OBJECTIVE—To examine the mechanisms of action, therapeutic potential, and challenges inherent in the use of incretin peptides and dipeptidyl peptidase-IV (DPP-IV) inhibitors for the treatment of type 2 diabetes.

RESEARCH DESIGN AND METHODS—The scientific literature describing the biological importance of incretin peptides and DPP-IV inhibitors in the control of glucose homeostasis has been reviewed, with an emphasis on mechanisms of action, experimental diabetes, human physiological experiments, and short-term clinical studies in normal and diabetic human subjects.

RESULTS—Glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) exert important effects on β-cells to stimulate glucose-dependent insulin secretion. Both peptides also regulate β-cell proliferation and cytoprotection. GLP-1, but not GIP, inhibits gastric emptying, glucagon secretion, and food intake. The glucose-lowering actions of GLP-1, but not GIP, are preserved in subjects with type 2 diabetes. However, native GLP-1 is rapidly degraded by DPP-IV after parenteral administration; hence, degradation-resistant, long-acting GLP-1 receptor (GLP-1R) agonists are preferable agents for the chronic treatment of human diabetes. Alternatively, inhibition of DPP-IV–mediated incretin degradation represents a complementary therapeutic approach, as orally available DPP-IV inhibitors have been shown to lower glucose in experimental diabetic models and human subjects with type 2 diabetes.

CONCLUSIONS—GLP-1R agonists and DPP-IV inhibitors have shown promising results in clinical trials for the treatment of type 2 diabetes. The need for daily injections of potentially immunogenic GLP-1–derived peptides and the potential for unanticipated side effects with chronic use of DPP-IV inhibitors will require ongoing scrutiny of the risk-benefit ratio for these new therapies as they are evaluated in the clinic.

After meal ingestion, nutrient entry into the stomach and transit through the proximal gastrointestinal (GI) tract stimulates activation of neural and hormonal signals that control gastric emptying and gut motility, nutrient absorption, and hormonal regulation of energy disposal and storage. The mucosal epithelium of the GI tract is one of the earliest integrators of information relevant to digestion and assimilation of nutrient loads. Highly specialized enteroendocrine cells dispersed along the length of the GI tract play an important role in controlling the rate of gastric emptying and small bowel motility, pancreatic enzyme secretion, and the growth and differentiated absorptive function of the small and large bowel epithelium. The aim of this review is to examine our current understanding of the physiological actions of two gut hormones, glucagon-like peptide (GLP)-1 and glucose-dependent insulinotropic polypeptide (GIP), with an emphasis on the biological importance and pharmaceutical potential of these peptides for the treatment of type 2 diabetes.

The development and application of the insulin radioimmunoassay to clinical investigation has permitted the assessment of β-cell secretory function after meal ingestion in normal and diabetic subjects. The observation that food ingestion or enteral glucose administration provoked a greater stimulation of insulin release compared with similar amounts of energy (glucose) infused intravenously (1,2) led to the development of the incretin concept. Hence, it was postulated that gut-derived signals stimulated by oral nutrient ingestion represent potent insulin secretagogues responsible for the augmentation of insulin release when energy is administered via the gut versus the parenteral route (3). Although several neurotransmitters and gut hormones possess incretin-like activity, the considerable evidence from immunoneutralization, antagonist, and knockout studies suggests that GIP and GLP-1 represent the dominant peptides responsible for the majority of nutrient-stimulated insulin secretion. The observation that patients with type 2 diabetes exhibit a significant reduction in the magnitude of meal-stimulated insulin release underlies the interest in determining whether defective incretin release or resistance to incretin action contributes to the pathophysiology of β-cell dysfunction in diabetic subjects.

GIP and GLP-1 are members of the glucagon peptide superfamily and share considerable amino acid identity. GIP is a single 42−amino acid peptide encoded within a larger 153−amino acid precursor (Fig. 1) (4). GIP-secreting enteroendocrine K-cells are concentrated in the duodenum and proximal jejunum; hence, these cells are anatomically situated in an ideal location for sensing and responding to nutrient ingestion. GLP-1 is derived from a larger proglucagon precursor that encodes not only GLP-1 but also the related proglucagon-derived peptides glucagon, GLP-2, oxyntomodulin, and glicentin (Fig. 1) (5). The two forms of GLP-1 secreted after meal ingestion, GLP-1(7-37) and GLP-1(7-36)amide differ by a single amino acid. Both peptides are equipotent and exhibit identical plasma half-lives and biological activities acting through the same receptor (6,7); however, the majority (∼80%) of circulating active GLP-1 appears to be GLP-1(7-36)amide (8). In contrast to the more proximal location of GIP-producing K-cells, the majority of GLP-1 is synthesized within L-cells located predominantly in the ileum and colon, although GLP-1–producing L-cells have also been identified more proximally in the duodenum and jejunum. Despite the more distal location of most L-cells, circulating levels of GLP-1 also increase rapidly within minutes of food ingestion. Hence, GLP-1 secretion from the distal gut is controlled by both neural and endocrine signals initiated by nutrient entry in the proximal GI tract, as well as by subsequent direct contact of open-type L-cells with digested nutrients. Ingestion of a mixed meal or a meal enriched with specific fats and complex carbohydrates is particularly effective in stimulating GIP and GLP-1 release in human subjects (9,10). Although the vagal nerve, via M1 muscarinic receptors, and several neuroendocrine peptides contribute to the regulation of GLP-1 release in rodents (11,12), the factors responsible for rapid nutrient-stimulated GLP-1 release in human subjects are largely unknown.

The levels of total circulating GIP and GLP-1 immunoreactivity reflect a combination of intact, full-length active and NH2-terminally truncated inactive peptides, with GIP(3-42) and GLP-1(9-36)amide contributing to >50% of total immunoreactive GIP and GLP-1 in both the fasting and the postprandial states (13,14). Plasma levels of both GIP and GLP-1 immunoreactivity are low in the fasting state and rise rapidly within minutes of food ingestion. Initial studies of circulating levels of GIP and GLP-1 relied principally on radioimmunoassays incapable of distinguishing the biologically active full-length peptides from inactive COOH-terminal peptide fragments generated as a result of proteolytic cleavage. Studies have demonstrated that both GIP and GLP-1 were cleaved at the position 2 alanine by the widely expressed aminopeptidase dipeptidyl peptidase IV (DPP-IV) (15,16). These findings have prompted a reanalysis of the circulating molecular forms of GIP and GLP-1 using newer radioimmunoassays more specific for the full-length bioactive peptides in normal and diabetic subjects.

The disappearance of exogenously administered GIP and GLP-1 has been studied in normal and diabetic human subjects using antisera capable of discriminating the full-length from the NH2-terminally cleaved peptides. The t1/2 of infused GIP is ∼7 and 5 min in normal and diabetic human subjects, respectively (14). In contrast, the t1/2 of exogenously infused intact GLP-1 is considerably shorter (13), with intravenously administered GLP-1 eliminated with a half-life of ∼2 min in both normal and obese diabetic human subjects (17). Although the NH2-terminally truncated peptides GIP(3-42) and GLP-1(9-36)amide function as weak antagonists of their respective receptors (18,19), there is little evidence that these truncated peptides exert physiologically important actions in human subjects in vivo. Despite observations that GLP-1(9-36)amide may function as an activator of insulin-independent glucose clearance in pigs (20), this peptide does not exert significant glucose-lowering properties in human subjects (21).

Circulating levels of GIP(1-42) are normal or slightly increased in type 2 diabetic subjects in the basal or postprandial states (22). In contrast, subjects with diabetes or impaired glucose tolerance exhibit modest but significant reductions in levels of meal-stimulated circulating GLP-1 (22,23). Furthermore, meal-induced increases in GIP and GLP-1 secretion are inversely correlated with the extent of insulin resistance detected in human subjects (24). The lower levels of circulating GLP-1 detected in diabetic subjects are not attributable to altered GLP-1 clearance (17). Whether levels of meal-stimulated GLP-1 may be restored toward normal with improved control of diabetes remains unknown.

GIP was originally observed to inhibit gastric acid secretion (gastric inhibitory polypeptide), predominantly at supraphysiological dosages. Subsequent studies have demonstrated potent glucose-dependent insulin stimulatory effects from GIP administration in dogs and rodents. GIP also regulates fat metabolism in adipocytes, including stimulation of lipoprotein lipase activity, fatty acid incorporation, and fatty acid synthesis (25). Unlike GLP-1, GIP does not inhibit glucagon secretion or gastric emptying. GIP does promote β-cell proliferation and cell survival in islet cell line studies (26,27); whether GIP also induces β-cell growth or survival in diabetic rodents remains unclear.

The physiological actions of GIP have been deduced using GIP peptide antagonists, GIP receptor antisera, and GIP receptor knockout mice. NH2-terminally truncated or modified GIP peptides such as GIP(6-30)amide, GIP(7-30)amide, or (Pro3)GIP block GIP binding to the GIP receptor with varying effectiveness, and attenuate the insulinotropic effects of exogenous GIP in vitro and endogenous GIP in vivo (2830). Similarly, immunopurified antisera against the extracellular domain of the GIP receptor block GIP binding and attenuate glucose-dependent insulin secretion after oral glucose loading in rats and mice (31,32). Complementary evidence for the incretin-like actions of GIP is derived from analysis of GIP receptor−null mice, which exhibit mild glucose intolerance after oral glucose loading (33). Surprisingly, GIPR−/− mice exhibit resistance to diet-induced obesity after months of high-fat feeding. Moreover, the GIPR−/− genotype attenuates obesity in the ob/ob mouse, possibly because of reduced fat storage and altered lipid metabolism as a direct result of absent GIP receptor (GIPR) action in adipocytes (34). Whether GIPR action significantly modulates adipocyte biology, lipoprotein synthesis, and weight accretion in humans is not known.

In contrast to the potent glucose-lowering actions of GIP in normal rodents, exogenous GIP administration is comparatively less insulinotropic in obese diabetic rodents. GIP levels are increased in some models of experimental rodent diabetes, and continuous GIP infusion for 4 h produces GIPR desensitization in normal rats (35). ZDF rats exhibit normal levels of GIP, absent insulinotropic responses to exogenous GIP and reduced expression of the GIPR in isolated islets (36). Recent studies with more potent GIP analogs engineered for resistance to DPP-IV have demonstrated improved insulinotropic and glucose-lowering properties after peptide administration to both normal and diabetic rodents (3739).

