81
Views
4
CrossRef citations to date
0
Altmetric
Review

Incretin-based therapies: new treatments for type 2 diabetes in the new millennium

, , &
Pages 683-698 | Published online: 20 Aug 2009

Abstract

The advent of ‘incretin-based therapies’ – GLP-1 agonists and dipeptidyl-peptidase-4 inhibitors – which result in improvements in glycemic control comparable to those with existing oral hypoglycemic agents, and potentially improve cardiovascular and pancreatic β-cell function, represents a major therapeutic advance in the management of type 2 diabetes. Gastrointestinal adverse effects occur commonly with GLP-1 agonists, and rarely with DPP-4 inhibitors, but are dose-dependent and usually transient. The low risk of hypoglycemia, and beneficial or neutral effects on body weight, render GLP-1 agonists and DPP-4 inhibitors suitable alternatives to insulin secretagogues and insulin in overweight and elderly patients. Incretin-based therapies also improve quality of life in patients with type 2 diabetes, and may be cost-effective in the long term.

Introduction

Type 2 diabetes mellitus (T2DM) is well-recognized as a major problem worldwide, with substantial impacts on morbidity, mortality, quality of life, and health care costs. Because current treatment regimens for T2DM do not effectively target the fundamental defects in glucose-mediated insulin secretion and beta-cell loss, an increasing proportion of T2DM patients progress to requiring insulin. Accordingly, the recent advent of so-called ‘incretin-based therapies’, the incretin hormones being glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP), which have the potential to address these defects, represents a major paradigm shift in management.

The prevalence of T2DM has been rising dramatically, reflecting aging populations and the increasing prevalence of obesity, so that by 2025 an estimated 350 million people worldwide will be affected.Citation1 T2DM is characterized by peripheral insulin resistance, impaired regulation of hepatic glucose production, and declining β-cell function. The last is evident initially as impaired first-phase insulin secretion in response to oral, or intravenous, glucose and progresses at a variable rate to absolute insulin deficiency, reflecting β-cell failure, which is present in a substantial number of T2DM patients at diagnosis. This defect, rather than insulin resistance, may be a primary abnormality in T2DM, particularly in Asian populations, in which postprandial hyperglycemia is often apparent before elevation of fasting plasma glucose.Citation1 The development, and progression, of the macrovascular (cardiovascular, cerebrovascular and peripheral vascular disease) and, particularly, microvascular (nephropathy, neuropathy, retinopathy) complications of diabetes can be reduced substantially by optimizing glycemic control.Citation2,Citation3 However, many patients fail to achieve the target glycated hemoglobin (HbA1c) of ≤7% suggested by the American Diabetes Association and European Association for the Study of Diabetes, despite use of maximal doses of oral hypoglycemic agents (OHAs) in combination.Citation4 Moreover, concerns have recently been raised over the risk of malignancy, particularly breast cancer, with the use of sulfonylureas and insulin (especially glargine).Citation5

Current therapy for type 2 diabetes

The majority of OHAs in common use are insulin sensitizers and/or insulin secretagogues. Older patients, in particular, are vulnerable to impaired awareness of hypoglycemia with consequent neuroglycopenia and adverse cardiovascular effects, dictating the need for particular caution with therapies that increase the risk of hypoglycemia. A history of severe hypoglycemia in older T2DM patients has been associated with a greater risk of dementia, which increases with the number of hypoglycemic episodes.Citation6 There has been considerable interest in the outcomes of the recent ADVANCECitation7 and ACCORDCitation8 trials, which failed to show any cardiovascular benefit of lowering HbA1c to below 7% in patients recently diagnosed with T2DM. Significantly, in the ACCORD trial, combination therapy using high doses of thiazolidinediones (TZD), sulfonylureas (SU), metformin, and insulin, was associated with an increase in cardiovascular and all-cause mortality, possibly because of hypoglycemia. Metformin and TZDs decrease insulin resistance and hepatic glucose output, but are contraindicated in patients with significant renal and/or cardiac dysfunction, both of which occur frequently in T2DM.

There is now compelling evidence that postprandial hyperglycemia (PPG) is a dominant contributor to overall glycemia, particularly when HbA1c is below 8.5%,Citation9 and that PPG increases cardiovascular risk.Citation10,Citation11 However, no current OHA specifically targets PPG, with the possible exception of α-glucosidase inhibitors such as acarbose, which decrease the rate of glucose absorption, but whose use is limited by a high prevalence of gastrointestinal adverse effects (GI AEs); and the meglitinides repaglinide and nateglinide, which are insulin secretagogues (though the risk of hypoglycemia is lower than that with sulfonylureas). Furthermore, higher doses, and combinations, of OHA are progressively required in the majority of patients.Citation4 The reasons for this are diverse and include difficulty in compliance with lifestyle modifications (diet, exercise) and medications; but perhaps, most importantly, the failure of these OHAs to target several underlying pathophysiologic mechanisms of T2DM, particularly inappropriately high glucagon secretion, impaired first-phase insulin secretion, and progressive β-cell failure. Hence, the availability of drugs that stimulate insulin secretion in a physiological fashion, ie, at elevated glucose levels but not during euglycemia, are weight-neutral or promote weight loss, and have beneficial effects to preserve β-cell function, would represent a major asset in the management of T2DM. These properties are evident with recently developed medications that target the incretin system, namely the glucagon-like peptide-1 agonists and the dipeptidyl peptidase-IV inhibitors, which form the major focus of the review.

The incretin effect – implications for pathophysiology and management of T2DM

Subsequent to the discovery of insulin, La Barre reported in 1932 that a substance produced in the upper intestine had the capacity to cause hypoglycemia without stimulating exocrine pancreatic secretion, which he termed ‘incrétine’ (incretin), and envisaged its use for the treatment of diabetes.Citation12 However, more than three decades would elapse before the ‘incretin effect’, ie, greater insulin release in response to an oral glucose load than an isoglycemic intravenous infusion, was demonstrated in 1964 by concurrent reports from both sides of the Atlantic.Citation13,Citation14 The known incretin hormones, glucose-dependent insulinotropic polypeptide(GIP), a 42-amino acid hormone produced by K-cells in the proximal small intestine, and glucagon-like peptide-1 (GLP-1), a 30-amino acid peptide synthesized and secreted by L-cells in the ileum and colon, were not isolated until 1970Citation15 and 1985,Citation16 respectively.

In healthy people, up to 70% of post-glucose insulin secretion is mediated by incretins.Citation17 However, in T2DM patients, the insulin response to oral glucose is blunted in comparison to non-diabetic control subjects,Citation18 suggesting impairment of the incretin effect. The actions of incretins on the defects in glucose metabolism, pancreatic function and energy intake in T2DM patients are shown in . In T2DM patients during hyperglycemic clamp studies, infusion of GLP-1, but not GIP, stimulates insulin secretion,Citation19 establishing that the insulinotropic effect of GLP-1 is relatively well-preserved in T2DM, despite possibly lower levels, when compared to non-diabetic subjects.Citation20 On the other hand, GIP levels are essentially normal in T2DM but GIP-stimulated second-phase insulin secretion is markedly diminshedCitation21 (although it has recently been reported that reversal of poor glycemic control in T2DM improves the insulin response to both GIP and GLP-1).Citation21 Hence, the development of incretin-based therapies for T2DM has hitherto focused on GLP-1, rather than GIP.

Table 1 Actions of incretins on the defects in glucose handling, βcell function, and energy intake in type 2 diabetes patients

In both T2DM and healthy humans, circulating levels of intact GLP-1 decrease rapidly (half-life ∼2 minutes) due to inactivation by the enzyme, dipeptidyl peptide-4 (DPP-4), such that biologically active GLP-1 represents only 10% to 20% of total plasma levels.Citation22 Among the truncated forms of GLP-1, GLP-1 (7–36) was found to be more potent compared to other metabolites, such as the 9–36 form.Citation23 The secretion of GLP-1 depends on the rate of delivery of carbohydrate to the small intestine, and is thus influenced by the rate of gastric emptying.Citation24 GLP-1 activates specific G-protein-coupled receptors on β-cells to stimulate insulin secretion at a threshold glucose concentration of 3.7 mmol/L, and also reduces glucose-dependent glucagon secretion, possibly in a paracrine fashion by insulin, or via GLP-1 receptors on α-cells,Citation25 although the precise mechanism for glucagon suppression is unknown. The role of endogenous GLP-1 in glucoregulation was established by animal and human studies – GLP-1-receptor knockout mice display impaired glucose tolerance and glucose-stimulated insulin secretion.Citation26 Administration of the GLP-1 receptor antagonist, exendin (9–39), inhibited postprandial insulin secretion and concomitantly increased plasma glucose in mice,Citation27 and accelerated gastric emptying in rats.Citation28 Similarly, treatment of non-diabetic human subjects with exendin 9–39 results in defective glucose-stimulated insulin secretion, reduced glucose uptake, increased glucagon levels, and, possibly, accelerated gastric emptying.Citation29Citation31 Slowing of gastric emptying may be as, if not more, important for postprandial glycemic control than stimulating insulin, given that variations in gastric emptying account for about 35% of the variance in the glycemic response to 75 g oral glucose loads in healthy subjects,Citation32 as well as T2DM patients.Citation33 The relationship of glycemia with gastric emptying is also evident for the ingestion of solid carbohydrate-containing food.Citation34,Citation35

Exogenous GLP-1 has diverse effects on pancreatic endocrine function and gut motility. Importantly, it has no significant insulinotropic effects below a glucose threshold ∼4 mmol/L,Citation36 and the counterregulatory release of glucagon in response to hypoglycemia is preserved, even when GLP-1 is administered. Exogenous GLP-1 also stimulates insulin gene transcription and other steps in insulin biosynthesis,Citation37 and, in in vitro studies, has a trophic effect on β-cells by increasing proliferation and neogenesis, and inhibiting apoptosis,Citation38,Citation39 which has the potential to retard or reverse β-cell failure, the fundamental defect in T2DM. In rats, GLP-1 infusion has been reported to increase insulin secretion in response to feeding, and reduce glucagon release and postprandial glycemia.Citation40 Exogenous GLP-1 has also been shown to inhibit energy intake and gastric emptying in rats.Citation41 In humans with T2DM, overnight GLP-1 infusions lowered blood glucose by restoring first-phase insulin secretion and improving β-cell function to levels that were comparable to non-diabetic subjects.Citation42 Preprandial boluses of GLP-1 were also effective in improving postprandial glucose levels.Citation43 Administration of exogenous GLP-1, resulting in supraphysiological levels in peripheral blood, stimulated glucose-mediated insulin secretion while suppressing glucagon and gastric emptying in T2DM patients with poor glycemic control.Citation44 In contrast, the insulin response to a carbohydrate-containing meal in healthy subjects was found to be decreased by exogenous GLP-1 in healthy subjects, because of slower gastric emptying causing lower post-load blood glucose levels.Citation45 In elderly T2DM patients whose OHA had been discontinued, continuous GLP-1 infusion for 12 weeks enhanced postprandial insulin secretion, and insulin-mediated glucose disposal, while maintaining satisfactory glycemic control (HbA1c ~7%).Citation46 In another study,Citation47 sustained improvements of insulin secretory capacity and insulin sensitivity were accompanied by reductions in HbA1c (∼1%) and weight (∼2 kg) with supraphysiological (60 to 70 pmol/L) levels of GLP-1 maintained by 6 weeks of subcutaneous infusion. Accordingly, the mechanisms by which exogenous GLP-1 reduces postprandial glycemia in both healthy subjects and T2DM patients include delaying the entry of nutrients into the intestine, as well as increasing the insulin, and suppressing the glucagon, response to carbohydrate.

