Glucoregulatory Hormones and GLP-1/GIP Receptor Agonists

Reviewed on August 08, 2024

Introduction

The quest for more physiologic approaches to augment insulin treatment prompted investigation of glucoregulatory hormones. These include the gut-derived hormones, such as the potent incretins glucagon-like polypeptide-1 (GLP-1) and gastric inhibitory peptide (GIP) and the β-cell hormone amylin.

Incretins

A potential role for intestinal peptides in the treatment of type 2 diabetes (T2D) was based on the observation that insulin responses to an oral glucose load exceeded those after IV glucose administration measured at the same blood glucose concentration. This so-called incretin effect is attributed to the insulinotropic action of gut hormones, particularly:

  • Glucose-dependent insulinotropic polypeptide (GIP)
  • Glucagon-like peptide 1 (GLP-1).

Since the glucose-lowering effects of GIP have been shown to be impaired in patients with T2D, interest has focused on developing antidiabetic therapies based upon the actions of GLP-1 and GIP, the response to which remains intact.…

Introduction

The quest for more physiologic approaches to augment insulin treatment prompted investigation of glucoregulatory hormones. These include the gut-derived hormones, such as the potent incretins glucagon-like polypeptide-1 (GLP-1) and gastric inhibitory peptide (GIP) and the β-cell hormone amylin.

Incretins

A potential role for intestinal peptides in the treatment of type 2 diabetes (T2D) was based on the observation that insulin responses to an oral glucose load exceeded those after IV glucose administration measured at the same blood glucose concentration. This so-called incretin effect is attributed to the insulinotropic action of gut hormones, particularly:

  • Glucose-dependent insulinotropic polypeptide (GIP)
  • Glucagon-like peptide 1 (GLP-1).

Since the glucose-lowering effects of GIP have been shown to be impaired in patients with T2D, interest has focused on developing antidiabetic therapies based upon the actions of GLP-1 and GIP, the response to which remains intact. Combination (dual) GLP-1 and GIP receptor agonism has recently emerged as a promising therapeutic avenue.

GLP-1 is produced by the tissue-specific cleavage of proglucagon into several smaller peptides in the L-cells of the intestinal mucosa and GIP is synthesized by K cells found in the intestinal mucosa (Figure 19-1). The insulino­tropic functions of GLP-1 are mediated through interaction with GLP-1 receptors expressed on pancreatic β cells. The release of GLP-1 in response to a meal is rapid (within 10 minutes) and highly correlated with the release of insulin and amylin. In healthy subjects, postprandial levels of GLP-1 rise, whereas patients with T2D or impaired glucose tolerance (IGT) exhibit reduced levels of meal-stimulated circulating GLP-1.

Exogenous GLP-1 has the potential to normalize fasting and PPG levels in patients with T2D. One study in patients with T2D demonstrated that intravenously administered GLP-1 significantly reduced fasting blood glucose (FBG) and, in a glucose-dependent manner, both enhanced insulin secretion and suppressed glucagon secretion (Figure 19-2). Another study found that subcutaneously (SC) administered GLP-1 normalized postprandial glucose (PPG), enhanced glucose-mediated insulin secretion and delayed gastric emptying. Importantly, the insulinotropic and glucagonostatic actions of GLP-1 are strictly glucose dependent. Therefore, GLP-1 should not induce hypoglycemia when administered alone or with other antidiabetic agents that do not cause hypoglycemia, such as metformin (MET), thiazolidinediones (TZDs) and alpha-glucosidase inhibitors (AGIs). Dipeptidyl peptidase-4 (DPP-4) inhibitors do not cause hypoglycemia; however, they are not used concomitantly with GLP-1 receptor agonists (GLP-1 RAs).

Significant acute reductions in appetite and food intake were initially observed after intravenous (IV) administration of GLP-1 both in healthy individuals and in patients with T2D. The weight effects of GLP-1 therapy are potentially significant for the treatment of T2D, in which excessive caloric intake and weight management are important issues that are difficult to control.

In addition to these well-characterized metabolic activities, a number of novel biologic functions of GLP-1 have been shown to occur in animals and include:

  • Regulation of β-cell genes involved in glucose sensing and insulin biosynthesis and secretion
  • Expansion of β-cell mass through the proliferation and neogenesis of pancreatic cells
  • Potentiation of glucose uptake in peripheral tissues.

Several of these effects occur in humans treated with GLP-1 agonists or analogues, and studies that looked at surrogate markers of β-cell health are promising. The collective glucoregulatory actions of GLP1 make this peptide a potential powerful agent for the treatment of diabetes. Despite these features, the therapeutic potential of GLP1 has been limited by its rapid and extensive degradation. In humans, the half-life of GLP-1 is <2 minutes due to rapid degradation as a result of N-terminal cleavage by the ubiquitous enzyme, DPP-4. Elimination is principally via the kidneys.

Enlarge  Figure 19-1: Schematic Model of the Multihormonal Control of Glucose Homeostasis. The multihormonal control of glucose homeostasis is regulated by a complex interplay of several gut and islet hormones, including insulin, amylin, glucagon, and incretins.
Figure 19-1: Schematic Model of the Multihormonal Control of Glucose Homeostasis. The multihormonal control of glucose homeostasis is regulated by a complex interplay of several gut and islet hormones, including insulin, amylin, glucagon, and incretins.
Enlarge  Figure 19-2: Plasma Glucose, Insulin, and Glucagon Responses to the IV Administration of GLP-1 or Placebo in Patients with Type 2 Diabetes. Data are mean ± SE. <sup>a</sup> <em>P</em> <0.05. Source: Nauck MA, et al. <em>Diabetologia</em>. 1993;36:741-744.
Figure 19-2: Plasma Glucose, Insulin, and Glucagon Responses to the IV Administration of GLP-1 or Placebo in Patients with Type 2 Diabetes. Data are mean ± SE. a P <0.05. Source: Nauck MA, et al. Diabetologia. 1993;36:741-744.

GLP-1 Analogues

One approach to utilizing the therapeutic potential of GLP-1 has been the development of incretin mimetics. This has resulted in the production of GLP-1 analogues and GLP-1–like compounds that exhibit increased resistance to DPP-4 degradation. Several GLP-1 analogs and a GLP-receptor agonist that exhibit increased resistance to DPP-4 degradation, as well as DPP-4 inhibitors, have been developed. The GLP-1 receptor agonist exenatide (synthetic exendin-4) was the first agent of the incretin mimetic class to be approved by the Food and Drug Administration (FDA). More recently, the GLP-1 analogs liraglutide, dulaglutide, lixisenatide, semaglutide (oral and injectable formulations) and tirzepatide have also been approved.

Exenatide (Byetta)

Exendin-4 is a naturally occurring incretin mimetic that was originally isolated from the salivary secretions of the lizard Heloderma suspectum (Gila monster). Exendin-4 and mammalian GLP-1 have an approximately 50% amino acid sequence identity. However, exendin-4 and GLP-1 are transcribed from distinct genes and, therefore, the exendin-4 gene is not the Gila monster homologue of the mammalian proglucagon gene from which GLP-1 is expressed. In mammals, exendin-4 exhibits antidiabetic activities similar to those of native GLP-1 but is resistant to degradation by DPP-4, and this contributes to its longer half-life compared with GLP-1.

Exenatide is indicated as an adjunct to diet and exercise to improve glycemic control in adults with T2D.

Phase 3 Clinical Trials

Three 30-week, double-blind, placebo-controlled trials were conducted to evaluate the safety and efficacy of exenatide in patients with T2D unable to achieve adequate glycemic control with MET alone, an SFU alone, or MET in combination with an SFU. A total of 1446 patients were randomized in these three trials. The mean A1C values at baseline for the trials ranged from 8.2% to 8.7%. After a 4-week placebo lead-in period, patients were randomly assigned to receive exenatide 5 mcg bid, exenatide 10 mcg bid, or placebo before the morning and evening meals, in addition to their current oral antidiabetic agent. All patients randomized to exenatide began a treatment initiation period with 5 mcg bid for 4 weeks. After 4 weeks, those patients either continued to receive exenatide 5 mcg bid or had their dose increased to 10 mcg bid. Patients assigned to placebo received placebo bid throughout the study. The primary end point in each study was mean change from baseline A1C at 30 weeks. The results of the 30-week pivotal trials are summarized in Table 19-1.

The average reduction in A1C across the three phase 3 pivotal trials in patients completing the 30-week studies on the highest dose of exenatide (10 mg twice daily) approached 1%. Additionally, approximately 35% of these patients achieved A1C measurements ≤7%. On average, subjects in the phase 3 trials on the highest dose of exenatide also showed reductions in body weight of approximately 2 kg. Exenatide was generally well tolerated and the most common adverse event reported was mild-to-moderate nausea, which occurred most frequently early in the study. Patients taking exenatide in combination with an sulfonylurea (SFU) had an increased risk of hypoglycemia. When exenatide was used in combination with MET, no increase in hypoglycemia was seen over that of placebo in combination with MET. In general, hypoglycemia rates were consistent with the glucose-dependent action of exenatide, with no difference between the placebo and drug arms. In all three pivotal trials, observed antibody formation to exenatide was consistent with data reported to date. These data do not suggest an influence of antibody formation on exenatide’s glucose-lowering effect.

A randomized, placebo-controlled, double-blind trial was performed in 233 subjects with elevated A1C (in spite of therapy) with a TZD alone (20%) or with a TZD and MET (80%). Patients were randomized to receive subcutaneous (SC) injections of placebo or exenatide 5 mcg for 4 weeks then 10 mcg bid for 16 weeks in addition to their previous regimen (Figure 19-3). Exenatide therapy decreased A1C by -0.8 from a baseline of 7.9% (Figure 19-3-A). With exenatide, 62% of subjects achieved A1C ≤7% vs 16% with placebo (Figure 19-3-B). Fasting serum glucose with exenatide was -1.5 mmol/L (-27 mg/dL) lower than with placebo). Exenatide also significantly reduced body weight from baseline compared with placebo (-1.5 kg vs -0.2 kg respectively) (Figure 19-3-C). Nausea was the most frequent adverse event (40% vs 15%, exenatide vs placebo). There was no significant difference in the incidence of hypoglycemia between the two treatments.

Comparative and Long-Term Open-Label Trials
Exenatide vs Insulin Glargine

A 26-week, randomized, active-controlled noninferiority study was performed to determine if exenatide could be used safely and effectively as an alternative to basal insulin glargine in 551 patients being treated with MET and an SFU. Patients were randomized to receive exenatide (5 mcg bid for the first 4 weeks, then 10 mcg bid for the remainder of the study) or insulin glargine qd (one daily dose titrated to maintain FBG levels of 5.6 mmol/L [<100 mg/dL]). At end point, similar A1C reductions were achieved with exenatide and glargine (-1.1% and -1.1%, respectively) consistent with the noninferiority study design (Figure 19-4-A). As measured by 7-point glucose monitoring, exenatide reduced postprandial excursions following breakfast and dinner, whereas the predominant effect of glargine was a reduction of fasting glucose. Mean weight changes from baseline were -2.5 kg (5.5 lb) with exenatide and +1.8 kg (3.9 lb) with glargine (Figure 19-4-B). GI symptoms were more common in the exenatide group than in the insulin glargine group, including nausea (57.1% vs 8.6%), vomiting (17.4% vs 3.7%), and diarrhea (8.5% vs 3.0%). Nausea (57%), generally mild to moderate with a decreasing incidence during the study, was the most common adverse event with exenatide and resulted in a 6% discontinuation rate. The rates of symptomatic hypoglycemia were similar with the two treatments except that nocturnal hypoglycemia was significantly less common with exenatide (0.9 vs 2.4 mean events/patient/year).

The ability of exenatide to improve glycemic control with minimal weight gain in overweight patients with T2D compared with insulin glargine was assessed in the Helping Evaluate Exenatide in Overweight Patients With Diabetes Compared With Long-Acting Insulin (HEELA) study. Patients (mean BMI 34.1 kg/m2) inadequately controlled on two or three OADs and with elevated CV risk factors were randomized to open-label add-on treatment with exenatide 5-10 mcg bid (n = 118) or insulin glargine qd titrated to fasting plasma glucose ≤5.6 mmol/L [100.8 mg/dL] (n = 117). The primary outcome was a composite of A1C ≤7.4% and weight gain ≤1 kg. After 26 weeks, a significantly greater proportion of patients in the exenatide group achieved the composite end point compared with those in the insulin glargine group (53.4% vs 19.8%, respectively). Although there was no significant difference in the reductions in A1C with exenatide and insulin glargine (-1.25% and -1.26%, respectively), whereas mean body weight decreased in the exenatide group but increased in the insulin glargine group (-2.71 vs +2.98 kg, respectively). There were more treatment-related adverse events with exenatide but a lower incidence of nocturnal hypoglycemia, with no differences in overall or severe hypoglycemia.

A 1-year, randomized, open-label study in 69 MET-treated patients with T2D found that exenatide significantly improved β-cell function (as assessed by arginine-stimulated hyperglycemic clamp) during 1 year of treatment compared with titrated insulin glargine. After cessation of both exenatide and insulin glargine therapy, the β-cell function, A1C and body weight returned to pretreatment values, suggesting that ongoing treatment is necessary to maintain the beneficial effects of either therapy.

82-Week Studies

Several published analyses reported the effects of exenatide on A1C and weight over 82 weeks in patients with T2D who had participated in prior 30-week phase 3 clinical trials and a subsequent 52-week open-label extension. One post hoc analysis examined changes in A1C and weight in 393 patients. During the open-label extensions, all patients received exenatide 5 mcg bid for 4 weeks followed by exenatide 10 mcg bid. All patients continued their MET, SFU, or MET plus SFU. In patients who were treated with exenatide 10 mcg bid for a total of 82 weeks, the reduction from baseline of A1C was -1.1% and the progressive reduction in weight was -4.5 kg or 9.9 lb (Figure 19-5). At week 82, patients who received placebo during the first 30 weeks prior to subsequent exenatide 10 mcg bid had reductions in A1C of -1.2% as well as reductions in weight of -3.3 kg from their 30-week baseline.

Another analysis examined pooled data from a larger cohort of 314 overweight patients (weight 99 ± 21 kg, BMI 34 ± 6 kg/m2) with T2D who received exenatide in addition to an SFU and/or MET in a 30-week placebo-controlled trial and subsequently in an additional 52-week open-label uncontrolled extension study. Patients continued their SFU and/or MET regimens throughout. In these 314 patients who completed 82 weeks of exenatide treatment, reduction in A1C from baseline to week 30 (-0.9%) was sustained to week 82 (-1.1%), with 48% of patients achieving A1C ≤7% at week 82. At week 30, exenatide reduced body weight from baseline (-2.1 kg), with progressive reduction at week 82 (-4.4 kg) (Figure 19-6). In addition, patients who completed 82 weeks of exenatide showed statistically significant improvement in some CV risk factors.

Dosage and Administration

Exenatide therapy should be initiated at 5 mcg admin­­istered twice daily at any time within the 60-minute period before the morning and evening meals (or before the two main meals of the day, approximately 6 hours or more apart) (Table 19-2). Exenatide should not be administered after a meal. Based on clinical response, the dose of exenatide can be increased to 10 mcg twice daily after 1 month. Each dose should be administered as an SC injection in the thigh, abdomen, or upper arm. Exenatide is not recommended in patients with severe GI disease, ESRD, or severe renal impairment (CrCl <30mL/min).

Exenatide Extended-Release Formulation

An extended-release (ER) formulation of exenatide (Bydureon) was recently approved by the FDA as an adjunct to diet and exercise to improve glycemic control in adults and pediatric patients aged 10 years and older with T2D in multiple clinical settings. Whereas the regular exenatide formulation is administered twice daily, this ER formulation is administered every 7 days.

ER exenatide has been studied as monotherapy and in combination with MET, an SFU, a TZD, a combination of MET and an SFU, or a combination of MET and a TZD. The concurrent use of ER exenatide with insulin has not been studied and cannot be recommended.

The efficacy and safety of ER exenatide 2 mg once every 7 days and regular exenatide 10 mcg twice daily were compared in a 24-week, randomized, open-label trial in 252 patients with T2D and inadequate glycemic control with diet and exercise alone (19%), a single OAD agent (47%), or a combination of OAD agents (35%). As shown in Table 19-3, the reductions in A1C and FPG were significantly greater with ER exenatide compared with regular exenatide. In addition, reductions from mean baseline (97/94 kg) in body weight were observed with both ER exenatide (-2.3 kg) and regular exenatide (-1.4 kg). In this study, the most common adverse events were nausea (ER exenatide 14.0%; regular exenatide 5.0%), diarrhea (ER exenatide 9.3%; regular exenatide 4.1%)cand injection site erythema (ER exenatide 5.4%; regular exenatide 2.4).

Prescribing Extended-Release Exenatide

ER exenatide (2 mg per dose) should be administered via SC injection once every 7 days (weekly). The dose can be administered at any time of day, with or without meals. Prior treatment with regular exenatide is not required when initiating the ER formulation. Patients changing from the ER to the immediate-release formulation of exenatide may experience transient (approximately 2 weeks) elevations in blood glucose concentrations. ER exenatide should not be used in patients with severe renal impairment (creatinine clearance (CrCl) <30 mL/min) or end-stage renal disease (ESRD). ER exenatide is contraindicated in patients with personal or family history of medullary thyroid carcinoma or in patients with Multiple Endocrine Neoplasia syndrome type 2.

