenta E, Akiyama TE, Lagakos WS, Lackey D, Pessentheiner AR, Sasik R, Hah N, Chi TJ, Cox JM, Powels MA, Di Salvo J, Sinz C, Watkins SM, Armando AM, Chung H, Evans RM, Quehenberger O, McNelis J, Bogner-Strauss JG and Olefsky JM (2014). “A Gpr120-selective agonist improves insulin resistance and chronic inflammation in obese mice.” Nat Med 20(8): 942–947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Ostergaard S, Kofoed J, Paulsson JF, Madsen KG, Jorgensen R and Wulff BS (2018). “Design of Y2 Receptor Selective and Proteolytically Stable PYY3–36 Analogues.” J Med Chem 61(23): 10519–10530. [DOI] [PubMed] [Google Scholar]
  173. Overton HA, Babbs AJ, Doel SM, Fyfe MC, Gardner LS, Griffin G, Jackson HC, Procter MJ, Rasamison CM, Tang-Christensen M, Widdowson PS, Williams GM and Reynet C (2006). “Deorphanization of a G protein-coupled receptor for oleoylethanolamide and its use in the discovery of small-molecule hypophagic agents.” Cell Metab 3(3): 167–175. [DOI] [PubMed] [Google Scholar]
  174. Pachanski MJ, Kirkland ME, Kosinski DT, Mane J, Cheewatrakoolpong B, Xue J, Szeto D, Forrest G, Miller C, Bunzel M, Plummer CW, Chobanian HR, Miller MW, Souza S, Thomas-Fowlkes BS, Ogawa AM, Weinglass AB, Di Salvo J, Li X, Feng Y, Tatosian DA, Howard AD, Colletti SL and Trujillo ME (2017). “GPR40 partial agonists and AgoPAMs: Differentiating effects on glucose and hormonal secretions in the rodent.” PLoS One 12(10): e0186033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Patel YC (1999). “Somatostatin and its receptor family.” Front Neuroendocrinol 20(3): 157–198. [DOI] [PubMed] [Google Scholar]
  176. Patel YC and Reichlin S (1978). “Somatostatin in hypothalamus, extrahypothalamic brain, and peripheral tissues of the rat.” Endocrinology 102(2): 523–530. [DOI] [PubMed] [Google Scholar]
  177. Pellicciari R, Gioiello A, Macchiarulo A, Thomas C, Rosatelli E, Natalini B, Sardella R, Pruzanski M, Roda A, Pastorini E, Schoonjans K and Auwerx J (2009). “Discovery of 6alpha-ethyl-23(S)-methylcholic acid (S-EMCA, INT-777) as a potent and selective agonist for the TGR5 receptor, a novel target for diabesity.” J Med Chem 52(24): 7958–7961. [DOI] [PubMed] [Google Scholar]
  178. Penick SB and Hinkle LE Jr. (1961). “Depression of food intake induced in healthy subjects by glucagon.” N Engl J Med 264: 893–897. [DOI] [PubMed] [Google Scholar]
  179. Peukert S, Hughes R, Nunez J, He G, Yan Z, Jain R, Llamas L, Luchansky S, Carlson A, Liang G, Kunjathoor V, Pietropaolo M, Shapiro J, Castellana A, Wu X and Bose A (2014). “Discovery of 2-Pyridylpyrimidines as the First Orally Bioavailable GPR39 Agonists.” ACS Med Chem Lett 5(10): 1114–1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Pi-Sunyer X, Astrup A, Fujioka K, Greenway F, Halpern A, Krempf M, Lau DC, le Roux CW, Violante Ortiz R, Jensen CB, Wilding JP, Obesity S and Prediabetes NNSG (2015). “A Randomized, Controlled Trial of 3.0 mg of Liraglutide in Weight Management.” N Engl J Med 373(1): 11–22. [DOI] [PubMed] [Google Scholar]
  181. Pittner RA, Moore CX, Bhavsar SP, Gedulin BR, Smith PA, Jodka CM, Parkes DG, Paterniti JR, Srivastava VP and Young AA (2004). “Effects of PYY[3–36] in rodent models of diabetes and obesity.” Int J Obes Relat Metab Disord 28(8): 963–971. [DOI] [PubMed] [Google Scholar]