In addition to glycemic control, GLP-1 may have beneficial cardiovascular effects. Continuous GLP-1 infusion increases myocardial insulin sensitivity and glucose uptake, leading to improved left ventricular (LV) contractility, as evidenced by a significant rise in LV ejection fraction from ∼30% to 40% in both non-diabetic and diabetic patients after acute myocardial infarction,Citation48 and in patients with chronic heart failure.Citation49 Perioperative (12 hours before and 48 hours after) GLP-1 infusion in T2DM patients who underwent coronary artery bypass grafting not only improved glycemic control, but also reduced hospital mortality and requirements for inotropic agents.Citation50 A shorter, purely postoperative (12 hours after), period of intravenous GLP-1 also improved glycemic control and reduced the need for inotropic support in a group of T2DM patients after bypass surgery.Citation51 Endothelial function is improved by GLP-1, which induces vasodilation in coronary artery grafts of patients with ischemic heart disease,Citation52 and increases flow-mediated dilatation in brachial arteries of healthy humans.Citation53 GLP-1 is thought to act by both GLP-1 receptor-dependent and-independent pathways, via its metabolites GLP-1 (7–36) and GLP-1 (9–36) respectively.Citation54

The encouraging outcomes of studies of GLP-1 administration in T2DM have led to the development of drugs that enhance GLP-1 activity: synthetic long-acting agonists (such as exenatide and liraglutide) that are resistant to degradation by DPP-IV and could thus be administered less frequently, and inhibitors of DPP-IV (such as sitagliptin and vildagliptin) that increase endogenous GLP-1 levels. GLP-1 agonists and DPP-IV inhibitors are discussed in the following sections.

GLP-1 agonists

Exenatide (synthetic exendin-4) and exenatide LAR

Exenatide (Byetta®; Eli Lilly and Co.), an injectable GLP-1 receptor agonist, was approved by the US Food and Drug Administration (FDA) in 2005 as an adjunct therapy for T2DM patients who fail to achieve satisfactory glycemic control on metformin in combination with SU and/or TZD, and is currently available in the United States, the European Union, Australia and several Asian countries. It was developed from exendin-4, found in the saliva of the gila monster lizard,Citation55 has approximately 50% homology with human GLP-1, but binds more avidly to GLP-1 receptors, and is resistant to degradation by DPP-4, thus prolonging its duration of effect.Citation56 Exenatide is administered twice daily in a dose of 5 or 10 μg by subcutaneous injection. Reductions in fasting and postprandial glucose are mediated by the combined effects of glucose-dependent insulin secretion, suppression of glucagon release, and, in the case of postprandial glycemia, perhaps predominantly by the slowing of gastric emptying and the resulting delayed entry of nutrients into the small intestine.Citation57 Acutely, postprandial glucose levels are decreased by up to 75% primarily as a result of the substantial, and dose-dependent, slowing of gastric emptying,Citation58 associated with a reduction in plasma insulin levels in absolute terms. As a result of delayed gastric emptying (the magnitude of which differs substantially between individuals), oral medication such as contraceptive pills and antibiotics, whose efficacy depends on reaching a threshold concentration, are best taken at least an hour before exenatide.Citation59 As exenatide is primarily cleared by the kidneys, its use is contraindicated in patients with end-stage renal disease.Citation59

Exenatide has been studied as monotherapy, as well as in combination with OHAs. In drug-naïve T2DM patients, a dose of 5 μg twice daily (bid), for 4 weeks, followed by 10 μg bid for 26 weeks, reduced mean HbA 1c (∼1%), fasting glucose (∼1 mmol/L,) and weight (∼3 kg), compared to non-significant reductions with placebo.Citation60 Three large, randomized, placebo-controlled, 30-week trials compared the efficacy and tolerability of exenatide 5 or 10 μg bid added to metformin,Citation61 sulfonylureas (SU),Citation62 or both,Citation63 in T2DM patients with HbA1c 7.5% to 11% and body mass index (BMI) 27 to 45 kg/m Citation2, who were inadequately controlled on metformin and/or SU. Exenatide 10 μg bid reduced HbA1c by 0.8% to 1.0%, 40% of patients achieving a HbA1c ≤ 7.0%. The weighted mean decrease in HbA1c in these studies was 1.0% (CI 0.8% to 1.2%)Citation64, equating to a 4.2 odds ratio for attaining HbA1c <7.0%, and reductions of ∼1.5 mmol/L in fasting glucose and 1.4 kg in body weight. An open-label 52-week extension of these trials in overweight patients indicated that exenatide has a sustained beneficial effect on glycemic control: ∼50% of these patients achieved HbA1c ≤7% and mean weight loss was 4.4 kg after 82 weeks.Citation65 Pancreatic β-cell function (assessed by the homeostasis model of assessment, HOMA) improved by 20% in T2DM patients on a TZD, with or without metformin, who were treated with exenatide, when compared to patients in whom placebo was added to their existing OHA;Citation66 however, this may reflect the improvement in glycemic control per se, rather than an absolute increase in β-cell mass or longevity. In these trials, the magnitude of the reduction in HbA1c with exenatide was greater with higher pre-treatment HbA1c,Citation64 indicating greater benefits of GLP-1 agonist treatment in patients with relatively worse glycemic control. Twice-daily exenatide was also compared with insulin in an open-label trial of patients with long-standing T2DM suboptimally controlled on stable doses of OHA. Patients on exenatide and insulin glargine had comparable decreases in HbA1c (∼1%), with lower postprandial excursions in the exenatide group,Citation67 and greater reduction in fasting and ‘premeal’ glucose in the insulin-treated group. In addition, body weight decreased (−2.3 kg) progressively over 26 weeks in patients treated with exenatide, with significant loss (−1.9 kg) even in those who did not report nausea, and increased (1.8 kg) in patients on insulin. In a cross-over trial, HbA1c fell by about 1.4% with both treatments,Citation68 while postprandial excursions were less and weight loss occurred with exenatide treatment. Similar trends were seen in another study comparing exenatide with premixed biphasic insulin aspart in overweight T2DM patients who were suboptimally controlled with OHA.Citation69 Hence, it appears that exenatide is non-inferior to insulin for glycemic control, and has the advantage of causing weight loss in obese T2DM patients.

Exenatide LAR, a long-acting release form, has been developed for once-weekly subcutaneous injection, but has not yet been marketed. In a 15-week trial of T2DM patients suboptimally controlled by metformin or diet and exercise, exenatide LAR 2 mg/week reduced mean HbA1c by 1.7%, fasting glucose by ∼2 mmol/L, and weight by 3.8 kg. Citation70 In T2DM patients with suboptimal control on diet and exercise or ≥1 OHA, this dose of exenatide LAR resulted in comparable reductions in HbA1c, fasting glucose and weight after 30 weeks of treatment, with a reduced incidence and severity of nausea and other GI AEs than, exenatide 10 μg bid.Citation71 Exenatide LAR may produce smaller effects in post-prandial glucose excursions, perhaps because inhibition of gastric emptying is less (the latter has hitherto only been assessed using the kinetics of paracetamol absorption), than twice-daily exenatide. This needs to be explored more fully, but may suggest that continuous GLP-1 receptor activation slows gastric emptying less than repeated, acute exposure to GLP-1 agonists, and may also mitigate the GI AEs. Exenatide LAR may potentially be a useful alternative for patients who are less tolerant of frequent injections, though its place in therapy remains to be determined.