Enlarge  Figure 19-3: Adjunctive Treatment With Exenatide or Placebo in Patients With Type 2 Diabetes Not Adequately Controlled With a Thiazolidinedione Alone or in Combination With Metformin. <sup>a</sup> ITT patient sample; mean + SE. <sup>b </sup>Per-protocol patient sample: randomized patients who had no protocol violations and either completed the protocol or received at least 16 weeks of treatment. Baseline A1C >7% or 6.5%. <sup>c </sup>Baseline body weight (mean ± SD): exenatide, 97.5 ± 18.9 kg; placebo, 96.9 ± 19.0 kg. Source: Zinman B, et al. <em>Ann Intern Med</em>. 2007;146:477-485.
Figure 19-3: Adjunctive Treatment With Exenatide or Placebo in Patients With Type 2 Diabetes Not Adequately Controlled With a Thiazolidinedione Alone or in Combination With Metformin. a ITT patient sample; mean + SE. b Per-protocol patient sample: randomized patients who had no protocol violations and either completed the protocol or received at least 16 weeks of treatment. Baseline A1C >7% or 6.5%. c Baseline body weight (mean ± SD): exenatide, 97.5 ± 18.9 kg; placebo, 96.9 ± 19.0 kg. Source: Zinman B, et al. Ann Intern Med. 2007;146:477-485.
Enlarge  Figure 19-4: Exenatide/Insulin Glargine Comparator Trial Intent-to-treat population; mean ± SE shown. <sup>a </sup><em>P</em> <0.0001, exenatide vs insulin glargine at same time point. Source: Heine RJ, et al. <em>Ann Intern Med</em>. 2005;143:559-569.
Figure 19-4: Exenatide/Insulin Glargine Comparator Trial Intent-to-treat population; mean ± SE shown. a P <0.0001, exenatide vs insulin glargine at same time point. Source: Heine RJ, et al. Ann Intern Med. 2005;143:559-569.
Enlarge  Figure 19-5: Open-Label Extension<sup>a</sup>: Sustained A1C Reductions and Progressive Body Weight Reductions With Exenatide Therapy Over 82 Weeks. Mean (SE); <em>N</em> = 393; completer population; 82-week data. <sup>a</sup>	All subjects received exenatide 10 mcg bid. <sup>b</sup> Baseline A1C 8.3% in all three groups. <sup>c</sup> Weight change was a secondary end point. Baseline weight: placebo, 98 kg; 5 mcg, 98 kg; 10 mcg, 100 kg. Source: Data on file: Amylin Pharmaceuticals, Inc.
Figure 19-5: Open-Label Extensiona: Sustained A1C Reductions and Progressive Body Weight Reductions With Exenatide Therapy Over 82 Weeks. Mean (SE); N = 393; completer population; 82-week data. a All subjects received exenatide 10 mcg bid. b Baseline A1C 8.3% in all three groups. c Weight change was a secondary end point. Baseline weight: placebo, 98 kg; 5 mcg, 98 kg; 10 mcg, 100 kg. Source: Data on file: Amylin Pharmaceuticals, Inc.
Enlarge  Figure 19-6: Changes in Body Weight Over 82 Weeks of Treatment With Exenatide and Metformin, a Sulfonylurea, or Both in Patients Who Previously Participated in 30-Week Phase 3 Trials. Source: Blonde L, et al. <em>Diabetes Obes Metab</em>. 2006:8:436-447.
Figure 19-6: Changes in Body Weight Over 82 Weeks of Treatment With Exenatide and Metformin, a Sulfonylurea, or Both in Patients Who Previously Participated in 30-Week Phase 3 Trials. Source: Blonde L, et al. Diabetes Obes Metab. 2006:8:436-447.

Liraglutide (Victoza)

Liraglutide is an acylated GLP-1 analog that shares 97% amino acid sequence identity with native GLP-1. Liraglutide self-associates into a heptameric structure that delays absorption from the SC injection site and slows its metabolism, thereby resulting in a prolonged elimination half-life compared with native GLP-1. This structural modification also results in resistance to GLP-1 inactivation by DPP-4. This pharmacokinetic profile allows for once-daily administration. Liraglutide reduces both FPG and PPG levels, an effect that lasts throughout a 24-hour dosing interval. It increases insulin secretion, reduces postprandial glucagon secretion, delays gastric emptying and improved β-cell function. Its use has been associated with reductions in body weight and SBP.

Liraglutide is indicated as an adjunct to diet and exercise to improve glycemic control in patients 10 years and older with T2D. It is also indicated to reduce the risk of major adverse cardiovascular (CV) events in adults with T2D and established cardiovascular disease (CVD). This latter indication was the result of the positive cardiovascular outcomes trial (CVOT) discussed below. It is administered once daily at any time of day, independently of meals. It should be initiated at a dose of 0.6 mg per day for 1 week, following by a dose increase to 1.2 mg per day. Results from phase 3 clinical trials have demonstrated that liraglutide can be used as monotherapy, or in combination with one or two oral antidiabetic drugs (OADs), such as MET, SFUs, or TZDs. When using liraglutide with insulin, administer as separate injections in non-adjacent areas. The concurrent use of liraglutide and prandial insulin has not been studied although it is commonly employed. In addition, liraglutide and other GLP-1 RAs can be used with sodium glucose cotransporter type 2 (SGLT2) inhibitors.

Phase 3 Clinical Trials

The efficacy and safety of liraglutide were assessed in the Liraglutide Effect and Action in Diabetes (LEAD) phase 3 program consisting of six large, multicenter, randomized trials of 26- or 52-weeks in duration that enrolled >4000 adult patients with T2D (Table 19-4). Three of the trials included both placebo and active control groups, while others were either placebo- or active-controlled. Four of the trials used double-blind, double-dummy methodology; however, LEAD-6 was open-label and the insulin glargine arm in LEAD-5 was open-label since the basal insulin dose was individually titrated. LEAD-3 compared liraglutide monotherapy with glimepiride monotherapy in patients with early-stage diabetes who had not achieved adequate glycemic control with diet and exercise or a single OAD at up to half the maximum approved dose. Change from baseline in A1C was the primary efficacy end point in all LEAD trials. Secondary end points included change from baseline in fasting plasma glucose (FPG), postprandial glucose ( PPG) and body weight.

Monotherapy

In the LEAD-3 trial, both doses of liraglutide as monotherapy demonstrated significant improvements in glycemic control in comparison with glimepiride (Table 19-5). In addition, A1C reductions from baseline were significantly greater with liraglutide 1.8 mg than with liraglutide 1.2 mg (between-treatment difference of -0.29%). By week 52, significantly more patients in each of the liraglutide groups achieved the target A1C <7%. A1C levels tended to decrease over the first 8 to 12 weeks of treatment with liraglutide. They then remained stable until 52 weeks in the liraglutide 1.8-mg group but increased slightly between week 12 and week 52 with liraglutide 1.2 mg or glimepiride treatment. Patients previously treated with diet and exercise had greater decreases in A1C than did those who were switched from an OAD to liraglutide. Significantly greater reductions in body weight with both doses of liraglutide were also observed compared an increase with glimepiride.

The LEAD-3 trial was extended for an additional 52 weeks of open-label treatment during which 440 of the original study population entered the extension period of whom 321 completed the full 2 years of monotherapy with liraglutide. Of these patients, 110 received liraglutide 1.2 mg, 114 received liraglutide 1.8 mg and 97 received glimepiride. After 2 years, the mean reductions in A1C with both doses of liraglutide were significantly greater that those of glimepiride (-0.9% and -1.1% vs -0.6, respectively). The proportion of patients achieving A1C <7% were significantly greater with liraglutide 1.2 mg and 1.8 mg than with glimepiride (53% and 58% vs 37%). Reductions in FPG also were significantly greater with both doses of liraglutide than with glimepiride. Whereas there was a mean increase in body weight with glimepiride (+1.1 kg), body weight decreased with both doses of liraglutide (-2.1 and -2.7 kg).

In Combination With One OAD

Liraglutide in combination with either rosiglitazone or glimepiride was evaluated in two 26-week, double-dummy, placebo-controlled and active-controlled studies (Table 19-4). In LEAD-1, liraglutide used in combination with glimepiride reduced A1C to a significantly greater degree than placebo plus glimepiride or rosiglitazone plus glimepiride (Figure 19-7-A). The reductions in A1C were greater for patients previously treated with monotherapy compared with combination therapy. However, because the increase with placebo was higher for individuals entering from combination therapy (0.7% vs 0.23%), the differences between treatment groups in favor of liraglutide were similar irrespective of whether individuals were treated previously with monotherapy or combination therapy. A significantly greater proportion of patients treated with liraglutide 0.6, 1.2, or 1.8 mg (24%, 35% and 42%, respectively) achieved A1C <7% compared with 8% of those who received placebo with glimepiride. The proportions were also significantly greater with liraglutide 1.2 or 1.8 mg (35% and 42%, respectively) compared with that in the rosiglitazone plus glimepiride group (22%). At week 26, all doses of liraglutide decreased FPG significantly more than did placebo (Figure 19-7-B), while only liraglutide 1.2 or 1.8 mg produced greater reductions than rosiglitazone. Changes in body weight with liraglutide 0.6, 1.2 and 5 mg were significantly less than with rosiglitazone (liraglutide +0.7, +0.3, -0.2 kg vs rosiglitazone +2.1 kg, respectively).

In the LEAD-2 trial, reductions in A1C with all three doses of liraglutide plus MET and glimepiride plus MET were significantly greater than with placebo plus MET (Table 19-6). The reductions with liraglutide were similar to that with glimepiride; specifically, they were statistically noninferior to glimepiride. The proportions of patients reaching A1C <7% were significantly greater with all doses of liraglutide plus MET compared with placebo plus MET. The proportion of patients in the liraglutide 1.8 mg plus MET group was significantly greater than in the glimepiride plus MET group. Although the proportion of patients with A1C <7% in the glimepiride plus MET group was similar to those receiving liraglutide, the P value compared with placebo plus MET was not reported. FPG levels decreased from baseline in the liraglutide plus MET groups as well as in the glimepiride group, whereas FPG increased with placebo plus MET. While body weight decreased in the liraglutide plus MET and placebo plus MET groups, there was an increase in the glimepiride plus MET group.

In Combination With Two OADs

The LEAD-4 and LEAD-5 trials assessed the efficacy of liraglutide as add-on to two OADs: MET and rosiglitazone or MET and glimepiride, respectively (Table 19-4).

In LEAD-4, A1C values decreased significantly more in the liraglutide 1.2 and 1.8 mg plus MET and rosiglitazone groups vs placebo plus MET and rosiglitazone. Mean A1C values with both doses of liraglutide decreased from baseline within the first 12 weeks of the study and thereafter remained steady throughout the rest of the trial (Figure 19-8-A). By week 26, 57.5% and 53.7% of patients in the 1.2- and 1.8-mg liraglutide groups, respectively, had an A1C of <7% compared with 28.1 % in the placebo group. FPG values decreased within 2 weeks of randomization with liraglutide and remained relatively stable thereafter, while there was a significantly smaller decrease with placebo (Figure 19-8-B). In addition, 90-minute PPG at the end of the study decreased from baseline in all treatment groups by -47 mg/dL for 1.2-mg liraglutide, -49 mg/dL 1.8-mg liraglutide and -14 mg/dL for placebo. Dose-dependent weight loss was observed with 1.2- and 1.8-mg liraglutide (-l.0 and -2.0 kg, respectively) compared with weight gain with placebo (+0.6 kg). The weight loss with liraglutide 1.8 mg was significantly greater than with liraglutide 1.2 mg.

The LEAD-5 trial compared liraglutide 1.8 mg qd, placebo, or open-label insulin glargine, all as add-on to MET and glimepiride. After 26 weeks, the addition of liraglutide 1.8 mg reduced A1C significantly compared with the addition of placebo or insulin glargine. Significantly more patients achieved an A1C <7% with liraglutide than with placebo. FPG levels decreased to a similar degree with liraglutide and insulin glargine, while they increased in the placebo group. There was greater weight loss with liraglutide vs placebo and insulin glargine.

Liraglutide Compared With Exenatide

Liraglutide 1.8 mg qd was compared with exenatide 10 mcg bid, both added to previous treatment with maximally tolerated, but inadequate, doses of MET, an SFU, or both, in a randomized, open-label trial (LEAD-6). By week 26, liraglutide reduced A1C significantly more than exenatide (1.0% vs 0.79%) (Figure 19-9-A), and more patients reached A1C <7% with liraglutide than with exenatide (54% vs 43%). Liraglutide treatment also resulted in a greater reduction in FPG than with exenatide (-1.61 mmol/L [-29.0 mg/dL] vs -0.60 mmol/L [-10.8 mg/dL]; P <0.0001) (Figure 19-9-B). Weight losses from baseline were similar with both liraglutide and exenatide (-3.24 kg and -2.87 kg).

Safety and Tolerability

Throughout the LEAD trials, liraglutide was generally well tolerated, with most adverse events reported to be mild to moderate in severity. GI events (eg, nausea, vomiting, diarrhea) were the most frequently reported adverse events with liraglutide monotherapy and combination therapy and were often dose related. For example, in the LEAD-3 trial with liraglutide monotherapy, 27%, 29% and 8% of patients who received liraglutide 1.2 mg, 1.8 mg, or glimepiride, respectively, reported treatment-emergent nausea. Nausea generally occurred early during treatment and <10% of patients in the liraglutide 1.8-mg group had experienced nausea by week 4. In the LEAD-4 trial in which liraglutide was added to MET and rosiglitazone, nausea was reported 29% and 40% in the liraglutide 1.2-mg and 1.8-mg groups. However, nausea tended to decrease in frequency over time; for example, there were 216 events during weeks 1 through 4 and 65 events during weeks 4 through 26. In general, serious adverse events were uncommon with liraglutide. In the LEAD-5 trial, patients treated with insulin glargine or placebo reported a 7% frequency of serious adverse events in comparison with a 4% frequency with liraglutide.

Few episodes of minor and major hypoglycemia were reported with liraglutide across the LEAD studies. In the LEAD-3 monotherapy trial, no major hypoglycemic episodes were reported and only 8% of patients treated with liraglutide 1.8 mg reported minor hypoglycemia compared with 24% of glimepiride-treated patients. There were no reports of major hypoglycemia in the LEAD-4 trial. The incidence of minor hypoglycemia also was low (9.0%, 4.9% and 5.1%) in patients who received liraglutide 1.8 mg, 1.2 mg combined with MET and rosiglitazone, or placebo and MET and rosiglitazone, respectively. Although data are limited, major hypoglycemia with liraglutide seems to occur only when liraglutide is used in combination with an SFU. This may be a result of both agents acting simultaneously to potentiate insulin secretion from β cells, and the fact that insulin secretion stimulated by an SFU is not glucose dependent. This phenomenon of increased hypoglycemic risk in combination with an SFU has also been reported with exenatide.

Cardiovascular Outcomes (LEADER)

Until the LEADER (Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results) trial, the effects of liraglutide or any other GLP-1 RA on CV outcomes were unknown. This large trial randomized a total of 9340 patients with T2D and high CV risk to receive either liraglutide or placebo plus standard of care. Additional antihyperglycemic agents (with the exception of GLP-1 receptor agonists, DPP-4 inhibitors, or pramlintide) were available to patients who did not meet the glycemic control target of ≤7% after randomization. Patients were then followed for 3.5 to 5.0 years.

The primary endpoint was a composite of the first occurrence of death from CV causes, nonfatal MI, or nonfatal stroke. This outcome occurred in statistically fewer patients in the liraglutide group compared with the placebo group (hazard ratio [HR], 0.87; 95% CI, 0.78 to 0.97; P <0.001 for noninferiority; P = 0.01 for superiority) (Figure 19-10-A). In addition, death from CV causes (HR, 0.78; 95% CI, 0.66 to 0.93; P = 0.007) (Figure 19-10-B), death from any cause (HR, 0.85; 95% CI, 0.74 to 0.97; P = 0.02), and microvascular outcomes, as measured by a composite of renal or retinal microvascular events (HR, 0.84; 95% CI, 0.73 to 0.97; P = 0.02), were significantly reduced in the liraglutide group. Adverse events leading to permanent discontinuation of treatment were more common with liraglutide. Overall, among patients with T2D on standard therapy and at high risk of CV events, those administered liraglutide experienced lower rates of CV events and death from any cause compared with those receiving placebo.

In similar trials, other agents—including insulin, thiazolidinediones and DPP-4 inhibitors—have had comparable effects on glycemic control as liraglutide. However, these other agents have not demonstrated significant benefits with respect to rates of CV events or death. LEADER is the first trial to demonstrate CV risk reduction with a GLP-1 receptor agonist and the second of any antihyperglycemic agent, after empagliflozin, to show CV benefit.

Dosage and Administration

Liraglutide should be initiated at 0.6 mg qd for 1 week in order to reduce GI symptoms during initiation of treatment. However, this dose is not effective for glycemic control. After 1 week, the dose should be increased 1.2 mg qd. If the 1.2-mg dose does not result in acceptable glycemic control, the dose can be increased to 1.8 mg. Liraglutide can be injected SC in the abdomen, thigh, or upper arm at any time of day, independently of meals. The timing and site of injection can be changed without dosage adjustment. In order to reduce the risk of hypoglycemia, reduction in the dose of a concomitant insulin secretagogue (eg, an SFU) should be considered.

Since liraglutide is not a substitute for insulin, it should not be used in patients with T1D or for the treatment of diabetic ketoacidosis, as it would not be effective in these settings.

It should be noted that since liraglutide causes thyroid C-cell tumors at clinically relevant exposures in rodents, it is contraindicated in patients with a personal or family history of medullary thyroid carcinoma or in patients with Multiple Endocrine Neoplasia syndrome type 2.

Enlarge  Figure 19-7: LEAD-1 Trial: Change in Glycemic Parameters After 24 Weeks With Liraglutide or Rosiglitazone Added to Glimepiride.<sup>a </sup><em>P</em> <0.0001 vs PBO. <sup>b</sup> <em>P</em> <0.0001 vs RSG. <sup>c </sup><em>P</em> <0.01 vs RSG. Source: Modified from Marre M, et al. <em>Diabet Med</em>. 2009;26(3):268-278.
Figure 19-7: LEAD-1 Trial: Change in Glycemic Parameters After 24 Weeks With Liraglutide or Rosiglitazone Added to Glimepiride.a P <0.0001 vs PBO. b P <0.0001 vs RSG. c P <0.01 vs RSG. Source: Modified from Marre M, et al. Diabet Med. 2009;26(3):268-278.
Enlarge  Figure 19-8: LEAD-4 Trial: Change in Glycemic Parameters After 24 Weeks With Liraglutide Added to Metformin and Rosiglitazone.  <sup>a </sup><em>P</em> <0.0001 vs PBO. Source: Zinman B, et al. <em>Diabetes Care</em>. 2009;32(7):1224-1230.
Figure 19-8: LEAD-4 Trial: Change in Glycemic Parameters After 24 Weeks With Liraglutide Added to Metformin and Rosiglitazone. a P <0.0001 vs PBO. Source: Zinman B, et al. Diabetes Care. 2009;32(7):1224-1230.
Enlarge  Figure 19-9: LEAD-6 Trial: Change in Glycemic Parameter After 24 Weeks With Liraglutide or Exenatide, Both Added to Previous Treatment With MET, SFU, or Both. Source: Buse JB, et al. <em>Lancet</em>. 2009;374:39-47.
Figure 19-9: LEAD-6 Trial: Change in Glycemic Parameter After 24 Weeks With Liraglutide or Exenatide, Both Added to Previous Treatment With MET, SFU, or Both. Source: Buse JB, et al. Lancet. 2009;374:39-47.
Enlarge  Figure 19-10: LEADER Trial Cardiovascular Outcomes. Primary composite outcome is the first occurrence of death from CV causes, nonfatal myocardial infarction, or non-fatal stroke. The insets show the same data on an enlarged y axis. Cumulative incidences estimated using the Kaplan–Meier method and hazard ratios with the Cox proportional-hazard regression model. Source: Marso SP, et al. <em>N Engl J Med</em>. 2016;375(4):311-322.
Figure 19-10: LEADER Trial Cardiovascular Outcomes. Primary composite outcome is the first occurrence of death from CV causes, nonfatal myocardial infarction, or non-fatal stroke. The insets show the same data on an enlarged y axis. Cumulative incidences estimated using the Kaplan–Meier method and hazard ratios with the Cox proportional-hazard regression model. Source: Marso SP, et al. N Engl J Med. 2016;375(4):311-322.