Infusion of porcine or human GIP into patients with type 2 diabetes has produced variable insulinotropic responses, ranging from preserved (40) to attenuated or near absent insulin secretion (4145). The potential for β-cell GIP responsivity to improve with treatment in type 2 diabetic subjects is intriguing, but has not been extensively examined (46). The GIP defect in insulin secretion seems most pronounced in the late phase of insulin secretion (47). Moreover, ∼50% of normoglycemic first-degree relatives of type 2 diabetic subjects exhibit reduced insulin secretion after exogenous GIP infusion (48). Hence the reduced insulinotropic action of GIP in diabetes likely reflects a combination of genetic and acquired defects. Whether the pancreatic effects of GIP on β-cell proliferation and survival are also diminished in experimental or clinical diabetes is not known.

Original observations elucidating a role for GLP-1 in the potentiation of glucose-dependent insulin secretion (4951) and insulin gene expression (52) were followed by experiments demonstrating that GLP-1 also inhibits glucagon secretion (53,54) and gastric emptying (55) (Fig. 2). Acute intracerebroventricular (56) injection of GLP-1 or GLP-1 receptor (GLP-1R) agonists produces transient reduction in food intake, whereas more prolonged intracerebroventricular or peripheral GLP-1R agonist administration is associated with weight loss in some (5760), but not all (61) studies. GLP-1 actions on food intake appear related in part to overlapping actions on central nervous system aversive signaling pathways, which remains a topic of intense interest (6266). In contrast to GIP, the spectrum of actions delineated for GLP-1 that promote glucose lowering (regulation of insulin and glucagon secretion, inhibition of gastric emptying, and reduction of food intake) appear comparable in diabetic versus nondiabetic animals of various ages.

GLP-1 exerts actions on β-cells independent of acute stimulation of insulin secretion. Incubation of isolated rat islet cells with GLP-1 recruited nonresponsive glucose-resistant β-cells to a functional state of glucose-responsive insulin secretion, designated glucose competence (67,68). GLP-1R agonists also promote insulin biosynthesis, β-cell proliferation, and survival (6971), and stimulate differentiation of exocrine cells or islet precursors toward a more differentiated β-cell phenotype (7274). The GLP-1R–dependent augmentation of β-cell mass has been demonstrated in diverse experimental models, including neonatal rats administered streptozotocin and exendin-4 (75) and normal Wistar rats ages 6 and 22 months infused with native GLP-1 for 5 days (76). Similarly, GLP-1R agonists promote β-cell proliferation and expansion of functional islet mass after partial pancreatectomy in rats aged 4–5 weeks (69) or in neonatal rat pups subjected to experimental intrauterine growth retardation (77). The expansion of β-cell mass after GLP-1R agonist administration prevents or delays the occurrence of diabetes in db/db mice (78) and GK diabetes-prone rats (79). Furthermore, the induction of islet proliferation after GLP-1R activation has been seen with a broad range of GLP-1R agonists, including native GLP-1 (76,79,80), exendin-4, NN2211 (81), and CJC-1131 (82).

GLP-1R agonists also activate anti-apoptotic pathways coupled to a reduction in β-cell death. db/db mice treated with exendin-4 for 2 weeks exhibited decreased numbers of apoptotic β-cells, reduced pancreatic caspase-3 activation, and increased Akt1 expression (78). Reduced islet apoptosis has been observed in GLP-1–treated Zucker diabetic rats (83) and in exendin-4–treated mice after streptozotocin-induced β-cell injury (70). The anti-apoptotic actions of GLP-1R agonists are likely direct, as GLP-1 reduced peroxide-induced apoptosis in Min6 insulinoma cells (84) and exendin-4 significantly attenuated cytokine-induced apoptosis in cultures of purified rat β-cells (70). Hence, the GLP-1R–dependent activation of both proliferative and anti-apoptotic pathways in the pancreas provides complementary mechanisms for preserving and enhancing functional β-cell mass.

The physiological importance of GLP-1 action has been studied using GLP-1R antagonists. Infusion of the peptide exendin(9-39) into rats, mice, baboons, and humans produces an increase in fasting glucose and glycemic excursion after oral glucose loading in association with reduced levels of circulating insulin (32,8587). Exendin(9-39) also produces abnormal glycemic excursion after nonenteral glucose loading in mice (32). These findings illustrate that transient disruption of GLP-1 action consistently perturbs the incretin and nonincretin actions of GLP-1 on glucoregulation. Acute intracerebroventricular injection of exendin(9-39) increases food intake in satiated rats (56), whereas repeated daily intracerebroventricular administration of exendin(9-39) increases food intake and weight gain (57). Similarly, acute exendin(9-39) administration increases gastric emptying after glucose ingestion in fistulized rats (88). Comparable studies with exendin(9-39) in humans have demonstrated the essential role of GLP-1 action for glucose control via regulation of glucagon and insulin secretion (89,90). Hence, the majority of actions observed after exogenous administration of GLP-1R agonists are also physiologically essential, as revealed by acute interruption of GLP-1 action.

Genetic disruption of GLP-1R expression in mice has produced comparable insights into the physiological importance of GLP-1 action. GLP-1R−/− mice exhibit abnormal glucose tolerance after both oral and intraperitoneal glucose challenge in association with diminished glucose-stimulated insulin secretion. In contrast, insulin sensitivity and the glucagon response to glucose loading or hypoglycemia are normal in the absence of GLP-1R signaling (91). Consistent with the cardiovascular effects of GLP-1 in rodents, GLP-1R−/− mice exhibit defective cardiovascular responses to stress (92). Despite the potential importance of GLP-1R circuits for transducing the anorectic action of leptin (93), GLP-1R−/− mice retain normal to enhanced leptin sensitivity (94,95). Similarly, food intake and body weight are not significantly perturbed in GLP-1R−/− mice in the CD1 genetic background (96,97). In contrast, GLP-1R−/− mice manifest subtle but detectable abnormalities in islet number and size (98) and exhibit a defective β-cell regenerative response to partial pancreatectomy (99). Hence, GLP-1R actions are physiologically important for the growth and adaptive regeneration of murine β-cells.

GLP-1R agonists and experimental models of diabetes

The glucose-lowering action of GLP-1 delineated in nondiabetic animals has been demonstrated in multiple models of experimental diabetes. A 48-h infusion of native GLP-1 lowered blood glucose in association with increased levels of circulating insulin, islet insulin content, and insulin mRNA in Wistar rats aged 22 months (100), and perfused pancreas studies have demonstrated GLP-1–dependent augmentation of insulin secretion in ZDF rats of diverse genetic backgrounds (101103). Similarly, exendin-4 lowered glucose in db/db and ob/ob mice, ZDF rats, and diabetic rhesus monkeys in acute and chronic experiments (78,104,105) and the GLP-1 analog NN211 improved glycemic control in pigs, rats, and mice (60,81,106). Remarkably, glucose tolerance remained significantly improved in ZDF rats for weeks after a 48-h infusion of native GLP-1 (107). This “memory effect” for sustained improvement of glycemic control was also observed in db/db mice after discontinuation of therapy with CJC-1131, an albumin-bound GLP-1R agonist (82).

GLP-1 action in human subjects

The majority of GLP-1 actions delineated in preclinical experiments have also been demonstrated in human studies. Infusion of GLP-1(7-36)amide into normal human subjects stimulated insulin secretion, reduced glucagon secretion, and significantly reduced blood glucose in the fasting state after glucose loading or meal ingestion (6,50,108). In contrast to GIP, the insulinotropic and glucose-lowering actions of GLP-1 are preserved in human subjects with type 2 diabetes (45,109) in both the fasting and the postprandial states (7). Similarly, GLP-1 inhibits gastric acid secretion (110,111) and gastric emptying in humans (55) and the GLP-1–dependent attenuation of gastric emptying contributes to decreased glycemic excursion and, consequently, reduced glucose-stimulated insulin secretion (112,113). Consistent with the importance of gastric emptying and glucagon secretion for glycemic control, GLP-1 also lowers blood glucose in type 1 diabetic subjects (114116). Analogous to studies demonstrating the induction of glucose competence in rodent β-cells, GLP-1 infusion enhances β-cell function and insulin secretory dynamics in human subjects with impaired glucose tolerance or type 2 diabetes (117119). GLP-1 may also enhance glucose clearance in humans (120,121); however, the majority of these actions are likely mediated indirectly through effects on insulin and glucagon (122124). Although several reports have described the effects of GLP-1 on muscle, liver, and fat cells, experimental evidence demonstrating expression of the GLP-1 receptor in these tissues in vivo is lacking. Hence, the indirect actions of GLP-1, leading to improvement in glycemic control and reduction in free fatty acids, may explain observations of improved insulin sensitivity in GLP-1–treated diabetic subjects (125).

The effect of GLP-1 in restoring glucose competence in rodent islets has prompted studies of GLP-1 action and β-cell function in type 2 diabetic patients. Insulin–treated diabetic subjects previously classified as “sulfonylurea nonresponders” exhibited β-cell GLP-1 responsivity, with lowering of fasting and postprandial glucose in association with enhanced insulin secretion (126). Patients treated with both GLP-1 and glibenclamide exhibited a greater degree of glucose reduction compared with the effect of either agent alone (127). Similarly, the combination of GLP-1 and metformin was shown in a 48-h crossover study to be more effective for lowering blood glucose than monotherapy with either agent alone (128).

The GLP-1–dependent suppression of glucagon secretion raises the possibility that GLP-1 therapy will be associated with an increased risk of hypoglycemia and potentially defective counterregulation if glucagon secretion remains suppressed in the face of GLP-1–linked hypoglycemia. Rapid gastric emptying may be associated with enhanced GLP-1 release and an increased risk of hypoglycemia in postgastrectomy patients (129). Similarly, acute administration of GLP-1 (80 nmol) to nondiabetic subjects in the fasted state produced mild relative hypoglycemia in some subjects (mean glucose ∼3.5 mmol/l) (130). Nevertheless, appropriate glucagon responses to hypoglycemia do not appear to be blunted in GLP-1–treated subjects (130), and GLP-1 infusion does not impair normal counterregulatory responses to hypoglycemia in healthy human subjects (131). Hence, the risk of hypoglycemia seems modest in type 2 diabetic subjects treated with GLP-1R agonists alone.