Liraglutide

Liraglutide has 97% homology with GLP-1, and is longer-acting than exenatide because of an attached free fatty acid derivative that increases non-covalent binding to albumin, and renders it more resistant to DPP-4 degradation, which slows renal clearance and absorption from the subcutaneous injection site.Citation72 Its half-life is ∼12 hours, allowing it to be administered once daily;Citation73 while its onset of action is slower than exenatide, it is effective and well tolerated in doses of up to 1.9 mg/day.Citation74,Citation75 Liraglutide has recently been recommended for marketing authorization (under the trade name Victoza®; Novo Nordisk) as combination treatment with metformin and/or SU, or metformin ± TZD in T2DM patients with suboptimal control, by the Committee for Medicinal Products for Human Use (CHMP) under the European Medicines Agency (EMEA), with final marketing authorization expected from the European Commission by June 2009.Citation76 Liraglutide improves glycemic control and increases insulin secretion in response to carbohydrate loads.Citation68Citation73 Effects on gastric emptying or appetite have not been observed thus far, despite its efficacy in reducing body weight. Liraglutide monotherapy for 14 weeks reduced HbA1c, fasting glucose, and weight, and increased the proportion of subjects achieving HbA1c < 7% compared to placebo, in a phase 2 randomized double-blind trial of patients with suboptimal control on diet or 1 OHA,Citation74 the greatest effects being seen with a daily dose of 1.9 mg. Phase 3 trials in the Liraglutide Effect and Action in Diabetes (LEAD) program investigated the effects of liraglutide as monotherapy, or in combination with various OHAs. A 52-week monotherapy trial (LEAD-3) demonstrated that liraglutide 1.2 mg and 1.8 mg daily led to greater reductions in HbA1c and fasting glucose than glimepiride 8 mg daily.Citation77 In addition, body weight decreased significantly by ∼2 kg in patients on liraglutide compared to a mean gain of 1 kg on glimepiride, and the incidence of minor hypoglycemia in the liraglutide group was ∼10%, compared to 24% of patients on glimepiride. A 26-week comparison of the efficacy and safety of liraglutide, glimepiride, and placebo, in combination with metformin, found that liraglutide resulted in a comparative improvement in glycemic control, with weight loss and a lower incidence of hypoglycemia.Citation78 A 26-week, double-blind, placebo-controlled study (LEAD-4 Met + TZD) demonstrated that liraglutide when combined with metformin and rosiglitazone was effective for glycemic control.Citation79 Another 26-week study found that when either liraglutide, placebo, or rosiglitazone was added to glimepiride, liraglutide produced the greatest reductions in HbA1c and fasting glucoseCitation80 and adding liraglutide to glimepiride stabilized body weight. The beneficial effect of liraglutide on body weight was also seen in a trial of patients on metformin and glimepiride, which compared the addition of liraglutide (mean weight loss of 1.8 kg) with that of insulin glargine (weight gain of 1.6 kg),Citation81 Liraglutide-induced weight loss reflected a reduction in visceral and subcutaneous fat mass, and lean mass increased slightly.Citation82 Markers of β-cell function, such as the proinsulin:insulin ratio,Citation74 HOMA,Citation80 and first- and second-phase insulin responses during arginine-stimulated hyperglycemia,Citation83 were also improved by liraglutide. Hence, like exenatide, liraglutide is effective in improving and maintaining glycemic control as monotherapy or in combination with OHA, and in reducing body weight, with the advantage over twice-daily exenatide of requiring less frequent injections.

Safety and tolerability of GLP-1 agonists

Hypoglycemia is clearly less common with the use of GLP-1 agonists than with SU or insulin, presumably reflecting the glucose-dependent effects of the former on endogenous insulin secretion. The frequency of minor hypoglycemic episodes (about 5%) was comparable to placebo in studies of exenatide with metformin as the background therapy, though the incidence of hypoglycemia increased when exenatide improved glycemic control in conjunction with SU, and decreased in tandem with reduction in SU dose.Citation64 Severe hypoglycemia was rare when exenatide was combined with either metformin or TZDs.Citation61,Citation63,Citation66 The incidence of severe hypoglycemia was about 1.5% in the exenatide/glargine parallel-group comparative study,Citation67 but none of the episodes was severe, required medical attention or necessitated withdrawal of treatment. Minor hypoglycemia has been reported in ∼10% of patients taking liraglutide as monotherapyCitation74 or in combination with metformin and/or TZD,Citation79 though the incidence is increased to ∼25% when liraglutide is added to SU.Citation78,Citation80

The adverse effects of GLP-1 agonists are chiefly gastrointestinal.Citation58Citation86 Nausea is the most common with both the short-acting and LAR formulations of exenatide,Citation58Citation68 with mild to moderate self-reported nausea in ∼35% of patients on exenatide 5 μg bid and up to half of the patients receiving 10 μg bid.Citation84 In most patients the frequency, and severity, peaks in the first 8 weeks of treatment and decreases thereafter,Citation64,Citation66 and is likely to be a central GLP-1 effect, unrelated to the slowing of gastric emptying.Citation84 Other GI AEs, such as diarrhea and constipation, are less common, and are also usually transient and mild. In clinical trials, treatment discontinuation because of GI AEs (nausea, vomiting, diarrhea, anorexia, abdominal pain) occurred in about 5% of patients. The risk of exenatide-induced nausea can apparently be minimized by progressive escalation in dosage.Citation85 The frequency of nausea may be less in patients on exenatide LAR.Citation70,Citation71 Liraglutide is also associated with GI AEs, which are dose-dependent, the most common being transient mild-to-moderate nausea, which occurs in 5% to 15% of patients.Citation73Citation77 A patient-reported evaluation of GI AEs using the Gastrointestinal System Rating Scale (GSRS),Citation84 as part of a 14-week phase 2 trial,Citation74 indicated that GI symptoms are not usually severe (maximum 2 on a 7-point scale), and occur mainly in the first 2 weeks of treatment, returning to baseline thereafter. In a 26-week study of T2DM patients with suboptimal glycemic control on OHA, once-daily liraglutide was associated with less persistent nausea than twice-daily exenatide, which may be attributable to smaller fluctuations in concentration of the GLP-1 receptor agonist.Citation86

Anti-exenatide antibodies are evident in about half to two-thirds of patients on exenatide,Citation64 or exenatide LAR,Citation70 but do not appear to affect either glycemic control or adverse events. A higher prevalence of injection-site discomfort and anti-exenatide antibodies were seen in patients on the LAR form. Anti-liraglutide antibodies have been detected in ∼10% of patients, but, again, do not appear to have adverse effects, or influence glycemic control.Citation81

GLP-1 agonists in development

New GLP-1 agonists have demonstrated improvement in fasting and postprandial glucose control in phase III trials, with apparently fewer GI AEs than currently marketed compounds. Albiglutide, a DPP-4-resistant GLP-1 analog which is administered weekly, reduced fasting and postprandial glucose levels without causing hypoglycemia in healthy subjectsCitation87 and T2DM patients.Citation88 A dose of 32 mg reduced 24-hour mean weighted glucose by 35 mg/dL (∼2 mmol/L) at day 2 and 56 mg/dL (∼3 mmol/L) at day 9 after treatment in T2DM subjects. Of note, the frequency and severity of GI AEs of albiglutide were comparable to placebo. Taspoglutide, another once-weekly GLP-1 agonist, reduced HbA1c (∼0.9% to 1.2%), as well as fasting and postprandial glucose levels at doses of 10 or 20 mg in T2DM, in combination with metformin for 8 weeks, with a low incidence of hypoglycemia. Citation89 Weight loss was also significant, especially in the group receiving 20 mg/week (−2.8 kg).

DPP-4 inhibitors

Prevention of GLP-1 degradation by pharmacological DPP-4 inhibition in pigs,Citation90 and genetic inactivation in DPP-4 knockout mice,Citation91 increase endogenous total GLP-1 levels, leading to increased insulin secretion and reduced fasting and postprandial glucose concentrations. These findings have led to the development of once-daily, orally-active DPP-4 inhibitors to increase the incretin effect. DPP-4 activity is reduced by almost 100% within 15 to 30 minutes of oral administration of the DPP-4 inhibitors sitagliptin or vildagliptin, producing a 2-fold increase in mean active GLP-1 levels (to 15 to 25 pmol/L), with a duration of inhibition in excess of 16 hours because of initial rapid binding to DPP-4, followed by a slow phase of tight binding,Citation92 so that effects persist for 24 hours after administration of a single dose of sitagliptinCitation93 and vildagliptin.Citation94 DPP-4 inhibition increases GLP-1 and GIP levels by 2- to 3-fold, while reducing glucagon,Citation92 although the magnitude of the rise in GLP-1 is dependent on the type of nutrient ingested. The pharmacokinetics of sitagliptin and vildagliptin are not affected by age, gender, ethnicity or body mass index, and no significant drug interactions have hitherto been noted.Citation95

Sitagliptin

Sitagliptin (Januvia®; Merck and Co, Inc.) was approved for use in a dose of 100 mg once daily, by the FDA in 2006 and subsequently in the EU, Australia and Asia, for the improvement of glycemic control in combination with metformin and/or a sulfonylurea when diet and exercise plus OHA do not result in adequate glycemic control.Citation96 In drug-naïve T2DM patients, sitagliptin monotherapy is more effective than placebo in reducing HbA1c (by up to 1%) and fasting glucose (by up to 18 mg/dL).Citation97 Greater benefits in glycemic control were seen with sitagliptin 100 mg daily, compared to placebo, over a period of 24 weeks in T2DM patients already on metforminCitation98,Citation99 and pioglitazone,Citation100 and when added to sulfonylureas (with or without metformin).Citation101 It was non-inferior to glipizide when added to ongoing metformin therapy.Citation102 Metformin stimulates GLP-1 release, increasing both active and total GLP-1 levels by 2-fold, but does not act as a DPP-4 inhibitor, and, accordingly, has a synergistic effect with sitagliptin, to increase both active GLP-1 (∼4-fold).Citation103 A fixed-dose combination of sitagliptin and metformin (Janumet®; Merck and Co, Inc.) has recently been developed. An improvement in β-cell function has been suggested by a reduction in the proinsulin ratio and increase in insulin levels with sitagliptin, compared to placebo.Citation97,Citation98,Citation100 The effect of sitagliptin on gastric emptying has not been reported. Unlike GLP-1 agonists, sitagliptin is not associated with changes in body weight.Citation97Citation102 Sitagliptin is thus a useful add-on therapy to OHA in T2DM patients who are not overweight, especially those with suboptimal control on maximum doses of metformin. Dose adjustment of sitagliptin is recommended in patients with renal insufficiency.Citation104

Vildagliptin

Vildagliptin (Galvus®; Novartis) was approved in the EU in 2008, for use in combination with metformin and/or TZD (50 mg twice daily), or with an SU (50 mg once daily), and is pending approval by the FDA.Citation105 Vildagliptin monotherapy was shown to be superior to placebo in reducing HbA1c (by 0.4% to 0.8%), fasting (by ∼10 to 20 mg/dL) and postprandial glucose,Citation106 and non-inferior compared to rosiglitazoneCitation107 in randomized, double-blinded 24-week trials. The addition of vildagliptin to metformin further improved glycemic control (HbA1c reduction 0.5% to 1.2%).Citation108,Citation109 The combination of vildagliptin with pioglitazone reduced fasting glucose to a greater extent than did vildagliptin or pioglitazone alone,Citation110 and vildagliptin is not inferior to pioglitazone when added to metformin.Citation111 Vildagliptin has been shown to increase insulin and C-peptide responses to glucose by up to 100%,Citation108,Citation112 suggesting improvements in β-cell function.Citation113 DPP-4 inhibition by vildagliptin does not apparently slow gastric emptying, possibly due to the relatively modest elevation in plasma GLP-1 levels, compared to administration of exogenous GLP-1 agonists.Citation114