Dulaglutide (Trulicity)

Dulaglutide has an amino acid sequence that is 90% homologous to endogenous human GLP-1 and is indicated as an adjunct to diet and exercise to improve glycemic control in adults with T2D. It is also indicated to reduce the risk of major adverse CV events in adults with T2D who have established CVD or multiple CV risk factors. Dulaglutide functions by lowering fasting glucose and reduces postprandial glucose concentrations in patients with T2D, the effect of which can be observed after a single dose. It also causes a delay in gastric motility, which subsides after the first dose with subsequent doses. Dulaglutide is administered as a once-weekly SC injection.

Efficacy

The efficacy of dulaglutide 0.75 mg and 1.5 mg once weekly has been studied as monotherapy and in combination with MET, MET and SFU, MET and TZD and prandial insulin with or without MET. The efficacy of dulaglutide 3.0 mg and 4.5 mg once weekly was assessed in combination with MET. In patients with T2D, dulaglutide produced reductions from baseline in A1C compared with placebo (Table 19.7). No overall differences in glycemic effectiveness were observed across demographic subgroups (age, gender, race/ethnicity, duration of diabetes).

Monotherapy

Monotherapy with dulaglutide was compared with MET-treated patients with T2D in a 52-week double-blind study which randomized patients to SC dulaglutide 1.5 mg, dulaglutide 0.75 mg, or MET. The primary objective compared change from baseline A1C at 26 weeks among treated groups.

At 26 weeks, changes from baseline A1C were greater in dulaglutide 1.5 and 0.75 mg vs MET (LS mean difference): -0.22% (-2.4 mmol/mol) and -0.15% (-1.6 mmol/mol) (one-sided P <0.025, both comparisons), respectively. Greater percentages reached A1C targets <7.0% with dulaglutide 1.5 and 0.75 mg compared with MET (P <0.05, all comparisons). No severe hypoglycemia was reported. Compared with MET, decrease in weight was similar with dulaglutide 1.5 mg and smaller with dulaglutide 0.75 mg.

When compared with sitagliptin, dulaglutide 0.75 mg and dulaglutide 1.5 mg were found to be superior in reducing the levels of A1C up to week 52 (P <0.001 for both comparisons). Mean weight changes to 52 weeks were greater with dulaglutide 1.5 mg (-3.03 ± 0.22 kg) and dulaglutide 0.75 mg (-2.60 ± 0.23 kg) compared with sitagliptin (-1.53 ± 0.22 kg) (P <0.001, both comparisons).

The safety and efficacy of dulaglutide was compared with that of liraglutide in MET-treated patients with uncontrolled T2D in a head-to-head phase 3 trial. 599 patients were randomized to receive once-weekly dulaglutide (1.5 mg) or once-daily liraglutide (1.8 mg). At 26 weeks, dulaglutide was noninferior to once-daily liraglutide for LS mean reduction in A1C, with a similar safety and tolerability profile. The LS mean reduction in A1C was -1·42% (SE 0.05) in the dulaglutide group and -1.36% (0.05) in the liraglutide group. Mean treatment difference in A1C was -0.06% (95% CI, -0.19, 0.07; Pnon-inferiority <0.0001) between the two groups. (Figure 19-11)

In Combination With One OAD

The efficacy of dulaglutide 0.75 mg and 1.5 mg as an add-on to MET was assessed in a 104-week placebo-controlled, double-blind study, in which 972 patients were randomized to placebo, dulaglutide 0.75 mg once weekly, dulaglutide 1.5 mg once weekly, or sitagliptin 100 mg/day, all as add-on to MET. Treatment with dulaglutide 0.75 mg and 1.5 mg once weekly resulted in a statistically significant reduction in A1C compared with placebo (at 26 weeks) and compared with sitagliptin (at 26 and 52 weeks), all in combination with MET. At 26 weeks, the percentage of patients who achieved A1C <7.0% was 22%, 56%, 62%, 39% and a mean weight reduction of 1.4 kg, 2.7 kg, 3.0 kg and 1.4 kg was observed for placebo, dulaglutide 0.75 mg, dulaglutide 1.5 mg and sitagliptin, respectively. There was a mean reduction of fasting glucose of 9 mg/dL, 35 mg/dL, 41 mg/dL and 18 mg/dL for placebo, dulaglutide 0.75 mg, dulaglutide 1.5 mg and sitagliptin, respectively.

The efficacy of dulaglutide 3.0 mg and 4.5 mg as an add-on to MET was assessed in AWARD-11, a 52-week active-controlled, double-blind trial. A total of 1,842 patients were randomized to dulaglutide 1.5 mg once weekly, dulaglutide 3.0 mg once weekly, and dulaglutide 4.5 mg once weekly; patients continued with MET therapy in all three groups. Two estimands were used in the analysis of 36-week data: a treatment regimen estimand (included all baseline and end point data regardless of initiation of new antihyperglycemic therapy or premature treatment discontinuation) and an efficacy estimand (included data from all study visits before either initiation of a new antihyperglycemic drug for >14 days or premature treatment discontinuation).

With respect to the primary endpoint of A1C reduction from baseline, dulaglutide 4.5 mg was superior to the 1.5 mg dose at Week 36 under the treatment regimen estimand (mean change from baseline [mean baseline 8.6%]: dulaglutide 1.5 mg -1.54%, dulaglutide 4.5 mg -1.77%; estimated treatment difference [ETD]: -0.24%; P <0.001) and under the efficacy estimand (mean change from baseline: dulaglutide 1.5 mg -1.53%, dulaglutide 4.5 mg -1.87%; ETD: -0.34%; P <0.001). Dulaglutide 3.0 mg was also superior to the 1.5 mg dose under the efficacy estimand (mean change from baseline: dulaglutide 1.5 mg -1.53%, dulaglutide 4.5 mg -1.71%; estimated treatment difference [ETD]: -0.17%; P = 0.003) but was not superior under the treatment regimen estimand (mean change from baseline: dulaglutide 1.5 mg -1.54%, dulaglutide 4.5 mg -1.64%; estimated treatment difference [ETD]: -0.10%; P = 0.096).

Additionally, dulaglutide 4.5 mg demonstrated superiority to the 1.5 mg dose for weight loss at Week 36 under both the treatment regimen estimand (mean change from baseline [mean baseline 95.7 kg]: dulaglutide 1.5 mg -3.0 kg, dulaglutide 4.5 mg -4.6 kg; ETD: -1.6 kg; P <0.001) and the efficacy estimand (mean change from baseline: dulaglutide 1.5 mg -3.1 kg, dulaglutide 4.5 mg -4.7 kg; ETD: -1.6 kg; P <0.001).

In Combination With Two OADs

The addition of dulaglutide to MET and a TZD was examined in a 52-week placebo-controlled study, where 976 patients were randomized to placebo, dulaglutide 0.75 mg once weekly, dulaglutide 1.5 mg once weekly, or exenatide 10 mcg BID, all as add-on to maximally tolerated doses of MET (≥1500 mg per day) and pioglitazone (up to 45 mg per day).

After 26 weeks, patients in the placebo treatment group were randomized to either dulaglutide 0.75 mg once weekly or dulaglutide 1.5 mg once weekly. At 26 weeks, treatment with dulaglutide 0.75 mg and 1.5 mg once weekly resulted in a statistically significant reduction in A1C compared with placebo and compared with exenatide. Over the 52-week study period, the percentage of patients who required glycemic rescue was 8.9% in the dulaglutide 0.75 mg once weekly plus MET and pioglitazone treatment group, 3.2% in the dulaglutide 1.5 mg once weekly plus MET and pioglitazone treatment group, and 8.7% in the exenatide BID + MET and pioglitazone treatment group.

Treatment with dulaglutide once weekly resulted in similar reductions in A1C from baseline at 52 weeks when used in combination with MET and SFU, as well as with postprandial insulin injections with or without MET.

Safety and Tolerability

The most common side effects observed in patients treated with dulaglutide were nausea, diarrhea, vomiting, abdominal pain and decreased appetite.

As with other GLP-1 agonists, pancreatitis-related adverse reactions were reported in patients exposed to dulaglutide. Patients should be observed carefully for signs and symptoms of pancreatitis after initiation of treatment with dulaglutide, including persistent severe abdominal pain. If pancreatitis is suspected, dulaglutide should be discontinued and other anti-diabetic therapies considered. Dulaglutide has not been evaluated in patients with a prior history of pancreatitis. Similarly to other GLP-1 agents, the risk of hypoglycemia is increased when dulaglutide is used in combination with insulin secretagogues or insulin. Patients may require a lower dose of SFU or insulin to reduce the risk of hypoglycemia.

Although dulaglutide has been shown to cause a dose-related and treatment-duration–dependent increase in the incidence of thyroid C-cell tumors (adenomas and carcinomas) after lifetime exposure in animal studies, it is unknown whether dulaglutide will cause thyroid C-cell tumors, including MTC in humans. Dulaglutide is contraindicated in patients with a personal or family history of MTC or in patients with MEN 2. Dulaglutide should not be used to treat people with T1Ds, diabetic ketoacidosis, or severe abdominal or intestinal problems, or as first-line therapy for patients who cannot be managed with diet and exercise.

Cardiovascular Outcomes

The effect of dulaglutide on CV outcomes were investigated in REWIND, a large, international, double-blind trial. A total of 9,901 eligible patients (aged at least 50 years with T2D and a previous CV event or CV risk factors) were randomized (1:1) to receive either a weekly subcutaneous injection of dulaglutide 1.5 mg or placebo. The primary (composite) outcome was the first occurrence of non-fatal myocardial infarction, non-fatal stroke, or death from CV causes (including unknown causes).

The primary outcome occurred in 12.0% and 13.4% of patients in the dulaglutide and placebo groups, respectively (hazard ratio 0.88; P = 0.026), demonstrating the superiority of dulaglutide over placebo in reducing the incidence of a composite of CV outcomes (Figure 19-12). While the frequencies of prespecified adverse events remained comparable between the dulaglutide and placebo groups, gastrointestinal adverse events were significantly more frequent in the dulaglutide group (47.4% compared to 34.1% with the placebo; P <0.0001).

Dosage and Administration

The recommended initiating dose of dulaglutide is 0.75 mg once weekly, any time of day regardless of food intake. The dose may be increased to 1.5 mg once weekly for additional glycemic control. If further glycemic control is needed, the dose can be increased to 3 mg once weekly after at least 4 weeks on the 1.5 mg dose and further to the maximum dose of 4.5 mg once weekly after at least 4 weeks on the 3 mg dose. Dulaglutide is available as injectable solutions of 0.75 mg/0.5 mL,1.5 mg/0.5 mL, 3.0 mg/0.5 mL and 4.5 mg/0.5 mL. It should be injected subcutaneously in the abdomen, thigh, or upper arm using a single dose pen or prefilled syringe. Injection is rapid and takes <5 seconds.

Dulaglutide should not be used in patients with T1D or for the treatment of diabetic ketoacidosis. It is not a substitute for insulin. Dulaglutide has not been studied in patients with severe GI disease, including severe gastroparesis and is therefore not recommended in patients with pre-existing severe GI disease.

Enlarge  Figure 19-11: Change From Baseline in  Glycemic Control and Body Weight at Week 26 for Dulaglutide vs Liraglutide. <sup>a </sup><em>P</em> <0.05. Source: Modified from Dungan KM, et al. <em>Lancet</em>. 2014;384(9951):1349-1357.
Figure 19-11: Change From Baseline in Glycemic Control and Body Weight at Week 26 for Dulaglutide vs Liraglutide. a P <0.05. Source: Modified from Dungan KM, et al. Lancet. 2014;384(9951):1349-1357.
Enlarge  Figure 19-12: Primary Cardiovascular Composite Outcome in REWIND. Source: Modified from Gerstein HC, et al. <em>Lancet</em>. 2019;394(10193):121-130.
Figure 19-12: Primary Cardiovascular Composite Outcome in REWIND. Source: Modified from Gerstein HC, et al. Lancet. 2019;394(10193):121-130.

Lixisenatide (Adlyxin)

Lixisenatide was approved by the FDA in 2016 and is used alone but also commonly in the fixed ratio combination with insulin glargine U-100 (Soliqua 100/33, see below). Lixisenatide is indicated as an adjunct to diet and exercise to improve glycemic control in adults with T2D. It functions by increasing glucose-dependent insulin release, decreasing glucagon secretion, and slowing gastric emptying.

Efficacy

The efficacy of lixisenatide once daily has been studied as monotherapy and in combination with MET, MET ± an SFU, an SFU ± MET, pioglitazone ± MET, and basal insulin ± OADs (Table 19-8). In patients with T2D, lixisenatide significantly reduced A1C from baseline compared with placebo. Treatment with lixisenatide also had weight neutral effects or lead to significant weight loss, an important finding considering that weight gain is commonly associated with treatment intensification.

Monotherapy

In a 12-week double-blind study, 241 patients with T2D inadequately controlled on diet and exercise were randomized to lixisenatide 20 mcg once daily or placebo. Compared with placebo, treatment with lixisenatide resulted in statistically significant reductions in A1C from baseline at Week 12 (treatment difference: -0.65% [P <0.0001]). A significant difference was not observed in adjusted mean weight change from baseline between trial arms.

In Combination With MET (Alone or in Combination With an SFU)

Three 24-week studies assessed the efficacy of lixisenatide 20 mcg once daily in patients with T2D. The first trial enrolled 323 patients inadequately controlled on diet, exercise and MET. The second trial enrolled 391 Asian patients inadequately controlled on diet, exercise and MET with or without an SFU. In both trials, treatment with lixisenatide resulted in statistically significant reductions in A1C from baseline at Week 24 compared with placebo (treatment difference: -0.46% [P <0.0001] and -0.27% [P = 0.0032], respectively).

In a third study, 634 patients with T2D inadequately controlled on diet, exercise and MET were randomized to lixisenatide 20 mcg once daily or exenatide 10 mcg twice daily. Lixisenatide met the prespecified noninferiority margin of 0.4% vs exenatide for the difference in A1C reduction from baseline; however, lixisenatide provided significantly less A1C reduction than exenatide (treatment difference: 0.17% [P = 0.0175]).

In Combination With an SFU or Pioglitazone (Alone or in Combination With MET)

The efficacy of lixisenatide 20 mcg once daily was assessed in combination with an SFU or pioglitazone, alone or in combination with MET, in two 24-week studies. The first study enrolled 859 patients with T2D inadequately controlled with diet, exercise and an SFU with or without MET. The second study enrolled 484 patients with T2D with inadequately controlled with diet, exercise and pioglitazone with or without MET. In both studies, treatment with lixisenatide resulted in statistically significant reductions in A1C from baseline at Week 24 compared with placebo (treatment difference: -0.58% [P <0.0001] and -0.48% [P <0.0001], respectively).

In Combination With Basal Insulin (Alone or in Combination With OADs)

The efficacy of lixisenatide in combination with basal insulin alone or in combination with OADs was assessed in four studies. The first study enrolled 496 patients with T2D inadequately controlled on diet, exercise and basal insulin with or without MET. The second study enrolled 311 Asian patients with T2D inadequately controlled on diet, exercise and basal insulin with or without an SFU. The third study enrolled 446 patients with T2D inadequately controlled on diet, exercise, insulin glargine and MET with or without TZDs. In all three studies, randomization to lixisenatide 20 mcg once daily resulted in statistically significant reductions in A1C from baseline at Week 24 compared with placebo (treatment difference: -0.36% [P = 0.0002], -0.76% [P <0.0001] and -0.28% [P = 0.0005], respectively).

A fourth study enrolled 894 patients with T2D inadequately controlled on diet, exercise and basal insulin combined with 1 to 3 OADs. In this 26-week open-label study, patients were randomized to lixisenatide 20 mcg once daily, insulin glulisine once daily, or insulin glulisine TID combined with insulin glargine with or without MET. Lixisenatide 20 mcg once daily met the prespecified noninferiority margin of 0.4% vs insulin glulisine QD and TID for the difference in A1C reduction from baseline. However, lixisenatide provided significantly less A1C reduction than insulin glulisine TID (treatment difference: 0.23% [P = 0.0002]).

Safety and Tolerability

Adverse reactions that were not present at baseline, occurred more commonly with lixisenatide than with placebo and occurred in ≥5% of patients treated with lixisenatide were nausea (25%), vomiting (10%), headache (9%), diarrhea (8%) and dizziness (7%). The majority of these adverse reactions occurred during the first 3 weeks after starting treatment.

As with other GLP-1 agonists, pancreatitis has been reported in patients exposed to lixisenatide. Patients should be observed carefully for signs and symptoms of pancreatitis after initiation of treatment with lixisenatide, including persistent severe abdominal pain. If pancreatitis is suspected, lixisenatide should be promptly discontinued and appropriate management initiated. Other antidiabetic therapies should be considered in patients with a history of pancreatitis, since lixisenatide has not been studied in patients with chronic pancreatitis or a history of unexplained pancreatitis.

Similar to other GLP-1 agents, the risk of hypoglycemia is increased when lixisenatide is used in combination with basal insulin or an SFU. Patients may require a lower dose of SFU or insulin to reduce the risk of hypoglycemia. Acute kidney injury and worsening of chronic renal failure, which may sometimes require hemodialysis, has been reported in patients exposed to GLP-1 receptor agonists. Monitor renal function when initiating or escalating doses of lixisenatide in patients with renal impairment and in patients reporting severe gastrointestinal reactions.

Clinical studies have not shown macrovascular risk reduction with lixisenatide.

Dosage and Administration

The recommended starting dose of lixisenatide is 10 mcg SC once daily for 14 days. On Day 15, the dose should be increased to the maintenance dose of 20 mcg once daily. Lixisenatide is administered by SC injection in the abdomen, thigh, or upper arm 1 hour prior to the first meal of the deal, preferably at the same time each day. If a dose is missed, lixisenatide should be administered within 1 hour prior to the next meal. Lixisenatide is available as solutions for SC injection in single-patient use prefilled pens as:

  • 50 mcg/mL in 3 mL of solution, for 14 doses at 10 mcg/dose
  • 100 mcg/mL in 3 mL solution, for 14 doses at 20 mcg/dose.

Lixisenatide is not a substitute for insulin and is not indicated for use in patients with T1D or for the treatment of diabetic ketoacidosis. The concurrent use of lixisenatide with short-acting insulin has not been studied and is not recommended.