  182. Plamboeck A, Veedfald S, Deacon CF, Hartmann B, Wettergren A, Svendsen LB, Meisner S, Hovendal C, Vilsboll T, Knop FK and Holst JJ (2013). “The effect of exogenous GLP-1 on food intake is lost in male truncally vagotomized subjects with pyloroplasty.” Am J Physiol Gastrointest Liver Physiol 304(12): G1117–1127. [DOI] [PubMed] [Google Scholar]
  183. Pocai A (2012). “Unraveling oxyntomodulin, GLP1’s enigmatic brother.” J Endocrinol 215(3): 335–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Pocai A, Carrington PE, Adams JR, Wright M, Eiermann G, Zhu L, Du X, Petrov A, Lassman ME, Jiang G, Liu F, Miller C, Tota LM, Zhou G, Zhang X, Sountis MM, Santoprete A, Capito E, Chicchi GG, Thornberry N, Bianchi E, Pessi A, Marsh DJ and SinhaRoy R (2009). “Glucagon-like peptide 1/glucagon receptor dual agonism reverses obesity in mice.” Diabetes 58(10): 2258–2266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Polonsky KS, Shoelson SE and Docherty HM (1983). “Plasma somatostatin 28 increases in response to feeding in man.” Journal of Clinical Investigation 71(5): 1514–1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Rangwala SM, D’Aquino K, Zhang YM, Bader L, Edwards W, Zheng S, Eckardt A, Lacombe A, Pick R, Moreno V, Kang L, Jian W, Arnoult E, Case M, Jenkinson C, Chi E, Swanson RV, Kievit P, Grove K, Macielag M, Erion MD, SinhaRoy R and Leonard JN (2019). “A Long-Acting PYY3–36 Analog Mediates Robust Anorectic Efficacy with Minimal Emesis in Nonhuman Primates.” Cell Metab 29(4): 837–843 e835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Rao RS and Kini S (2011). “GIP and bariatric surgery.” Obes Surg 21(2): 244–252. [DOI] [PubMed] [Google Scholar]
  188. Ratner C, He Z, Grunddal KV, Skov LJ, Hartmann B, Zhang F, Feuchtinger A, Bjerregaard A, Christoffersen C, Tschop MH, Finan B, DiMarchi RD, Leinninger GM, Williams KW, Clemmensen C and Holst B (2019). “Long-Acting Neurotensin Synergizes With Liraglutide to Reverse Obesity Through a Melanocortin-Dependent Pathway.” Diabetes 68(6): 1329–1340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Ratner C, Skov LJ, Raida Z, Bachler T, Bellmann-Sickert K, Le Foll C, Sivertsen B, Dalboge LS, Hartmann B, Beck-Sickinger AG, Madsen AN, Jelsing J, Holst JJ, Lutz TA, Andrews ZB and Holst B (2016). “Effects of Peripheral Neurotensin on Appetite Regulation and Its Role in Gastric Bypass Surgery.” Endocrinology 157(9): 3482–3492. [DOI] [PubMed] [Google Scholar]
  190. Rigamonti AE, Cella SG, Bonomo SM, Mancia G, Grassi G, Perotti M, Agosti F, Sartorio A, Muller EE and Pincelli AI (2011). “Effect of somatostatin infusion on peptide YY secretion: studies in the acute and recovery phase of anorexia nervosa and in obesity.” Eur J Endocrinol 165(3): 421–427. [DOI] [PubMed] [Google Scholar]
  191. Ritter K, Buning C, Halland N, Poverlein C and Schwink L (2016). “G Protein-Coupled Receptor 119 (GPR119) Agonists for the Treatment of Diabetes: Recent Progress and Prevailing Challenges.” J Med Chem 59(8): 3579–3592. [DOI] [PubMed] [Google Scholar]
  192. Rubino F, Nathan DM, Eckel RH, Schauer PR, Alberti KG, Zimmet PZ, Del Prato S, Ji L, Sadikot SM, Herman WH, Amiel SA, Kaplan LM, Taroncher-Oldenburg G, Cummings DE and S. Delegates of the 2nd Diabetes Surgery (2016). “Metabolic Surgery in the Treatment Algorithm for Type 2 Diabetes: A Joint Statement by International Diabetes Organizations.” Diabetes Care 39(6): 861–877. [DOI] [PubMed] [Google Scholar]