Safety and tolerability of DPP-4 inhibitors

Neither sitagliptinCitation97Citation102 nor vildagliptinCitation106Citation114 appear to increase the risk of hypoglycemia when used as monotherapy, or in combination with metformin or TZD, though the incidence of mild-moderate hypoglycemia was increased by approximately 2-fold to ∼4% to 5% when sitagliptin was added to SU.Citation101,Citation102 DPP-4 inhibitors have not been associated with significant GI AEs, and appear to be safe and well-tolerated in patients with moderate to severe renal insufficiency (including those with end-stage disease on hemodialysis) if doses are adjusted according to creatinine clearance.Citation115 An increased risk (odds ratio 1.34, 95% CI 1.10 to 1.64) of nasopharyngitis and all-cause infections (sinusitis, viral upper respiratory tract infections, urinary tract infections) has been observed with the use of sitagliptin,Citation95 and hypersensitivity reactions (anaphylaxis, angioedema and exfoliative dermatitis) have been reported within the first 3 months of sitagliptin therapy, Citation96 while cases of vildagliptin-associated severe dermatological allergic reactions and elevated liver transaminases, have led to a delay in FDA approval.Citation105

DPP-4 inhibitors in development

Alogliptin and saxagliptin are DPP-4 inhibitors that may be available in the near future: alogliptin is currently under FDA review, while saxagliptin is under review by the EMEA for potential marketing in late 2009. Alogliptin monotherapy is reportedly effective in reducing HbA1c, as well as fasting and postprandial glucose levels in T2DM patients in doses of 25 to 400 mg daily.Citation116,Citation117 In randomized, placebo-controlled, double-blinded 26-week trials, the addition of alogliptin to metforminCitation118 or glyburideCitation119 improved HbA1c and fasting glucose without an increased incidence of GI AEs or hypoglycemia. Alogliptin also improved glycemic control and β-cell function in combination with pioglitazoneCitation120 or insulinCitation121 in animal models. Saxagliptin reduced HbA1c and both fasting and postprandial glucose levels in drug-naïve T2DM patients in a placebo-controlled, randomized, double-blind study,Citation122 and evidence from ongoing trials suggests that it may be effective in combination with metformin,Citation123 glyburideCitation124 or TZD.Citation125 Daily doses of alogliptin 12.5 to 25 mg and saxagliptin 2.5 to 10 mg lower HbA 1c by ∼1%, which is comparable to sitagliptin and vildagliptin in doses of 50 to 100 mg daily, implying greater potency. In addition, no adverse effects attributable to disordered immune function have been reported to date for alogliptin or saxagliptin,Citation126 although it should be recognized that most of the clinical data available on the newer DPP-4 inhibitors have been published only in abstract form, and there is less information available about their safety profiles, in comparison with sitagliptin and vildagliptin.

Extraglycemic effects of incretin-based therapies

In addition to improving glycemic control, GLP-1 agonists may have additional beneficial effects on blood pressure (BP) and plasma lipids, in part, by effects that are apparently independent of weight loss. Exenatide 5 μg bid reduced BP by ∼9 mmHg from baseline when added to existing antihypertensive therapy and OHA in obese T2DM patients during 26 weeks of treatment,Citation127 while mean diastolic and systolic BP (in another trial of obese T2DM patients on 5 to 10 μg bid for 82 weeksCitation128) was reduced by ∼4 mmHg and ∼6 mmHg respectively, with the greatest reduction seen in patients who lost the most weight. Patients treated with exenatide 5 to 10 μg bid for an average of 3.5 years demonstrated significant reductions in triglycerides (12%), total cholesterol (5%), low-density lipoprotein (LDL) cholesterol (6%), and increases in high-density lipoprotein (HDL) cholesterol (24%);Citation129 again, these beneficial effects were related to the magnitude of weight loss. However, a recent study using exenatide LARCitation71 found that the mean reductions in systolic BP over 30 weeks (−4.7 mmHg) and diastolic BP (−1.7 mmHg) were independent of body weight. In the LEAD studiesCitation77Citation80 liraglutide 1.8 mg daily for 2 weeks reduced mean systolic BP by ∼2 to 4.5 mmHg, which was also apparently independent of weight loss. Liraglutide was found to lower triglyceride (TG) levels in association with reductions in weight and HbA1c.Citation74 Other than weight reduction, the mechanisms by which GLP-1 agonists improve BP and lipids remain unclear. As discussed previously, GLP-1 analogues also have direct beneficial effects on the heart, as shown by animal studies. Both exenatide and GLP-1 (9–36) improve left ventricular performance and decreased infarct size in rat hearts during reperfusion after ischemia.Citation130 Liraglutide also induces significant infarct shrinkage in an ischemia-reperfusion injury murine heart preparations.Citation131 In T2DM patients, liraglutide reduces levels of the inflammatory markers plasminogen activator inhibitor 1, B-type natriuretic peptide and high-sensitivity C-reactive protein, which are associated with increased cardiovascular risk.Citation132

There is also evidence that DPP-4 inhibitors also have beneficial effects on BP and lipids. In non-diabetic patients with moderate hypertension, sitagliptin reduced systolic and diastolic BP by ∼2 mmHg, compared to placebo, within 5 days at doses of 50 to 100 mg daily. Citation133 Sitagliptin also reduced plasma TG by 10% to 15% and increased HDL by ∼5% in doses of 25 to 100 mg daily as monotherapy over 12 weeks in T2DM patients. Citation134 Vildagliptin lowered total and LDL cholesterol by ∼10%Citation135 and TG by up to 15%Citation122 when added to TZD in metformin-resistant patients. Possible mechanisms for the lipid-lowering effects of DPP-4 inhibitors include reduced production of intestinal TG-rich particles after fat-rich mealsCitation136 and augmentation of lipid mobilization and oxidation,Citation137 although the exact mechanisms remain unclear.

compares GLP-agonists and DPP-inhibitors.

Table 2 Comparison of GLP-1 agonists and DPP-4 inhibitors

Unresolved issues and long-term safety of incretin-based therapies

The slowing of gastric emptying induced by exogenous GLP-1 and GLP-1 agonists may cause concerns over use in T2DM patients with gastroparesis. It is now well recognized that gastric emptying (GE) is delayed in perhaps 30% of patients with long-standing T2DM,Citation32 and correlates poorly with the presence and severity of upper GI symptoms.Citation138 In healthy subjects the magnitude of the slowing of GE by exogenous GLP-1 is sufficient to cause ‘gastroparesis’ in ∼50%,Citation45 and in T2DM patients the slowing of GE by exenatide is greater when GE is relatively faster.Citation58 It is also known that in diabetics with vagal neuropathy, unlike healthy subjects, exogenous GLP-1 fails to relax the proximal stomach.Citation139 The implications are that the magnitude of the reduction in postprandial glycemia induced by GLP-1 is likely to depend on GE, and that in patients with autonomic neuropathy-associated gastroparesis (whether symptomatic or not), any further slowing of gastric emptying is likely to be minimal. Both of these hypotheses warrant formal evaluation. Patients with symptomatic gastroparesis have been excluded from the majority of trials of GLP-1 agonists – hence, the issue of whether these drugs exacerbate, or have no effect on symptoms, also remains unresolved.

Post-marketing cases of acute pancreatitis have rarely been reported with exenatide;Citation140 however, T2DM patients have a ∼3-fold increased risk of pancreatitis,Citation141 as well as other risk factors for pancreatitis including gallstones, obesity, ethanol abuse and severe hypertriglyceridemia. Eight out of the nine reported cases of pancreatitis in the LEAD program occurred in patients on liraglutide.Citation142 Studies in animals to evaluate this issue have hitherto failed to clarify whether there is a causal association between pancreatitis and the use of incretin-based therapies. Administration of metformin, sitagliptin, or liraglutide did not increase transcription of genes associated with pancreatitis in mice, and GLP-1 receptor knockout mice did not differ from normal mice in the severity of experimentally induced pancreatitis when treated with exenatide.Citation143 However, treatment with sitagliptin and metformin was associated with increased pancreatic ductal turnover, ductal metaplasia, and one case of pancreatitis in rats.Citation144

The non-GI AEs associated with sitagliptin and vildagliptin may be related to the immunological properties of DPP-4, which is expressed in many tissues, and has numerous substrates, including GI hormones, neuropeptides, and cytokines.Citation145 As DPP-4 is known to be involved in immunoregulation as a T-cell costimulator, and in breakdown of cytokines such as bradykinin and interleukin-2 and-1β,Citation146 inhibition of DPP-4 may have adverse effects on immune function. Indeed, the severity of rheumatoid arthritis has been reported to be inversely related to DPP-4 activity,Citation147 and DPP-4 levels are reduced in nasal tissue of patients with chronic rhinosinusitis.Citation146 Although DPP-4 inhibition is known to reduce the levels of the cardioprotective GLP-1 metabolite GLP-1 (9–36) which is cleaved from native intact GLP-1, there is hitherto no evidence of adverse cardiovascular effects.Citation101Citation118 As clinical trials involving sitagliptin and vildagliptin evaluated use for a maximum of only 52 weeks, more long-term safety data are required, particularly in view of the need for chronic treatment in T2DM, and the presence of DPP-4 in multiple organ systems.

Cost-effectiveness and patient-centered outcomes of incretin-based therapies

The cost-effectiveness of exenatide, compared to OHA or insulin, has been evaluated in several studies. As add-on therapy to metformin, it is cost-effective compared to pioglitazone and glibenclamide,Citation59 and also as monotherapy in drug-naïve patients compared to metformin alone. An analysis, based on data from 314 overweight T2DM patients who completed an 82-week trial of exenatide,Citation65 projected that 30 years of treatment with exenatide added to OHA would be cost-effective in comparison to a hypothetical placebo arm,Citation148 in terms of improvements in clinical outcomes (glycemic control, weight, BP, and lipids) with concomitant reductions in the risk of micro- and macrovascular complications, increased life expectancy, and improved quality of life. These were associated with incremental cost-effectiveness ratios (ICER) of US$35,571/life-years gained, and US$36,133/quality-adjusted life-year, below the threshold ICER of US$50,000. Using data from the same cohort, a study projected models of long-term complications, life expectancy, quality-adjusted life expectancy, and direct medical costs of patients on exenatide versus those on insulin glargine in the United Kingdom (UK),Citation149 and concluded that exenatide was associated with a lower cumulative incidence of most cardiovascular (CVD) complications, including CVD-related death, and was more cost-effective than glargine. However, this study based its analyses on estimated costs of exenatide which ranged from 20% to 100% of the US price (US$161/28 days). Another UK study, which used the actual UK National Health System price of exenatide (∼£68/28 days), rather than an estimated cost, found that exenatide was less cost-effective than glargine in a 40-year projection.Citation150 It is evident that the cost-effectiveness of exenatide in relation to insulin glargine is highly dependent on the relative prices of these medications, which vary substantially between countries.