Ozempic (Semaglutide)

Semaglutide was approved by the FDA in 2017 as an adjunct to diet and exercise to improve glycemic control in adults with T2D. It has also received approval to reduce the risk of major adverse CV events in adults with T2D mellitus and established CVD.

Efficacy

The safety and efficacy of semaglutide has been evaluated in eleven clinical trials (SUSTAIN-1 to -11) in patients with T2D as monotherapy or in combination with metformin, metformin and sulfonylureas, metformin and/or thiazolidinedione, basal insulin, or metformin and basal insulin. In these trials, treatment with semaglutide had clinically relevant effects on reducing A1C, FPG, and weight from baseline (Table 19-9).

Monotherapy

SUSTAIN-1 was a 30-week trial than enrolled 388 patients with T2D inadequately controlled with diet and exercise. Eligible patients were randomized to 0.5 mg semaglutide or 1.0 mg semaglutide once weekly or placebo. Monotherapy with 0.5 mg or 1.0 mg semaglutide resulted in statistically significantly reductions in A1C compared with placebo (baseline mean A1C level: placebo group 8.0%, semaglutide 0.5 mg group 8.1%, semaglutide 1.0 mg group 8.1%; change: placebo -0.1%, semaglutide 0.5 mg -1.4%, semaglutide 1.0 mg -1.6%; difference from placebo: semaglutide 0.5 mg -1.2%, semaglutide 1.0 mg -1.4%; P <0.0001 for both).

Combination With Metformin and/or Thiazolidinediones

SUSTAIN-2 was a 56-week trial that randomized 1,231 patients with T2D to 0.5 mg semaglutide or 1.0 mg semaglutide once weekly or sitagliptin 100 mg once daily, all in combination with metformin and/or thiazolidinediones. Treatment with semaglutide 0.5 mg or 1.0 mg once weekly resulted in a significant reduction in A1C compared to sitagliptin (baseline mean A1C level: sitagliptin group 8.2%, semaglutide 0.5 mg group 8.0%, semaglutide 1.0 mg group 8.0%; change: sitagliptin -0.7%, semaglutide 0.5 mg -1.3%, semaglutide 1.0 mg -1.5%; difference from sitagliptin: semaglutide 0.5 mg -0.6%, semaglutide 1.0 mg -0.8%; P <0.0001 for both).

Combination With Metformin or Metformin With Sulfonylurea

SUSTAIN-3 was a 56-week open-label trial that enrolled 813 patients with T2D, with the majority (96%) of patients on metformin alone or metformin with sulfonylurea. Eligible patients were randomized to receive 1.0 mg semaglutide or 2.0 mg exenatide once weekly. Treatment with semaglutide resulted in a significant reduction in A1C compared to exenatide (baseline mean A1C level: exenatide group 8.3%, semaglutide group 8.4%; change: exenatide -0.9%, semaglutide -1.4%; difference from exenatide: -0.5%; P <0.0001).

SUSTAIN-4 was a 30-week trial of 1089 patients with T2D randomized to receive 0.5 mg semaglutide or 1.0 mg semaglutide once weekly or insulin glargine once daily. All patients received background metformin or metformin and sulfonylurea. Patients who received insulin glargine started at a dose of 10U once daily, with dose adjustments being made throughout the trial period based on self-measured fasting plasma glucose before breakfast, with a target of 71 mg/dL to <100 mg/dL. Insulin glargine could also be titrated between study visits at the discretion of the investigator. Treatment with 0.5 mg or 1.0 mg semaglutide resulted in a significant reduction in A1C compared with insulin glargine (baseline mean A1C level: insulin glargine group 8.1%, semaglutide 0.5 mg group 8.1%, semaglutide 1.0 mg group 8.2%; change: insulin glargine -0.9%, semaglutide 0.5 mg -1.2%, semaglutide 1.0 mg -1.5%; difference from insulin glargine: semaglutide 0.5 mg -0.3%, semaglutide 1.0 mg -0.6%; P <0.0001 for both).

SUSTAIN-FORTE was a 40-week randomized, active-controlled, double-blind trial involving a total of 961 patients with T2D inadequately controlled on metformin with or without sulfonylurea. The goal of SUSTAIN-FORTE was to assess the safety and efficacy of a higher semaglutide dose compared to the maximum then-approved weekly dose. Participants were randomized (1:1) to receive either semaglutide 1.0 mg weekly or semaglutide 2.0 mg weekly. The 2.0 mg weekly dose was superior to the led to a significantly lower A1C level at 40 weeks (baseline mean A1C level: semaglutide 1.0 mg group 8.8%, semaglutide 2.0 mg group 8.9%; mean change: semaglutide 1.0 mg -1.9%, semaglutide 2.0 mg -2.1%; difference in favor of semaglutide 2.0 mg: -0.2%; P <0.01).

Combination With Basal Insulin

SUSTAIN-5 was a 30-week trial of 397 patients with T2D inadequately controlled with basal insulin, with or without metformin. Enrolled patients were randomized to 0.5 mg semaglutide or 1.0 mg semaglutide once weekly or placebo. To reduce the risk of hypoglycemia, those patients with A1C <8.0% at screening reduced their insulin dose by 20% at trial initiation. Treatment with semaglutide resulted in a significant reduction in A1C after 30 weeks compared to placebo (baseline mean A1C level: placebo group 8.4%, semaglutide 0.5 mg group 8.4%, semaglutide 1.0 mg group 8.3%; change: placebo -0.2%, semaglutide 0.5 mg -1.3%, semaglutide 1.0 mg -1.7%; difference from placebo: semaglutide 0.5 mg, -1.1%; semaglutide 1.0 mg, -1.6%; P <0.0001).

Combination With Metformin: Semaglutide vs Dulaglutide

SUSTAIN-7 was a 40-week, head-to-head, open-label, parallel-group trial that compared the safety and efficacy of semaglutide and dulaglutide in T2D treatment. A total of 1201 eligible patients (adults with T2D uncontrolled [A1C 7.0–10.5%] on metformin monotherapy) were randomized (1:1:1:1) to one of the following once-weekly regimens: 1) semaglutide 0.5 mg; 2) dulaglutide 0.75 mg; 3) semaglutide 1.0 mg; or 4) dulaglutide 1.5 mg. Semaglutide demonstrated superiority over dulaglutide with respect to the primary endpoint of A1C change from baseline (mean baseline: semaglutide 0.5 mg group 8.3%, dulaglutide 0.75 mg group 8.2%, semaglutide 1.0 mg group 8.2%, dulaglutide 1.5 mg group 8.2%; mean change: semaglutide 0.5 mg -1.5%, dulaglutide 0.75 mg -1.1%, semaglutide 1.0 mg -1.8%, dulaglutide 1.5 mg -1.4%; ETD in favor of semaglutide 0.5 mg over dulaglutide 0.75 mg: -0.40%; ETD in favor of semaglutide 1.0 mg over dulaglutide 1.5 mg: -0.41%; P <0.0001 for both). Semaglutide was also superior to dulaglutide in reducing bodyweight (mean baseline: semaglutide 0.5 mg group 96.4 kg, dulaglutide 0.75 mg group 95.6 kg, semaglutide 1.0 mg group 95.5 kg, dulaglutide 1.5 mg group 93.4 kg; mean change: semaglutide 0.5 mg -4.6 kg, dulaglutide 0.75 mg -2.3 kg, semaglutide 1.0 mg -6.5 kg, dulaglutide 1.5 mg -3.0 kg; ETD in favor of semaglutide 0.5 mg over dulaglutide 0.75 mg: -2.26 kg; ETD in favor of semaglutide 1.0 mg over dulaglutide 1.5 mg: -3.55 kg; P <0.0001 for both).

Combination With Metformin: Semaglutide vs Canagliflozin

In SUSTAIN-8, a 52-week, head-to-head, double-blind trial, the safety and efficacy of semaglutide 1.0 mg once weekly in patients with T2D uncontrolled on metformin was compared to that of canagliflozin 300 mg once daily. A total of 788 eligible patients was randomized (1:1) to either semaglutide 1.0 mg weekly or canagliflozin 300 mg daily. Patients receiving semaglutide had a significantly greater reduction in A1C at Week 52 (mean baseline: canagliflozin group 8.2%, semaglutide group 8.3%; mean change: canagliflozin -1.0%, semaglutide -1.5%; ETD: -0.49%; P <0.0001), the primary endpoint of the trial. Semaglutide also led to a significantly greater reduction in bodyweight compared to canagliflozin (mean baseline: canagliflozin group 89.8 kg, semaglutide group 90.6 kg; mean change: canagliflozin -4.2 kg, semaglutide -5.3 kg; ETD: -1.06 kg; P = 0.0029).

Combination With SGLT2 Inhibitor

SUSTAIN-9 was a 30-week trial that compared to safety and efficacy of semaglutide to a placebo in patients with T2D uncontrolled despite existing SGLT2 inhibitor therapy. A total of 302 patients were randomized (1:1) to semaglutide 1.0 mg or a volume-matched placebo and continued taking existing therapy. Patients in the semaglutide group achieved greater reductions in A1C (mean baseline: placebo group 8.1%, semaglutide group 8.0%; mean change: placebo -0.1%, semaglutide -1.5%; ETD: -1.42%; P <0.0001) at Week 30, the primary endpoint of the trial. Semaglutide was also superior to the placebo with regard to mean change in bodyweight (mean baseline: placebo group 93.8 kg, semaglutide group 89.6 kg; mean change: placebo -0.9 kg, semaglutide -4.7 kg; ETD: -3.81 kg; P <0.0001).

Combination With Metformin, Sulfonylurea, or SGLT Inhibitor: Semaglutide vs Liraglutide

The objective of SUSTAIN-10, a 30-week, head-to-head, open-label European trial was to compare the efficacy of semaglutide to liraglutide in their most frequently prescribed doses (reflecting real-world clinical practice). A total of 577 eligible patients (with T2D uncontrolled on 1-3 oral antidiabetic drugs) were randomized (1:1) to either semaglutide 1.0 mg once weekly or liraglutide 1.2 mg. Patients receiving semaglutide had significantly reduced A1C levels at Week 30 (the primary endpoint) compared to those receiving liraglutide (mean baseline: liraglutide group 8.3%, semaglutide group 8.2%; mean change: liraglutide -1.0%, semaglutide -1.7%; ETD: -0.69%; P <0.0001). Semaglutide also demonstrated superior results in mean bodyweight change (mean baseline: liraglutide group 97.2 kg, semaglutide group 96.6 kg; mean change: liraglutide -1.9 kg, semaglutide -5.8 kg; ETD: -3.83 kg; P <0.0001).

Combination With Metformin and Basal Insulin: Semaglutide vs Insulin Aspart

In SUSTAIN-11, a 52-week, parallel, open-label trial, 1748 adult patients with T2D uncontrolled on insulin glargine 100 units/mL and metformin were randomized (1:1) to semaglutide 1.0 mg once a week or insulin aspart 100 units/mL three times a day. At treatment initiation, insulin aspart was distributed at 4 units/mL three times a day. Both insulin glargine and insulin aspart doses were adjusted based on self-measured plasma glucose and individualized treatment goals. At week 52, semaglutide demonstrated a greater improvement in A1C levels compared to insulin aspart (mean baseline: 8.6% in both semaglutide and insulin aspart groups; mean change: -1.5% for semaglutide, and -1.2% for insulin aspart; ETD: -0.29%; P<0.0001). Additionally, semaglutide provided greater weight loss than insulin aspart (mean baseline: 87.9 kg for both groups; mean change: -4.1 kg for semaglutide, and +2.8 kg for insulin aspart; ETD: -6.99 kg).

Safety and Tolerability

The prescribing information for semaglutide includes a boxed warning for risk of thyroid C-cell tumors. Although semaglutide causes thyroid C-cell tumors, including medullary thyroid carcinoma, in rodents, the relevance of this risk in humans has not been determined. Regardless, semaglutide is contraindicated in patients with a personal or family history of medullary thyroid carcinoma or in patients with Multiple Endocrine Neoplasia syndrome type 2. The prescribing information for semaglutide also contains warnings and precautions for pancreatitis, diabetic retinopathy complications, hypoglycemia, acute kidney injury and hypersensitivity.

The most common adverse reactions, excluding hypoglycemia, associated with the use of 1.0 mg semaglutide and occurring in ≥5% of semaglutide-treated patients include nausea (20.3%), vomiting (9.2%), diarrhea (8.8%), abdominal pain (5.7%) and constipation (3.1%).

Cardiovascular Outcome Trial of Semaglutide (SUSTAIN-6)

SUSTAIN-6 was a 104-week trial that enrolled 3297 patients with T2D at a high risk for CV events. Eligible patients were randomized to 0.5 mg semaglutide once weekly, 1.0 mg semaglutide once weekly, or placebo in addition to standard-of-care. Eighty-three percent of patients had a history of CV disease and 17% were at high risk, but without known CV disease. The composite primary endpoint was the first occurrence of death from CV causes, nonfatal MI, or nonfatal stroke.

The composite primary outcome occurred in 6.6% of semaglutide-treated patients compared to 8.9% of the placebo group (HR=0.74; P <0.001 for noninferiority; P = 0.02 for superiority; Figure 19-13-A). Of the individual composite components, only nonfatal stroke occurred significantly less frequently in semaglutide-treated patients compared to placebo (1.6% vs 2.7%; HR=0.61; P = 0.04; Figure 19-13-B). No significant differences were observed between treatment groups for components of nonfatal MI or CV death. Similar results were observed for both doses of semaglutide.

Complications of diabetic retinopathy occurred more frequently in semaglutide-treated patients compared to the placebo group (3.0% vs 1.8%; HR=1.76; P = 0.02), whereas new or worsening nephropathy occurred less frequently in semaglutide-treated patients (3.8% vs 6.1%; HR=0.64; P = 0.005).

Enlarge  Figure 19-13: Primary Composite Outcome and Nonfatal Stroke Component of the SUSTAIN-6 Trial. Source: Modified from Marso SP, et al. <em>N Engl J Med</em>. 2016;375:1834-1844.
Figure 19-13: Primary Composite Outcome and Nonfatal Stroke Component of the SUSTAIN-6 Trial. Source: Modified from Marso SP, et al. N Engl J Med. 2016;375:1834-1844.

Dosage and Administration

Semaglutide should be initiated with a 0.25 mg SC injection once weekly for 4 weeks, followed by an increase to 0.5 mg once weekly. The 0.25 mg dose is not effective for glycemic control and is only intended for treatment initiation. Dosage may be increased to 1.0 mg once weekly if additional glycemic control is needed after at least 4 weeks on the 0.5 mg dose, and to a maximum of 2.0 mg once weekly if additional glycemic control is required after at least 4 weeks on the 1.0 mg dose.

Semaglutide should be administered on the same day each week, at any time of day, with or without food. The day of weekly administration can be changed, but the time between two consecutive doses should be at least 48 hours. If a patient misses a dose, they should administer their dose as soon as soon as possible, but within 5 days of the missed dose. If more than 5 days have passed, the patient should skip the missed dose and resume the next dose on the next scheduled day. If the patient is also receiving insulin, injections should not be administered adjacent to one another.

Switching from Injectable to Oral Semaglutide

Patients treated with SC semaglutide 0.5 mg once weekly can be transitioned to a once daily dose of oral semaglutide (7 mg or 14 mg; see the Oral Semaglutide section below). Patients can start taking oral semaglutide 7 days after the last dose of injectable semaglutide. There is no equivalent dose of oral semaglutide for SC semaglutide 1 mg or 2 mg.

Summary

The introduction of GLP-1 receptor agonists represents a significant advance in the treatment of T2D. Not only are they effective in improving overall glycemic control as indicated by the FBG and PPG values and A1C, but at the same time they lead to weight loss. These effects on weight are very important since weight gain associated with intensification of therapy in an effort to normalize or near normalize A1C is a significant challenge in clinical practice. The positive effects on ASCVD also adds another important clinical dimension to this class of agents. The combination of GLP-1 RAs with MET appears to be not only effective in terms of metabolic outcomes but also safe in terms of a lower risk of hypoglycemia. It also appears that the weight gain seen with SFUs, TZDs and insulin is blunted when these agents are combined with these new classes of antidiabetic agents. Finally, activation of the GLP-1 receptors by these agents may also present the potential to improve β-cell function and, therefore, result in slowing down or halting the natural history of T2D. Thus, these novel medications have an important role in the prevention of T2D if used early in the course of glucose intolerance.

Rybelsus (Oral Semaglutide)

The oral (tablet) formulation of semaglutide received FDA approval in 2019 as an adjunct to diet and exercise to improve glycemic control in adults with T2D. This marks the first approval of an oral GLP-1 receptor agonist.

Efficacy

The safety and efficacy of the oral formulation of semaglutide has been evaluated in eleven clinical trials of the PIONEER series (PIONEER-1 to PIONEER-10 and PIONEER PLUS). The PIONEER series assessed oral semaglutide in patients with T2D as monotherapy (PIONEER-1 and -9) or in combination with metformin (PIONEER-2), sulfonylureas and/or metformin (PIONEER-3), SGLT2 inhibitors and/or metformin (PIONEER-4), insulin and/or metformin (PIONEER-8), or diverse glucose-lowering medications (PIONEER-7, PIONEER-10, and PIONEER PLUS). PIONEER-5 assessed the safety and efficacy of oral semaglutide in patients with moderate renal impairment, while PIONEER-6 assessed the CV outcomes of oral semaglutide. Treatment with oral semaglutide had clinically relevant effects on reducing A1C, FPG, and weight from baseline in the PIONEER trial series (Table 19-10).

Monotherapy

PIONEER-1 was a 26-week trial that randomized (1:1:1:1) 703 patients with T2D inadequately controlled with diet and exercise to a once-daily regimen of: 1) oral semaglutide 3 mg; 2) oral semaglutide 7 mg; 3) oral semaglutide 14 mg; or 4) placebo. Monotherapy with any of the three semaglutide doses resulted in statistically significant reductions in A1C compared to placebo under both the treatment policy estimand (ie, regardless of trial product discontinuation or rescue medication use; mean baseline: 7.9% in the semaglutide 3 mg group, 8.0% in the semaglutide 7 mg group, 8.0% in the semaglutide 14 mg group, and 7.9% in the placebo group; mean changes from baseline: -0.9% with semaglutide 3 mg, -1.2% with semaglutide 7 mg, -1.4% with semaglutide 14 mg, and -0.3% with placebo; ETD from the placebo: oral semaglutide 3 mg, -0.6%; oral semaglutide 7 mg, -0.9%; oral semaglutide 14 mg, -1.1%; P <0.001 for all) and the trial product estimand (ie, assuming no trial product discontinuation or rescue medication use; mean changes from baseline: -0.8% with semaglutide 3 mg, -1.3% with semaglutide 7 mg, -1.5% with semaglutide 14 mg, and -0.1% with placebo; ETD from the placebo: oral semaglutide 3 mg, -0.7%; oral semaglutide 7 mg, -1.2%; oral semaglutide 14 mg, -1.4%; P <0.001 for all).