  193. Rudenko O, Shang J, Munk A, Ekberg JP, Petersen N, Engelstoft MS, Egerod KL, Hjorth SA, Wu M, Feng Y, Zhou YP, Mokrosinski J, Thams P, Reimann F, Gribble F, Rehfeld JF, Holst JJ, Treebak JT, Howard AD and Schwartz TW (2019). “The aromatic amino acid sensor GPR142 controls metabolism through balanced regulation of pancreatic and gut hormones.” Mol Metab 19: 49–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Ryan KK, Tremaroli V, Clemmensen C, Kovatcheva-Datchary P, Myronovych A, Karns R, Wilson-Perez HE, Sandoval DA, Kohli R, Backhed F and Seeley RJ (2014). “FXR is a molecular target for the effects of vertical sleeve gastrectomy.” Nature 509(7499): 183–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Salehi M and D’Alessio DA (2016). “Mechanisms of surgical control of type 2 diabetes: GLP-1 is the key factor-Maybe.” Surg Obes Relat Dis 12(6): 1230–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Salehi M, Gastaldelli A and D’Alessio DA (2019). “Beta-cell sensitivity to insulinotropic gut hormones is reduced after gastric bypass surgery.” Gut. [DOI] [PubMed] [Google Scholar]
  197. Salem V, Izzi-Engbeaya C, Coello C, Thomas DB, Chambers ES, Comninos AN, Buckley A, Win Z, Al-Nahhas A, Rabiner EA, Gunn RN, Budge H, Symonds ME, Bloom SR, Tan TM and Dhillo WS (2016). “Glucagon increases energy expenditure independently of brown adipose tissue activation in humans.” Diabetes Obes Metab 18(1): 72–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Schalla MA and Stengel A (2019). “Pharmacological Modulation of Ghrelin to Induce Weight Loss: Successes and Challenges.” Curr Diab Rep 19(10): 102. [DOI] [PubMed] [Google Scholar]
  199. Schauer PR, Bhatt DL, Kirwan JP, Wolski K, Aminian A, Brethauer SA, Navaneethan SD, Singh RP, Pothier CE, Nissen SE, Kashyap SR and Investigators S (2017). “Bariatric Surgery versus Intensive Medical Therapy for Diabetes - 5-Year Outcomes.” N Engl J Med 376(7): 641–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Schellekens H, Dinan TG and Cryan JF (2010). “Lean mean fat reducing “ghrelin” machine: hypothalamic ghrelin and ghrelin receptors as therapeutic targets in obesity.” Neuropharmacology 58(1): 2–16. [DOI] [PubMed] [Google Scholar]
  201. Schilperoort M, van Dam AD, Hoeke G, Shabalina IG, Okolo A, Hanyaloglu AC, Dib LH, Mol IM, Caengprasath N, Chan YW, Damak S, Miller AR, Coskun T, Shimpukade B, Ulven T, Kooijman S, Rensen PC and Christian M (2018). “The GPR120 agonist TUG-891 promotes metabolic health by stimulating mitochondrial respiration in brown fat.” EMBO Mol Med 10(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Schmidt JB, Gregersen NT, Pedersen SD, Arentoft JL, Ritz C, Schwartz TW, Holst JJ, Astrup A and Sjodin A (2014). “Effects of PYY3–36 and GLP-1 on energy intake, energy expenditure, and appetite in overweight men.” Am J Physiol Endocrinol Metab 306(11): E1248–1256. [DOI] [PubMed] [Google Scholar]
  203. Schmitt C, Portron A, Jadidi S, Sarkar N and DiMarchi R (2017). “Pharmacodynamics, pharmacokinetics and safety of multiple ascending doses of the novel dual glucose-dependent insulinotropic polypeptide/glucagon-like peptide-1 agonist RG7697 in people with type 2 diabetes mellitus.” Diabetes Obes Metab 19(10): 1436–1445. [DOI] [PubMed] [Google Scholar]