Patient-reported measures, such as quality of life and treatment satisfaction, have also been analyzed, using data from a 26-week randomized, open-label study comparing exenatide to insulin glargine in T2DM patients with suboptimal glycemic control on metformin and sulfonylurea.Citation67 This analysis included 228 patients on exenatide and 227 on insulin, with outcomes measured by 5 scales: 1) Diabetes Symptom Checklist (frequency and perceived discomfort of physical and psychological symptoms associated with diabetes); 2) Diabetes Treatment Flexibility Scale (focusing on patient choices in meals and daily activities); 3) Diabetes Treatment Satisfaction Questionnaire (satisfaction with current treatment regimen); 4) EuroQol-EQ5D (overall health status); and 5) vitality scale of the SF-36 (energy level and fatigue). Both exenatide and insulin groups showed significant improvement from baseline in symptoms, satisfaction with treatment, overall health status, and energy levels.Citation151 The authors noted that although exenatide was associated with GI AEs and increased frequency of injections, these did not result in less patient satisfaction, which they attributed to the benefits of weight loss in this treatment group. Hence, the benefits of exenatide therefore appear to outweigh adverse effects, weight loss in particular conferring an advantage over insulin. At present, there are no cost-benefit analyses available for liraglutide.

In relation to the DPP-4 inhibitors, a cost-effectiveness analysis compared the addition of sitagliptin, rosiglitazone or SU to metformin in patients with HbA1c > 6.5%. Citation152 Local health surveys in Austria, Finland, Portugal, Scotland, Spain, and Sweden were used to generate average patient profiles for the analysis, using data on clinical and adverse effects from two recent trials of sitagliptin.Citation102,Citation153 Adding sitagliptin was found to be a more cost-effective alternative to the addition of rosiglitazone or an SU, incremental cost-effectiveness ratio values ranging from 5949/QALY to 20 350/QALY, depending on the individual country. However, these studies may not be generalizable to countries outside the EU, because of differences in health care costs and prices of medication. Analysis of the cost-effectiveness of vildagliptin is not currently available.

Given the comparable outcomes of the use of GLP-1 agonists and DPP-4 inhibitors in T2DM for many parameters, their cost-effectiveness is an important issue. An analysis compared estimated six-month total, and diabetes-related, medical costs among 2482 patients on sitagliptin, with 1885 patients on exenatide, in the US.Citation154 Exenatide was associated with lower total 6-month direct medical costs (US$9340 vs US$9995), despite some component costs being higher with exenatide, ie, those associated with diabetes-related drugs, and diabetes-related medical care including emergency room attendance. Sitagliptin was associated with higher outpatient costs. This study concluded that use of the GLP-1 agonist was associated with higher diabetes-related costs, but lower total medical costs than the DPP-4 inhibitor; however, as with comparisons of incretin therapies with conventional injectable and oral medications, the results of this cost-benefit analysis cannot be extrapolated to outside the US.

The place of incretin-based therapies in the treatment of T2DM and impaired glucose tolerance

The American Association of Clinical Endocrinologists (AACE) currently advocates DPP-4 inhibitor monotherapy for T2DM patients with HbA1c 6% to 7%, or in combination with metformin or TZD if target HbA1c (≤6.5%) is not achieved, and adding a GLP-1 agonist to SU, metformin, and/or TZD in patients who do not achieve target HbA1c.Citation155 In contrast, the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD) did not recommend, in their combined consensus statements on standards of medical care in diabetesCitation156 and initiation and adjustment of treatment in T2DM,Citation4 either GLP-1 agonists or DPP-4 inhibitors as first- or second-tier treatments, despite acknowledging the weight-lowering effect of exenatide, and the utility of exenatide and sitagliptin for improved postprandial glycemic control with an associated low risk of hypoglycemia. It should be recognized that selected groups of patients may benefit from early, or even first-line, use of incretin-based therapies. The weight-lowering effect of GLP-1 agonists is beneficial in the overweight and obese, especially since many add-on therapies (sulfonylurea, TZD and insulin) promote weight gain. As discussed, trials using exenatide reported a dose-dependent reduction in body weight by up to 4.5 kg in at least 30 weeks.Citation55Citation65

GLP-1 agonists may also have a role in the management of critically ill patients with T2DM, in whom both hyperglycemia and hypoglycemia are risk factors for a poor outcome. GLP-1 infusion appears to reduce plasma glucose significantly in association with increased insulin and suppressed glucagon concentrations, without the risk of hypoglycemia in severely ill patients who were hyperglycemic while receiving total parenteral nutrition,Citation157 as well as in fasted diabetic patients who had undergone major surgery.Citation158 More recently, GLP-1 infusion has been shown to markedly attenuate the glycemic response to enteral nutrition (arguably the preferred route of nutritional support) in non-diabetic critically ill patients, reflecting its insulinotropic and glucagonostatic properties.Citation159 These observations suggest that GLP-1 and/or its agonists have a potential role in the management of hyperglycemia in the critically ill, without the attendant risks of insulin-induced hypoglycemia. The latter were emphasized by the results of the NICE-SUGAR study, in which the use of insulin infusions to achieve target blood glucose 4.5 to 6.0 mmol/L was shown to increase both hypoglycemia and mortality in critically ill patients.Citation160

Although DPP-4 inhibitors are weight-neutral and do not significantly reduce appetite, their lack of GI side effects compared to GLP-1 agonists and metformin, and low risk of hypoglycemia, render them especially suitable for the management of T2DM in older patients, in whom the polypharmacy often required for glycemic control is accompanied by increased risks of hypoglycemia and other adverse effects due to age-related changes in drug metabolism, reduced energy intake, and comorbidities such as cardiovascular and renal impairment. In particular, sulfonylureas are associated with a high risk of hypoglycemia accounting for substantial morbidity and health-care costs in this group.Citation161 Studies of sitagliptinCitation97,Citation98,Citation162 and vildagliptinCitation106,Citation107,Citation111,Citation163 including subjects ≥65 years of age, and patients with moderately severe hepaticCitation164 and renalCitation165 impairment, suggest that the DPP-4 inhibitors are as effective (HbA1c reduction ∼1%) and well-tolerated as in younger patients. The DPP-4 inhibitors were also associated with a low incidence (∼1%) of severe hypoglycemia, and GI and other adverse events such as peripheral edema (2% to 10%).

Conclusions

Despite the relatively recent advent of GLP-1 agonists and DPP-4 inhibitors, evidence has rapidly accumulated to support their efficacy and safety in the management of T2DM, as well as their potential for improving cardiovascular and β-cell function. The low risk of inducing hypoglycemia, and beneficial or neutral effects on body weight, render them attractive alternatives to insulin secretagogues and insulin as add-on therapy to metformin, or even as first-line therapy in selected groups of patients, especially in the overweight and the elderly. Incretin-based medications are likely to be increasingly at the forefront of therapy of T2DM in the new millennium.

Disclosures

The authors declare no conflicts of interest.