PIONEER-9 was a 52-week phase 2/3a trial that randomized (1:1:1:1:1) 243 Japanese patients (≥20 years or age) with uncontrolled T2D managed by diet, exercise, or oral glucose-lowering drug monotherapy (washed out) to receive a once daily dose of: 1) oral semaglutide 3 mg; 2) oral semaglutide 7 mg; 3) oral semaglutide 14 mg; 4) placebo; or 5) open-label SC liraglutide 0.9 mg. With regard to the primary endpoint (change in A1C from baseline to Week 26 with the trial product estimand), all three doses of oral semaglutide demonstrated superiority to the placebo (mean baseline: 8.1% in the semaglutide 3 mg group, 8.3% in the semaglutide 7 mg group, 8.0% in the semaglutide 14 mg group and 8.3% in the placebo group; mean changes from baseline: -1.1% with semaglutide 3 mg, -1.5% with semaglutide 7 mg, -1.7% with semaglutide 14 mg and -0.1% with placebo; ETD from the placebo: oral semaglutide 3 mg, -1.1%; oral semaglutide 7 mg, -1.5%; oral semaglutide 14 mg, -1.7%; P <0.0001 for all). Oral semaglutide also showed non-inferiority to SC liraglutide (mean baseline: 8.1% in the semaglutide 3 mg group, 8.3% in the semaglutide 7 mg group, 8.0% in the semaglutide 14 mg group and 8.3% in the liraglutide group; mean changes from baseline: -1.1% with semaglutide 3 mg, -1.5% with semaglutide 7 mg, -1.7% with semaglutide 14 mg and -1.4% with liraglutide 0.9 mg; ETD from liraglutide: oral semaglutide 3 mg, -1.1% [P = 0.0799]; oral semaglutide 7 mg, -1.5% [P = 0. 3942]; oral semaglutide 14 mg, -1.7% [P = 0.0272]).

Combination With Metformin: Oral Semaglutide Vs Empagliflozin

PIONEER-2 was a 52-week trial that randomized (1:1) 822 eligible patients with T2D uncontrolled with metformin monotherapy to either once-daily oral semaglutide 14 mg or empagliflozin 25 mg (while continuing metformin treatment). Oral semaglutide provided a superior reduction in A1C compared with empagliflozin at Week 26 both with the treatment policy estimand (mean baseline: 8.1% in both the semaglutide 14 mg group and the empagliflozin 25 mg group; mean changes from baseline [mean baseline 8.1%]: -1.3% with semaglutide 14 mg and -0.9% with empagliflozin; ETD in favor of semaglutide: -0.4%; P <0.0001) and the trial product estimand (mean changes from baseline: -1.4% with semaglutide 14 mg and -0.9% with empagliflozin; ETD: -0.5%; P <0.0001). Semaglutide was also superior at Week 52 for both the treatment policy estimand (mean changes from baseline: -1.3% with semaglutide 14 mg and -0.9% with empagliflozin; ETD in favor of semaglutide: -0.4%; P <0.0001) and the trial product estimand (mean changes from baseline: -1.3% with semaglutide 14 mg and -0.8% with empagliflozin; ETD: -0.5%; P <0.0001).

Combination With Sulfonylureas and/or Metformin: Oral Semaglutide Vs Sitagliptin

PIONEER-3 was a 78-week trial that randomized (1:1:1:1) 1864 eligible patients with T2D uncontrolled with metformin with or without sulfonylurea to a once-daily oral regimen of: 1) oral semaglutide 3 mg; 2) oral semaglutide 7mg; 3) oral semaglutide 14 mg; or 4) sitagliptin 100 mg. The 7 mg and 14 mg semaglutide dosages were superior to sitagliptin with respect to the primary endpoint of A1C reduction from baseline at Week 26 with the treatment policy estimand (mean baseline: 8.3% in the semaglutide 3 mg group, 8.4% in the semaglutide 7 mg group, 8.3% in the semaglutide 14 mg group and 8.3% in the sitagliptin group; mean changes from baseline: -0.6% with semaglutide 3 mg, -1.0% with semaglutide 7 mg, -1.3% with semaglutide 14 mg and -0.8% with sitagliptin; ETDs against sitagliptin: 7 mg semaglutide: -0.3%; 14 mg semaglutide: -0.5%; P <0.001 for both). The superiority was maintained at Week 78 (ETDs against sitagliptin: 7 mg semaglutide: -0.1%; 14 mg semaglutide: -0.4%; P <0.001 for both).

Combination With SGLT2 Inhibitors and/or Metformin: Oral Semaglutide Vs SC Liraglutide

PIONEER-4 was a 52-week trial that randomized (2:2:1) 711 eligible patients with T2D uncontrolled on metformin with or without a SGLT2 inhibitor to a once daily regimen of: 1) oral semaglutide (dose escalated to 14 mg); 2) SC liraglutide (dose escalated to 1.8 mg); or 3) placebo. The patients continued their existing background glucose-lowering medication. With regard to the primary endpoint (mean change from baseline in A1C at Week 26 under the treatment policy estimand), oral semaglutide demonstrated superiority to the placebo and non-inferiority to SC liraglutide (mean baseline: 8.0% in the semaglutide 14 mg group, 8.0% in the liraglutide 1.8 mg group and 7.9% in the placebo group; mean changes from baseline: -1.2% with semaglutide 14 mg, -1.1% with liraglutide 1.8 mg and -0.2% with placebo; ETD against placebo -1.1% [P <0.0001], ETD against liraglutide -0.1% [P =0.0645]). With the trial product estimand, semaglutide was superior to both the placebo and SC liraglutide (mean changes from baseline: oral semaglutide -1.3%, liraglutide -1.1%, placebo -0.1%; ETDs: -1.2% against placebo [P <0.0001] and -0.2% against liraglutide [P = 0.0056]).

Combination With Insulin and/or Metformin

In the 52-week PIONEER-8 study, 731 eligible patients with T2D uncontrolled on insulin with or without metformin were randomized (1:1:1:1) to supplement their existing regiment with a once daily dose of: 1) oral semaglutide 3 mg; 2) oral semaglutide 7 mg; 3) oral semaglutide 14 mg; or 4) placebo. At all three doses, semaglutide demonstrated superiority to the placebo with respect to the primary endpoint of A1C reduction at Week 26 under the treatment policy estimand (mean baseline: 8.2% in all groups; mean changes from baseline: semaglutide 3 mg -0.6%, semaglutide 7 mg -0.9%, semaglutide 14 mg -1.3%, placebo -0.1%; ETD from the placebo: semaglutide 3 mg, -0.5%; semaglutide 7 mg, -0.9%; semaglutide 14 mg, -1.2%; P <0.001 for all). Semaglutide also showed a significantly greater reduction in A1C at Week 26 for the trial product estimand (mean changes from baseline: semaglutide 3 mg -0.6%, semaglutide 7 mg -1.0%, semaglutide 14 mg -1.4%, placebo -0.0%; ETD from the placebo: semaglutide 3 mg, -0.6%; semaglutide 7 mg, -1.0%; semaglutide 14 mg, -1.4%; P <0.001 for all). Semaglutide was also superior at Week 52 for both the treatment policy estimand (mean changes from baseline: semaglutide 3 mg -0.6%, semaglutide 7 mg -0.8%, semaglutide 14 mg -1.2%, placebo -0.2%; ETD from the placebo: semaglutide 3 mg -0.4%, semaglutide 7 mg -0.6%, semaglutide 14 mg -0.9%; P <0.001 for all) and the trial product estimand (mean changes from baseline: semaglutide 3 mg -0.5%, semaglutide 7 mg -0.8%, semaglutide 14 mg -1.2%, placebo 0.0%; ETD from the placebo: semaglutide 3 mg -0.5%, semaglutide 7 mg -0.9%, semaglutide 14 mg -1.3%; P <0.001 for all).

Combination With Background Oral Glucose-Lowering Medications: Oral Semaglutide Vs Sitagliptin

In the 52-week PIONEER-7 trial, 504 eligible patients with T2D inadequately controlled on stable daily doses of 1-2 oral glucose-lowering drugs were randomized (1:1) to add one of the following once-daily doses to their existing regimen: 1) semaglutide with flexible dose adjustments to 3, 7, or 14 mg; or 2) sitagliptin 100 mg. At Week 52, with the treatment policy estimand, a greater proportion of patients in the semaglutide group (58%) achieved the primary endpoint (A1C <7% [53 mmol/mol]) compared to those in the sitagliptin group (25%; odds ratio [OR] = 4.40; P <0.001). Semaglutide was also superior to sitagliptin with the trial product estimand, with 63% of patients achieving the primary endpoint, compared to 28% in the sitagliptin group (OR = 5.54; P <0.001).

Combination With Background Oral Glucose-Lowering Medications: Oral Semaglutide Vs SC Dulaglutide

PIONEER-10 was a Japan-based 52-week trial that randomized (2:2:2:1) 458 eligible patients (≥20 years or age) with uncontrolled T2D despite background medication to receive: 1) oral semaglutide 3 mg once-daily; 2) oral semaglutide 7 mg once-daily; 3) oral semaglutide 14 mg once-daily; or 4) SC dulaglutide 0.75 mg once-weekly. Under the primary (treatment policy) estimand, oral semaglutide 14 mg demonstrated superiority in A1C reduction compared to SC dulaglutide at Week 26 (mean baseline: 8.2% in the semaglutide 3 mg group, 8.3% in the semaglutide 7 mg group, 8.4% in the semaglutide 14 mg group and 8.4% in the dulaglutide 0.75 mg group; mean changes from baseline: semaglutide 3 mg -1.1%, semaglutide 7 mg -1.7%, semaglutide 14 mg -2.0%, dulaglutide 0.75 mg -1.5%; ETD from dulaglutide: oral semaglutide 3 mg 0.4% [P = 0.0026], oral semaglutide 7 mg -0.1% [P = 0.2710], oral semaglutide 14 mg -0.3% [P = 0.0006]) and at Week 52 (mean changes from baseline: semaglutide 3 mg -0.9%, semaglutide 7 mg -1.4%, semaglutide 14 mg -1.7%, dulaglutide 0.75 mg -1.4%; ETD from dulaglutide: oral semaglutide 3 mg 0.5% [P = 0.0005], oral semaglutide 7 mg -0.1% [P = 0.6580], oral semaglutide 14 mg -1.7% [P = 0.0170]).

Combination With Background Oral Glucose-Lowering Medications

PIONEER PLUS was a 68-week long trial that randomized (1:1:1) 1606 adult patients with T2D that were on stable daily doses of oral glucose-lowering medication, to receive either oral semaglutide at a daily dose of 14 mg (the currently approved dose), 25 mg, or 50 mg. Oral semaglutide 25 mg and 50 mg demonstrated superiority in lowering A1C compared to semaglutide 14 mg at week 52 (mean baseline: 9.0% for all three groups; mean A1C changes for the treatment policy estimand: -1.5% for semaglutide 14 mg, -1.8% for semaglutide 25 mg [ETD: -0.27%; P=0.0006], and -2.0 for semaglutide 50 mg [ETD -0.53%; P<0.0001]). Both 25 mg and 50 mg doses of semaglutide also reduced bodyweight to a greater extent than the 14 mg dose (mean baseline: 96.4 kg for all three groups; mean changes for the treatment policy estimand: -4.4 kg for semaglutide 14 mg, -6.7 kg for semaglutide 25 mg [ETD: -2.32 kg; P<0.0001], and -8.0 kg for semaglutide 50 mg [ETD: -3.63 kg; P<0.0001]).

Safety and Tolerability

Like the label for SC semaglutide, the prescribing information for oral semaglutide includes a boxed warning for risk of thyroid C-cell tumors, based on the increased risk of these malignancies in rodents. However, whether this risk applies to humans has not been determined. Oral semaglutide is contraindicated in patients with a personal or family history of medullary thyroid carcinoma or in patients with Multiple Endocrine Neoplasia syndrome type 2. The prescribing information for oral semaglutide also contains warnings and precautions for pancreatitis, diabetic retinopathy complications, hypoglycemia, acute kidney injury and hypersensitivity reactions.

The most common adverse reactions, excluding hypoglycemia, occurring in ≥5% of patients treated with oral semaglutide, include nausea (11% with 7 mg; 20% with 14 mg), abdominal pain (10% with 7 mg; 11% with 14 mg), diarrhea (9% with 7 mg; 10% with 14 mg), decreased appetite (9% with 7 mg; 6% with 14 mg), vomiting (8% with 7 mg; 6% with 14 mg) and constipation (6% with 7 mg; 5% with 14 mg).

Oral Semaglutide in Patients with Renal Impairment (PIONEER-5)

PIONEER-5 was a 26-week trial that investigated the safety and efficacy of oral semaglutide for the treatment of T2D in patients with moderate renal impairment. The trial randomized (1:1) 324 eligible patients with an estimated glomerular filtration rate of 30–59 mL/min per 1.73 m² and T2D uncontrolled on metformin, sulfonylureas, or both, or basal insulin with or without metformin, to once-daily oral semaglutide (escalated up to 14 mg) or placebo, in addition to their background medication. Oral semaglutide 14 mg demonstrated superiority in the primary endpoint (reduction from baseline in HbA1 at Week 26) assessed with either the treatment policy estimand (mean baseline: 8.0% in the semaglutide 14 mg group and 7.9% in the placebo group; mean changes from baseline: semaglutide 14 mg -1.0%, placebo -0.2%; ETD: -0.8%; P <0.0001) or the trial product estimand (mean changes from baseline: semaglutide 14 mg -1.1%, placebo -0.1%; ETD: -1.0%; P <0.0001). Oral semaglutide demonstrated an overall safety and renal safety profile consistent with that seen for other GLP-1 receptor agonists.

Cardiovascular Outcome Trial of Oral Semaglutide (PIONEER-6)

The CV safety of oral semaglutide was investigated in PIONEER-6, a trial that included 3183 patients with T2D and an established CVD. Patients were randomized (1:1) to receive once-daily oral semaglutide (dose escalation up to 14 mg) or placebo. Randomization was stratified according to evidence of CVD or the presence of CV risk factors (84.7% of patients had established CVD and 15.3% had CV risk factors). The primary outcome was the time from randomization to the first occurrence of a major adverse CV event (MACE), a composite of death from CV causes (including death with undetermined cause), nonfatal MI, or nonfatal stroke.

The primary outcome occurred in 3.8% of patients receiving oral semaglutide and in 4.8% of patients receiving the placebo (HR 0.79; P <0.001 for inferiority; P = 0.17 for superiority). The incidence of the individual components of the primary outcome was similar between the semaglutide and placebo groups, including death from CV causes (0.9% vs 1.9% with semaglutide and placebo, respectively; HR 0.49), nonfatal MI (2.3% vs 1.9%; HR 0.73) and nonfatal stroke (0.8% vs 1.0%; HR 0.74).

Based on the results from SUSTAIN-6, diabetic retinopathy was an adverse event of interest in PIONEER-6. The incidence of diabetic retinopathy was comparable between the oral semaglutide (7.1%) and the placebo (6.3%) groups. Almost all cases of diabetic retinopathy were nonproliferative and did not require additional treatment in PIONEER-6. A long-term study investigating the effects of semaglutide on the development and progression of diabetic retinopathy is currently ongoing.

Dosage and Administration

Oral semaglutide is provided in three tablet strengths: 3 mg, 7 mg and 14 mg. Oral semaglutide should be initiated at a 3 mg once-daily dose and increased to 7 mg once-daily after 30 days in all patients. The dosage may be increased to 14 mg once daily if target glycemic control is not achieved after 30 days on the 7 mg once-daily dose. It is not recommended to use two 7 mg tablets to achieve a 14 mg dose.

To optimize absorption, semaglutide tablets should be taken at least 30 minutes before the first food, beverage, or other oral medications of the day, with no more than 4 ounces of plain water only. Tablets should be swallowed whole, not split, crushed, or chewed. If a dose of semaglutide dose is missed, it should be skipped and the next dose taken the following day.

Switching from Oral to Injectable Semaglutide

Patients taking 14 mg of oral semaglutide once daily can be transitioned to 0.5 mg of injectable semaglutide once weekly. SC semaglutide can be started the day after the last oral semaglutide dose.

Summary

Semaglutide is the first GLP-1 RA to receive approval as an oral formulation. Across the phase 3 PIONEER trials, oral semaglutide has demonstrated a favorable safety profile consistent with SC GLP-1 RAs. It also demonstrated superiority to sitagliptin and empagliflozin and non-inferiority to other SC GLP-1 RAs in A1C reduction.

Mounjaro (Tirzepatide)

Tirzepatide is a newly (2022) approved GIP/GLP-1 RA indicated as an adjunct to diet and exercise for improving glycemic control in adults with T2D. Based on the sequence of endogenous GIP, tirzepatide is a 39-amino-acid peptide and the first dual GIP receptor and GLP-1 receptor agonist to receive approval for the treatment of T2D. Although GIP appears to be the dominant incretin in healthy individuals, its role in patients with T2D is not as prominent as that of GLP-1 (see the Incretins section above); GIP stimulates glucagon secretion under hypoglycemia and has lipogenic effects. Early data on the potential therapeutic use of GIP were not encouraging: no incretin effect in T2D was demonstrated with GIP infusion, administration of GIP increased glucagon secretion even under hyperglycemia, it was associated with fat mass accumulation and GIP receptor antagonism resulted in substantial weight loss in preclinical studies in mice and non-human primates.

Nevertheless, subsequent preclinical and early clinical studies on GIP receptor agonism reported promising results. The need for additional glycemic control in some patients on GLP-1 RAs led to the therapeutic utilization of GIP receptor agonism in combination incretin RAs. Even though the relative contribution of GIP receptor agonism to the overall tirzepatide mechanism of action is still not understood, the dual RA has demonstrated remarkable glycemic control and weight loss efficacy in phase 3 trials (see the Efficacy sub-section below). Like semaglutide and liraglutide, tirzepatide has been associated with dose-dependent and treatment-duration-dependent thyroid C-cell tumor development in rodents, although the relevance of these findings in human patients is not yet determined.

Efficacy

The efficacy of tirzepatide for improved glycemic control in adults with T2D was established in 6 phase 3 trials (SURPASS-1 to -6). Tirzepatide has been studied as monotherapy (SURPASS-1), as an add-on to metformin, sulfonylureas and/or SGLT2 inhibitors (SURPASS-2, -3, and -4), as well as in combination with basal insulin with or without metformin (SURPASS-5 and -6). In the SURPASS program, the efficacy of tirzepatide was compared to placebo, semaglutide (1 mg), insulin degludec, insulin glargine, or insulin lispro; tirzepatide was superior to all comparators at the tested doses with respect to A1C and body weight reductions from baseline (Table 19-11). The efficacy was unaffected by age, gender, race, ethnicity, or geographic area, nor baseline BMI, A1C level, diabetes duration, or renal function.