The demonstration that both intracerebroventricular and peripheral administration of GLP-1R agonists induces weight loss in preclinical experiments has fostered interest in the potential actions of these agents to diminish appetite and reduce weight gain in overweight human subjects. The majority of human studies have examined appetite and food ingestion over short (24-h) time periods after single-dose injection or continuous infusion of GLP-1. Small but statistically significant reductions in appetite and meal ingestion have been recorded in studies of normal, obese, and diabetic GLP-1–treated subjects (132136). A meta-analysis of available data from 115 subjects demonstrated significant GLP-1–dependent reductions in energy consumption in lean and overweight subjects (137). The acute reduction in food consumption and inhibition of gastric emptying has been detected even with physiological increases in levels of circulating GLP-1 (136). Administration of GLP-1 via continuous subcutaneous infusion for 6 weeks to obese diabetic subjects was associated with reduced appetite and a small but significant mean 1.9-kg weight loss (125). Hence, GLP-1 therapy in human subjects appears associated with prevention of weight gain or modest weight loss; however, long-term data are not yet available.

Although single or repeated subcutaneous injections of native GLP-1 decrease blood glucose in human subjects (138,139), the glucose-lowering effects are transient and no longer evident 1–2 h after peptide injection (140,141). Furthermore, continuous enhancement of GLP-1 action for 24 h/day appears superior for glucose control compared with peptide infusion for 16 h (142). Continuous intravenous or subcutaneous infusion of GLP-1 in short- and long-term studies has been shown to be highly effective in lowering blood glucose in diabetic subjects (125,143,144), but this intensive and expensive approach has major limitations for the treatment of large numbers of diabetic patients. The rapid degradation and clearance of native endogenous and exogenously administered GLP-1 (145) have spurred the clinical development of degradation-resistant GLP-1 analogs with longer durations of action in vivo.

Exendin-4 is a naturally occurring 39−amino acid GLP-1R agonist isolated from the salivary gland venom of the lizard Heloderma suspectum (146). Exendin-4 exhibits 53% amino acid identity to mammalian GLP-1 (146,147), yet binds to and activates the GLP-1 receptor. Furthermore, exendin-4 is highly resistant to the proteolytic activity of DPP-IV and exhibits a longer duration of action in vivo. Intravenous infusion of exendin-4 lowered fasting and postprandial blood glucose in normal healthy volunteers and was associated with a 19% reduction in calorie consumption assessed during a single test meal (148). Exendin-4 exerted similar effects on insulin secretion after acute intravenous infusion in diabetic subjects (149), and subcutaneous daily administration of exendin-4 to subjects with type 2 diabetes significantly reduced blood glucose and HbA1c (a decline from 9.1 to 8.3%) over a 1-month treatment period (150). Exendin-4 has been evaluated in eight phase 2 trials in 323 individuals with type 2 diabetes who received dosages of 0.05–2.0 μg/kg subcutaneously. Nausea and vomiting were the principal side effects observed (151). A 4-week treatment period produced a significant reduction in HbA1c levels, with sustained reduction in postprandial glycemia maintained over the 28-day treatment period.

Exendin-4 treatment (0.08 μg/kg s.c., b.i.d. or t.i.d.) over 1 month was evaluated in 109 patients treated with sulfonylureas or metformin, alone or in combination. The treatment was generally well tolerated, with three subjects withdrawing in the first 12 days because of nausea. At the end of the study period, a significant reduction was observed in levels of serum fructosamine, HbA1c, and mean postprandial glucose, but no significant change was noted in body weight or serum lipids (152). Antibodies against exendin-4 were detected in 19% of treated subjects; however, the antibodies did not affect treatment responses. In all, 15% of patients experienced hypoglycemia; all of these subjects received sulfonylureas plus exendin-4 (152). Exendin-4, recently renamed exenatide, is currently being evaluated for the treatment of type 2 diabetes in phase 3 trials in combination with metformin, sulfonylurea agents, or both.

NN2211 (liraglutide) is a fatty acid–linked DPP-IV–resistant derivative of GLP-1 designed for subcutaneous administration that exhibits a pharmacokinetic profile compatible with once-daily injection (153). NN2211 reduced fasting and postprandial glycemia in diabetic subjects after a single 10 μg/kg subcutaneous injection at 11:00 p.m., in association with inhibition of gastric emptying and reduced levels of circulating glucagon (154). NN2211 has been tested in phase 2 clinical trials. Additional approaches for prolonging the duration of action of GLP-1 derivatives include the use of albumin-bound GLP-1 molecules (82) and sustained release exendin-4 preparations; however, human data with these pharmaceutical approaches is currently limited.

Inhibition of DPP-IV for the treatment of type 2 diabetes

The observation that GLP-1 and GIP are rapidly cleaved at the position 2 alanine leading to inactivation of their biological activity (15,16) has fostered interest in the development of inhibitors of DPP-IV, the principal enzyme responsible for incretin inactivation (155,156). DPP-IV is but one member of a large family of related enzymes with overlapping enzyme specificity; however, adenosine deaminase affinity chromatography that specifically binds DPP-IV removes 95% of DPP-IV–like activity from human sera, consistent with the dominant role for DPP-IV as the major circulating enzyme exhibiting DPP-IV–like enzymatic activity in vivo (157). Complementary evidence supporting the importance of DPP-IV as a pharmaceutical target for lowering glucose levels is derived from analysis of rodents with inactivating DPP-IV mutations. DPP-IV knockout mice and the Fischer DPP-IV mutant rat exhibit reduced levels of glycemic excursion after glucose loading in association with increased levels of circulating GLP-1 and insulin (158,159). Remarkably, DPP-IV knockout mice exhibit resistance to obesity and display improved insulin sensitivity after high-fat feeding (160). Hence, both pharmacological and genetic attenuation of DPP-IV activity is associated with enhanced incretin action, increased insulin, and lower glucose in vivo.

DPP-IV inhibitors lowered blood glucose after acute and chronic administration in preclinical studies through mechanisms predominantly dependent on incretin action, leading to potentiation of glucose-stimulated insulin secretion (161163). Treatment of ZDF rats for 3 months with the inhibitor P32/98 resulted in progressive improvement in glycemic control, enhanced insulin secretory responses, increased insulin-stimulated muscle glucose uptake, and improved hepatic and peripheral insulin sensitivity (164,165). In one intriguing result, daily DPP-IV inhibitor therapy for 7 weeks in Wistar rats with streptozotocin-induced diabetes increased the numbers of islets and β-cells (166), consistent with the actions of GIP and GLP-1 in promoting islet neogenesis and cytoprotection (71).

Clinical experience with DPP-IV inhibitors in diabetic subjects is limited. A single-dose escalation study of P32/98 in healthy male volunteers demonstrated improved oral glucose tolerance in association with enhanced circulating levels of GLP-1 (167). A 4-week trial of NVP DPP728 administered several times a day to subjects with mild type 2 diabetes (mean entry HbA1c of ∼7.6%) produced significant glucose lowering in mean HbA1c to 6.9% (168). A second-generation DPP-IV inhibitor, LAF237, is currently in phase 2 clinical trials and additional DPP-IV inhibitors are in clinical development. Although inhibition of DPP-IV activity is a promising approach for enhancing incretin action in diabetic subjects, DPP-IV exhibits catalytic activity against a broad number of peptide substrates (155,169). Furthermore, DPP-IV, also known as the lymphocyte cell surface transmembrane-signaling molecule CD26, is activated by external stimuli and modulates T-cell activation, producing pleiotropic effects in experimental inflammatory and neoplastic disorders (155,170). Global genetic inactivation of CD26 in mice is associated with subtle but detectable abnormalities in cytokine and immunoglobulin secretion after mitogen stimulation (171). Whether highly selective inhibition of the catalytic activity of DPP-IV will adversely perturb immune-related activity in human subjects is unclear; hence, the long-term safety of sustained DPP-IV/CD26 inhibition merits careful scrutiny.

GLP-1R agonists and DPP-IV inhibitors: unanswered questions

Although GIP and GLP-1 exhibit both overlapping and unique mechanisms of action, GLP-1 exhibits several distinct advantages desirable in a therapeutic agent for treating type 2 diabetes (Table 1). Currently, there are few clinical data to support the development of injectable GIP agonists to treat human subjects with type 2 diabetes. Chronic GLP-1R agonist administration lowers blood glucose and HbA1c in diabetic subjects and has not yet been associated with receptor downregulation or tachyphylaxis, but few clinical reports are available that address this issue. Because nausea limits the dosage of GLP-1 administered in human studies, the potential for long-term prevention of weight gain or, ideally, induction of weight loss versus lack of compliance from unwanted gastrointestinal side effects will require scrutiny. Similarly, whether subsets of patients with type 2 diabetes will exhibit preferential GLP-1 responsivity or, alternatively, relative resistance to the glucose-lowering effects of GLP-1 is not known. As is the case with intensive insulin administration, the potent glucose-lowering properties of GLP-1R agonists may increase the likelihood of treatment-associated hypoglycemia in susceptible patients concomitantly treated with insulin secretagogues such as sulfonylureas. Although GLP-1R agonists produce remarkable effects on β-cell proliferation and cytoprotection in rodent studies, and human β-cells exhibit proliferative and cytoprotective responses to GLP-1 in vitro, the potential for GLP-1R agonists to prevent progression to β-cell failure in diabetic subjects is intriguing, but largely undocumented. Moreover, the current need for once or twice daily injections of GLP-1–based pharmaceutical agents raises acceptance and compliance issues for prolonged therapy with these agents. Whether the newer generation of long-acting GLP-1R agonists currently designed for weekly administration will be as potent as agents given once or twice daily remains unknown. Furthermore, GLP-1R agonists in the clinic exhibit <100% amino acid identity with the native peptide. The known immunogenic potential of even identical recombinant human therapeutic proteins (172) raises the specter of immunoneutralizing antibodies in some patients, which may lead to reduction in therapeutic efficacy or potential exacerbation of diabetes if the antibodies cross-react with endogenous GLP-1.

DPP-IV inhibitors represent a complementary approach for enhancing incretin action through orally available tablets. Whether these inhibitors should ideally aim for 24 h/day inhibition of DPP-IV activity is uncertain. Similarly, the observations that subjects with type 2 diabetes exhibit reduced levels of meal-stimulated circulating GLP-1 poses theoretical limitations for drugs acting in part through GLP-1–dependent mechanisms. DPP-IV inhibitors will be unable to achieve the same pharmaceutical elevation in levels of circulating GLP-1 compared with injectable GLP-1–based drugs, and are likely to be less potent compared with injectable GLP-1R agonists. A comparison of the advantages and disadvantages of DPP-IV inhibitors versus GLP-1R agonists is shown in Table 2. The broad spectrum of DPP-IV activity and the large number of potential bioactive peptide substrates pose important questions regarding unanticipated side effects associated with the long-term use of DPP-IV inhibitors. Taken together, the urgent need for diabetes therapeutic agents exhibiting new mechanisms of action and the preliminary efficacy of GLP-1R agonists and DPP-IV inhibitors in ongoing clinical trials suggest that one or both classes of agents may ultimately be approved for the treatment of type 2 diabetes. Furthermore, there remains intense interest in developing GLP-1 secretagogues or GLP-1 receptor activators (Fig. 3); hence, strategies focused on enhancing incretin action are likely to receive increasing attention if the first generation of GLP-1R agonists and DPP-IV inhibitors is approved for the treatment of type 2 diabetes.

Figure 1—

Structure of preproglucagon and preproGIP encoding GLP-1 and GIP, respectively, is shown. The arrow designates the position of the DPP-IV–mediated cleavage after the position 2 alanine residue. GRPP, glicentin-related pancreatic peptide; IP, intervening peptide.

Figure 1—

Structure of preproglucagon and preproGIP encoding GLP-1 and GIP, respectively, is shown. The arrow designates the position of the DPP-IV–mediated cleavage after the position 2 alanine residue. GRPP, glicentin-related pancreatic peptide; IP, intervening peptide.

Close modal
Figure 2—

The major biological actions of GLP-1.

Figure 2—

The major biological actions of GLP-1.

Close modal
Figure 3—

Potential mechanisms for potentiation of incretin action for the treatment of type 2 diabetes.

Figure 3—

Potential mechanisms for potentiation of incretin action for the treatment of type 2 diabetes.

Close modal
Table 1—

Properties and biological actions of GIP and GLP-1

GIPGLP-1
42–Amino acid peptide 30/31–Amino acid peptide 
Released from duodenum Released from distal small bowel and colon 
NH2-terminal inactivation by DPP-IV NH2-terminal inactivation by DPP-IV 
Stimulates insulin secretion Stimulates insulin secretion 
Minimal effect on gastric emptying Inhibits gastric emptying 
No effect on glucagon secretion Inhibits glucagon secretion 
No regulation of satiety and body weight Inhibits food intake and weight gain 
Promotes expansion of β-cell mass Promotes expansion of β-cell mass 
Normal GIP secretion in diabetic subjects Reduced GLP-1 secretion in diabetic subjects 
Defective GIP response in type 2 diabetes Preserved GLP-1 response in type 2 diabetes 
GIPGLP-1
42–Amino acid peptide 30/31–Amino acid peptide 
Released from duodenum Released from distal small bowel and colon 
NH2-terminal inactivation by DPP-IV NH2-terminal inactivation by DPP-IV 
Stimulates insulin secretion Stimulates insulin secretion 
Minimal effect on gastric emptying Inhibits gastric emptying 
No effect on glucagon secretion Inhibits glucagon secretion 
No regulation of satiety and body weight Inhibits food intake and weight gain 
Promotes expansion of β-cell mass Promotes expansion of β-cell mass 
Normal GIP secretion in diabetic subjects Reduced GLP-1 secretion in diabetic subjects 
Defective GIP response in type 2 diabetes Preserved GLP-1 response in type 2 diabetes 
Table 2—

Characteristics of DPP-IV inhibitors and GLP-1R agonists

DPP-IV inhibitorsGLP-1R agonists
Orally available Injectable 
Multiple targets Single known GPCR target 
GLP-1 PK favorable Higher levels of GLP-1 achievable, but narrow PK profile 
Short- versus long-acting Longer acting—days to weeks 
Less potent agents More potent glucose lowering 
Drug overdose nontoxic Drug overdose problematic 
No central nervous system side effects Nausea and vomiting 
Less defined side effect profile Well-described and tolerable side effect profile 
DPP-IV inhibitorsGLP-1R agonists
Orally available Injectable 
Multiple targets Single known GPCR target 
GLP-1 PK favorable Higher levels of GLP-1 achievable, but narrow PK profile 
Short- versus long-acting Longer acting—days to weeks 
Less potent agents More potent glucose lowering 
Drug overdose nontoxic Drug overdose problematic 
No central nervous system side effects Nausea and vomiting 
Less defined side effect profile Well-described and tolerable side effect profile 

GPCR, G protein–coupled receptor; PK, pharmacokinetic.

D.J.D. is supported in part by a Senior Scientist Award from the Canadian Institutes for Health Research and operating grants from the Juvenile Diabetes Research Foundation International and the Canadian Diabetes Association.