  204. Secher A, Jelsing J, Baquero AF, Hecksher-Sorensen J, Cowley MA, Dalboge LS, Hansen G, Grove KL, Pyke C, Raun K, Schaffer L, Tang-Christensen M, Verma S, Witgen BM, Vrang N and Bjerre Knudsen L (2014). “The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss.” J Clin Invest 124(10): 4473–4488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Seeley RJ, Chambers AP and Sandoval DA (2015). “The role of gut adaptation in the potent effects of multiple bariatric surgeries on obesity and diabetes.” Cell Metab 21(3): 369–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Semple G, Ren A, Fioravanti B, Pereira G, Calderon I, Choi K, Xiong Y, Shin YJ, Gharbaoui T, Sage CR, Morgan M, Xing C, Chu ZL, Leonard JN, Grottick AJ, Al-Shamma H, Liang Y, Demarest KT and Jones RM (2011). “Discovery of fused bicyclic agonists of the orphan G-protein coupled receptor GPR119 with in vivo activity in rodent models of glucose control.” Bioorg Med Chem Lett 21(10): 3134–3141. [DOI] [PubMed] [Google Scholar]
  207. Shankar SS, Shankar RR, Mixson LA, Miller DL, Pramanik B, O’Dowd AK, Williams DM, Frederick CB, Beals CR, Stoch SA, Steinberg HO and Kelley DE (2018). “Native Oxyntomodulin Has Significant Glucoregulatory Effects Independent of Weight Loss in Obese Humans With and Without Type 2 Diabetes.” Diabetes 67(6): 1105–1112. [DOI] [PubMed] [Google Scholar]
  208. Shen LP, Pictet RL and Rutter WJ (1982). “Human somatostatin I: sequence of the cDNA.” Proc Natl Acad Sci U S A 79(15): 4575–4579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Sheng R, Yang L, Zhang Y, Xing E, Shi R, Wen X, Wang H and Sun H (2018). “Discovery of novel selective GPR120 agonists with potent anti-diabetic activity by hybrid design.” Bioorg Med Chem Lett 28(15): 2599–2604. [DOI] [PubMed] [Google Scholar]
  210. Shi J, Gu Z, Jurica EA, Wu X, Haque LE, Williams KN, Hernandez AS, Hong Z, Gao Q, Dabros M, Davulcu AH, Mathur A, Rampulla RA, Gupta AK, Jayaram R, Apedo A, Moore DB, Liu H, Kunselman LK, Brady EJ, Wilkes JJ, Zinker BA, Cai H, Shu YZ, Sun Q, Dierks EA, Foster KA, Xu C, Wang T, Panemangalore R, Cvijic ME, Xie C, Cao GG, Zhou M, Krupinski J, Whaley JM, Robl JA, Ewing WR and Ellsworth BA (2018). “Discovery of Potent and Orally Bioavailable Dihydropyrazole GPR40 Agonists.” J Med Chem 61(3): 681–694. [DOI] [PubMed] [Google Scholar]
  211. Sjostrom L (2013). “Review of the key results from the Swedish Obese Subjects (SOS) trial - a prospective controlled intervention study of bariatric surgery.” J Intern Med 273(3): 219–234. [DOI] [PubMed] [Google Scholar]
  212. Sloop KW, Briere DA, Emmerson PJ and Willard FS (2018). “Beyond Glucagon-like Peptide-1: Is G-Protein Coupled Receptor Polypharmacology the Path Forward to Treating Metabolic Diseases?” ACS Pharmacology & Translational Science 1(1): 3–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Sparks SM, Aquino C, Banker P, Collins JL, Cowan D, Diaz C, Dock ST, Hertzog DL, Liang X, Swiger ED, Yuen J, Chen G, Jayawickreme C, Moncol D, Nystrom C, Rash V, Rimele T, Roller S and Ross S (2017). “Exploration of phenylpropanoic acids as agonists of the free fatty acid receptor 4 (FFA4): Identification of an orally efficacious FFA4 agonist.” Bioorg Med Chem Lett 27(5): 1278–1283. [DOI] [PubMed] [Google Scholar]
  214. Srivastava G and Apovian C (2018). “Future Pharmacotherapy for Obesity: New Anti-obesity Drugs on the Horizon.” Curr Obes Rep 7(2): 147–161. [DOI] [PubMed] [Google Scholar]