Monotherapy

SURPASS-1 was a 40-week double-blind trial of 478 adult patients with T2D. Patient were naïve to injectable diabetes medication and managed glycemic levels with diet and exercise. Eligible patients were randomized (1:1:1:1) to receive a once-weekly injection of 5 mg, 10 mg, or 15 mg of tirzepatide, or placebo. At 40 weeks, monotherapy with tirzepatide resulted in significant reduction in A1C (mean baseline: 8.1% in the placebo group, 8.0% in the tirzepatide 5-mg group, 7.9% in the tirzepatide 10 mg group, and 7.9% in the tirzepatide 15 mg group; mean changes from baseline: -0.1% with placebo, -1.8% with tirzepatide 5 mg, -1.7% with tirzepatide 10 mg, and -1.7% with tirzepatide 15 mg; difference from the placebo: tirzepatide 5 mg -1.7%, tirzepatide 10 mg -1.6%, tirzepatide 15 mg -1.6%; P <0.001 for all).

Tirzepatide treatment also resulted in a progressive reduction in body weight with increasing dose. At 40 weeks, body weight significantly decreased from baseline in the tirzepatide groups compared to the placebo group (mean baseline: 84.5 kg in the placebo group, 87.0 kg in the tirzepatide 5 mg, 86.2 kg in the tirzepatide 10 mg, and 85.5 kg in the tirzepatide 15 mg group; mean changes from baseline: -1.0 kg with placebo, -6.3 kg with tirzepatide 5 mg, -7.0 kg with tirzepatide 10 mg, and -7.8 kg with tirzepatide 15 mg; difference from the placebo: tirzepatide 5 mg -5.3 kg, tirzepatide 10 mg -6.0 kg, tirzepatide 15 mg -6.8 kg; P <0.001 for all).

Combination With Metformin: Tirzepatide vs Semaglutide

In SURPASS-2, a 40-week open-label trial (double-blinded for tirzepatide dose), tirzepatide was compared to semaglutide 1 mg once-weekly. A total of 1879 adult patients with T2D and inadequate glycemic control on stable doses of metformin alone were randomized (1:1:1:1) to a weekly dose of semaglutide 1 mg or tirzepatide 5 mg, 10 mg, or 15 mg.

At all three doses, tirzepatide was superior to semaglutide 1 mg with respect to A1C reduction (mean baseline: 8.3% in all groups; mean changes from baseline: -1.9% with semaglutide 1 mg, -2.0% with tirzepatide 5 mg, -2.2% with tirzepatide 10 mg, and -2.3% with tirzepatide 15 mg; difference from semaglutide: tirzepatide 5 mg -0.2%, tirzepatide 10 mg -0.4%, tirzepatide 15 mg -0.5%; P <0.05 for tirzepatide 5 mg, P <0.001 for tirzepatide 10 mg and 15 mg).

All three doses of tirzepatide also demonstrated superiority to semaglutide 1 mg with respect to body weight reduction at 40 weeks (mean baseline weight: 93.7 kg in the semaglutide 1 mg group, 92.5 kg in the tirzepatide 5 mg, 94.8 kg in the tirzepatide 10 mg, and 93.8 kg in the tirzepatide 15 mg group; mean changes from baseline: -5.7 kg with semaglutide 1 mg, -7.6 kg with tirzepatide 5 mg, -9.3 kg with tirzepatide 10 mg, and -11.2 kg with tirzepatide 15 mg; difference from semaglutide 1 mg: tirzepatide 5 mg -1.9 kg, tirzepatide 10 mg -3.6 kg, tirzepatide 15 mg -5.5 kg; P <0.05 for tirzepatide 5 mg, P <0.001 for tirzepatide 10 mg and 15 mg).

Combination With Metformin With or Without SGLT2 Inhibitors: Comparison to Insulin Degludec

SURPASS-3 was a 52-week open-label trial that randomized 1444 adult patients with T2D who were on stable doses of metformin with or without SGLT2 inhibitor to add tirzepatide 5 mg, 10 mg, or 15 mg once weekly, or insulin degludec 100 units/mL once daily. Insulin degludec was initiated at 10 units once daily and was modified weekly throughout the trial using a treat-to-target algorithm based on self-measured fasting blood glucose levels. At week 52, 26% of insulin degludec patients met their fasting blood glucose goal of 90 mg/dL and the average daily insulin degludec dosage was 49 U (0.5 U per kilogram).

Compared to daily doses of insulin degludec, treatment with tirzepatide 5 mg, 10 mg and 15 mg once weekly for 52 weeks resulted in a statistically significant reduction in A1C (mean baseline: 8.1% in the insulin degludec group and 8.2% in the tirzepatide 5 mg, 10 mg and 15 mg groups; mean changes from baseline: -1.3% with insulin degludec, -1.9% with tirzepatide 5 mg, -2.0% with tirzepatide 10 mg and -2.1% with tirzepatide 15 mg; difference from insulin degludec: tirzepatide 5 mg -0.6%, tirzepatide 10 mg -0.8%, tirzepatide 15 mg -0.9%; P <0.001 for all).

All three once-weekly doses of tirzepatide also demonstrated superiority to once-daily insulin degludec with regard to weight loss at 52 weeks (mean baseline weight: 94.0 kg in the insulin degludec group, 94.4 kg in the tirzepatide 5 mg, 93.8 kg in the tirzepatide 10 mg and 94.9 kg in the tirzepatide 15 mg group; mean changes from baseline: 1.9 kg with insulin degludec, -7.0 kg with tirzepatide 5 mg, -9.6 kg with tirzepatide 10 mg and -11.3 kg with tirzepatide 15 mg; difference from insulin degludec: tirzepatide 5 mg -8.9 kg, tirzepatide 10 mg -11.5 kg, tirzepatide 15 mg -13.2 kg; P <0.001 for all).

Combination With Metformin and/or Sulfonylurea and/or SGLT2 Inhibitors: Comparison to Insulin Glargine

SURPASS-4 was a 104-week open-label trial (primary endpoint assessed at week 52) in which 2002 adult patients with T2D and increased CV risk were randomized (1:1:1:3) to tirzepatide 5 mg, 10 mg, or 15 mg once weekly, or insulin glargine 100 units/mL once daily on a background of metformin and/or sulfonylureas and/or SGLT2 inhibitors. Insulin glargine was initiated at 10 U per day and increased weekly throughout the trial using a treat-to-target algorithm based on self-measured fasting blood glucose levels. At week 52, 30% of insulin glargine patients met the fasting serum glucose goal of less than 100 mg/dL and the average daily insulin glargine dosage was 44 U (0.5 U per kilogram).

Treatment with all three doses of tirzepatide resulted in a statistically significant reduction in A1C at 52 weeks, compared to insulin glargine once daily (mean baseline: 8.6% in the tirzepatide 10 mg group and 8.5% in all other groups; mean changes from baseline: -1.4% with insulin glargine, -2.1% with tirzepatide 5 mg, -2.3% with tirzepatide 10 mg and -2.4% with tirzepatide 15 mg; difference from the insulin glargine: tirzepatide 5 mg -0.7%, tirzepatide 10 mg -0.9%, tirzepatide 15 mg -1.0%; P <0.001 for all).

Tirzepatide treatment at all three doses also resulted in superior weight loss at 52 weeks, compared to once-daily insulin glargine (mean baseline weight: 90.2 kg in the insulin glargine group, 90.3 kg in the tirzepatide 5 mg, 90.6 kg in the tirzepatide 10 mg and 90.0 kg in the tirzepatide 15 mg group; mean changes from baseline: 1.7 kg with insulin glargine, -6.4 kg with tirzepatide 5 mg, -8.9 kg with tirzepatide 10 mg and -10.6 kg with tirzepatide 15 mg; difference from insulin degludec: tirzepatide 5 mg -8.1 kg, tirzepatide 10 mg -10.6 kg, tirzepatide 15 mg -12.2 kg; P <0.001 for all).

Combination With Basal Insulin With or Without Metformin

In SURPASS-5, a 40-week double-blind trial, in which 475 patients with T2D and inadequate glycemic control on insulin glargine 100 units/mL with or without metformin were assigned to tirzepatide 5 mg, 10 mg, or 15 mg once weekly, or placebo. A treat-to-target algorithm based on self-measured fasting blood glucose readings was used to alter the dose of background insulin glargine, with a goal of less than 100 mg/dL. At baseline, patients receiving tirzepatide 5 mg, 10 mg, 15 mg and placebo received 34, 32, 35 and 33 units of insulin glargine, respectively. In patients with a A1C of less than 8.0%, the baseline insulin glargine dose was lowered by 20% at randomization. At week 40, patients receiving tirzepatide 5 mg, 10 mg, 15 mg and placebo received 38, 36, 29 and 59 units of insulin glargine per day, respectively.

At week 40, tirzepatide 5 mg, 10 mg and 15 mg once weekly resulted in a statistically significant reduction in A1C compared to the placebo (mean baseline: 8.4% in the placebo group, 8.3% in the tirzepatide 5 mg group, 8.4% in the tirzepatide 10 mg group and 8.2% in the tirzepatide 15 mg group; mean changes from baseline -0.9% with placebo, -2.1% with tirzepatide 5 mg, -2.4% with tirzepatide 10 mg and -2.3% with tirzepatide 15 mg; difference from the placebo: tirzepatide 5 mg -1.2%, tirzepatide 10 mg -1.5%, tirzepatide 15 mg -1.5%; P <0.001 for all).

At all three doses, tirzepatide treatment also resulted in significant body weight reduction from baseline compared to the placebo (mean baseline weight: 94.2 kg in the placebo group, 95.8 kg in the tirzepatide 5 mg, 94.6 kg in the tirzepatide 10 mg and 96.0 kg in the tirzepatide 15 mg group; mean changes from baseline: + 1.6 kg with placebo, -5.4 kg with tirzepatide 5 mg, -7.5 kg with tirzepatide 10 mg and -8.8 kg with tirzepatide 15 mg; difference from the placebo: tirzepatide 5 mg -7.1 kg, tirzepatide 10 mg -9.1 kg, tirzepatide 15 mg -10.5 kg; P <0.001 for all).

Combination With Basal Insulin With or Without Metformin: Tirzepatide vs Insulin Lispro

SURPASS-6 was a 52-week, open-label trial with a 4-week safety follow-up, which randomized (1:1:1:3) 1428 patients with T2D inadequately controlled with basal insulin (insulin NPH, insulin glargine, insulin detemir, or insulin degludec), with or without any combination of up to 2 oral glucose-lowering medications (metformin, sulfonylurea, or DPP-4 inhibitors), to receive one of the following: 1) tirzepatide 5 mg once weekly; 2) tirzepatide 10 mg once weekly; 3) tirzepatide 15 mg once weekly; or 4) prandial insulin lispro three times a day. Prior to randomization, individuals eligible to enter the trial discontinued their previous diabetes treatment, with the exception of metformin, and were put on a standardized therapy of insulin glargine 100 units/mL for an insulin stabilization period. During this period, the dosage of insulin glargine was adjusted to achieve a fasting blood glucose target range of 100 to 125 mg/dL, and at randomization, the doses were reduced by 30% in all groups to lower the risk of hypoglycemia with the initiation of tirzepatide. Insulin lispro was initiated at lower doses (4 units/mL) and titrated throughout the trial. The dose was adjusted twice a week until week 24, and at least once a week thereafter, to achieve a target prelunch, predinner, and bedtime blood glucose of 100-125 mg/dL.

At week 52, all three doses of tirzepatide showed statistical superiority in lowering A1C compared to insulin lispro (mean baseline: 8.80% in the insulin lispro group, 8.88% in the tirzepatide 5 mg group, 8.78% in the tirzepatide 10 mg group, and 8.74% in the tirzepatide 15 mg group; mean changes from baseline: -1.13% with insulin lispro, -1.92% with tirzepatide 5 mg, -2.15% with tirzepatide 10 mg, and -2.27% with tirzepatide 15 mg; P<0.001 for all).

Tirzepatide 5 mg, 10 mg, and 15 mg demonstrated superiority to insulin lispro in body weight reduction from baseline as well (mean baseline weight: 90.3 kg in the insulin lispro group, 91.7 kg in the tirzepatide 5 mg group, 89.1 kg in the tirzepatide 10 mg group, and 91.2 kg in the tirzepatide 15 mg group; mean changes from baseline: +3.2 kg with insulin lispro, -6.7 kg with tirzepatide 5 mg, -9.2 kg with tirzepatide 10 mg, -11.0 kg with tirzepatide 15 mg; P<0.001 for all).

Safety and Tolerability

Tirzepatide is contraindicated in patients with a history of medullary thyroid carcinoma or in patients with Multiple Endocrine Neoplasia syndrome type 2 because it causes dose-dependent and treatment-duration-dependent thyroid C-cell tumors in both male and female rodents. Although the human relevance of tirzepatide-induced rodent thyroid C-cell tumors has not been determined, tirzepatide should be administered with caution and alongside routine monitoring of serum calcitonin, neck imaging and other evaluation tests. In addition to a black box warning about the possible development of C-cell tumors, the prescribing information for tirzepatide includes warnings and precautions for pancreatitis, hypoglycemia, acute kidney injury, gastrointestinal disease, diabetic retinopathy complications, acute gallbladder disease and hypersensitivity reactions.

The most commonly reported adverse reactions of 5 mg tirzepatide which occurred in ≥5% of treated adult patients were nausea (12%), diarrhea (12%), decreased appetite (5%), vomiting (5%), constipation (6%), dyspepsia (8%) and abdominal pain (6%).

Dosage and Administration

Tirzepatide is given as a subcutaneous injection once a week. It is introduced at a 2.5 mg dose; the dosage should be increased to 5 mg once weekly after 4 weeks. If additional glycemic control is needed, the dosage can be increased by 2.5 mg every 4 weeks after at least 4 weeks on the current dose, to a maximum of 15 mg once a week.

If a dose is missed, the patient should take the tirzepatide injection as soon as possible, preferably within 4 days (96 hours). If more than 4 days since the last dose have elapsed, the missed dose should be skipped and the following dose taken on the next regularly scheduled day. Patients can then continue their usual once weekly dose plan.

Fixed-Ratio Combination of a GLP-1 Agonist
and Basal Insulin

Two once-daily, single-injection, fixed-combination products consisting of basal insulin and a GLP-1 receptor agonist have been approved by the FDA: Xultophy 100/3.6 (100 units insulin degludec per mL and 3.6 mg liraglutide per mL) and Soliqua 100/33 (100 units insulin glargine per mL and 33 mcg lixisenatide per mL). These products were developed for patients initiating or intensifying insulin therapy who could benefit from a convenient dosing regimen.

Insulin Degludec/Liraglutide (Xultophy 100/3.6)

Xultophy 100/3.6 is indicated as an adjunct to diet and exercise to improve glycemic control in adults with T2D. It is not recommended as first-line therapy in this patient population. It is available as a clear solution in a 3 mL pre-filled, disposable, single-patient-use pen injector.

Efficacy

The approval of Xultophy was based on data from the DUAL phase 3 clinical trial program. The first studies in the series were DUAL-I and DUAL-II, both 26-week, randomized trials. DUAL-I was an open-label study enrolling 1663 insulin-naïve patients with T2D; it compared Xultophy with its components given alone, while continuing pretrial doses of OADs. DUAL-II was a double-blind study enrolling 413 basal insulin-experienced patients with T2D; it compared the safety and efficacy of Xultophy to insulin degludec, both with metformin. The primary endpoint in both trials was change from baseline in A1C after 26 weeks of treatment.

In the DUAL-I study, A1C decreased by 1.9% with Xultophy, 1.4% with insulin degludec, and 1.3% with liraglutide after 26 weeks. Xultophy met the criteria for noninferiority compared with insulin degludec (-0.47%; P <0.0001), and the criteria for superiority compared with liraglutide (-0.64%; P <0.0001). A greater proportion of patients in the Xultophy treatment group (81%) achieved <7.0% A1C at week 26 compared with patients receiving either insulin degludec (65%) or liraglutide alone (60%). A 26-week extension of DUAL-I assessed the sustained efficacy and safety of Xultophy. At 52 weeks, mean A1C was reduced by 1.84% for the Xultophy group, 1.40% for the insulin degludec group and 1.21% for the liraglutide group. Xultophy was also associated with a significant decrease in body weight compared with insulin degludec alone (estimated treatment difference [ETD] -2.80 kg, P <0.0001).

Results from the DUAL-II trial, which enrolled insulin-experienced patients, were similar. Mean A1C decreased over time in both Xultophy and insulin degludec treatment groups, but the decrease was more pronounced and observed earlier with Xultophy. At 26 weeks, A1C decreased by 1.9% with Xultophy and by 0.9% with insulin degludec, demonstrating the superiority of Xultophy (ETD -1.1%; P <0.0001). At week 26, more patients in the Xultophy group achieved <7.0% A1C (60% vs 23%). Additionally, Xultophy was associated with significantly greater weight loss compared with insulin degludec (ETD -2.5 kg; P <0.0001).

More recently, the DUAL-V trial assessed whether Xultophy was noninferior to continued insulin glargine treatment in patients with T2D inadequately controlled on insulin glargine and metformin. The trial randomized 557 patients to receive either Xultophy or insulin glargine, with twice-weekly titration to a glucose target of 72 to 90 mg/dL. The primary outcome of the trial was change in A1C level after 26 weeks. Mean A1C decreased by 1.81% for the Xultophy group compared with 1.13% for the insulin glargine group, demonstrating both the noninferiority and superiority of Xultophy (ETD -0.59; P <0.001). Additionally, patients in the Xultophy group experienced weight loss, whereas patients in the insulin glargine group experienced weight gain (ETD -3.20 kg; P <0.001).

Overall, for A1C reduction, Xultophy demonstrated noninferiority to insulin degludec in insulin-naïve patients (DUAL-I), superiority to insulin degludec in insulin-experienced patients (DUAL-II), and superiority to continued insulin glargine titration in patients inadequately controlled on insulin glargine and metformin (DUAL-V).

Safety and Tolerability

In the DUAL trials, Xultophy was well tolerated, with a safety profile reflected by its component parts, and no new adverse events or tolerability issues. In some trials, the rate of hypoglycemia was more favorable with Xultophy. In DUAL-I, the number of confirmed hypoglycemic events was lower with Xultophy than with degludec alone (estimated rate ratio 0.68; P = 0.0023). This observation was maintained in the DUAL-1 extension, where Xultophy was associated with a 37% lower rate of hypoglycemia. In DUAL-II, hypoglycemic incidence was comparable between patients receiving Xultophy or insulin degludec (24% vs 25%, respectively). Lastly, in DUAL-V, Xultophy was associated with fewer confirmed hypoglycemic episodes per patient year (2.23 vs 5.05; estimated rate ratio 0.43; P <0.001), but more nonserious gastrointestinal adverse events (79 vs 18) compared with insulin glargine.

Due to its liraglutide component, Xultophy is contraindicated in patients with a personal or family history of medullary thyroid carcinoma and in patients with Multiple Endocrine Neoplasia syndrome type 2.

Dosing and Administration

Prior to initiating Xultophy 100/3.6, therapy with liraglutide or basal insulin should be discontinued. Administer Xultophy once daily at the same time each day with or without food. The recommended starting dosage is 16 units (16 units of insulin degludec and 0.58 mg of liraglutide) given subcutaneously once daily. The daily dose is then titrated from 1 to 50 units based on FPG, with the maximum dose corresponding to 50 units of insulin degludec and 1.8 mg of liraglutide.

Insulin Glargine/Lixisenatide (Soliqua 100/33)

Soliqua 100/33 is indicated as an adjunct to diet and exercise to improve glycemic control in adults with T2D. It is available as a clear solution in a 3 mL prefilled, disposable, single-patient use SoloStar pen.

Efficacy

The approval of Soliqua was based on the efficacy outcomes from four major clinical trials: GetGoal-Duo 1, the LixiLan Proof-of-Concept (LixiLan PoC) trial, LixiLan-O and LixiLan-L. The former three trials enrolled patients with T2D inadequately controlled on metformin-based oral therapies, whereas LixiLan-L enrolled patients with T2D inadequately controlled on basal insulin, alone or in combination with OADs. In these trials, treatment with metformin was continued throughout the study period. The efficacy of switching to Soliqua from a GLP-1 RA was assessed in LixiLan-G.

The first trial, GetGoal-Duo 1, initiated patients on insulin glargine during a 12-week run-in period. The safety and efficacy of adding lixisenatide to the treatment regimen was then evaluated over 24 weeks while insulin titration continued. A1C decreased during the run-in period from 8.6% to 7.6%. The addition of lixisenatide decreased A1C significantly more than placebo: 0.7% vs 0.4%, respectively (P <0.0001), and more patients in the lixisenatide group achieved target A1C <7.0% (56% vs 39%; P = 0.0001).

LixiLan PoC was similar to GetGoal-Duo 1, but it compared the efficacy and safety Soliqua 100/33, the fixed-ratio combination of insulin glargine and lixisenatide, with insulin glargine. Three hundred twenty-three patients were randomized 1:1 to received either once-daily Soliqua or insulin glargine for 24 weeks, and both treatments were titrated based on FPG levels. At week 24, reductions in A1C were greater with Soliqua (-1.82%) than with insulin glargine (-1.64%), meeting the criteria for both noninferiority and superiority (treatment difference -0.17%; P = 0.01), and numerically more Soliqua-treated patients reached target A1C <7.0% (84 vs 78%; nonsignificant). Weight loss was also experienced by patients in the Soliqua group (-1.16 kg) while patients in the insulin glargine group gained weight (0.39 kg); treatment difference -1.44 kg; (P <0.0001).

Unlike the previous two trials, LixiLan-O compared the safety and efficacy of Soliqua vs treatment with each component alone, in 1170 patients over 30 weeks. Results from LixiLan-O were comparable to previous studies: Soliqua showed significantly greater reductions in A1C compared with insulin glargine or lixisenatide (-1.6%, -1.3%, -0.9%, respectively; P <0.0001), and more patients achieved target A1C <7.0% (74%, 59%, 33%, respectively). Soliqua-treated patients also lost significantly more weight (-0.3 kg) compared with insulin glargine-treated patients (who gained 1.1 kg; treatment difference -1.4 kg; [P <0.0001]), but not lixisenatide-treated patients (-2.3 kg).

LixiLan-L assessed the efficacy and safety of Soliqua vs insulin glargine over 30 weeks in 736 patients inadequately controlled on basal insulin. At 30 weeks, Soliqua showed significantly greater reduction in A1C compared with insulin glargine (-1.1% vs -0.6%; P <0.0001), and more patients reached target A1C <7.0% (55% vs 30%; P <0.0001). Consistent with previous trials, body weight increased with insulin glargine (0.7 kg) and decreased with Soliqua (-0.7 kg; difference -1.4 kg; [P <0.0001]).

LixiLan-G was a 26-week randomized double-blind trial designed to assess the efficacy and safety of switching to Soliqua in patients with inadequate glycemic control on a GLP-1 RA. Patients were randomized (1:1) to either switch to Soliqua or to continue GLP-1 RA therapy; both groups continued an existing background regimen of metformin with or without a pioglitazone and/or SGLT2 inhibitor. The baseline A1C level was 7.8% in both groups. At Week 26, patients in the Soliqua group had a significantly greater decrease in the A1C level (-1.0%) compared to those in the GLP-1 RA group (-0.4%; treatment difference: -0.6%; P <0.0001). The proportion of patients achieving an A1C level of <7% was significantly greater in the Soliqua-switch group (62%) than in the continuous GLP-1 RA group (26%; treatment difference 36%; P <0.0001).

The LixiLan-G study also included a 26-week open-label extension for patients in the Soliqua-switch group. At Week 52, the level of glycemic control achieved at Week 26 was maintained: the mean change in A1C level from baseline was -1.0% (compared to -1.0% at Week 26) and the proportion of patients with an A1C level <7% was 64% (compared to 62% at Week 26).

SoliComplex, a real-world, retrospective, observational analysis of the US Optum Clinformatics claims database, compared the treatment persistence (the primary outcome), adherence, efficacy, healthcare resource utilization, and costs of Soliqua and basal-bolus insulin (BBI) in patients with T2D who previously received basal insulin. After 12 months of follow-up, more patients in the Soliqua group persisted with treatment (43.7%) compared to patients in the BBI group (22.3%; P<0.001). A marked difference in treatment persistence was also observed among patients 65 years of age or older (the secondary outcome), with a rate of 43.8% in the Soliqua group and 17.6% in the BBI group. Thus, Soliqua may represent an option with reduced treatment complexity suitable for both younger and older patients interested in a lower burden of management.

Safety and Tolerability

In the GetGoal-Duo 1 trial, where lixisenatide was added to insulin glargine therapy, symptomatic glucose-confirmed hypoglycemia was higher in the lixisenatide-treated group compared with placebo (20.2% vs 11.7%). However, in trials administering Soliqua (LixiLan PoC, LixiLan-O, and LixiLan-L), the fixed-ratio combination of insulin glargine and lixisenatide, documented symptomatic hypoglycemia occurred in a similar proportion of patients between treatment groups. In LixiLan-G, the hypoglycemic event rate was expectedly higher in the Soliqua-switch group than in continued GLP-1 RA group; however, only one severe symptomatic hypoglycemic event was reported in the Soliqua-switch group. The overall safety results recorded by Week 52 in the LixiLan-G extension study were similar to those observed until Week 24, with no new safety signals emerging.

In all trials, Soliqua-treated patients experienced more gastrointestinal events (nausea and vomiting) compared with insulin-glargine treated patients. In the LixiLan-O trial, nausea and vomiting occurred in 9.6% and 3.2% of Soliqua-treated patients, 3.6% and 1.5% of insulin glargine-treated patients, and 24.0% and 6.4% of lixisenatide-treated patients, respectively.

Dosing and Administration

Prior to initiating Soliqua 100/33, therapy with lixisenatide or basal insulin should be discontinued. Administer Soliqua subcutaneously once a day within the hour prior to the first meal of the day. In patients inadequately controlled on <30 units of basal insulin or on lixisenatide, the recommended starting dosage of Soliqua is 15 units (15 units insulin glargine/5 mcg lixisenatide) given subcutaneously once daily. In patients inadequately controlled on 30 to 60 units of basal insulin, the recommended starting dosage of Soliqua is 30 units once daily. The maximum dosage of Soliqua is 60 units.

Amylin Analogue

Pramlintide (Symlin)

Pramlintide is a synthetic analog of human amylin, a naturally occurring neuroendocrine hormone synthesized by pancreatic beta cells that contributes to glucose control during the postprandial period. In healthy individuals, amylin secretion follows the same pattern as insulin, whereby it surges into the bloodstream in response to nutrient uptake (Figure 19-14-A). Amylin modulates the rate of gastric emptying, most likely via the vagus nerve, to regulate the inflow of nutrients into the small intestine and thereby reduce the postprandial rise in glucose. Like insulin, amylin secretion is abnormal in patients with T2D and is deficient in patients with T1D (Figure 19-14-B). The rate of gastric emptying is frequently accelerated in people with diabetes with early increases in PPG directly proportional to the rate of gastric emptying. Studies in patients with T1D utilizing both solid and liquid meals, showed that amylin prolonged the half-gastric emptying time by 60 to 90 minutes and did not influence gastric emptying rates of subsequent meals.

Pramlintide slows the rate at which food is released from the stomach to the small intestine following a meal and, thus, it reduces the initial postprandial increase in plasma glucose. This effect lasts for approximately 3 hours following pramlintide. Pramlintide does not alter the net absorption of ingested carbohydrate or other nutrients.

In patients with diabetes, glucagon concentrations are abnormally elevated during the postprandial period, contributing to hyperglycemia. Pramlintide has been shown to decrease postprandial glucagon concentrations in insulin-using patients with diabetes.

When administered prior to a meal, pramlintide has been shown to induce satiety and reduce total caloric intake. This effect appears to be independent of the nausea that can accompany pramlintide.

Enlarge  Figure 19-14: Amylin: Cosecreted With Insulin and Deficient in Diabetes. Source: Data from: Kruger DF, et al. <em>Diabetes Educ</em>. 1999;25:389-397.
Figure 19-14: Amylin: Cosecreted With Insulin and Deficient in Diabetes. Source: Data from: Kruger DF, et al. Diabetes Educ. 1999;25:389-397.

Clinical Efficacy

Postprandial Glucose

The effect of pramlintide on PPG excursions was assessed in a five-way crossover study in patients with T2D using the rapid-acting insulin analog, insulin lispro. Following four separate standardized meals, each subject received pramlintide at different times relative to the meal (-15, 0, +15 and +30 minutes) and, on one occasion, they received placebo 15 minutes before the meal. Insulin lispro was administered immediately before meals. Administration of pramlintide either at or just prior to a meal caused a greater reduction in PPG than either placebo or postmeal pramlintide. Results from this study confirmed that pramlintide, as an adjunct to mealtime insulin therapy, significantly reduced PPG excursions compared with insulin therapy alone. Pramlintide achieved its effects with an average reduction in mealtime insulin dose of approximately 30%.

Long-Term Glycemic Control With Pramlintide

Several long-term clinical trials have shown the benefit of adding pramlintide to an existing regimen of insulin therapy to improve glycemic control in insulin-using patients with T2D. A 52-week, randomized, placebo-controlled, double-blind, dose-ranging study in 538 insulin-treated patients compared the efficacy and safety of 30-, 75-, or 150-mcg subcutaneous (SC) doses of pramlintide tid with placebo. Compared with placebo, at week 13 there were significant reductions in A1C in patients receiving the 75-mcg and 150-mcg pramlintide doses. (The 30-mcg dose was subtherapeutic.) The mean A1C reduction from baseline to week 52 of 0.6% in the 150-mcg–dose group was also significantly greater than in the placebo group (P = 0.0068). The greater reduction in A1C with pramlintide was achieved without increases in insulin use or severe hypoglycemia. Moreover, the proportion of patients who were able to achieve reductions in both A1C and body weight was 3-fold greater with the 150-mcg dose compared with placebo (48% vs 16%). In addition, there was a significant reduction in body weight in all pramlintide-dose groups compared with placebo. It is important to note that due to pH differences between formulations, the 75- and 150-mcg pramlintide doses used in this study were bioequivalent to the 60- and 120-mcg doses used in other studies.

Another 52-week, randomized, placebo-controlled, double-blind, dose-ranging study in 656 insulin-treated patients with T2D compared the efficacy and safety of 60-, 90-, or 120-mcg doses of SC pramlintide tid to placebo. Treatment with pramlintide 120 mcg resulted in a sustained reduction from baseline in A1C (-0.68% and -0.62% at weeks 26 and 52, respectively), which was significantly greater than seen with placebo (Figure 19-15-A). The proportion of patients who achieved an A1C <8% was approximately 2-fold greater in patients receiving the 120-mcg dose compared with those receiving placebo (48% vs 28%). Once again, the glycemic improvement with pramlintide 120 mcg was accompanied by a reduction in body weight (-1.4 kg) while the placebo group experienced weight gain (+0.7 kg) (Figure 19-15-B).

Clinical Practice Study

In order to assess efficacy and safety of pramlin­tide in a typical clinical practice setting, an open-label study was performed in 166 insulin-using patients with T2D who were not able to achieve glycemic control using insulin alone. In this study, pramlintide was initiated at a dose of 120 mcg at major meals. Patients were instructed to reduce their mealtime insulin dose by 30% to 50% upon initiation of pramlintide, then they subsequently adjusted their insulin regimen according to premeal and postmeal glucose monitoring once pramlintide therapy was established. At 6 months, there was a baseline-subtracted A1C reduction of -0.56% from baseline and a body weight reduction of -2.76 kg from baseline. Pramlintide significantly reduced both fasting and PPG (Figure 19-16). These changes were achieved with dose reductions of total, short-acting, and long-acting insulin (-6.4%, -10.3% and -4.20%, respectively).

Enlarge  Figure 19-15: Change in A1C and Body Weight Over 52 Weeks in 656 Patients With Type 2 Diabetes Treated With Insulin Plus Pramlintide. Change from baseline in mean A1C <em>(A)</em> and body weight (kg) <em>(B)</em>. <sup>a </sup><em>P</em> < 0.05 compared with placebo. Source: Adapted from Hollander PA, et al. <em>Diabetes Care</em>. 2003;26:784-790.
Figure 19-15: Change in A1C and Body Weight Over 52 Weeks in 656 Patients With Type 2 Diabetes Treated With Insulin Plus Pramlintide. Change from baseline in mean A1C (A) and body weight (kg) (B). a P < 0.05 compared with placebo. Source: Adapted from Hollander PA, et al. Diabetes Care. 2003;26:784-790.
Enlarge  Figure 19-16: 7-pt Glucose Profile in Patients With Type 2 Diabetes After 6 months of pramlintide therapy as an adjunct to mealtime insulin, postprandial glucose excursions and glucose fluctuations throughout the day were reduced in patients with type 2 diabetes. <sup>a  </sup><em>P</em> <0.05. Source: Karl D, et al. <em>Diabetes Technol Ther</em>. 2007;9:191-199.
Figure 19-16: 7-pt Glucose Profile in Patients With Type 2 Diabetes After 6 months of pramlintide therapy as an adjunct to mealtime insulin, postprandial glucose excursions and glucose fluctuations throughout the day were reduced in patients with type 2 diabetes. a P <0.05. Source: Karl D, et al. Diabetes Technol Ther. 2007;9:191-199.

Weight-Loss Effects of Pramlintide

Several long-term, placebo-controlled studies have shown that improved glycemic control in pramlintide-treated patients was not accompanied by increased body weight. The greater reduction in A1C with pramlintide compared with placebo has been associated with a sustained and statistically significant reduction in body weight. These findings are further supported by the results of a pooled post hoc analysis of data from 498 overweight/obese (BMI <25 kg/m2) pramlintide- or placebo-treated type 2 diabetic patients who participated in two placebo-controlled long-term trials. At week 26, pramlintide treatment resulted in significant reductions compared with placebo in A1C and body weight (placebo-corrected -0.41% and -1.8 kg, respectively). The greatest reductions in body weight were seen in pramlintide-treated patients with a BMI >40 kg/m2 and in those treated with MET.

Tolerability

Pramlintide treatment has been generally well tolerated in patients with diabetes. No evidence of cardiac, hepatic, or renal toxicity, changes in serum lipids, or clinically relevant changes in laboratory parameters, vital signs, electrocardiograms, or abnormal findings upon physical examinations have been observed.

In patients with T2D, the most common adverse events, other than hypoglycemia, were GI, including nausea (30%) and vomiting (7%). These symptoms, particularly nausea, appeared early after initiation of therapy, were mostly of mild-to-moderate intensity, were dose dependent and resolved over time. Slow titration of pramlintide has been shown to significantly reduce the incidence of nausea.

Hypoglycemia

The use of pramlintide with insulin has been associated with an increased risk of insulin-induced severe hypoglycemia in both T1D and T2D. When severe hypoglycemia associated with pramlintide occurs, it is usually seen within the first 3 hours after pramlintide injection. Pramlintide alone (without concomitant insulin use) does not cause hypoglycemia.

It should be emphasized that the addition of any antihyperglycemic agent to a patient’s current insulin therapy has the potential to increase the risk of insulin-induced hypoglycemia, particularly at the start of therapy. In one of the initial long-term pivotal studies of patients with type 1 and insulin-requiring T2D where full-dose pramlintide was added to existing insulin regimens in double-blind fashion without titration, it was observed that the severe hypoglycemia event rate during the first 4 weeks of therapy was higher in the pramlintide group compared with the placebo group. In another study in patients with T1D, it was shown that this risk was short-term and manageable with adequate glucose monitoring, a 30% to 50% reduction of premeal insulin doses at initiation of pramlintide and gradual upward titration of the pramlintide dose during its initiation. In the previously discussed clinical practice study, in patients with T2D, proactive reduction in premeal insulin doses at initiation of pramlintide was also shown to reduce the incidence of hypoglycemia.

Dosing of Pramlintide

Pramlintide is available in a 60 mcg or 120 mcg pen. Patients with T2D generally use the 120 mcg pen which delivers doses of either 60 or 120 mcg. Pramlintide should always be administered at a distinct injection site >2 inches away from concomitant insulin injections. All patients should reduce premeal insulin by 50% to lessen the risk of insulin-induced hypoglycemia, monitor blood glucose frequently, and contact their health care provider if symptoms of nausea and/or hypoglycemia are severe or unusually persistent and when the dosage of pramlintide or insulin is changed. General guidelines and dosing for patients with T2D:

  • Initiate pramlintide at the 60 mcg dose taken immediately prior to major meals
  • Titrate pramlintide to 120 mcg dose after 3 or more days with no clinically significant nausea
  • Optimize insulin to achieve glycemic targets once the maintenance dose of pramlintide is reached and blood glucose concentrations are stable

Practical Tips for Patients On Pramlintide

Start With a Low Dose and Titrate Slowly

Pramlintide can cause nausea, anorexia and vomiting, especially in T1D where a more gradual titration is required. It is extremely important not to rush the dose titration. Patients with T2D generally experience fewer GI side effects. In patients with T2D, start with a dose of 60 mcg, followed by escalation to the final dose of 120 mcg if the patient is asymptomatic. If the patient experiences nausea or other GI side effects, do not increase the dose of pramlintide until the GI side effects dissipate.

Take Pramlintide Just Prior to the Meal

In a dose-timing study of pramlintide, PPG concentrations were lowered most effectively when pramlintide was administered just before the meal. This reduction of PPG occurred whether the patient was using insulin lispro or regular insulin.

When Initiating Pramlintide, Decrease Dose of Mealtime Insulin by 50%

Pramlintide not only works to reduce glucose appearance via the mechanisms previously described but it may also lead to a reduction in food intake greater than anticipated by a patient newly starting pramlintide. Further adjustments of the insulin dose either up or down should be based on home glucose monitoring results and experience with pramlintide.

Timing of the Insulin Dose May Be Important

Many patients who have experience with pramlintide take a fast-acting insulin analog as they approach the end of a meal. The reason is because they will know how much and what types of food they have eaten, so the insulin-dose calculation using carbohydrate counting or other means will be more accurate. Pramlintide delays gastric emptying and thus the peak in PPG may overlap with the peak action of the fast-acting analogs when given a little later than the beginning of the meal.

References

  • Edelman SV. Diagnosis and Management of Type 2 Diabetes. 14th ed. Professional Communications Inc. 2022
  • Adlyxin (lixisenatide) [package insert]. Bridgewater, NJ: Sanofi-Aventis U.S. LLC; July 2021.
  • Aroda VR, Rosenstock J, Terauchi Y, et al; PIONEER 1 Investigators. PIONEER 1: randomized clinical trial of the efficacy and safety of oral semaglutide monotherapy in comparison with placebo in patients with type 2 diabetes. Diabetes Care. 2019;42(9):1724-1732.
  • Aroda VR, Aberle J, Bardtrum L, et al. Efficacy and safety of once-daily oral semaglutide 25 mg and 50 mg compared with 14 mg in adults with type 2 diabetes (PIONEER PLUS): a multicentre, randomised, phase 3b trial. Lancet. 2023;402(10403):693-704.
  • Blonde L, Klein EJ, Han J, et al. Interim analysis of the effects of exenatide treatment on A1C, weight and cardiovascular risk factors over 82 weeks in 314 overweight patients with type 2 diabetes. Diabetes Obes Metab. 2006;8(4):436-447.
  • Buse JB, Henry RR, Han J, Kim DD, Fineman MS, Baron AD; Exenatide-113 Clinical Study Group. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in sulfonylurea-treated patients with type 2 diabetes. Diabetes Care. 2004;27:2628-2635.
  • Buse JB, Rosenstock J, Sesti G et al. Liraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6). Lancet. 2009;374:39-47.
  • Buse JB, Vilsbøll T, Thurman J, et al; NN9068-3912 (DUAL-II) Trial Investigators. Contribution of liraglutide in the fixed-ratio combination of insulin degludec and liraglutide (IDegLira). Diabetes Care. 2014;37(11):2926-2933.
  • Buse JB, Weyer C, Maggs DG. Amylin replacement with pramlintide in type 1 and type 2 diabetes: a physiological approach to overcome barriers with insulin therapy. Clin Diabetes. 2002;20:137-144.
  • Bydureon (exenatide extended-release) [package insert]. Wilmington, DE: AstraZeneca Pharmaceuticals LP; June 2022.
  • Byetta (exenatide) [package insert]. Wilmington, DE: AstraZeneca Pharmaceuticals LP; June 2022.
  • Capehorn MS, Catarig AM, Furberg JK, et al. Efficacy and safety of once-weekly semaglutide 1.0mg vs once-daily liraglutide 1.2mg as add-on to 1-3 oral antidiabetic drugs in subjects with type 2 diabetes (SUSTAIN 10). Diabetes Metab. 2020;46(2):100-109.
  • Chapman I, Parker B, Doran S, et al. Effect of pramlintide on satiety and food intake in obese subjects and subjects with type 2 diabetes. Diabetologia. 2005;48:838-848.
  • Croom KF, McCormack PL. Liraglutide. A review of its use in type 2 diabetes mellitus. Drugs. 2009;69:1985-2004.
  • DeFronzo RA, Ratner RE, Han J, et al. Effects of exenatide (exendin-4) on glycemic control and weight over 30 weeks in metformin-treated patients with type 2 diabetes. Diabetes Care. 2005;28:1092-1100.
  • Del Prato S, Kahn SE, Pavo I, et al; SURPASS-4 Investigators. Tirzepatide versus insulin glargine in type 2 diabetes and increased cardiovascular risk (SURPASS-4): a randomised, open-label, parallel-group, multicentre, phase 3 trial. Lancet. 2021;398(10313):1811-1824.
  • Dungan KM, Povedano ST, Forst T, et al. Once-weekly dulaglutide versus once-daily liraglutide in metformin-treated patients with type 2 diabetes (AWARD-6): a randomised, open-label, phase 3, non-inferiority trial. Lancet. 2014;384(9951):1349-1357.
  • Edelman SV, Weyer C. Unresolved challenges with insulin therapy in type 1 and type 2 diabetes: potential benefit of replacing amylin, a second beta-cell hormone. Diabetes Technol Ther. 2002;4:175-189.
  • Frías JP. Tirzepatide: a glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) dual agonist in development for the treatment of type 2 diabetes. Expert Rev Endocrinol Metab. 2020;15(6):379-394.
  • Frías JP, Davies MJ, Rosenstock J, et al; SURPASS-2 Investigators. Tirzepatide versus semaglutide once weekly in patients with type 2 diabetes. N Engl J Med. 2021;385(6):503-515.
  • Garber A, Henry R, Ratner R et al. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): randomized, 52-week, phase III, double-blind, parallel-treatment trial. Lancet. 2009; 373:473-481.
  • Gerstein HC, Colhoun HM, Dagenais GR, et al; REWIND Investigators. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet. 2019394(10193):121-130.
  • Gough SC, Bode B, Woo V, et al; NN9068-3697 (DUAL-I) trial investigators. Efficacy and safety of a fixed-ratio combination of insulin degludec and liraglutide (IDegLira) compared with its components given alone: results of a phase 3, open-label, randomised, 26-week, treat-to-target trial in insulin-naive patients with type 2 diabetes. Lancet Diabetes Endocrinol. 2014;2(11):885-893.
  • Gough SC, Bode BW, Woo VC, et al. One-year efficacy and safety of a fixed combination of insulin degludec and liraglutide in patients with type 2 diabetes: results of a 26-week extension to a 26-week main trial. Diabetes Obes Metab. 2015;17(10):965-973.
  • Heine RJ, Van Gaal LF, Johns D, et al. Exenatide versus insulin glargine in patients with suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med. 2005;143:559-569.
  • Henry RR, Ratner RE, Stonehouse AH, et al. Exenatide maintained glycemic control with associated weight reduction over 2 years in patients with type 2 diabetes. Presented at: 66th Scientific Sessions of the American Diabetes Association; June 9-13, 2006; Washington, DC. Poster 485-P.
  • Hollander P, Maggs DG, Ruggles JA, et al. Effect of pramlintide on weight in overweight and obese insulin-treated type 2 diabetes patients. Obes Res. 2004;12:661-668.
  • Hollander PA, Levy P, Fineman MS, et al. Pramlintide as an adjunct to insulin therapy improves long-term glycemic and weight control in patients with type 2 diabetes: a 1-year randomized controlled trial. Diabetes Care. 2003;26:784-790.
  • Husain M, Birkenfeld AL, Donsmark M, et al; PIONEER 6 Investigators. Oral semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2019;381(9):841-851.
  • Karl D, Philis-Tsimikas A, Darsow T, et al. Pramlintide as an adjunct to insulin in patients with type 2 diabetes in a clinical practice setting reduced HbA1c, postprandial glucose excursions, and weight. Diabetes Technol Ther. 2007;9:191-199.
  • Kendall DM, Riddle MC, Rosenstock J, et al. Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea. Diabetes Care. 2005;28:1083-1091.
  • Kellerer M, Kaltoft MS, Lawson J, et al. Effect of once‐weekly semaglutide versus thrice‐daily insulin aspart, both as add‐on to metformin and optimized insulin glargine treatment in participants with type 2 diabetes (SUSTAIN 11): A randomized, open‐label, multinational, phase 3b trial. Diabetes Obes Metab. 2022;24(9):1788-1799.
  • Lingvay I, Catarig AM, Frias JP, et al. Efficacy and safety of once-weekly semaglutide versus daily canagliflozin as add-on to metformin in patients with type 2 diabetes (SUSTAIN 8): a double-blind, phase 3b, randomised controlled trial. Lancet Diabetes Endocrinol. 2019;7(11):834-844.
  • Lingvay I, Manghi FP, García-Hernández P,et al; DUAL V Investigators. Effect of insulin glargine up-titration vs insulin degludec/liraglutide on glycated hemoglobin levels in patients with uncontrolled type 2 diabetes: The DUAL V Randomized Clinical Trial. JAMA. 2016;315(19):2125.
  • Ludvik B, Giorgino F, Jódar E, et al. Once-weekly tirzepatide versus once-daily insulin degludec as add-on to metformin with or without SGLT2 inhibitors in patients with type 2 diabetes (SURPASS-3): a randomised, open-label, parallel-group, phase 3 trial. Lancet. 2021;398(10300):583-598.
  • Maggs DG, Fineman M, Kornstein J, et al. Pramlintide reduces postprandial glucose excursions when added to insulin lispro in subjects with type 2 diabetes: a dose-timing study. Diabetes Metab Res Rev. 2004;20:55-60.
  • Marso SP, Daniels GH, Brown-Frandsen K, et al; LEADER Steering Committee on behalf of the LEADER Trial Investigators. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375(4):311-322.
  • Marre M, Shaw J, Brändle M, et al. Liraglutide, 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 Med. 2009;26(3):268-278.
  • Matthews DR, Marre M, Le-Thi TD, et al. Liraglutide, a human GLP-1 analogue, significantly improves beta cell function in subjects with type 2 diabetes. Diabetologia. 2008;51(suppl 1):S356.
  • Mosenzon O, Blicher TM, Rosenlund S, et al; PIONEER 5 Investigators. Efficacy and safety of oral semaglutide in patients with type 2 diabetes and moderate renal impairment (PIONEER 5): a placebo-controlled, randomised, phase 3a trial. Lancet Diabetes Endocrinol. 2019;7(7):515-527.
  • Mounjaro (tirzepatide) [package insert]. Indianapolis, IN: Lilly USA LLC; May 2022.
  • Nauck M, Weinstock RS, Umpierrez GE, Guerci B, Skrivanek Z, Milicevic Z. Efficacy and safety of dulaglutide versus sitagliptin after 52 weeks in type 2 diabetes in a randomized controlled trial (AWARD-5). Diabetes Care. 2014;37(8):2149-2158.
  • Nauck MA, Frid A, Hermansen K, et al. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin in type 2 diabetes. Diabetes Care. 2009;32:84-90.
  • Ozempic (semaglutide) [package insert]. Bagsvaerd, Denmark: Novo Nordisk A/D; March 2022.
  • Pieber TR, Bode B, Mertens A, et al; PIONEER 7 investigators. Efficacy and safety of oral semaglutide with flexible dose adjustment versus sitagliptin in type 2 diabetes (PIONEER 7): a multicentre, open-label, randomised, phase 3a trial. Lancet Diabetes Endocrinol. 2019;7(7):528-539.
  • Pratley R, Amod A, Hoff ST; PIONEER 4 investigators. Oral semaglutide versus subcutaneous liraglutide and placebo in type 2 diabetes (PIONEER 4): a randomised, double-blind, phase 3a trial. Lancet. 2019;394(10192):39-50.
  • Ratner RE, Maggs D, Nielsen LL, et al. Long-term effects of exenatide therapy over 82 weeks on glycemic control and weight in over-weight metformin-treated patients with type 2 diabetes mellitus. Diabetes Obes Metab. 2006;8:419-428.
  • Ratner RE, Want LL, Fineman MS, et al. Adjunctive therapy with the amylin analogue pramlintide leads to a combined improvement in glycemic and weight control in insulin-treated subjects with type 2 diabetes. Diabetes Technol Ther. 2002;4:51-61.
  • Riddle MC, Forst T, Aronson R, et al. Adding once-daily lixisenatide for type 2 diabetes inadequately controlled with newly initiated and continuously titrated basal insulin glargine: a 24-week, randomized, placebo-controlled study (GetGoal-Duo 1). Diabetes Care. 2013;36(9):2497-2503.
  • Riddle MC, Henry RR, Poon TH, et al. Exenatide elicits sustained glycemic control and progressive reduction of body weight in patients with type 2 diabetes inadequately controlled by sulphonylureas with or without metformin. Diabetes Metab Res Rev. 2006;22:483-491.
  • Rodbard HW, Rosenstock J, Canani LH, et al; PIONEER 2 Investigators. Oral semaglutide versus empagliflozin in patients with type 2 diabetes uncontrolled on metformin: the PIONEER 2 trial. Diabetes Care. 2019;42(12):2272-2281.
  • Rosenstock J, Allison D, Birkenfeld AL, et al; PIONEER 3 Investigators. Effect of additional oral semaglutide vs sitagliptin on glycated hemoglobin in adults with type 2 diabetes uncontrolled with metformin alone or with sulfonylurea: the PIONEER 3 randomized clinical trial. JAMA. 2019;321(15):1466-1480.
  • Rosenstock J, Diamant M, Aroda VR, et al; LixiLan PoC Study Group. Efficacy and safety of LixiLan, a titratable fixed-ratio combination of lixisenatide and insulin glargine, versus insulin glargine in type 2 diabetes inadequately controlled on metformin monotherapy: the LixiLan Proof-of-Concept Randomized Trial. Diabetes Care. 2016;39(9):1579-1586.
  • Rosenstock J, Wysham C, Frías JP, et al. Efficacy and safety of a novel dual GIP and GLP-1 receptor agonist tirzepatide in patients with type 2 diabetes (SURPASS-1): a double-blind, randomised, phase 3 trial. Lancet. 2021;398(10295):143-155.
  • Rosenstock J, Frías JP, Rodbard HW, et al. Tirzepatide vs Insulin Lispro Added to Basal Insulin in Type 2 Diabetes: The SURPASS-6 Randomized Clinical Trial. JAMA. 2023;330(17):1631.
  • Russell-Jones D, Vaag A, Schmitz O, et al. Liraglutide vs insulin glargine and placebo in combination with metformin and sulphonylurea therapy in type 2 diabetes mellitus: a randomized controlled trial (LEAD-5 met þ SU). Diabetologia. 2009;52:4026-4055.
  • Rybelsus (semaglutide) [package insert]. Bagsvaerd, Denmark: Novo Nordisk A/S; June 2022.
  • Schmitz O, Russell-Jones D, Shaw J, et al. Liraglutide, a human GLP-1 analogue, reduces bodyweight in subjects with type 1 diabetes, irrespective of body mass index at baseline. Diabetologia. 2008;51(suppl 1):S354.
  • Seino Y, Rasmussen MF, Zdravkovic M, et al. Dose-dependent improvement in glycemia with once-daily liraglutide without hypoglycemia or weight gain: a double-blind, randomized, controlled trial in Japanese patients with type 2 diabetes. Diabetes Res Clin Pract. 2008:81:161-168.
  • Soliqua 100/33 (insulin glargine and lixisenatide injection) [package insert]. Bridgewater, NJ: Sanofi-Aventis U.S. LLC; July 2021.
  • Trulicity (dulaglutide) [package insert]. Indianapolis, IN: Eli Lily and Company; June 2022.
  • Umpierrez G, Tofé Povedano S, Pérez Manghi F, Shurzinske L, Pechtner V. Efficacy and safety of dulaglutide monotherapy versus metformin in type 2 diabetes in a randomized controlled trial (AWARD-3). Diabetes Care. 2014;37(8):2168-2176.
  • Victoza (liraglutide) [package insert]. Bagsvaerd, Denmark: Novo Nordisk A/S; June 2022.
  • Vilsbøll T, Zdravkovic M, Le-Thi T, et al. Liraglutide, 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 diabetes. Diabetes Care. 2007;30:1608-1610.
  • Weyer C, Gottlieb A, Kim DD, et al. Pramlintide reduces postprandial glucose excursions when added to regular insulin or insulin lispro in subjects with type 1 diabetes: a dose-timing study. Diabetes Care. 2003;26:3074-3079.
  • Whitehouse F, Kruger DF, Fineman M, et al. A randomized study and open-label extension evaluating the long-term efficacy of pramlintide as an adjunct to insulin therapy in type 1 diabetes. Diabetes Care. 2002;25:724-730.
  • Wysham C, Blevins T, Arakaki R, et al. Efficacy and safety of dulaglutide added onto pioglitazone and metformin versus exenatide in type 2 diabetes in a randomized controlled trial (AWARD-1). Diabetes Care. 2014;37(8):2159-2167.
  • Xultophy 100/3.6 (insulin degludec and liraglutide injection) [package insert]. Bagsvaerd, Denmark: Novo Nordisk A/S; June 2022.
  • Yabe D, Nakamura J, Kaneto H, et al; PIONEER 10 Investigators. Safety and efficacy of oral semaglutide versus dulaglutide in Japanese patients with type 2 diabetes (PIONEER 10): an open-label, randomised, active-controlled, phase 3a trial. Lancet Diabetes Endocrinol. 2020;8(5):392-406.
  • Yamada Y, Katagiri H, Hamamoto Y, et al; PIONEER 9 investigators. Dose-response, efficacy, and safety of oral semaglutide monotherapy in Japanese patients with type 2 diabetes (PIONEER 9): a 52-week, phase 2/3a, randomised, controlled trial. Lancet Diabetes Endocrinol. 2020;8(5):377-391.
  • Zander M, Madsbad S, Madsen JL, Holst JJ. Effect 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 study. Lancet. 2002;359:824-830.
  • Zinman B, Aroda VR, Buse JB, et al; PIONEER 8 Investigators. Efficacy, safety, and tolerability of oral semaglutide versus placebo added to insulin with or without metformin in patients with type 2 diabetes: the PIONEER 8 trial. Diabetes Care. 2019;42(12):2262-2271.
  • Zinman B, Bhosekar V, Busch R, et al. Semaglutide once weekly as add-on to SGLT-2 inhibitor therapy in type 2 diabetes (SUSTAIN 9): a randomised, placebo-controlled trial. Lancet Diabetes Endocrinol. 2019;7(5):356-367.
  • Zinman B, Gerich J, Buse J, et al. Efficacy 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 Care. 2009;32:1224-1230.
  • Zinman B, Hoogwerf B, Durant Garcia S, et al. The effect of adding exenatide to a thiazolidinedione in suboptimally controlled type 2 diabetes: a randomized trial. Ann Intern Med. 2007;146:477-485.