References

  • IDF Clinical Guidelines Task ForceGlobal guideline for type 2 diabetes: recommendations for standard, comprehensive and minimal careDiabet Med20062357959316759299
  • UKPDS GroupIntensive blood-glucose control with sulphonylureas or inslin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33)Lancet19983528378539742976
  • ShichiriMKishikawaHOhkuboYLong-term results of the Kumamoto Study on Optimal Diabetes Control in type 2 diabetic patientsDiabetes Care200023B21B2910860187
  • NathanDMBuseJBDavidsonMBMedical Management of Hyperglycemia in Type 2 Diabetes: A Consensus Algorithm for the Initiation and Adjustment of Therapy: A consensus statement of the American Diabetes Association and the European Association for the Study of DiabetesDiabetes Care20093219320318945920
  • CurrieCJPooleCDGaleEAThe influence of glucose-lowering therapies on cancer risk in type 2 diabetesDiabetologia200972 (Epub ahead of print)
  • WhitmerRAKarterAJYaffeKQuesenberryCPJrSelbyJVHypoglycemic episodes and risk of dementia in older patients with type 2 diabetes mellitusJAMA20093011565157219366776
  • The ADVANCE Collaborative GroupIntensive blood glucose control and vascular outcomes in patients with type 2 diabetesN Engl J Med20083582560257218539916
  • The Action to Control Cardiovascular Risk in Diabetes Study GroupEffects of intensive glucose lowering in type 2 diabetesN Engl J Med20083582545255918539917
  • MonnierLLapinskiHColetteCContributions of fasting and postprandial plasma glucose increments to the overall diurnal hyperglycemia of type 2 diabetic patients: variations with increasing levels of HbA1cDiabetes Care20032688188512610053
  • CerielloAHanefeldMLeiterLPostprandial glucose regulation and diabetic complicationsArch Intern Med20041642090209515505121
  • RazIWilsonPWFStrojekKEffects of prandial versus fasting glycemia on cardiovascular outcomes in type 2 diabetes: the HEART2D trialDiabetes Care20093238138619246588
  • La BarreJSur les possibilities d’un traitement du diabète par l’incrétineBull Acad R Med Belg193212620634
  • ElrickHStimmlerLHladCJAraiYPlasma insulin response to oral and intravenous glucose administrationJ Clin Endocrinol Metab1964241076108214228531
  • McIntyreNHoldsworthCDTurnerDANew interpretation of oral glucose toleranceLancet1964II202114149200
  • BrownJCMuttVPedersonRAFurther purification of polypeptide demonstrating enterogastrone activityJ Physiol197020957645499047
  • SchmidtWESiegelEFCreutzfeldtWGlucagon-like peptide-1 but not glucagon-like peptide-2 stimulates insulin release from isolated rat pancreatic isletsDiabetologia1985287047073905480
  • NauckMAHombergerESiegelEGIncretin effects of increasing glucose loads in man calculated from venous insulin and C-peptide responsesJ Clin Endocrinol Metab1986634924983522621
  • NauckMAStockmannFEbertRCreutzfeldtWReduced incretin effect in type 2 (non-insulin-dependent) diabetesDiabetologia19862946523514343
  • VilsbollTKrarupTMadsbadSHolstJJDefective amplification of the late phase insulin response to glucose by GIP in obese type II diabetic patientsDiabetologia2002451111111912189441
  • KjemsLLHolstJJVolundAMadsbadSThe influence of GLP-1 on glucose-stimulated insulin secretion effects on beta-cell sensitivity in type 2 and non-diabetic subjectsDiabetes20035238038612540611
  • HøjbergPVVilsbøllTRabølRFour weeks of nearnormalisation of blood glucose improves the insulin response to glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide in patients with type 2 diabetesDiabetologia20095219920719037628
  • DeaconCFNauckMAToft-NielsenMPridalLWillmsBHolstJJBoth subcutaneous and intravenously administered glucagon-like peptide-1 are rapidly degraded from the NH2-terminus in type 2-diabetic patients and in healthy subjectsDiabetes199544112611317657039
  • KreymannBWilliamsGGhateiMABloomSRGlucagon-like peptide 1 7–36 a physiological incretin in manLancet19872130013042890903
  • O’DonovanDGDoranSFeinle-BissetCEffect of variations in small intestinal glucose delivery on plasma glucose, insulin, and incretin hormones in healthy subjects and type 2 diabetesJ Clin Endocrinol Metab2004893431341515240627
  • NauckMAHeimesaatMMBehleKEffects of glucagon-like peptide 1 on counterregulatory hormone responses, cognitive function, and insulin secretion during hyperinsulinemic, stepped hypoglycemic clamp experiments in healthy volunteersJ Clin Endocrinol Metab2002871239124611889194
  • ScrocchiLABrownTJMaCluskyNGlucose intolerance but normal satiety in mice with a null mutation in the glucagon-like peptide 1 receptor geneNat Med19962125412588898756
  • TsengCCZhangXYWolfeMMEffect of GIP and GLP-1 antagonists on insulin release in the ratAm J Physiol Endocrinol Metab1999276E1049E1054
  • ImeryuzNYeganBCBozkurtACoskunTVillanueva-PenacarriloMLUlusonNBGlucagon-like peptide-1 inhibits gastric emptying via vagal afferent-mediated central mechanismsAm J Physiol Gastrointest Liver Physiol1997273G920G927
  • SchirraJSturmKLeichtPArnoldRGokeBKatschinskiMExendin (9–39) amide is an antagonist of glucagon-like peptide-1 (7–36) amide in humansJ Clin Invest1998101142114309525985
  • EdwardsCMToddJFMahmoudiMGlucagon-like peptide 1 has a physiological role in the control of postprandial glucose in humans: studies with the antagonist exendin 9–39Diabetes1999486939892226
  • SchirraJNicolausMRoggelPEndogenous glucagon-like peptide 1 controls endocrine pancreatic secretin and antro-pyloroduodenal motility in humansGut20065524325115985560
  • HorowitzMEdelbroekMAWishartJMRelationship between oral glucose tolerance and gastric emptying in normal healthy subjectsDiabetologia1993368578628405758
  • JonesKLHorowitzMCarneyBJGastric emptying in early non-insulin diabetes mellitusJ Nucl Med199637164316488862300
  • MourotJThouvenotPCouetCAntoineJMKrobickaADebryGRelationship between the rate of gastric emptying and glucose and insulin responses to starchy foods in young healthy adultsAm J Clin Nutr198848103510403048076
  • O’DonovanDHorowitzMRussoAEffects of lipase inhibition on gastric emptying of, and on the glycaemic, insulin and cardiovascular responses to, a high-fat/carbohydrate meal in type 2 diabetesDiabetologia2004472208221415662558
  • PerfettiRMerkelPGlucagon-like peptide-1: a major regulator of pancreatic beta-cell functionEur J Endocrinol200014371777511124853
  • DruckerDJPhillippeJMojsovSChickWLHaenerJFGlucagon-like peptide 1 stimulates gene expression and increases cyclic AMP level in a rat islet cell lineProc Natl Acad Sci U S A198784343434383033647
  • FarillaLBulottaAHirshbergBGlucagon-like peptide-1 inhibits cell apoptosis and improves glucose responsiveness of freshly isolated human isletsEndocrinology20031445149518812960095
  • LiLEl-KholyWRhodesCJBrubakerPLGlucagon-like peptide-1 protectsbeta cells from cytokine-induced apoptosis and necrosis: role of protein kinase BDiabetologia2005481339134915902400
  • Van DijkGLindskogSHolstJJSteffensABAhrenBEffects of glucagon-like peptide-I on glucose turnover in ratsAm J Physiol Endocrinol Metab1996270E1015E1021
  • ChelikaniPKHaverACReidelbergerRDIntravenous infusion of glucagon-like peptide-1 potently inhibits food intake, sham feeding, and gastric emptying in ratsAm J Physiol Regul Integr Comp Physiol1997288R1695R170615718384
  • RachmanJGribbleFMBarrowBALevyJCBuchananKDTurnerRCNormalization of insulin responses to glucose by overnight infusion of glucagon-like peptide 1 7–36 amide in patients with NIDDMDiabetes199645152415308866556
  • Juntti-BerggrenLPigonJKarpeFThe antidiabetogenic effect of GLP-1 is maintained during a 7-day period and improves diabetic dyslipoproteinemia in NIDDM patientsDiabetes Care199619120012068908380
  • WillmsBWernerJHolstJJOrskovCCreutzfeldtWNauckMAGastric emptying, glucose responses, and insulin secretion after a liquid test meal: effects of exogenous glucagon-like peptide-1 (GLP-1)-7–36 amide in type 2 (noninsulin-dependent) diabetic patientsJ Clin Endocrinol Metab1996813273328550773
  • LittleTJPilichiewiczANRussoAEffects of intravenous GLP-1 on gastric emptying and intragastric distribution in healthy subjects – relationships with postprandial glycemic and insulinemic responsesJ Clin Endocrinol Metab2006911916192316492694
  • MeneillyGSGreigNTildesleyHHabenerJFEganJMElahiDEffects of 3 months of continuous subcutaneous administration of glucagon-like peptide-1 in elderly patients with type 2 diabetesDiabetes Care2003262835284114514588
  • ZanderMMadsbadSMadsenJLHolstJJEffect of 6-week course of glucagon-like peptide 1 on glycemic control, insulin sensitivity, and beta-cell function in type 2 diabetes: a parallel-group studyLancet200235982483011897280
  • NikolaidisLAMankadSSokosGGEffects of glucagon-like peptide-1 in patients with acute myocardial infarction and left ventricular dysfunction after successful reperfusionCirculation200410996296514981009
  • SokosGGNikolaidisLAMankadSElahiDShannonRPGlucagon-like peptide-1 infusion improves left ventricular ejection fraction and functional status in patients with chronic heart failureJ Card Fail20061269469917174230
  • SokosGGBolokogluHGermanJEffect of glucagon-like peptide-1 (GLP-1) on glycemic control and left ventricular function in patients undergoing coronary artery bypass graftingAm J Cardiol200710082482917719327
  • MüssigKOncuALindauerPEffects of intravenous glucagon-like peptide 1 on glucose control and hemodynamics after coronary artery bypass surgery in patients with type 2 diabetesAm J Cardiol200810264664718721530
  • NystromTGutniakMKZhangQEffects of glucagon-likep peptide 1 on endothelial function in type 2 diabetes patients with stable coronary artery diseaseAm J Physiol Endocrinol Metab2004287E1209E121515353407
  • BasuACharkoudianNSchrageWRizzaRABasuRJoynerMJBeneficial effects of GLP-1 on endothelial function in humans: dampening by glyburide but not by glimepirideAm J Physiol Endocrinol Metab2007293E1289E129517711996
  • BanKNoyan-AshrafMHHoeferJCardioprotective and vasodilatory actions of glucagon-like peptide 1 receptor are mediated through both glucagon-like peptide-1 receptor-dependent and-independent pathwaysCirculation20081172340235018427132
  • EngJKleinmanWASinghLSinghGRaufmanJPIsolation and characterization of exendin-4, an exendin-3 analogue, from Heloderma suspectum venom. Further evidence for an exendin receptor on dispersed acini from guinea pig pancreasJ Biol Chem199226741084115
  • ThorensBPorretABuhlerLDengSPMorelPWidmannCCloning and functional expression of the human islet GLP-1 receptor. Demonstration that exendin-4 is an agonist and exendin (9–39) an antagonist of the receptorDiabetes19934216786828405712