  215. Sumida Y and Yoneda M (2018). “Current and future pharmacological therapies for NAFLD/NASH.” J Gastroenterol 53(3): 362–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Sun EWL, Martin AM, Young RL and Keating DJ (2018). “The Regulation of Peripheral Metabolism by Gut-Derived Hormones.” Front Endocrinol (Lausanne) 9: 754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Sundstrom L, Myhre S, Sundqvist M, Ahnmark A, McCoull W, Raubo P, Groombridge SD, Polla M, Nystrom AC, Kristensson L, Nagard M and Winzell MS (2017). “The acute glucose lowering effect of specific GPR120 activation in mice is mainly driven by glucagon-like peptide 1.” PLoS One 12(12): e0189060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Talsania T, Anini Y, Siu S, Drucker DJ and Brubaker PL (2005). “Peripheral exendin-4 and peptide YY(3–36) synergistically reduce food intake through different mechanisms in mice.” Endocrinology 146(9): 3748–3756. [DOI] [PubMed] [Google Scholar]
  219. Tan T, Behary P, Tharakan G, Minnion J, Al-Najim W, Albrechtsen NJW, Holst JJ and Bloom SR (2017). “The Effect of a Subcutaneous Infusion of GLP-1, OXM, and PYY on Energy Intake and Expenditure in Obese Volunteers.” J Clin Endocrinol Metab 102(7): 2364–2372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Tan TM, Vanderpump M, Khoo B, Patterson M, Ghatei MA and Goldstone AP (2004). “Somatostatin infusion lowers plasma ghrelin without reducing appetite in adults with Prader-Willi syndrome.” J Clin Endocrinol Metab 89(8): 4162–4165. [DOI] [PubMed] [Google Scholar]
  221. Tatemoto K (1982). “Isolation and characterization of peptide YY (PYY), a candidate gut hormone that inhibits pancreatic exocrine secretion.” PNAS 79(8): 2514–2518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Tatemoto K and Mutt V (1980). “Isolation of two novel candidate hormones using a chemical method for finding naturally occurring polypeptides.” Nature 285(5764): 417–418. [DOI] [PubMed] [Google Scholar]
  223. Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, Macchiarulo A, Yamamoto H, Mataki C, Pruzanski M, Pellicciari R, Auwerx J and Schoonjans K (2009). “TGR5-mediated bile acid sensing controls glucose homeostasis.” Cell Metab 10(3): 167–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Tillner J, Posch MG, Wagner F, Teichert L, Hijazi Y, Einig C, Keil S, Haack T, Wagner M, Bossart M and Larsen PJ (2019). “A novel dual glucagon-like peptide and glucagon receptor agonist SAR425899: Results of randomized, placebo-controlled first-in-human and first-in-patient trials.” Diabetes Obes Metab 21(1): 120–128. [DOI] [PubMed] [Google Scholar]
  225. Toda N, Hao X, Ogawa Y, Oda K, Yu M, Fu Z, Chen Y, Kim Y, Lizarzaburu M, Lively S, Lawlis S, Murakoshi M, Nara F, Watanabe N, Reagan JD, Tian H, Fu A, Motani A, Liu Q, Lin YJ, Zhuang R, Xiong Y, Fan P, Medina J, Li L, Izumi M, Okuyama R and Shibuya S (2013). “Potent and Orally Bioavailable GPR142 Agonists as Novel Insulin Secretagogues for the Treatment of Type 2 Diabetes.” ACS Med Chem Lett 4(8): 790–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM, Diakogiannaki E, Cameron J, Grosse J, Reimann F and Gribble FM (2012). “Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2.” Diabetes 61(2): 364–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Tremaroli V, Karlsson F, Werling M, Stahlman M, Kovatcheva-Datchary P, Olbers T, Fandriks L, le Roux CW, Nielsen J and Backhed F (2015). “Roux-en-Y Gastric Bypass and Vertical Banded Gastroplasty Induce Long-Term Changes on the Human Gut Microbiome Contributing to Fat Mass Regulation.” Cell Metab 22(2): 228–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Tsilingiris D, Koliaki C and Kokkinos A (2019). “Remission of Type 2 Diabetes Mellitus after Bariatric Surgery: Fact or Fiction?” Int J Environ Res Public Health 16(17). [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Tuttle KR, Lakshmanan MC, Rayner B, Busch RS, Zimmermann AG, Woodward DB and Botros FT (2018). “Dulaglutide versus insulin glargine in patients with type 2 diabetes and moderate-to-severe chronic kidney disease (AWARD-7): a multicentre, open-label, randomised trial.” Lancet Diabetes Endocrinol 6(8): 605–617. [DOI] [PubMed] [Google Scholar]