1.
Elrick H, Stimmler L, Hlad CJ, Arai Y: Plasma insulin responses to oral and intravenous glucose administration.
J Clin Endocrinol Metab
24
:
1076
–1082,
1964
2.
McIntyre N, Holdsworth CD, Turner DS: Intestinal factors in the control of insulin secretion.
J Clin Endocrinol Metab
25
:
1317
–1324,
1965
3.
Dupre J, Beck JC: Stimulation of release of insulin by an extract of intestinal mucosa.
Diabetes
15
:
555
–559,
1966
4.
Takeda J, Seino Y, Tanaka K, Fukumoto H, Kayano T, Takahashi H, Mitani T, Kurono M, Suzuki T, Tobe T, et al: Sequence of an intestinal cDNA encoding human gastric inhibitory polypeptide precursor.
Proc Natl Acad Sci U S A
84
:
7005
–7008,
1987
5.
Bell GI, Sanchez-Pescador R, Laybourn PJ, Najarian RC: Exon duplication and divergence in the human preproglucagon gene.
Nature
304
:
368
–371,
1983
6.
Orskov C, Wettergren A, Holst JJ: Biological effects and metabolic rates of glucagon-like peptide 1(7–36)amide and glucagon-like peptide 1(7–37) in healthy subjects are indistinguishable.
Diabetes
42
:
658
–661,
1993
7.
Nauck MA, Kleine N, Orskov C, Holst JJ, Willms B, Creutzfeldt W: Normalization of fasting hyperglycaemia by exogenous glucagon-like peptide 1 (7–36 amide) in type 2 (non-insulin-dependent) diabetic patients.
Diabetologia
36
:
741
–744,
1993
8.
Orskov C, Rabenhoj L, Wettergren A, Kofod H, Holst JJ: Tissue and plasma concentrations of amidated and glycine-extended glucagon-like peptide 1 in humans.
Diabetes
43
:
535
–539,
1994
9.
Herrmann C, Goke R, Richter G, Fehmann HC, Arnold R, Goke B: Glucagon-like peptide-1 and glucose-dependent insulin-releasing polypeptide plasma levels in response to nutrients.
Digestion
56
:
117
–126,
1995
10.
Elliott RM, Morgan LM, Tredger JA, Deacon S, Wright J, Marks V: Glucagon-like peptide-1(7–36)amide and glucose-dependent insulinotropic polypeptide secretion in response to nutrient ingestion in man: acute post-prandial and 24-h secretion patterns.
J Endocrinol
138
:
159
–166,
1993
11.
Rocca AS, Brubaker PL: Role of the vagus nerve in mediating proximal nutrient-induced glucagon-like peptide-1 secretion.
Endocrinology
140
:
1687
–1694,
1999
12.
Anini Y, Hansotia T, Brubaker PL: Muscarinic receptors control postprandial release of glucagon-like peptide-1: in vivo and in vitro studies in rats.
Endocrinology
143
:
2420
–2426,
2002
13.
Deacon CF, Nauck MA, Toft-Nielsen M, Pridal L, Willms B, Holst JJ: Both subcutaneously and intravenously administered glucagon-like peptide 1 are rapidly degraded from the NH2-terminus in type 2 diabetic patients and in healthy subjects.
Diabetes
44
:
1126
–1131,
1995
14.
Deacon CF, Nauck MA, Meier J, Hucking K, Holst JJ: Degradation of endogenous and exogenous gastric inhibitory polypeptide in healthy and in type 2 diabetic subjects as revealed using a new assay for the intact peptide.
J Clin Endocrinol Metab
85
:
3575
–3581,
2000
15.
Kieffer TJ, McIntosh CH, Pederson RA: Degradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IV.
Endocrinology
136
:
3585
–3596,
1995
16.
Mentlein R, Gallwitz B, Schmidt WE: Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon-like peptide-1(7–36)amide, peptide histidine methionine and is responsible for their degradation in human serum.
Eur J Biochem
214
:
829
–835,
1993
17.
Vilsboll T, Agerso H, Krarup T, Holst JJ: Similar elimination rates of glucagon-like peptide-1 in obese type 2 diabetic patients and healthy subjects.
J Clin Endocrinol Metab
88
:
220
–224,
2003
18.
Gault VA, Parker JC, Harriott P, Flatt PR, O’Harte FP: Evidence that the major degradation product of glucose-dependent insulinotropic polypeptide, GIP(3–42), is a GIP receptor antagonist in vivo.
J Endocrinol
175
:
525
–533,
2002
19.
Knudsen LB, Pridal L: Glucagon-like peptide-1-(9–36) amide is a major metabolite of glucagon-like peptide-1-(7–36)amide after in vivo administration to dogs and it acts as an antagonist on the pancreatic receptor.
Eur J Pharmacol
318
:
429
–435,
1996
20.
Deacon CF, Plamboeck A, Moller S, Holst JJ: GLP-1-(9–36) amide reduces blood glucose in anesthetized pigs by a mechanism that does not involve insulin secretion.
Am J Physiol
282
:
E873
−E879, 2002
21.
Vahl TP, Paty BW, Fuller BD, Prigeon RL, D’Alessio DA: Effects of GLP-1-(7–36)NH(2), GLP-1-(7–37), and GLP-1- (9–36)NH(2) on intravenous glucose tolerance and glucose-induced insulin secretion in healthy humans.
J Clin Endocrinol Metab
88
:
1772
–1779,
2003
22.
Vilsboll T, Krarup T, Deacon CF, Madsbad S, Holst JJ: Reduced postprandial concentrations of intact biologically active glucagon-like peptide 1 in type 2 diabetic patients.
Diabetes
50
:
609
–613,
2001
23.
Lugari R, Dei Cas A, Ugolotti D, Finardi L, Barilli AL, Ognibene C, Luciani A, Zandomeneghi R, Gnudi A: Evidence for early impairment of glucagon-like peptide 1-induced insulin secretion in human type 2 (non insulin-dependent) diabetes.
Horm Metab Res
34
:
150
–154,
2002
24.
Rask E, Olsson T, Soderberg S, Johnson O, Seckl J, Holst JJ, Ahren B: Impaired incretin response after a mixed meal is associated with insulin resistance in nondiabetic men.
Diabetes Care
24
:
1640
–1645,
2001
25.
Yip RG, Wolfe MM: GIP biology and fat metabolism.
Life Sci
66
:
91
–103,
2000
26.
Trumper A, Trumper K, Trusheim H, Arnold R, Goke B, Horsch D: Glucose-dependent insulinotropic polypeptide is a growth factor for beta (INS-1) cells by pleiotropic signaling.
Mol Endocrinol
15
:
1559
–1570,
2001
27.
Trumper A, Trumper K, Horsch D: Mechanisms of mitogenic and anti-apoptotic signaling by glucose-dependent insulinotropic polypeptide in beta(INS-1)-cells.
J Endocrinol
174
:
233
–246,
2002
28.
Gelling RW, Coy DH, Pederson RA, Wheeler MB, Hinke S, Kwan T, McIntosh CH: GIP(6–30amide) contains the high affinity binding region of GIP and is a potent inhibitor of GIP1–42 action in vitro.
Regul Pept
69
:
151
–154,
1997
29.
Tseng CC, Zhang XY, Wolfe MM: Effect of GIP and GLP-1 antagonists on insulin release in the rat.
Am J Physiol
276
:
E1049
−E1054, 1999
30.
Gault VA, O’Harte FP, Harriott P, Mooney MH, Green BD, Flatt PR: Effects of the novel (Pro[3])GIP antagonist and exendin(9-39)amide on GIP- and GLP-1-induced cyclic AMP generation, insulin secretion and postprandial insulin release in obese diabetic (ob/ob) mice: evidence that GIP is the major physiological incretin.
Diabetologia
46
:
222
–230,
2003
31.
Lewis JT, Dayanandan B, Habener JF, Kieffer TJ: Glucose-dependent insulinotropic polypeptide confers early phase insulin release to oral glucose in rats: demonstration by a receptor antagonist.
Endocrinology
141
:
3710
–3716,
2000
32.
Baggio L, Kieffer TJ, Drucker DJ: GLP-1 but not GIP regulates fasting and non-enteral glucose clearance in mice.
Endocrinology
141
:
3703
–3709,
2000
33.
Miyawaki K, Yamada Y, Yano H, Niwa H, Ban N, Ihara Y, Kubota A, Fujimoto S, Kajikawa M, Kuroe A, Tsuda K, Hashimoto H, Yamashita T, Jomori T, Tashiro F, Miyazaki J, Seino Y: Glucose intolerance caused by a defect in the entero-insular axis: a study in gastric inhibitory polypeptide receptor knockout mice.
Proc Natl Acad Sci U S A
96
:
14843
–14847,
1999
34.
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, Seino Y: Inhibition of gastric inhibitory polypeptide signaling prevents obesity.
Nat Med
8
:
738
–742,
2002
35.
Tseng CC, Boylan MO, Jarboe LA, Usdin TB, Wolfe MM: Chronic desensitization of the glucose-dependent insulinotropic polypeptide receptor in diabetic rats.
Am J Physiol
270
:
E661
−E666, 1996
36.
Lynn FC, Pamir N, Ng EH, McIntosh CH, Kieffer TJ, Pederson RA: Defective glucose-dependent insulinotropic polypeptide receptor expression in diabetic fatty Zucker rats.
Diabetes
50
:
1004
–1011,
2001
37.
O’Harte FP, Mooney MH, Kelly CM, Flatt PR: Improved glycaemic control in obese diabetic ob/ob mice using N-terminally modified gastric inhibitory polypeptide.
J Endocrinol
165
:
639
–648,
2000
38.
O’Harte FP, Gault VA, Parker JC, Harriott P, Mooney MH, Bailey CJ, Flatt PR: Improved stability, insulin-releasing activity and antidiabetic potential of two novel N-terminal analogues of gastric inhibitory polypeptide: N-acetyl-GIP and pGlu-GIP.
Diabetologia
45
:
1281
–1291,
2002
39.
Hinke SA, Gelling RW, Pederson RA, Manhart S, Nian C, Demuth HU, McIntosh CH: Dipeptidyl peptidase IV-resistant [D-Ala(2)]glucose-dependent insulinotropic polypeptide (GIP) improves glucose tolerance in normal and obese diabetic rats.
Diabetes
51
:
652
–661,
2002
40.
Jones IR, Owens DR, Luzio S, Hayes TM: Glucose dependent insulinotropic polypeptide (GIP) infused intravenously is insulinotropic in the fasting state in type 2 (non-insulin dependent) diabetes mellitus.
Horm Metab Res
21
:
23
–26,
1989
41.
Nauck M, Stockmann F, Ebert R, Creutzfeldt W: Reduced incretin effect in type 2 (non-insulin-dependent) diabetes.
Diabetologia
29
:
46
–52,
1986
42.
Krarup T, Saurbrey N, Moody AJ, Kuhl C, Madsbad S: Effect of porcine gastric inhibitory polypeptide on beta-cell function in type I and type II diabetes mellitus.
Metabolism
36
:
677
–682,
1987
43.
Jones IR, Owens DR, Vora J, Luzio SD, Hayes TM: A supplementary infusion of glucose-dependent insulinotropic polypeptide (GIP) with a meal does not significantly improve the beta cell response or glucose tolerance in type 2 diabetes mellitus.
Diabetes Res Clin Pract
7
:
263
–269,
1989
44.
Jones IR, Owens DR, Moody AJ, Luzio SD, Morris T, Hayes TM: The effects of glucose-dependent insulinotropic polypeptide infused at physiological concentrations in normal subjects and type 2 (non-insulin-dependent) diabetic patients on glucose tolerance and B-cell secretion.
Diabetologia
30
:
707
–712,
1987
45.
Elahi D, McAloon-Dyke M, Fukagawa NK, Meneilly GS, Sclater AL, Minaker KL, Habener JF, Andersen DK: The insulinotropic actions of glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (7–37) in normal and diabetic subjects.
Regul Pept
51
:
63
–74,
1994
46.
Meneilly GS, Bryer-Ash M, Elahi D: The effect of glyburide on beta-cell sensitivity to glucose-dependent insulinotropic polypeptide.
Diabetes Care
16
:
110
–114,
1993
47.
Vilsboll T, Krarup T, Madsbad S, Holst JJ: Defective amplification of the late phase insulin response to glucose by GIP in obese type II diabetic patients.
Diabetologia
45
:
1111
–1119,
2002
48.
Meier JJ, Hucking K, Holst JJ, Deacon CF, Schmiegel WH, Nauck MA: Reduced insulinotropic effect of gastric inhibitory polypeptide in first-degree relatives of patients with type 2 diabetes.
Diabetes
50
:
2497
–2504,
2001
49.
Mojsov S, Weir GC, Habener JF: Insulinotropin: Glucagon-like peptide I (7–37)co-encoded in the glucagon gene is a potent stimulator of insulin release in the perfused rat pancreas.
J Clin Invest
79
:
616
–619,
1987
50.
Kreymann B, Ghatei MA, Williams G, Bloom SR: Glucagon-like peptide-1 7–36: a physiological incretin in man.
Lancet
ii
:
1300
–1304,
1987
51.
Orskov C, Holst JJ, Nielsen OV: Effect of truncated glucagon-like peptide-1 [proglucagon-(78–107) amide] on endocrine secretion from pig pancreas, antrum, and nonantral stomach.
Endocrinology
123
:
2009
–2013,
1988
52.
Drucker DJ, Philippe J, Mojsov S, Chick WL, Habener JF: Glucagon-like peptide I stimulates insulin gene expression and increases cyclic AMP levels in a rat islet cell line.
Proc Natl Acad Sci U S A
84
:
3434
–3438,
1987
53.
Matsuyama T, Komatsu R, Namba M, Watanabe N, Itoh H, Tarui S: Glucagon-like peptide-1 (7–36 amide): a potent glucagonostatic and insulinotropic hormone.
Diabetes Res Clin Pract
5
:
281
–284,
1988
54.
Weir GC, Mojsov S, Hendrick GK, Habener JF: Glucagon-like peptide 1(7–37) actions on endocrine pancreas.
Diabetes
38
:
338
–342,
1989
55.
Wettergren A, Schjoldager B, Mortensen PE, Myhre J, Christiansen J, Holst JJ: Truncated GLP-1 (proglucagon 78–107-amide) inhibits gastric and pancreatic functions in man.
Dig Dis Sci
38
:
665
–673,
1993
56.
Turton MD, O’Shea D, Gunn I, Beak SA, Edwards CMB, Meeran K, Choi SJ, Taylor GM, Heath MM, Lambert PD, Wilding JPH, Smith DM, Ghatei MA, Herbert J, Bloom SR: A role for glucagon-like peptide-1 in the central regulation of feeding.
Nature
379
:
69
–72,
1996
57.
Meeran K, O’Shea D, Edwards CM, Turton MD, Heath MM, Gunn I, Abusnana S, Rossi M, Small CJ, Goldstone AP, Taylor GM, Sunter D, Steere J, Choi SJ, Ghatei MA, Bloom SR: Repeated intracerebroventricular administration of glucagon-like peptide-1-(7–36) amide or exendin-(9-39) alters body weight in the rat.
Endocrinology
140
:
244
–250,
1999
58.
Davis HR Jr, Mullins DE, Pines JM, Hoos LM, France CF, Compton DS, Graziano MP, Sybertz EJ, Strader CD, Van Heek M: Effect of chronic central administration of glucagon-like peptide-1 (7–36) amide on food consumption and body weight in normal and obese rats.
Obes Res
6
:
147
–156,
1998
59.
Szayna M, Doyle ME, Betkey JA, Holloway HW, Spencer RG, Greig NH, Egan JM: Exendin-4 decelerates food intake, weight gain, and fat deposition in Zucker rats.
Endocrinology
141
:
1936
–1941,
2000
60.
Larsen PJ, Fledelius C, Knudsen LB, Tang-Christensen M: Systemic administration of the long-acting GLP-1 derivative NN2211 induces lasting and reversible weight loss in both normal and obese rats.
Diabetes
50
:
2530
–2539,
2001
61.
Donahey JCK, Van Dijk G, Woods SC, Seeley RJ: Intraventricular GLP-1 reduces short- but not long-term food intake or body weight in lean and obese rats.
Brain Res
779
:
75
–83,
1998
62.
Thiele TE, Van Dijk G, Campfield LA, Smith FJ, Burn P, Woods SC, Bernstein H, Seeley RJ: Central infusion of GLP-1, but not leptin, produces conditioned taste aversion in rats.
Am J Physiol
272
:
R726
–R730,
1997
63.
Seeley RJ, Blake K, Rushing PA, Benoit S, Eng J, Woods SC, D’Alessio D: The role of CNS glucagon-like peptide-1 (7–36) amide receptors in mediating the visceral illness effects of lithium chloride.
J Neurosci
20
:
1616
–1621,
2000
64.
Rinaman L: A functional role for central glucagon-like peptide-1 receptors in lithium chloride-induced anorexia.
Am J Physiol
277
:
R1537
–R1540,
1999
65.
van Dijk G, Thiele TE: Glucagon-like peptide-1 (7–36) amide: a central regulator of satiety and interoceptive stress.
Neuropeptides
33
:
406
–414,
1999
66.
Kinzig KP, D’Alessio DA, Seeley RJ: The diverse roles of specific GLP-1 receptors in the control of food intake and the response to visceral illness.
J Neurosci
22
:
10470
–10476,
2002
67.
Holz GGt, Kuhtreiber WM, Habener JF: Pancreatic beta-cells are rendered glucose-competent by the insulinotropic hormone glucagon-like peptide-1(7–37).
Nature
361
:
362
–365,
1993
68.
Dachicourt N, Serradas P, Bailbe D, Kergoat M, Doare L, Portha B: Glucagon-like peptide-1(7–36)-amide confers glucose sensitivity to previously glucose-incompetent beta-cells in diabetic rats: in vivo and in vitro studies.
J Endocrinol
155
:
369
–376,
1997
69.
Xu G, Stoffers DA, Habener JF, Bonner-Weir S: Exendin-4 stimulates both β-cell replication and neogenesis, resulting in increased β-cell mass and improved glucose tolerance in diabetic rats.
Diabetes
48
:
2270
–2276,
1999
70.
Li Y, Hansotia T, Yusta B, Ris F, Halban PA, Drucker DJ: Glucagon-like peptide-1 receptor signaling modulates beta cell apoptosis.
J Biol Chem
278
:
471
–478,
2003
71.
Drucker DJ: Glucagon-like peptides: regulators of cell proliferation, differentiation and apoptosis.
Mol Endocrinol
17
:
161
–171,
2003
72.
Abraham EJ, Leech CA, Lin JC, Zulewski H, Habener JF: Insulinotropic hormone glucagon-like peptide-1 differentiation of human pancreatic islet-derived progenitor cells into insulin-producing cells.
Endocrinology
143
:
3152
–3161,
2002
73.
Hardikar AA, Wang XY, Williams LJ, Kwok J, Wong R, Yao M, Tuch BE: Functional maturation of fetal porcine beta-cells by glucagon-like peptide 1 and cholecystokinin.
Endocrinology
143
:
3505
–3514,
2002
74.
Movassat J, Beattie GM, Lopez AD, Hayek A: Exendin 4 up-regulates expression of PDX 1 and hastens differentiation and maturation of human fetal pancreatic cells.
J Clin Endocrinol Metab
87
:
4775
–4781,
2002
75.
Tourrel C, Bailbe D, Meile M-J, Kergoat M, Portha B: Glucagon-like peptide 1 and exendin-4 stimulate β-cell neogenesis in streptozotocin–treated newborn rats resulting in persistently improved glucose homeostasis at adult age.
Diabetes
50
:
1562
–1570,
2001
76.
Perfetti R, Zhou J, Doyle ME, Egan JM: Glucagon-like peptide-1 induces cell proliferation and pancreatic-duodenum homeobox-1 expression and increases endocrine cell mass in the pancreas of old, glucose-intolerant rats.
Endocrinology
141
:
4600
–4605,
2000
77.
Stoffers DA, Desai BM, DeLeon DD, Simmons RA: Neonatal exendin-4 prevents the development of diabetes in the intrauterine growth retarded rat.
Diabetes
52
:
734
–740,
2003
78.
Wang Q, Brubaker PL: Glucagon-like peptide-1 treatment delays the onset of diabetes in 8 week-old db/db mice.
Diabetologia
45
:
1263
–1273,
2002
79.
Tourrel C, Bailbe D, Lacorne M, Meile MJ, Kergoat M, Portha B: Persistent improvement of type 2 diabetes in the Goto-Kakizaki rat model by expansion of the β-cell mass during the prediabetic period with glucagon-like peptide 1 or exendin-4.
Diabetes
51
:
1443
–1452,
2002
80.
Edvell A, Lindstrom P: Initiation of increased pancreatic islet growth in young normoglycemic mice (Umea +/?).
Endocrinology
140
:
778
–783,
1999
81.
Rolin B, Larsen MO, Gotfredsen CF, Deacon CF, Carr RD, Wilken M, Knudsen LB: The long-acting GLP-1 derivative NN2211 ameliorates glycemia and increases beta-cell mass in diabetic mice.
Am J Physiol Endocrinol Metab
283
:
E745
−E752, 2002
82.
Kim J-G, Baggio LL, Bridon DP, Castaigne J-P, Robitaille MF, Jette L, Benquet C, Drucker DJ: Development and characterization of a GLP-1-albumin conjugate which retains the ability to activate the GLP-1 receptor in vivo.
Diabetes
52
:
751
–759,
2003
83.
Farilla L, Hui H, Bertolotto C, Kang E, Bulotta A, Di Mario U, Perfetti R: Glucagon-like peptide-1 promotes islet cell growth and inhibits apoptosis in Zucker diabetic rats.
Endocrinology
143
:
4397
–4408,
2002
84.
Hui H, Nourparvar A, Zhao X, Perfetti R: Glucagon-like peptide-1 inhibits apoptosis of insulin-secreting cells via a cyclic 5′-adenosine monophosphate-dependent protein kinase A- and a phosphatidylinositol 3-kinase-dependent pathway.
Endocrinology
144
:
1444
–1455,
2003
85.
Kolligs F, Fehmann H-C, Goke R, Goke B: Reduction of the incretin effect in rats by the glucagon-like peptide 1 receptor antagonist exendin (9-39) amide.
Diabetes
44
:
16
–19,
1995
86.
Wang Z, Wang RM, Owji AA, Smith DM, Ghatei MA, Bloom SR: Glucagon-like peptide 1 is a physiological incretin in rat.
J Clin Invest
95
:
417
–421,
1995
87.
D’alessio DA, Vogel R, Prigeon R, Laschansky E, Koerker D, Eng J, Ensinck JW: Elimination of the action of glucagon-like peptide 1 causes an impairment of glucose tolerance after nutrient ingestion by healthy baboons.
J Clin Invest
97
:
133
–138,
1996
88.
Imeryuz N, Yegen BC, Bozkurt A, Coskun T, Villanueva-Pennacarrillo ML, Ulusoy NB: Glucagon-like peptide-1 inhibits gastric emptying via vagal afferent-mediated central mechanisms.
Am J Physiol
273
:
G920
–G927,
1997
89.
Schirra J, Sturm K, Leicht P, Arnold R, Goke B, Katschinski M: Exendin(9-39)amide is an antagonist of glucagon-like peptide-1(7-36)amide in humans.
J Clin Invest
101
:
1421
–1430,
1998
90.
Edwards CM, Todd JF, Mahmoudi M, Wang Z, Wang RM, Ghatei MA, Bloom SR: Glucagon-like peptide 1 has a physiological role in the control of postprandial glucose in humans: studies with the antagonist exendin 9-39.
Diabetes
48
:
86
–93,
1999
91.
Scrocchi LA, Marshall BA, Cook SM, Brubaker PL, Drucker DJ: Glucose homeostasis in mice with disruption of GLP-1 receptor signaling.
Diabetes
47
:
632
–639,
1998
92.
Gros R, You X, Baggio LL, Kabir MG, Sadi A-M, Mungrue IN, Parker TG, Huang Q, Drucker DJ, Husain M: Cardiac function in mice lacking the glucagon-like peptide-1 receptor.
Endocrinology
144
:
2242
–2252,
2003
93.
Goldstone AP, Mercer JG, Gunn I, Moar KM, Edwards CMB, Rossi M, Howard JK, Rasheed S, Turton MD, Small C, Heath MM, O’Shea D, Steere J, Meeran K, Ghatei MA, Hoggard N, Bloom SR: Leptin interacts with glucagon-like peptide-1 neurons to reduce food intake and body weight in rodents.
FEBS Lett
415
:
134
–138,
1997
94.
Scrocchi LA, Brown TJ, Drucker DJ: Leptin sensitivity in non-obese GLP-1 receptor −/− mice.
Diabetes
46
:
2029
–2034,
1997
95.
Scrocchi LA, Hill ME, Saleh J, Perkins B, Drucker DJ: Elimination of GLP-1R signaling does not modify weight gain and islet adaptation in mice with combined disruption of leptin and GLP-1 action.
Diabetes
49
:
1552
–1560,
2000
96.
Scrocchi LA, Brown TJ, MacLusky N, Brubaker PL, Auerbach AB, Joyner AL, Drucker DJ: Glucose intolerance but normal satiety in mice with a null mutation in the glucagon-like peptide receptor gene.
Nat Med
2
:
1254
–1258,
1996
97.
Scrocchi LA, Drucker DJ: Effects of aging and a high fat diet on body weight and glucose control in GLP-1R−/− mice.
Endocrinology
139
:
3127
–3132,
1998
98.
Ling Z, Wu D, Zambre Y, Flamez D, Drucker DJ, Pipeleers DG, Schuit FC: Glucagon-like peptide 1 receptor signaling influences topography of islet cells in mice.
Virchows Arch
438
:
382
–387, 2001
99.
De Leon DD, Deng S, Madani R, Ahima RS, Drucker DJ, Stoffers DA: Role of endogenous glucagon-like peptide-1 in islet regeneration following partial pancreatectomy.
Diabetes
52
:
365
–371,
2003
100.
Wang Y, Perfetti R, Greig NH, Holloway HW, DeOre KA, Montrose-Rafizadeh C, Elahi D, Egan JM: Glucagon-like peptide-1 can reverse the age-related decline in glucose tolerance in rats.
J Clin Invest
99
:
2883
–2889,
1997
101.
Shen HQ, Roth MD, Peterson RG: The effect of glucose and glucagon-like peptide-1 stimulation on insulin release in the perfused pancreas in a non-insulin-dependent diabetes mellitus animal model.
Metabolism
47
:
1042
–1047,
1998
102.
Hargrove DM, Nardone NA, Persson LM, Parker JC, Stevenson RW: Glucose-dependent action of glucagon-like peptide-1(7–37) in vivo during short- or long-term administration.
Metabolism
44
:
1231
–1237,
1995
103.
Jia X, Elliott R, Kwok YN, Pederson RA, McIntosh CHS: Altered glucose dependence of glucagon-like peptide 1(7–36)-induced insulin secretion from the Zucker (fa/fa) rat pancreas.
Diabetes
44
:
495
–500,
1995
104.
Young AA, Gedulin BR, Bhavsar S, Bodkin N, Jodka C, Hansen B, Denaro M: Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta).
Diabetes
48
:
1026
–1034,
1999
105.
Greig NH, Holloway HW, De Ore KA, Jani D, Wang Y, Zhou J, Garant MJ, Egan JM: Once daily injection of exendin-4 to diabetic mice achieves long-term beneficial effects on blood glucose concentrations.
Diabetologia
42
:
45
–50,
1999
106.
Ribel U, Larsen M, Rolin B, Carr R, Wilken M, Sturis J, Westergaard L, Deacon C, Knudsen L: NN2211: a long-acting glucagon-like peptide-1 derivative with anti-diabetic effects in glucose-intolerant pigs.
Eur J Pharmacol
451
:
217
–225,
2002
107.
Hui H, Farilla L, Merkel P, Perfetti R: The short half-life of glucagon-like peptide-1 in plasma does not reflect its long-lasting beneficial effects.
Eur J Endocrinol
146
:
863
–869,
2002
108.
Qualmann C, Nauck MA, Holst JJ, Orskov C, Creutzfeldt W: Insulinotropic actions of intravenous glucagon-like peptide-1 (GLP-1) [7–36 amide] in the fasting state in healthy subjects.
Acta Diabetol
32
:
13
–16,
1995
109.
Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W: 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
:
301
–307,
1993
110.
Schjoldager BTG, Mortensen PE, Christiansen J, Orskov C, Holst JJ: GLP-1 (glucagon-like peptide 1) and truncated GLP-1, fragments of human proglucagon, inhibit gastric acid secretion in humans.
Dig Dis Sci
34
:
703
–708,
1989
111.
O’Halloran DJ, Nikou GC, Kreymann B, Ghatei MA, Bloom SR: Glucagon-like peptide-1 (7–36)-NH2: a physiological inhibitor of gastric acid secretion in man.
J Endocrinol
126
:
169
–173,
1990
112.
Nauck MA, Niedereichholz U, Ettler R, Holst JJ, Orskov C, Ritzel R, Schmiegel WH: Glucagon-like peptide 1 inhibition of gastric emptying outweighs its insulinotropic effects in healthy humans.
Am J Physiol
273
:
E981
–E988,
1997
113.
Nauck MA: Is glucagon-like peptide 1 an incretin hormone?
Diabetologia
42
:
373
–379,
1999
114.
Gutniak M, Orskov C, Holst JJ, Ahren B, Efendic S: Antidiabetogenic effect of glucagon-like peptide-1 (7–36)amide in normal subjects and patients with diabetes mellitus.
N Engl J Med
326
:
1316
–1322,
1992
115.
Dupre J, Behme MT, Hramiak IM, McFarlane P, Williamson MP, Zabel P, McDonald TJ: Glucagon-like peptide 1 reduces postprandial glycemic excursions in IDDM.
Diabetes
44
:
626
–630,
1995
116.
Creutzfeld WO, Kleine N, Willms B, Orskov C, Holst JJ, Nauck MA: Glucagonostatic actions and reduction of fasting hyperglycemia by exogenous glucagon-like peptide I(7–36) amide in type 1 diabetic patients.
Diabetes Care
19
:
580
–586,
1996
117.
Byrne MM, Gliem K, Wank U, Arnold R, Katschinski M, Polonsky KS, Goke B: Glucagon-like peptide 1 improves the ability of the β-cell to sense and respond to glucose in subjects with impaired glucose tolerance.
Diabetes
47
:
1259
–1265,
1998
118.
Ritzel R, Schulte M, Porksen N, Nauck MS, Holst JJ, Juhl C, Marz W, Schmitz O, Schmiegel WH, Nauck MA: Glucagon-like peptide 1 increases secretory burst mass of pulsatile insulin secretion in patients with type 2 diabetes and impaired glucose tolerance.
Diabetes
50
:
776
–784,
2001
119.
Kjems LL, Holst JJ, Volund A, Madsbad S: The influence of GLP-1 on glucose-stimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects.
Diabetes
52
:
380
–386,
2003
120.
D’Alessio DA, Kahn SE, Leusner CR, Ensinck JW: Glucagon-like peptide 1 enhances glucose tolerance both by stimulation of insulin release and by increasing insulin-independent glucose disposal.
J Clin Invest
93
:
2263
–2266,
1994
121.
D’Alessio DA, Prigeon RL, Ensinck JW: Enteral enhancement of glucose disposition by both insulin-dependent and insulin-independent processes: a physiological role of glucagon-like peptide I.
Diabetes
44
:
1433
–1437,
1995
122.
Toft-Nielson M, Madsbad S, Holst JJ: The effect of glucagon-like peptide 1 (GLP-1) on glucose elimination in healthy subjects depends on the pancreatic glucoregulatory hormones.
Diabetes
45
:
552
–556,
1996
123.
Ryan AS, Egan JM, Habener JF, Elahi D: Insulinotropic hormone glucagon-like peptide-1-(7–37) appears not to augment insulin-mediated glucose uptake in young men during euglycemia.
J Clin Endocrinol Metab
83
:
2399
–2404,
1998
124.
Vella A, Shah P, Basu R, Basu A, Holst JJ, Rizza RA: Effect of glucagon-like peptide 1(7–36) amide on glucose effectiveness and insulin action in people with type 2 diabetes.
Diabetes
49
:
611
–617,
2000
125.
Zander M, Madsbad S, Madsen JL, Holst JJ: Effect of 6-week course of glucagon-like peptide 1 on glycaemic control, insulin sensitivity, and beta-cell function in type 2 diabetes: a parallel-group study.
Lancet
359
:
824
–830,
2002
126.
Nauck MA, Sauerwald A, Ritzel R, Holst JJ, Schmiegel W: Influence of glucagon-like peptide 1 on fasting glycemia in type 2 diabetic patients treated with insulin after sulfonylurea secondary failure.
Diabetes Care
21
:
1925
–1931,
1998
127.
Gutniak MK, Juntti-Berggren L, Hellstrom PM, Guenifi A, Holst JJ, Efendic S: Glucagon-like peptide I enhances the insulinotropic effect of glibenclamide in NIDDM patients and in the perfused rat pancreas.
Diabetes Care
19
:
857
–863,
1996
128.
Zander M, Taskiran M, Toft-Nielsen MB, Madsbad S, Holst JJ: Additive glucose-lowering effects of glucagon-like peptide-1 and metformin in type 2 diabetes.
Diabetes Care
24
:
720
–725,
2001
129.
Miholic J, Orskov C, Holst JJ, Kotzerke J, Meyer HJ: Emptying of the gastric substitute, glucagon-like peptide-1 (GLP-1), and reactive hypoglycemia after total gastrectomy.
Dig Dis Sci
36
:
1361
–1370,
1991
130.
Edwards CM, Todd JF, Ghatei MA, Bloom SR: Subcutaneous glucagon-like peptide-1 (7–36) amide is insulinotropic and can cause hypoglycaemia in fasted healthy subjects.
Clin Sci (Colch)
95
:
719
–724,
1998
131.
Nauck MA, Heimesaat MM, Behle K, Holst JJ, Nauck MS, Ritzel R, Hufner M, Schmiegel WH: Effects of glucagon-like peptide 1 on counterregulatory hormone responses, cognitive functions, and insulin secretion during hyperinsulinemic, stepped hypoglycemic clamp experiments in healthy volunteers.
J Clin Endocrinol Metab
87
:
1239
–1246,
2002
132.
Flint A, Raben A, Astrup A, Holst JJ: Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans.
J Clin Invest
101
:
515
–520,
1998
133.
Gutzwiller JP, Drewe J, Goke B, Schmidt H, Rohrer B, Lareida J, Beglinger C: Glucagon-like peptide-1 promotes satiety and reduces food intake in patients with diabetes mellitus type 2.
Am J Physiol
276
:
R1541
−R1544, 1999
134.
Toft-Nielsen MB, Madsbad S, Holst JJ: Continuous subcutaneous infusion of glucagon-like peptide 1 lowers plasma glucose and reduces appetite in type 2 diabetic patients.
Diabetes Care
22
:
1137
–1143,
1999
135.
Naslund E, Barkeling B, King N, Gutniak M, Blundell JE, Holst JJ, Rossner S, Hellstrom PM: Energy intake and appetite are suppressed by glucagon-like peptide-1 (GLP-1) in obese men.
Int J Obes Relat Metab Disord
23
:
304
–311,
1999
136.
Flint A, Raben A, Ersboll AK, Holst JJ, Astrup A: The effect of physiological levels of glucagon-like peptide-1 on appetite, gastric emptying, energy and substrate metabolism in obesity.
Int J Obes Relat Metab Disord
25
:
781
–792,
2001
137.
Verdich C, Flint A, Gutzwiller JP, Naslund E, Beglinger C, Hellstrom PM, Long SJ, Morgan LM, Holst JJ, Astrup A: A meta-analysis of the effect of glucagon-like peptide-1 (7–36) amide on ad libitum energy intake in humans.
J Clin Endocrinol Metab
86
:
4382
–4389,
2001
138.
Nauck MA, Wollschlager D, Werner J, Holst JJ, Orskov C, Creutzfeldt W, Willms B: Effects of subcutaneous glucagon-like peptide 1 (GLP-1[7–36 amide]) in patients with NIDDM.
Diabetologia
39
:
1546
–1553,
1996
139.
Juntti-Berggren L, Pigon J, Karpe F, Hamsten A, Gutniak M, Vignati L, Efendic S: The antidiabetogenic effect of GLP-1 is maintained during a 7-day treatment period and improves diabetic dyslipoproteinemia in NIDDM patients.
Diabetes Care
19
:
1200
–1206,
1996
140.
Gutniak MK, Linde B, Holst JJ, Efendic S: Subcutaneous injection of the incretin hormone glucagon-like peptide 1 abolishes postprandial glycemia in NIDDM.
Diabetes Care
17
:
1039
–1044,
1994
141.
Ritzel R, Orskov C, Holst JJ, Nauck MA: Pharmacokinetic, insulinotropic, and glucagonostatic properties of GLP-1 [7–36 amide] after subcutaneous injection in healthy volunteers: dose-response relationships.
Diabetologia
38
:
720
–725,
1995
142.
Larsen J, Hylleberg B, Ng K, Damsbo P: Glucagon-like peptide-1 infusion must be maintained for 24 h/day to obtain acceptable glycemia in type 2 diabetic patients who are poorly controlled on sulphonylurea treatment.
Diabetes Care
24
:
1416
–1421,
2001
143.
Rachman J, Barrow BA, Levy JC, Turner RC: Near normalization of diurnal glucose concentrations by continuous administration of glucagon-like peptide 1 (GLP-1) in subjects with NIDDM.
Diabetologia
40
:
205
–211,
1997
144.
Todd JF, Wilding JP, Edwards CM, Ghatei MA, Bloom SR: Glucagon-like peptide-1 (GLP-1): a trial of treatment in non-insulin-dependent diabetes mellitus.
Eur J Clin Invest
27
:
533
–536,
1997
145.
Deacon CF, Johnsen AH, Holst JJ: Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo.
J Clin Endocrinol Metab
80
:
952
–957,
1995
146.
Eng J, Kleinman WA, Singh L, Singh G, Raufman JP: Isolation and characterization of exendin 4, an exendin 3 analogue from Heloderma suspectum venom.
J Biol Chem
267
:
7402
–7405,
1992
147.
Chen YE, Drucker DJ: Tissue-specific expression of unique mRNAs that encode proglucagon-derived peptides or exendin 4 in the lizard.
J Biol Chem
272
:
4108
–4115,
1997
148.
Edwards CM, Stanley SA, Davis R, Brynes AE, Frost GS, Seal LJ, Ghatei MA, Bloom SR: Exendin-4 reduces fasting and postprandial glucose and decreases energy intake in healthy volunteers.
Am J Physiol
281
:
E155
−E161, 2001
149.
Egan JM, Clocquet AR, Elahi D: The insulinotropic effect of acute exendin-4 administered to humans: comparison of nondiabetic state to type 2 diabetes.
J Clin Endocrinol Metab
87
:
1282
–1290,
2002
150.
Egan JM, Meneilly GS, Elahi D: Effects of one month bolus subcutaneous administration of exendin-4 in type 2 diabetes.
Am J Physiol Endocrinol Metab
34
:
E1072
–E1079,
2003
151.
Nielsen LL, Baron AD: Pharmacology of exenatide (synthetic exendin-4) for the treatment of type 2 diabetes.
Curr Opin Investig Drugs
4
:
401
–405,
2003
152.
Fineman MS, Bicsak TA, Shen LZ, Taylor K, Gaines E, Varns A, Kim DW, Baron AD: Effect on glycemic control of synthetic exendin-4 (AC2993) additive to existing metformin and/or sulfonylurea treatment in patients with type 2 diabetes.
Diabetes Care
26
:
2370
–2377,
2003
153.
Agerso H, Jensen LB, Elbrond B, Rolan P, Zdravkovic M: The pharmacokinetics, pharmacodynamics, safety and tolerability of NN2211, a new long-acting GLP-1 derivative, in healthy men.
Diabetologia
45
:
195
–202,
2002
154.
Juhl CB, Hollingdal M, Sturis J, Jakobsen G, Agerso H, Veldhuis J, Porksen N, Schmitz O: Bedtime administration of NN2211, a long-acting GLP-1 derivative, substantially reduces fasting and postprandial glycemia in type 2 diabetes.
Diabetes
51
:
424
–429,
2002
155.
Drucker DJ: Therapeutic potential of dipeptidyl peptidase IV inhibitors for the treatment of type 2 diabetes.
Expert Opin Investig Drugs
12
:
87
–100,
2003
156.
Holst JJ, Deacon CF: Inhibition of the activity of dipeptidyl-peptidase IV as a treatment for type 2 diabetes.
Diabetes
47
:
1663
–1670,
1998
157.
Durinx C, Lambeir AM, Bosmans E, Falmagne JB, Berghmans R, Haemers A, Scharpe S, De Meester I: Molecular characterization of dipeptidyl peptidase activity in serum: soluble CD26/dipeptidyl peptidase IV is responsible for the release of X-Pro dipeptides.
Eur J Biochem
267
:
5608
–5613,
2000
158.
Marguet D, Baggio L, Kobayashi T, Bernard AM, Pierres M, Nielsen PF, Ribel U, Watanabe T, Drucker DJ, Wagtmann N: Enhanced insulin secretion and improved glucose tolerance in mice lacking CD26.
Proc Natl Acad Sci U S A
97
:
6874
–6879,
2000
159.
Nagakura T, Yasuda N, Yamazaki K, Ikuta H, Yoshikawa S, Asano O, Tanaka I: Improved glucose tolerance via enhanced glucose-dependent insulin secretion in dipeptidyl peptidase IV-deficient Fischer rats.
Biochem Biophys Res Commun
284
:
501
–506,
2001
160.
Conarello SL, Li Z, Ronan J, Roy RS, Zhu L, Jiang G, Liu F, Woods J, Zycband E, Moller DE, Thornberry NA, Zhang BB: Mice lacking dipeptidyl peptidase IV are protected against obesity and insulin resistance.
Proc Natl Acad Sci U S A
100
:
6825
–6830,
2003
161.
Ahren B, Holst JJ, Martensson H, Balkan B: Improved glucose tolerance and insulin secretion by inhibition of dipeptidyl peptidase IV in mice.
Eur J Pharmacol
404
:
239
–245,
2000
162.
Balkan B, Kwasnik L, Miserendino R, Holst JJ, Li X: Inhibition of dipeptidyl peptidase IV with NVP-DPP728 increases plasma GLP-1 (7–36 amide) concentrations and improves oral glucose tolerance in obese Zucker rats.
Diabetologia
42
:
1324
–1331,
1999
163.
Sudre B, Broqua P, White RB, Ashworth D, Evans DM, Haigh R, Junien JL, Aubert ML: Chronic inhibition of circulating dipeptidyl peptidase IV by FE 999011 delays the occurrence of diabetes in male Zucker diabetic fatty rats.
Diabetes
51
:
1461
–1469,
2002
164.
Pospisilik JA, Stafford SG, Demuth HU, Brownsey R, Parkhouse W, Finegood DT, McIntosh CH, Pederson RA: Long-term treatment with the dipeptidyl peptidase IV inhibitor P32/98 causes sustained improvements in glucose tolerance, insulin sensitivity, hyperinsulinemia, and β-cell glucose responsiveness in VDF (fa/fa) Zucker rats.
Diabetes
51
:
943
–950,
2002
165.
Pospisilik JA, Stafford SG, Demuth HU, McIntosh CH, Pederson RA: Long-term treatment with dipeptidyl peptidase IV inhibitor improves hepatic and peripheral insulin sensitivity in the VDF Zucker rat: a euglycemic-hyperinsulinemic clamp study.
Diabetes
51
:
2677
–2683,
2002
166.
Pospisilik JA, Martin J, Doty T, Ehses JA, Pamir N, Lynn FC, Piteau S, Demuth HU, McIntosh CH, Pederson RA: Dipeptidyl peptidase IV inhibitor treatment stimulates β-cell survival and islet neogenesis in streptozotocin-induced diabetic rats.
Diabetes
52
:
741
–750,
2003
167.
Hoffmann T, Glund K, McIntosh CHS, Pederson RA, Hanefeld M, Rosenkranz B, Demuth H-U: DPPIV inhibition as treatment of type II diabetes. In
Cell-Surface Aminopeptidases: Basic and Clinical Aspects
. Mitzutani S, Turner AJ, Ino K, Nomura S, Eds. Tokyo, Elsevier Science,
2001
, p.
381
–387
168.
Ahren B, Simonsson E, Larsson H, Landin-Olsson M, Torgeirsson H, Jansson PA, Sandqvist M, Bavenholm P, Efendic S, Eriksson JW, Dickinson S, Holmes D: Inhibition of dipeptidyl peptidase IV improves metabolic control over a 4-week study period in type 2 diabetes.
Diabetes Care
25
:
869
–875,
2002
169.
Mentlein R: Dipeptidyl-peptidase IV (CD26): role in the inactivation of regulatory peptides.
Regul Pept
85
:
9
–24,
1999
170.
Gorrell MD, Gysbers V, McCaughan GW: CD26: a multifunctional integral membrane and secreted protein of activated lymphocytes.
Scand J Immunol
54
:
249
–264,
2001
171.
Yan S, Marguet D, Dobers J, Reutter W, Fan H: Deficiency of CD26 results in a change of cytokine and immunoglobulin secretion after stimulation by pokeweed mitogen.
Eur J Immunol
33
:
1519
–1527,
2003
172.
Casadevall N, Nataf J, Viron B, Kolta A, Kiladjian JJ, Martin-Dupont P, Michaud P, Papo T, Ugo V, Teyssandier I, Varet B, Mayeux P: Pure red-cell aplasia and antierythropoietin antibodies in patients treated with recombinant erythropoietin.
N Engl J Med
346
:
469
–475,
2002

D.J.D. is on the scientific advisory board for Amylin Pharmaceuticals, Conjuchem, and Transition Therapeutics and a consultant for Merck, Forest Labs, Bristol Myers Squibb, Triad Pharmaceuticals, Aventis, Novartis, Amylin Pharmaceuticals, and Conjuchem.

A table elsewhere in this issue shows conventional and Système International (SI) units and conversion factors for many substances.