  • KoltermanOGBuseJBFinemanMSGainesEHeintzSSynthetic exendin-4 (exenatide) significantly reduces postprandial and fasting plasma glucose in subjects with type 2 diabetesJ Clin Endocrinol Metab2003883082308912843147
  • LinnebjergHParkSKotharePAEffect of exenatide on gastric emptying and relationship to postprandial glycemia in type 2 diabetesRegul Pept200815112312918675854
  • CvetkovicRSPloskerGLExenatide: a review of its use in patients with type 2 diabetes mellitus (as an adjunct to metformin and/or a sulfonylurea)Drugs20076793595417428109
  • NelsonPPoonTGuanXSchnabelCWintleMFinemanMThe incretin mimetic exenatide as a monotherapy in patients with type 2 diabetesDiabetes Technol Ther2007931732617705687
  • DeFronzoRARatnerREHanJKimDDFinemanMSBaronADEffects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetesDiabetes Care2005281092110015855572
  • BuseJBHenryRRHanJKimDDFinemanMSBaronADEffects of exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea-treated patients with type 2 diabetesDiabetes Care2004272628263515504997
  • KendallDMRiddleMCRosenstockJEffects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylureaDiabetes Care2005281083109115855571
  • AmoriRELauJPittasAGEfficacy and safety of incretin therapy in type 2 diabetes: systematic review and meta-analysisJAMA20072585258917579226
  • BlondeLKleinEJHanJInterim analysis of the effects of exenatide treatment on A1c, weight and cardiovascular risk factors over 82 weeks in 314 overweight patients with type 2 diabetesDiabetes Obes Metab2006843644716776751
  • ZinmanBHoogwerfBJDuranGSThe effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetesAnn Intern Med200714647748517404349
  • HeineRJVan GaalLFJohnsDMihmMJWidelMHBrodowsRGExenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trialAnn Int Med200514355956916230722
  • BarnettAHBurgerJJohnsDTolerability and efficacy of exenatide and titrated insulin glargine in adult patients with type 2 diabetes previously uncontrolled with metformin or a sulfonylurea: a multinational, randomized, open-label, two-period crossover non-inferiority trialClin Ther2007292333234818158075
  • NauckMADuranSKimDA comparison of twice-daily exenatide and biphasic insulin aspart in patients with type 2 diabetes who were suboptimally controlled with sulfonylurea and metformin: a non-inferiority studyDiabetologia20075025926717160407
  • KimDMacConnellLZhuangDEffects of once-weekly dosing of a long-acting release formulation of exenatide on glucose control and body weight in subjects with type 2 diabetesDiabetes Care2007301487149317353504
  • DruckerDJBuseJBTaylorKDURATION-1 Study Group Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open-label, non-inferiority studyLancet20083721240125018782641
  • KnudsenLBKnudsenSMWilkemMPlasma protein binding of NN2211, a long-acting derivative of GLP-1, is important for its efficacyDiabetes2003A321A322
  • KnudsenLBNielsenPFHuusfeldtPOThe pharmacokinetics, pharmacodynamics, safety and tolerability of NN2211, a new long-acting GLP-1 derivative, in healthy menDiabetologia20024519520211935150
  • VilsbollTZdravokovicMLe-ThiTLiraglutide, a long-acting human glucagon-like peptide 1 analog, given as monotherapy significantly improves glycemic control and lowers body weight without risk of hypoglycemia in patients with type 2 diabetesDiabetes Care2007301608161017372153
  • IrieSMatsumuraYZdravkovicMJacobsenLVKageyamaSTolerability, pharmacokinetics and pharmacodynamics of the once-daily human GLP-1 analog liraglutide in Japanese healthy subjects: a randomised, double-blind, placebo-controlled dose-escalation studyInt J Clin Pharmacol Ther20084627327918541123
  • Liraglutide given positive opinion by EMEA. http://www.novonordisk.com/ Accessed May 7, 2009.
  • GarberAHenryRRatnerRLiraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomized, 52-week, phase III, double-blind, parallel-treatment trialLancet200937347348118819705
  • NauckMFridAHermansenKfor the Lead-2 Study GroupEfficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetesDiabetes Care200932849018931095
  • ZinmanBGerichKBuseJBEfficacy and safety of the human GLP-1 analog liraglutide in combination with metformin and TZD in patients with type 2 diabetes mellitus (LEAD-4 Met+TZD)Diabetes Care2009321224123019289857
  • MarreMShawKBrandleMon behalf of the LEAD-1 SU study groupLiraglutide, a once-daily human GLP-1 analogue, added to a sulphonylurea over 26 weeks produces greater improvements in glycemic and weight control compared with adding rosiglitazone or placebo in subjects with type 2 diabetes (LEAD-1 SU)Diabet Med20092626827819317822
  • Russell-JonesDMolecular, pharmacological and clinical aspects of liraglutide, a once-daily human GLP-1 analogueMol Cell Endocrinol200929713714019041364
  • HarderHNielsenLTuDTAstrupAThe effect of liraglutide, a long-acting glucagon-like peptide 1 derivative, on glycemic control, body composition, and 24-h energy expenditure in patients with type 2 diabetesDiabetes Care2004271915192115277417
  • VilsbollTBrockBPerrildHLiraglutide, a once-daily human GLP-1 analogue, improves pancreatic β-cell function and arginine-stmulated insulin secretion during hyperglycaemia in patients with type 2 diabetesDiabet Med20082515215618201212
  • HorowitzMVilsbøllTZdravkovicMHammerMMadsbadSPatient-reported rating of gastrointestinal adverse effects during treatment of type 2 diabetes with the once-daily human GLP-1 analogue, liraglutideDiab Obes Metab200810593602
  • FinemanMSShenLZTaylorKKimDDBaronADEffectiveness of progressive dose-escalation of exenatide (exendin-4) in reducing dose-limiting side effects in subjects with type 2 diabetesDiabetes Metab Res Rev20042041141715343588
  • BuseJBRosenstockJSestiGThe LEAD-6 Study GroupLiraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6)Lancet2009374394719515413
  • BushMAMatthewsJEDe BoeverEHSafety, tolerability, pharmacodynamics and pharmacokinetics of albiglutide, a long-acting glucagon-like peptide-1 mimetic, in healthy subjectsDiab Obes Metab200911498505
  • MatthewsJEStewartMWDe BoeverEHPharmacodynamics, pharmacokinetics, safety and tolerability of albiglutide, a long-activ glucagon-like peptide-1 mimetic, in patients with type 2 diabetesJ Clin Endocrinol Metab2008934810481718812476
  • NauckMARatnerREKapitzaCBerriaRBoldrinMBalenaRTreatment with the human once-weekly GLP-1 analogue taspoglutide in combination with metformin improves glycemic control and lowers body weight in patients with type 2 diabetes mellitus inadequately controlled with metformin alone: a double-blind placebo-controlled studyDiabetes Care2009321237124319366970
  • DeaconCFHughesTEHolstJJDipeptidyl peptidase IV inhibition potentiates the insulinotropic effect of glucagon-like peptide 1 in the anesthetized pigDiabetes1998477647699588448
  • MarguetDBaggioLKobayashiTEnhanced insulin secretion and improved glucose tolerance in mice lacking CD26Proc Natl Acad Sci U S A2000976874687910823914
  • ÅhrenBLandin-OlssonMJansenPAInhibition of dipeptidyl peptidase-4 reduces glycemia, sustains insulin levels and reduces glucagon levels in type 2 diabetesJ Clin Endocrinol Metab2004892078208415126524
  • HermanGAStevensCVan DyckKPharmacokinetics and pharmacodynamics of sitagliptin, an inhibitor of dipeptidyl peptidase IV, in healthy subjects: results from two randomized, double-blind, placebo-controlled studies with single oral dosesClin Pharmacol Ther20057867568816338283
  • MariASallasWMHeYLVildagliptin, a dipeptidyl peptidase-IV inhibitor, improves model-assessed beta-cell function in patients with type 2 diabetesJ Clin Endocrinol Metab2005904888489415886245
  • RichterBBandeira-EchtlerEBergerhoffKLerchCEmerging role of dipeptidyl peptidase-4 inhibitors in the management of type 2 diabetesVasc Health Risk Man20084753768
  • Januvia Summary of Product Characteristics Merck Sharp and Dohme Ltd. www.medicines.org.uk Accessed January 31, 2009.
  • RazIHanefeldMXuLEfficacy and tolerability of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetesDiabetologia2006492564257117001471
  • CharbonnelBKarasikALiuJWuMMeinitngerGEfficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing metformin therapy in patients with type 2 diabetes inadequately controlled with metformin aloneDiabetes Care2006292638264317130197
  • GoldsteinBFeinglosMNLuncefordJKJohnsonKWilliams-HermanDEEffect of initial combination therapy with sitagliptin, a dipeptidyl peptidase-4 inhibitor, and metformin on glycemic control in patients with type 2 diabetesDiabetes Care2007301979198717485570
  • RosenstockJBrazagRAndryukPJLuKSteinPEfficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin added to ongoing pioglitazone therapy in patients with type 2 diabetes: a 24-week, multicenter, randomized, double-blind, placebo-controlled, parallel-group studyClin Ther20061556156817157112
  • HermansenKKipnesMLuoEFanurikDKhatamiHSteinPEfficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, in patients with type 2 diabetes mellitus inadequately controlled on glimepiride alone or on glimepiride and metforminDiabetes Obes Metab2007973374517593236
  • NauckMAMeiningerGShengDTerranellaLSteinPPEfficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, compared with the sulfonylurea, glipizide, in patients with type 2 diabetes inadequately controlled on metformin alone: a randomized double-blind, non-inferiority trialDiabetes Obes Metab2007919420517300595
  • MigoyaEMMillerJLarsonPSitagliptin, a selective DPP-4 inhibitor, and metformin have complementary effects to increase active GLP-1 concentrationsDiabetes200756A74
  • BergmanAJCoteJYiBEffect of renal insufficiency on the pharmacokinetics of sitagliptin, a dipeptidyl peptidase-4 inhibitorDiabetes Care2007301862186417468348
  • Galvus® Summary of Product CharacteristicsNovartis Pte Ltd. http://emc.medicines.org.uk/ Accessed February 28, 2009.
  • Pi-SunyerFXSchweizerAMillsDDejagerSEfficacy and tolerability of vildagliptin monotherapy in drug-naïve patients with type 2 diabetesDiabetes Res Clin Pract20077613213817223217
  • RosenstockJBaronMADejagerSMillsDSchweizerComparison of vildagliptin and rosiglitazone monotherapy in patients with type 2 diabetes: a 24-week, double-blind, randomized trialDiabetes Care20073021722317259484
  • AhrenBGomisRStandlEtwelve- and 52-week efficacy of the dipeptidyl peptidase IV inhibitor LAF237 in metformin-treated patients with type 2 diabetesDiabetes Care2004272874288015562200
  • BosiECamisascaRPColloberCRochotteEGarberAJEffects of vildagliptin on glucose control over 24 weeks in patients with type 2 diabetes inadequately controlled with metforminDiabetes Care20073089089517277036
  • GarberAJSchweizerABaronMARochotteEDejagerSVildagliptin in combination with pioglitazone improves glycemic control in patients with type 2 diabetes failing thiazolidinedione monotherapy: a randomized, placebo-controlled studyDiabetes Obes Metab2007916617417300592
  • RosenstockJBaronMACamisascaRPEfficacy and tolerability of initial combination therapy with vildagliptin and pioglitzone compared with component monotherapy in patients with type 2 diabetesDiabetes Obes Metab2007917518517300593
  • D’AlessioDADenneyAMHermillerLMTreatment with the dipeptidyl peptidase-4 inhibitor vildagliptin improves fasting islet-cell function in subjects with type 2 diabetesJ Clin Endocrinol Metab200994818818957505
  • MariAScherbaumWANilssonPMCharacterization of the influence of vildagliptin on model-assessed-cell function in patients with type 2 diabetes and mild hyperglycemiaJ Clin Endocrinol Metab20089310310917925336
  • VellaABockGGieslerPDThe effect of dipeptidyl peptidase-4 inhibition on gastric volume, satiation and enteroendocrine secretion in type 2 diabetes: a double-blind, placebo-controlled crossover studyClin Endocrinol (Oxf)20086973774418331607
  • ChanJCScottRArjona FerreiraJCSafety and efficacy of sitagliptin in patients with type 2 diabetes and chronic renal insufficiencyDiabetes Obes Metab20081054555518518892
  • CovingtonPChristopherRDavenportMPharmacokinetic, pharmacodynamic, and tolerability profiles of the dipeptidyl peptidase-4 inhibitor alogliptin: a randomized, double-blind, placebo-controlled, multiple-dose study in adult patients with type 2 diabetesClin Ther20083099512
  • DeFronzoRAFleckPRWilsonCAMekkiQAlogliptin Study 010 Group. Efficacy and safety of the dipeptidyl peptidase-4 inhibitor alogliptin in patients with type 2 diabetes and inadequate glycemic control: a randomized, double-blind, placebo-controlled studyDiabetes Care2008312315231718809631
  • NauckMAEllisGCFleckPRWilsonCAMekkiQAlogliptin Study 008 Group. Efficacy and safety of adding the dipeptidyl peptidase-4 inhibitor alogliptin to metformin therapy in patients with type 2 diabetes inadequately controlled with metformin monotherapy: a multicentre, randomised, double-blind, placebo-controlled studyInt J Clin Pract200963465519125992
  • PratleyREKipnesMSFleckPRWilsonCMekkiQAlogliptin Study 007 GroupDiabetes Obes Metab20091116717619125778
  • MoritohYTakeuchiKAsakawaTKataokaOOdakaHCombining a dipeptidyl peptidase-4 inhibitor, alogliptin, with pioglitazone improves glycaemic control, lipid profiles and beta-cell function in db/db miceBr J Pharmacol200915741542619371350
  • PratleyJEAlogliptin: a new, highly selective dipeptidyl peptidase-4 inhibitor for the treatment of type 2 diabetesExpert Opin Pharmacother20091050351219191685
  • RosenstockJSankohSListJFGlucose-lowering activity of the dipeptidyl peptidase-4 inhibitor saxagliptin in drug-naïve patients with type 2 diabetesDiabetes Obes Metab20081037638818355324
  • DeFronzoRAHissaMBlauwetMBChenRSSaxagliptin added to metformin improves glycemic control in patients with type 2 diabetes [abstract]Diabetes200756Suppl 1A74
  • RavichandranSChacraARTanGHApanovitchAMChenRSaxagliptin added to a sub-maximal dose sulfonylurea is safe and more efficacious than up-titrating a sulfonylurea in patients with type 2 diabetes [abstract]Diabetologia200851Suppl 1S342
  • HollanderPAllenELiJChenRSaxagliptin added to a thiazolidinedione improves glycemic control in patients with inadequately controlled type 2 diabetes [abstract]Diabetologia200851Suppl 1S342
  • DeaconCFHolstJJSaxagliptin: a new dipeptidyl peptidase-4 inhibitor for the treatment of type 2 diabetesAdv Ther20092648849919444391
  • ViswanathanPChaudhuriABhatiaRExenatide therapy in obese patients with type 2 diabetes mellitus treated with insulinEndocr Pract20071344445017872344
  • RatnerREMaggsDNielsenLLLong-term effects of exenatide therapy over 82 weeks on glycaemic control and weight in overweight metformin-treated patients with type 2 diabetes mellitusDiabetes Obes Metab2006841942816776749
  • KlonoffDCBuseJBNielsenLLExenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 yearsCurr Med Res Opin20082427528618053320
  • SonneDPEngstromTTreimanMProtective effects of GLP-1 analogues exendin-4 and GLP-1 (9–36) amide against ischemia-reperfusion injury in rat heartRegul Pept200814624324917976835
  • Noyan-AshrafMHBanKSadiAThe GLP-1R agonist liraglutide protects cardeomyocytes and improved survival and cardiac function after experimental murine infarction [abstract 190-OR]Presented at the 68th Scientific Sessions of the American Diabetes AssociationSan Francisco, CA66–102008
  • CourregesJPVilsbollTZdravkovicMBeneficial effects of once-daily liraglutide, a human glucagon-like peptide-1 analogue, on cardiovascular risk biomarkers in patients with Type 2 diabetesDiabet Med2008251129113119183322
  • MistryGCMaesALLasseterKCEffect of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on blood pressure in nondiabetic patients with mild to moderate hypertensionJ Clin Pharmacol20084859259818353996
  • ScottRWuMSanchezMEfficacy and tolerability of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy over 12 weeks in patients with type 2 diabetesInt J Clin Pract20076117118017156104
  • BolliGDottaFRochotteEEfficacy and tolerability of vildagliptin vs. pioglitazone when added to metformin: a 24-week, randomized, double-blind studyDiabetes Obes Metab200810829018034842
  • MatikainenNMänttäriSSchweizerAVildagliptin therapy reduces postprandial intestinal triglyceride-rich lipoprotein particles in patients with type 2 diabetesDiabetologia2006492049205716816950
  • BoschmannMEngeliSDobbersteinKDipeptidyl-peptidase-IV inhibition augments postprandial lipid mobilization and oxidation in type 2 diabetic patientsJ Clin Endocrinol Metab20099484685219088168
  • JonesKLRussoABerryMKA longitudinal study of gastric emptying and upper gastrointestinal symptoms in patients with diabetes mellitusAm J Med200211344945512427492
  • Delgado-ArosSVellaACamilleriMEffects of glucagon-like peptide 1 and feeding on gastric volumes in diabetes mellitus with cardio-vagal dysfunctionNeurogastroenterol Motil20071543544312846732
  • Food and Drug Administration Information for Healthcare Professionals Exenatide (marketed as Byetta)www.fda.gov/cder/drug/InfoSheets/HCP/exenatideHCP.htm Accessed August 3, 2009.
  • NoelRABraunDKPattersonREBloomgrenGIncreased risk of acute pancreatitis and biliary disease observed in patients with type 2 diabetes: a retrospective cohort studyDiabetes Care20093283483819208917
  • http://www.fda.gov/ohrms/dockets/ac/09/briefing/2009-4422b-2-02-NovoNordisk.pdf8995Accessed May 8, 2009
  • KoehlerJABaggioLLLamontBJAliSDruckerDJGLP-1 receptor activation modulates pancreatitis-associated gene expression but does not modify the susceptibility to experimental pancreatitis in miceDiabetes200969[Epub ahead of print]
  • MatveyenkoAVDrySCoxHIBeneficial endocrine but adverse exocrine effects of sitagliptin in the human islet amyloid polypeptide transgenic rat model of type 2 diabetes: interactions with metforminDiabetes2009581604161519403868
  • KiefferTJMcIntoshCHPedersonRADegradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IVEndocrinology1995136358535967628397
  • De MeesterILambeirAMProostPDipeptidyl peptidase IV substrates: an update on in vitro peptide hydrolysis by human DPPIVAdv Exp Med Biol200352431712675218
  • BussoNWagtmannNHerlingCCirculating CD26 is negatively associated with inflammation in human and experimental arthritisAm J Pathol200516643344215681827
  • MinshallMEOglesbyAKWintleMEEstimating the long-term cost-effectiveness of exenatide in the United States: an adjunctive treatment for type 2 diabetesValue Health200811223318237357
  • RayJABoyeKSYurginNExenatide versus insulin glargine in patients with type 2 diabetes in the UK: a model of long-term clinical and cost outcomesCurr Med Res Opin20072360962217355742
  • WoehlAEvansMTetlowAPMcEwanPEvaluation of the cost effectiveness of exenatide versus insulin glargine in patients with sub-optimally controlled type 2 diabetes in the United KingdomCardiovasc Diabetol200872418694484
  • BoyeKSMatzaLSOglesbyAPatient-reported outcomes in a trial of exenatide and insulin glargine for the treatment of type 2 diabetesHealth Qual Life Outcomes200648017034640
  • SchwarzBGouveiaMChenJCost-effectiveness of sitagliptin-based treatment regimens in European patients with type 2 diabetes and haemoglobin A1c above target on metformin monotherapyDiab Obes Metab2008104355
  • ScottRLoeysTDaviesMJEngelSSEfficacy and safety of sitagliptin when added to ongoing metformin therapy in patients with type 2 diabetesDiabetes Obes Metab20081095996918201203
  • LageMJFabunmiRBoyeKSMisurskiDAComparison of costs among patients with type 2 diabetes treated with exenatide or sitagliptin therapyAdv Ther20092621722919219409
  • JellingerPSDavidsonJABlondeLRoad maps to achieve glycemic control n type 2 diabetes mellitusEndocr Pract200713261268
  • NathanDMBuseJBDavidsonMBMedical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapyDiabetes Care200831111
  • NauckMAWalbergJVethackeABlood glucose control in healthy subject and patients receiving intravenous glucose infusion or total parenteral nutrition using glucagon-like peptide 1Regul Pept2004118899714759561
  • MeierJJWeyheDMichaelyMIntravenous glucagon-like peptide 1 normalizes blood glucose after major surgery in patients with type 2 diabetesCrit Care Med20043284885115090972
  • DeaneAMChapmanMJFraserRJBurgstadCBesankoLKHorowitzMThe effect of exogenous glucagon-like peptide-1 on the glycaemic response to small intestinal nutrient in the critically ill: a randomised double-blind placebo controlled cross over studyCrit Care200913R67(Epub May 14)19439067
  • NICE-SUGAR Study InvestigatorsFinferSChittockDRSuSYIntensive versus conventional glucose control in critically ill patientsN Engl J Med20093601283129719318384
  • AscheCVMcAdam-MarxCShane-McWhorterLEvaluation of adverse events of oral antihyperglycemic monotherapy experienced by a geriatric population in a real-world setting: a retrospective cohort analysisDrugs Aging20082561162218582148
  • HanefeldMHermanGAWuMOnce-daily sitagliptin, a dipeptidyl peptidase-4 inhibitor, for the treatment of patients with type 2 diabetesCurr Med Res Opin2007231329133917559733
  • PratleyRERosenstockJPi-SunyerFXManagement of type 2 diabetes in treatment-naive elderly patients: benefits and risks of vildagliptin monotherapyDiabetes Care2007303017302217878242
  • MigoyaEMStevensCHBergmanAJEffect of moderate hepatic insufficiency on the pharmacokinetics of sitagliptinCan J Clin Pharmacol200916e165e17019221403
  • ChanJCScottRArjona FerreiraJCSafety and efficacy of sitagliptin in patients with type 2 diabetes and chronic renal insufficiencyDiabetes Obes Metab20081054555518518892