  230. van den Hoek AM, Heijboer AC, Corssmit EPM, Voshol PJ, Romijn JA, Havekes LM and Pijl H (2004). “PYY3–36 Reinforces Insulin Action on Glucose Disposal in Mice Fed a High-Fat Diet.” Diabetes 53(8): 1949–1952. [DOI] [PubMed] [Google Scholar]
  231. Vilsboll T, Knop FK, Krarup T, Johansen A, Madsbad S, Larsen S, Hansen T, Pedersen O and Holst JJ (2003). “The pathophysiology of diabetes involves a defective amplification of the late-phase insulin response to glucose by glucose-dependent insulinotropic polypeptide-regardless of etiology and phenotype.” J Clin Endocrinol Metab 88(10): 4897–4903. [DOI] [PubMed] [Google Scholar]
  232. Vu MK, van Oostayen JA, Biemond I and Masclee AAM (2001). “Effect of somatostatin on postprandial gallbladder relaxation.” Clinical Physiology 21(1): 25–31. [DOI] [PubMed] [Google Scholar]
  233. Wacker DA, Wang Y, Broekema M, Rossi K, O’Connor S, Hong Z, Wu G, Malmstrom SE, Hung CP, LaMarre L, Chimalakonda A, Zhang L, Xin L, Cai H, Chu C, Boehm S, Zalaznick J, Ponticiello R, Sereda, Han SP, Zebo R, Zinker B, Luk CE, Wong R, Everlof G, Li YX, Wu CK, Lee M, Griffen S, Miller KJ, Krupinski J and Robl JA (2014). “Discovery of 5-chloro-4-((1-(5-chloropyrimidin-2-yl)piperidin-4-yl)oxy)-1-(2-fluoro-4-(methyls ulfonyl)phenyl)pyridin-2(1H)-one (BMS-903452), an antidiabetic clinical candidate targeting GPR119.” J Med Chem 57(18): 7499–7508. [DOI] [PubMed] [Google Scholar]
  234. Watanabe M, Houten SM, Mataki C, Christoffolete MA, Kim BW, Sato H, Messaddeq N, Harney JW, Ezaki O, Kodama T, Schoonjans K, Bianco AC and Auwerx J (2006). “Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation.” Nature 439(7075): 484–489. [DOI] [PubMed] [Google Scholar]
  235. Wewer Albrechtsen NJ, Hornburg D, Albrechtsen R, Svendsen B, Torang S, Jepsen SL, Kuhre RE, Hansen, Janus C, Floyd A, Lund A, Vilsboll T, Knop FK, Vestergaard H, Deacon CF, Meissner F, Mann M, Holst JJ and Hartmann B (2016). “Oxyntomodulin Identified as a Marker of Type 2 Diabetes and Gastric Bypass Surgery by Mass-spectrometry Based Profiling of Human Plasma.” EBioMedicine 7: 112–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Willard FS, Wootten D, Showalter AD, Savage EE, Ficorilli J, Farb TB, Bokvist K, Alsina-Fernandez J, Furness SG, Christopoulos A, Sexton PM and Sloop KW (2012). “Small molecule allosteric modulation of the glucagon-like Peptide-1 receptor enhances the insulinotropic effect of oxyntomodulin.” Mol Pharmacol 82(6): 1066–1073. [DOI] [PubMed] [Google Scholar]
  237. Williams EK, Chang RB, Strochlic DE, Umans BD, Lowell BB and Liberles SD (2016). “Sensory Neurons that Detect Stretch and Nutrients in the Digestive System.” Cell 166(1): 209–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Wilson-Perez HE, Chambers AP, Ryan KK, Li B, Sandoval DA, Stoffers D, Drucker DJ, Perez-Tilve D and Seeley RJ (2013). “Vertical sleeve gastrectomy is effective in two genetic mouse models of glucagon-like Peptide 1 receptor deficiency.” Diabetes 62(7): 2380–2385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Wilson-Perez HE, Chambers AP, Sandoval DA, Stefater MA, Woods SC, Benoit SC and Seeley RJ (2013). “The effect of vertical sleeve gastrectomy on food choice in rats.” Int J Obes (Lond) 37(2): 288–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Wilson JE, Kurukulasuriya R, Sinz C, Lombardo M, Bender K, Parker D, Sherer EC, Costa M, Dingley K, Li X, Mitelman S, Tong S, Bugianesi R, Ehrhardt A, Priest B, Ratliff K, Ujjainwalla F, Nargund R, Hagmann WK and Edmondson S (2016). “Discovery and development of benzo-[1,2,4]-triazolo-[1,4]-oxazepine GPR142 agonists for the treatment of diabetes.” Bioorganic & Medicinal Chemistry Letters 26(12): 2947–2951. [DOI] [PubMed] [Google Scholar]
  241. Winters MP, Sui Z, Wall M, Wang Y, Gunnet J, Leonard J, Hua H, Yan W, Suckow A, Bell A, Clapper W, Jenkinson C, Haug P, Koudriakova T, Huebert N and Murray WV (2018). “Discovery of N-arylpyrroles as agonists of GPR120 for the treatment of type II diabetes.” Bioorg Med Chem Lett 28(5): 841–846. [DOI] [PubMed] [Google Scholar]
  242. Wynne K, Park AJ, Small CJ, Meeran K, Ghatei MA, Frost GS and Bloom SR (2006). “Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomised controlled trial.” Int J Obes (Lond) 30(12): 1729–1736. [DOI] [PubMed] [Google Scholar]
  243. Wynne K, Park AJ, Small CJ, Patterson M, Ellis SM, Murphy KG, Wren AM, Frost GS, Meeran K, Ghatei MA and Bloom SR (2005). “Subcutaneous oxyntomodulin reduces body weight in overweight and obese subjects: a double-blind, randomized, controlled trial.” Diabetes 54(8): 2390–2395. [DOI] [PubMed] [Google Scholar]
  244. Yamada T, Shojima N, Noma H, Yamauchi T and Kadowaki T (2018). “Weekly Versus Daily Dipeptidyl Peptidase 4 Inhibitor Therapy for Type 2 Diabetes: Systematic Review and Meta-analysis.” Diabetes Care 41(4): e52–e55. [DOI] [PubMed] [Google Scholar]
  245. Yamada Y, Terauchi Y, Watada H, Nakatsuka Y, Shiosakai K, Washio T and Taguchi T (2018). “Efficacy and Safety of GPR119 Agonist DS-8500a in Japanese Patients with Type 2 Diabetes: a Randomized, Double-Blind, Placebo-Controlled, 12-Week Study.” Adv Ther 35(3): 367–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Yousseif A, Emmanuel J, Karra E, Millet Q, Elkalaawy M, Jenkinson AD, Hashemi M, Adamo M, Finer N, Fiennes AG, Withers DJ and Batterham RL (2014). “Differential effects of laparoscopic sleeve gastrectomy and laparoscopic gastric bypass on appetite, circulating acyl-ghrelin, peptide YY3–36 and active GLP-1 levels in non-diabetic humans.” Obes Surg 24(2): 241–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Yu M, Lizarzaburu M, Motani A, Fu Z, Du X, Liu JJ, Jiao X, Lai S, Fan P, Fu A, Liu Q, Murakoshi M, Nara F, Oda K, Okuyama R, Reagan JD, Watanabe N, Yamazaki M, Xiong Y, Zhang Y, Zhuang R, Lin DC, Houze JB, Medina JC and Li L (2013). “Aminopyrazole-Phenylalanine Based GPR142 Agonists: Discovery of Tool Compound and in Vivo Efficacy Studies.” ACS Med Chem Lett 4(9): 829–834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Zhang X, Cai C, Sui Z, Macielag M, Wang Y, Yan W, Suckow A, Hua H, Bell A, Haug P, Clapper W, Jenkinson C, Gunnet J, Leonard J and Murray WV (2017). “Discovery of an Isothiazole-Based Phenylpropanoic Acid GPR120 Agonist as a Development Candidate for Type 2 Diabetes.” ACS Med Chem Lett 8(9): 947–952. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES