10,889
Views
36
CrossRef citations to date
0
Altmetric
Supplement: introducing oral semaglutide and the PIONEER program to primary care

GLP-1 receptor agonists in the treatment of type 2 diabetes: role and clinical experience to date

&
Pages 3-14 | Received 01 Jun 2020, Accepted 16 Jul 2020, Published online: 08 Sep 2020
2

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a hormone of the incretin system responsible for a variety of glucoregulatory effects, including glucose-dependent secretion of insulin and inhibition of glucagon release, the effects of which are impaired in people with type 2 diabetes (T2D). Targeting this deficiency using GLP-1 receptor agonists (GLP-1RAs) is a well-established approach in T2D, with over a decade of clinical experience now accrued. This article reviews the evidence for subcutaneous GLP-1RAs and their role in T2D treatment, and explores the rationale for an oral GLP-1RA from a primary care perspective. Clinical trials and real-world studies with subcutaneous GLP-1RAs indicate that these agents have good glycated hemoglobin (HbA1c)-lowering efficacy, an inherently low potential for hypoglycemia, and reduce body weight. Cardiovascular outcomes trials have established cardiovascular safety, and three GLP-1RAs have been proven to reduce the risk of major adverse cardiovascular events (MACE) in patients with established cardiovascular disease or at high cardiovascular risk. The most common adverse events associated with GLP-1RAs are gastrointestinal effects, which tend to occur soon after initiation and decline over time. T2D treatment guidelines recommend GLP-1RAs as a therapeutic option in various settings, including in those patients: i) not achieving HbA1c targets after first-line metformin and lifestyle modifications; ii) at high risk of/with established atherosclerotic cardiovascular disease (regardless of HbA1c; GLP-1RAs of proven benefit); iii) not achieving HbA1c targets on basal insulin if not already receiving a GLP-1RA. Despite the known benefits of GLP-1RAs, adherence and persistence rates are suboptimal, potentially due in part to injection-related concerns. With some patients having a preference for oral medications, the development of an oral GLP-1RA is a logical approach to improving treatment options for patients with T2D. Co-formulation of semaglutide with an absorption enhancer has enabled the development and recent approval of the first oral GLP-1RA, oral semaglutide, which has the potential to expand use of GLP-1RAs in clinical practice.

Article overview and relevance to your clinical practice

  • Glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1RAs) are an efficacious class of therapeutic agents for type 2 diabetes (T2D), with well-characterized safety and tolerability profiles that can be considered in many clinical scenarios, ranging from patients who are not reaching their glycemic goal with metformin, to those with comorbidities or in whom glycemic control is not achieved despite treatment with several oral antihyperglycemic agents.

  • Until recently, these agents were only available in injectable form, but the 2019 US Food and Drug Administration approval of oral semaglutide now provides the option to prescribe a GLP-1RA that is administered as a once-daily tablet.

  • The availability of oral semaglutide may create the opportunity for primary care providers to consider GLP-1RAs earlier in the T2D treatment pathway.

  • This article will provide a brief overview of the mechanism of action and clinical evidence for currently available GLP-1RAs, and discuss their role in treating people with T2D in the USA.

  • We will introduce oral semaglutide and explain how it was possible to develop an oral formulation of a peptide-based drug, before illustrating how the efficacy and safety of this medication has been evaluated in the extensive, global, PIONEER phase 3a clinical trial program.

1. Intensification of type 2 diabetes therapy in primary care

Primary care is central to the management of type 2 diabetes (T2D), and ≥90% of patients in the USA are treated in this setting [Citation1,Citation2]. With several therapeutic classes from which to choose and various comorbidities to address, the management of T2D in primary care is becoming increasingly complex. While diet and exercise remain important in managing the disease, pharmacological intervention is usually required. Treatment plans should be tailored to the individual patient and take various factors into consideration, including treatment goals, presence of comorbidities, and patient preference [Citation3].

Despite advancements in the quality of care and number of therapeutic options available, only around 50% of people with T2D in the US are estimated to achieve glycated hemoglobin (HbA1c) levels below 7.0% [Citation4], the target generally recommended by the American Diabetes Association (ADA) [Citation5]. Among the many potential factors that may contribute to this, a key element is the delay in intensifying therapy in order to reach glycemic goals [Citation6], particularly for those requiring initiation of injectable therapies [Citation7].

Since the 2005 approval of the first glucagon-like peptide-1 (GLP-1) receptor agonist (GLP-1RA), exenatide, this class has amassed a wealth of evidence from clinical trials and real-world studies demonstrating efficacy for achieving both glycemic control and weight loss in patients with T2D [Citation8–10]. Moreover, some of these agents offer the additional benefit of reducing the risk of major adverse cardiovascular events (MACE) in patients with both T2D and established cardiovascular disease (CVD) [Citation11–13] or multiple cardiovascular risk factors [Citation12].

Within this article, we will explore the background of GLP-1RAs and review the clinical experience with these agents to date, moving on to consider where this therapeutic class fits in the treatment paradigm for T2D and how the approval of the first oral GLP-1RA – oral semaglutide – might impact this.

2. Pathophysiology of T2D and the role of GLP-1RAs

T2D is a complex and multifactorial disease, with fundamental defects in at least eight tissues and organ systems known to contribute to the development and progression of hyperglycemia – the so-called ‘ominous octet’ [Citation14,Citation15]. Although impaired insulin secretion by pancreatic β-cells and insulin resistance in the liver and muscle are well-recognized as the core defects involved in the etiology of T2D, the ominous octet also comprises increased glucagon secretion, neurotransmitter dysfunction in the brain, enhanced glucose reabsorption in the kidneys, a decreased incretin effect, and increased lipolysis [Citation14,Citation15]. Targeting these pathophysiological defects is therefore fundamental to delaying the progression of the disease and should be a primary goal of T2D management [Citation14,Citation15].

A variety of glucoregulatory effects, including secretion of insulin, inhibition of glucagon release, and promotion of satiety, are mediated by peptide hormones of the incretin system, including GLP-1 [Citation16,Citation17]. In healthy individuals, GLP-1 is secreted by L-cells in the small intestine in response to the presence of food in the gastrointestinal tract [Citation18,Citation19]. In people with T2D, however, the effects of GLP-1 are significantly impaired, thereby greatly reducing the capacity for insulin release in response to food intake [Citation20].

The use of native GLP-1 as a therapeutic option for people with T2D has been explored, but is limited by its very short half-life and rapid degradation by the enzyme dipeptidyl peptidase-4 (DPP-4); for this reason, synthetic GLP-1RAs with enhanced protein binding and slower degradation have been developed [Citation18,Citation21]. By mimicking the effects of native GLP-1 through stimulation of the GLP-1 receptor, GLP-1RAs directly or indirectly target at least six of the eight core defects implicated in the pathogenesis of T2D (excluding increased lipolysis and increased glucose reabsorption in the kidney), more than any other class of antihyperglycemic medication, and ultimately promote insulin secretion and inhibit glucagon secretion [Citation14,Citation15,Citation22] (). Given that the effects of GLP-1 on insulin secretion are glucose-dependent, GLP-1RAs have an inherently low risk of hypoglycemia [Citation23]. The benefits of GLP-1RAs extend beyond their well-established effects on glycemic control. Indeed, these agents have demonstrated a variety of nonglycemic clinical effects, including weight loss and decreased systolic blood pressure [Citation21,Citation24–26].

Figure 1. Impact of various classes of antihyperglycemic medications on the ‘ominous octet’ of pathophysiological defects contributing to hyperglycemia in patients with T2D [Citation14].American Diabetes Association [Novel Agents for the Treatment of Type 2 Diabetes, American Diabetes Association, 2014]. Copyright and all rights reserved. Material from this publication has been used with the permission of American Diabetes Association.*Reduced satiety and appetite suppression.DPP-4i, dipeptidyl peptidase-4 inhibitor; GI, gastrointestinal; GLP-1RA, glucagon-like peptide-1 receptor agonist; HGP, hepatic glucose production; MET, metformin; SGLT2i, sodium-glucose co-transporter-2 inhibitor; T2D, type 2 diabetes; TZD, thiazolidinedione

Figure 1. Impact of various classes of antihyperglycemic medications on the ‘ominous octet’ of pathophysiological defects contributing to hyperglycemia in patients with T2D [Citation14].American Diabetes Association [Novel Agents for the Treatment of Type 2 Diabetes, American Diabetes Association, 2014]. Copyright and all rights reserved. Material from this publication has been used with the permission of American Diabetes Association.*Reduced satiety and appetite suppression.DPP-4i, dipeptidyl peptidase-4 inhibitor; GI, gastrointestinal; GLP-1RA, glucagon-like peptide-1 receptor agonist; HGP, hepatic glucose production; MET, metformin; SGLT2i, sodium-glucose co-transporter-2 inhibitor; T2D, type 2 diabetes; TZD, thiazolidinedione

Until the approval of oral semaglutide in September 2019, only subcutaneous GLP-1RAs were available for treating people with T2D in the USA, either alone (dulaglutide [Citation12], exenatide [Citation27], exenatide extended-release [Citation28,Citation29], liraglutide [Citation13], lixisenatide [Citation30], and subcutaneous semaglutide [Citation11]) or in combination with insulin (liraglutide + insulin degludec [Citation31] and lixisenatide + insulin glargine [Citation32]); oral semaglutide is the first in this class to be administered in tablet form [Citation33]. While these agents share a common mechanism of action, there are a number of key differences between them, which can include duration of action, dosing frequency and regimen, and administration device (), allowing for individualization of treatment for patients. In terms of the duration of action on the GLP-1 receptor, agents in this class can be categorized as either short-acting (exenatide and lixisenatide) or long-acting (dulaglutide, exenatide extended-release, liraglutide, and semaglutide) [Citation23,Citation25,Citation34]. Short-acting agents primarily reduce postprandial glucose by delaying gastric emptying, whereas long-acting agents have a greater effect than short-acting agents on fasting glucose, and offer the advantages of smaller fluctuations in plasma drug concentrations, improved gastrointestinal tolerability, and more convenient dosing regimens [Citation23,Citation34].

Table 1. Overview of the key characteristics of GLP-1RAs currently available for treating adults with T2D in the USA [Citation11–13,Citation23,Citation25,Citation27–30,Citation33–35]

3. GLP-1RAs in the T2D treatment paradigm

Individualized treatment plans for T2D are based on the requirements of each patient, including glycemic control and weight loss considerations, as well as the impact of comorbidities (e.g., atherosclerotic CVD, and chronic kidney disease [CKD]) and risk of hypoglycemia [Citation3]. GLP-1RAs are recommended by the ADA as a second-line therapy option after metformin and lifestyle management in patients across the treatment continuum, including those at high risk of or with established atherosclerotic CVD, CKD, or heart failure, and in patients without these comorbidities who have a compelling need to reduce the risk of hypoglycemia, minimize weight gain, or promote weight loss () [Citation3].

Figure 2. Current ADA recommendations for pharmacological treatment of adults with T2D [Citation3].American Diabetes Association [9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes–2020, American Diabetes Association, 2020]. Copyright and all rights reserved. Material from this publication has been used with the permission of American Diabetes Association.A1C, glycated hemoglobin; ADA, American Diabetes Association; ASCVD, atherosclerotic cardiovascular disease; CKD, chronic kidney disease; CV, cardiovascular; CVD, cardiovascular disease; CVOT, cardiovascular outcomes trial; DPP-4i, dipeptidyl peptidase-4 inhibitor; eGFR, estimated glomerular filtration rate; GLP-1RA, glucagon-like peptide-1 receptor agonist; HF, heart failure; HFrEF, heart failure reduced ejection fraction; LVEF, left ventricular ejection fraction; LVH, left ventricular hypertrophy; SGLT2i, sodium-glucose co-transporter-2 inhibitor; SU, sulfonylurea; T2D, type 2 diabetes; TZD, thiazolidinedione; UACR, urine albumin-to-creatinine ratio

Figure 2. Current ADA recommendations for pharmacological treatment of adults with T2D [Citation3].American Diabetes Association [9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes–2020, American Diabetes Association, 2020]. Copyright and all rights reserved. Material from this publication has been used with the permission of American Diabetes Association.A1C, glycated hemoglobin; ADA, American Diabetes Association; ASCVD, atherosclerotic cardiovascular disease; CKD, chronic kidney disease; CV, cardiovascular; CVD, cardiovascular disease; CVOT, cardiovascular outcomes trial; DPP-4i, dipeptidyl peptidase-4 inhibitor; eGFR, estimated glomerular filtration rate; GLP-1RA, glucagon-like peptide-1 receptor agonist; HF, heart failure; HFrEF, heart failure reduced ejection fraction; LVEF, left ventricular ejection fraction; LVH, left ventricular hypertrophy; SGLT2i, sodium-glucose co-transporter-2 inhibitor; SU, sulfonylurea; T2D, type 2 diabetes; TZD, thiazolidinedione; UACR, urine albumin-to-creatinine ratio

Irrespective of baseline HbA1c or individual HbA1c targets, a GLP-1RA or sodium-glucose co-transporter-2 (SGLT2) inhibitor with a proven CVD benefit is recommended as a component of the therapeutic regimen for patients in whom atherosclerotic CVD predominates [Citation3]. There are currently three GLP-1RAs indicated for reducing the risk of MACE in patients with T2D and established CVD: dulaglutide, liraglutide, and subcutaneous semaglutide [Citation11–13]; dulaglutide is also indicated for reducing this risk in patients with T2D and multiple cardiovascular risk factors [Citation12]. For those in whom CKD or heart failure predominate, GLP-1RAs are recommended if SGLT2 inhibitors are contraindicated or not tolerated, or if estimated glomerular filtration rate is less than adequate [Citation3] ().

Further along the treatment pathway, GLP-1RAs can also be considered for treatment intensification in patients with HbA1c above target despite dual or triple therapy with other antihyperglycemic agents, and the ADA recommends that GLP-1RAs should be initiated in most patients prior to insulin [Citation3]. For patients above HbA1c target despite therapy with basal insulin, the addition of a GLP-1RA can be considered [Citation3] ().

Key clinical take-home points

  • Individualized treatment plans are recommended for patients with T2D, based on several factors, including comorbidities, hypoglycemia risk, and impact on weight, as well as patient preference.

  • If patients are not at goal with metformin, or if metformin is contraindicated, GLP-1RAs can be used throughout the T2D treatment pathway, including in patients with:

    • – a need to minimize hypoglycemia;

    • – a need to minimize weight gain or promote weight loss;

    • – high risk or established atherosclerotic CVD, CKD, or heart failure (agents with proven CVD or renal benefit should be used).

4. Clinical experience with subcutaneous GLP-1RAs

Based on a wealth of data from both clinical trials and real-world studies gathered through more than 10 years of clinical use, subcutaneous GLP-1RAs are well established in the treatment of T2D [Citation8–10,Citation23,Citation37]. The potential direct and indirect effects of GLP-1RAs on glycemic control, body weight, and the composite MACE outcome are summarized in .

Table 2. Overview of key clinical outcomes with GLP-1RAs [Citation11–13,Citation25,Citation27,Citation28,Citation30]

Pivotal clinical studies have explored the efficacy and safety of GLP-1RAs as monotherapy versus placebo and – in the majority of studies – as add-on to other therapies, with comparators including placebo, insulin, other GLP-1RAs, SGLT-2 inhibitors, DPP-4 inhibitors, sulfonylureas, and thiazolidinediones [Citation10,Citation37,Citation38]. Real-world data from studies with a variety of GLP-1RAs suggest that the results seen in clinical practice, including in the primary care setting, are consistent with those seen in the clinical trial programs [Citation39–45]. However, it should be noted that differences between clinical outcomes in these settings may be obscured due to a limited number of studies and potential problems of confounding and selection bias in observational studies [Citation39]. There is also some evidence that the efficacy of GLP-1RAs is maintained through long-term use, with a recently published study reporting sustained improvements in glycemic control and body weight with the use of exenatide once weekly for up to 7 years [Citation46].

4.1. Effects of GLP-1RAs on glycemic control and body weight

4.1.1. Monotherapy or add-on compared with placebo

As monotherapy or add-on therapy, GLP-1RAs have demonstrated greater reductions in HbA1c compared with placebo, with more patients achieving their glycemic targets with GLP-1RAs [Citation37]. Beyond glycemic control, one of the main potential benefits of GLP-1RAs for patients with T2D is their effect on body weight, with weight loss versus placebo commonly reported with GLP-1RAs in studies of monotherapy or add-on therapy [Citation37].

4.1.2. Add-on therapy compared with other oral antihyperglycemic agents or insulin

A 2016 meta-analysis of 57 studies evaluating the comparative efficacy of GLP-1RAs and other agents (metformin, thiazolidinediones, sulfonylureas, DPP-4 inhibitors, and insulin products) concluded that GLP-1RAs: a) provide reductions in HbA1c at least equivalent to those seen with insulin; b) provide greater reductions in body weight compared with most other oral antihyperglycemic agents and insulin; and c) have lower risk of hypoglycemia than insulin or sulfonylureas [Citation10]. In addition, a more recent meta-analysis has indicated that GLP-1RAs provide greater reductions in HbA1c and body weight compared with DPP-4 inhibitors, without an increase in the risk of hypoglycemia [Citation47]. Data from head-to-head clinical trials comparing GLP-1RAs with SGLT2 inhibitors are scarce; however, subcutaneous semaglutide has demonstrated superior HbA1c and body weight reductions to canagliflozin in patients with T2D uncontrolled on metformin in the SUSTAIN 8 study [Citation38]. These results are consistent with a network meta-analysis of clinical trials with GLP-1RAs and/or SGLT2 inhibitors, which suggested that long-acting GLP-1RAs are superior in reducing HbA1c and body weight to SGLT2 inhibitors [Citation48]. Combining GLP-1RAs with SGLT2 inhibitors has been reported to provide additional reductions in HbA1c and body weight [Citation49,Citation50].

Another meta-analysis concluded slightly greater glycemic control with the addition of GLP-1RAs to oral therapies compared with the addition of insulin, together with weight reductions and lower risk of hypoglycemia [Citation51], although a separate meta-analysis suggested a greater effect on HbA1c compared with insulin was evident with once-weekly but not once-/twice-daily injectable GLP-1RAs [Citation52]. When it is necessary to intensify therapy for patients not achieving their glycemic goal with oral antihyperglycemic agents, the ADA recommends initiating a GLP-1RA prior to insulin in most cases [Citation3] ().

4.1.3. Add-on therapy compared with other GLP-1RAs

Several head-to-head studies of GLP-1RAs have now been conducted, and indicate variations in clinical efficacy between the agents in this class, as reviewed in detail elsewhere [Citation25,Citation37]. For example, once-weekly subcutaneous semaglutide has been shown to provide greater reductions in HbA1c compared with dulaglutide (add-on to metformin) [Citation53], exenatide extended-release (add-on to one to two oral antihyperglycemic agents) [Citation54], and liraglutide (add-on to one to three oral antihyperglycemic agents) [Citation55], while in turn, exenatide extended-release, administered once weekly, provides significant improvements in HbA1c relative to exenatide twice daily (add-on to diet and exercise or at least one oral antihyperglycemic agent) [Citation56]. A network meta-analysis of studies in patients with inadequate glycemic control on one to two oral antihyperglycemic agents reported similar findings, with subcutaneous semaglutide 1.0 mg found to provide greater reductions in HbA1c and body weight compared with other GLP-1RAs [Citation57].

4.1.4. Add-on to insulin

Several studies have compared the addition of a GLP-1RA to insulin versus intensifying the insulin regimen. Meta-analyses indicate similar or improved glycemic control with the addition of a GLP-1RA, with benefits compared with insulin intensification including reduction in body weight and lower incidence of hypoglycemia [Citation58–60].

4.2. Cardiovascular outcomes with GLP-1RAs

The elevated risk of CVD associated with T2D is the leading cause of morbidity and mortality in this patient population, and since 2008 the US Food and Drug Administration (FDA) has required all new antihyperglycemic therapies for T2D to demonstrate an absence of an increased cardiovascular risk [Citation61]. Cardiovascular outcomes trials (CVOTs) have been conducted for all approved subcutaneous GLP-1RAs (except for exenatide twice daily, which was approved prior to the FDA requirement), typically in populations with established CVD or cardiovascular risk factors [Citation27,Citation62–66]. These studies have universally shown that subcutaneous GLP-1RAs do not increase the risk of MACE [Citation67]. Moreover, the CVOTs for dulaglutide, liraglutide, and once-weekly subcutaneous semaglutide demonstrated significant reductions in the incidence of such events with these agents relative to placebo [Citation63,Citation65,Citation66]. As a result, dulaglutide, liraglutide, and subcutaneous semaglutide are indicated by the FDA for reducing cardiovascular risk in patients with T2D and established CVD or multiple cardiovascular risk factors (dulaglutide only) [Citation11–13]. These GLP-1RAs with proven CVD benefit are recommended by the ADA as a preferred treatment option, alongside SGLT2 inhibitors with proven CVD benefit, for patients at high risk for or with established atherosclerotic CVD, CKD, or heart failure [Citation3] ( and ).

4.3. Safety and tolerability of GLP-1RAs

The most common adverse events with GLP-1RAs are gastrointestinal, primarily nausea, vomiting, and diarrhea [Citation3,Citation23]. These adverse events are generally more likely to occur with GLP-1RAs than with other classes of antihyperglycemic medication, including DPP-4 inhibitors, sulfonylureas, thiazolidinediones, and insulins [Citation10]; however, they are typically most common during the first weeks of treatment, are mild-to-moderate in severity [Citation8,Citation23,Citation37,Citation68], and can be mitigated through approaches such as gradual dose escalation and patient counseling regarding the nature of these effects and how to prevent or relieve them [Citation37,Citation68,Citation69]. Such strategies include advising patients to avoid high-fat foods, stop eating when they feel full, and eat a small portion (perhaps half the usual amount) and wait 20–30 minutes to let satiety occur, before considering consuming any more [Citation68,Citation69].

Although a risk of thyroid C-cell tumors is listed in the prescribing information as a boxed warning for most GLP-1RAs based on observations in rodent studies, clinical studies have failed to establish a risk of thyroid C-cell tumors in patients treated with these agents [Citation8,Citation37,Citation70,Citation71]. In addition, while cases of pancreatitis [Citation8] and pancreatic cancer [Citation8,Citation72,Citation73] have been reported with GLP-1RAs, a causal relationship has not been found [Citation8,Citation37] and recent meta-analyses of clinical trials report a lack of increased risk of such events with GLP-1RAs [Citation70,Citation74]. Cases of acute gallbladder disease, such as cholelithiasis and cholecystitis, have been reported with GLP-1RAs [Citation11–13,Citation28,Citation29,Citation33], and if such events are suspected, gallbladder investigations are required and GLP-1RAs should be discontinued. GLP-1RAs have been associated with serious hypersensitivity reactions, including angioedema and anaphylactic reactions, but these events are uncommon (≥1 event per 1,000 patients treated to <1 event per 100 patients treated) or rare (≥1/10,000 to <1/1,000) [Citation23,Citation75–80]. For a more detailed discussion of the clinical relevance of these adverse events, see Brunton et al. [Citation81].

Diabetic retinopathy is a common microvascular complication that occurs as a result of vascular damage in the eye [Citation82]. A recent meta-analysis of trials of all currently available subcutaneous GLP-1RAs concluded no increased risk of diabetic retinopathy (based on Medical Dictionary for Regulatory Activities coding) for any subcutaneous GLP-1RA [Citation83]. An increased incidence of diabetic retinopathy complications was observed in patients treated with subcutaneous semaglutide compared with placebo (3.0% versus 1.8%) in the large-scale CVOT, SUSTAIN-6 [Citation66]. However, 84% and 83% of these events occurred in patients with diabetic retinopathy at baseline in the semaglutide and placebo groups, respectively [Citation66]. This finding appears to be related to these patients having had large and rapid declines in HbA1c in the first 16 weeks of therapy – a phenomenon that has previously been associated with temporary worsening of diabetic retinopathy during treatment with other antihyperglycemic treatments and in other settings, such as pregnancy [Citation84]. There was no unexpected safety signal for diabetic retinopathy in the other SUSTAIN trials [Citation38,Citation53–55,Citation85–90], and a subsequent analysis indicated no increased risk among those without diabetic retinopathy at baseline [Citation84]. Screening for diabetic retinopathy is recommended as part of standard care for all patients with T2D [Citation91] and patients with a history of diabetic retinopathy who are treated with subcutaneous semaglutide should be monitored for progression of this condition [Citation11]. Furthermore, to reduce the risk or slow the progression of diabetic retinopathy, optimization of glycemic control is imperative [Citation91].

GLP-1RAs are associated with increases in heart rate, typically of 2–3 beats per minute [Citation11–13,Citation24,Citation28,Citation29,Citation33], although the clinical relevance of such effects is uncertain [Citation24]. It has been suggested that increases in heart rate may imply a need for caution in patients with cardiac arrhythmias or heart failure [Citation34]. In a relatively small study of 241 patients with chronic heart failure, left ventricular ejection fraction ≤45% and in New York Heart Association functional class I–III, liraglutide increased heart rate and there were more cardiac adverse events with liraglutide than placebo, including a small number of events relating to tachycardia and atrial fibrillation (in 8 patients in the liraglutide group and 3 with placebo) [Citation92]. However, a subsequent analysis did not identify an association between heart rate changes and cardiac adverse events in this study [Citation93]. Furthermore, a recent meta-analysis of seven CVOTs with GLP-1RAs reported a small reduction in the risk of heart failure hospitalization (9–24% of patients in these studies had heart failure at enrollment) [Citation94]. The ADA standards of care recommend the use of an SGLT2 inhibitor with evidence of reducing heart failure progression (e.g., dapagliflozin) in patients with heart failure, with GLP-1RAs an alternative for those in whom SGLT2 inhibitors are not appropriate [Citation3].

Key clinical take-home points

  • GLP-1RAs have demonstrated efficacy for glycemic control and weight loss when studied as a monotherapy and as an adjunctive therapy alongside insulin or oral antihyperglycemic agents.

  • Results from CVOTs have universally shown no increased risk of cardiovascular events with GLP-1RAs, with some agents (dulaglutide, liraglutide, and once-weekly subcutaneous semaglutide) specifically indicated for reducing cardiovascular risk in patients with T2D and established CVD.

  • GLP-1RAs have a well-characterized safety and tolerability profile following over a decade of clinical use in people with T2D. The most common adverse events with these agents are gastrointestinal, namely nausea, vomiting, and diarrhea, but these events are typically transient and mild-to-moderate in severity.

5. Therapeutic adherence and persistence with injectable GLP-1RAs

Although GLP-1RAs have demonstrated effective glycemic control with a low risk of hypoglycemia and a reduction in body weight [Citation3,Citation8], reported adherence and persistence rates with injectable GLP-1RAs are suboptimal [Citation95–97]. This has been partly addressed by the advent of once-weekly injectable GLP-1RAs, which are typically associated with improved adherence and persistence compared with once- or twice-daily agents [Citation96,Citation98–100]. Nevertheless, in a US claims-based analysis, approximately 40% of patients were not fully adherent to the once-weekly GLP-1RA dulaglutide after 6 months [Citation96], and suboptimal adherence therefore remains a concern with these therapies. It is important to note that this issue is not unique to GLP-1RA therapy; poor adherence and persistence are well-established barriers to effective treatment with several chronic disease medications, including other antihyperglycemic agents, antihypertensives, antidepressants, and lipid-lowering drugs [Citation101].

Lack of medication adherence and persistence is a significant concern, given that patients who are more adherent to GLP-1RA treatment achieve greater HbA1c reductions [Citation96,Citation102]. Indeed, adequate diabetes control during the first year after diagnosis has been associated with a lower risk of long-term diabetes complications such as retinopathy and cardiovascular events, and mortality [Citation103–105].

Suboptimal adherence and persistence across the GLP-1RA class may be attributed to a variety of factors of importance to both patients and prescribers, including frequency of injections, administration device, needle size, efficacy, gastrointestinal side effects, and safety [Citation106], as well as the more general consideration of affordability [Citation107]. Although the variation across this class does allow for individualization of treatment for people with T2D, prescribers are faced with the challenge of familiarizing themselves with the dosing regimens and devices for several agents, with limited time and resources to educate patients on the correct administration and titration of these medications [Citation68,Citation108]. As oral medications may be preferred by some patients and potentially considered less burdensome by both patients and healthcare providers, the availability of an oral GLP-1RA may, therefore, expand the utilization of this class and lead to earlier use in the treatment continuum [Citation109].

6. Development of oral semaglutide

As semaglutide is a protein-based drug, oral bioavailability is restricted by degradation in the stomach and limited absorption across the gastrointestinal barrier [Citation110,Citation111]. To overcome this challenge, oral semaglutide was co-formulated with the absorption enhancer sodium N‑(8‑[2‑hydroxybenzoyl] amino) caprylate (SNAC) for administration in tablet form [Citation110,Citation112]. The buffering action of SNAC raises the local pH surrounding the tablet, thereby helping to protect semaglutide from proteolytic degradation, and promoting localized transcellular absorption of semaglutide across the gastric mucosa [Citation110]. Although oral semaglutide has a half-life of around one week, once-daily administration of this medication is required to achieve therapeutic steady state due to low bioavailability [Citation110,Citation112].

Oral semaglutide was approved by the FDA in 2019 as the first oral formulation of a GLP-1RA [Citation35], and is now also approved in Canada, Japan, Europe, Switzerland, and the UK [Citation113–116]. Approval was based on a suite of phase 3a clinical trials referred to as the Peptide InnOvatioN for Early diabEtes tReatment (PIONEER) program. The PIONEER program included eight multinational studies (PIONEER 1–8) () [Citation117–124] and two studies in people from Japan with T2D (PIONEER 9 and 10) [Citation125,Citation126]. Over 9,500 adults with T2D were randomized in these studies to evaluate the effect of oral semaglutide as monotherapy and as add-on to various background therapies, including metformin, sulfonylureas, SGLT2 inhibitors, thiazolidinediones, and insulin. Comparators included placebo, injectable GLP-1RAs, a DPP-4 inhibitor, and an SGLT2 inhibitor. In addition to studies in uncomplicated T2D, the PIONEER program also assessed the effect of oral semaglutide in those with moderate renal impairment (PIONEER 5 [Citation121]) and in those with high cardiovascular risk (PIONEER 6 [Citation122]).

Figure 3. Overview of the global PIONEER clinical trial program for oral semaglutide [Citation117–124].Text in italics indicates allowed background medications.*In PIONEER 8, patients could receive basal, basal bolus, or premixed insulin regimens.CV, cardiovascular; CVOT, cardiovascular outcomes trial; DPP-4i, dipeptidyl peptidase 4 inhibitor; GLP-1RA, glucagon-like peptide-1 receptor agonist; met, metformin; SGLT2i, sodium-glucose co-transporter-2 inhibitor; SU, sulfonylurea; TZD, thiazolidinedione; vs, versus

Figure 3. Overview of the global PIONEER clinical trial program for oral semaglutide [Citation117–124].Text in italics indicates allowed background medications.*In PIONEER 8, patients could receive basal, basal bolus, or premixed insulin regimens.CV, cardiovascular; CVOT, cardiovascular outcomes trial; DPP-4i, dipeptidyl peptidase 4 inhibitor; GLP-1RA, glucagon-like peptide-1 receptor agonist; met, metformin; SGLT2i, sodium-glucose co-transporter-2 inhibitor; SU, sulfonylurea; TZD, thiazolidinedione; vs, versus

Within the subsequent articles in this supplement, the results of the multinational PIONEER trials are reviewed in detail, closing with a discussion of their significance for clinical practice in primary care.

 Key clinical take-home points

  • Specially formulated to facilitate absorption in the stomach, oral semaglutide is the first oral formulation of a GLP-1RA to be approved in T2D.

  • Oral semaglutide was evaluated in an extensive phase 3a clinical trial program, PIONEER, in patients from across the spectrum of T2D and against several key comparators.

  • By overcoming some of the challenges with subcutaneous formulations, the availability of an oral GLP-1RA creates an opportunity to expand utilization and enable earlier use of this treatment class across a wide spectrum of patients with T2D.

Declaration of interest

S.A.B. has received speaker honoraria from AstraZeneca, Bayer, Eli Lilly and Company, Janssen, and Novo Nordisk; consultant honoraria from Abbott Diabetes, AstraZeneca, Bayer, Eli Lilly and Company, Janssen, Novo Nordisk, and Sanofi US.

C.H.W. has acted as an advisor, consultant, and/or speaker for Abbott, AstraZeneca, Boehringer Ingelheim, Eli Lilly and Company, Insulet, Janssen, Novo Nordisk, and Sanofi.

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Acknowledgments

Under the direction of the authors, medical writing and editorial support were provided by Nicola Beadle of Axis, a division of Spirit Medical Communications Group Ltd. (funded by Novo Nordisk Inc.). The authors were involved with drafting and/or critically editing all drafts during the development of the article, and all authors provided their final approval for submission.

Additional information

Funding

This peer reviewed article was supported by Novo Nordisk Inc.; the company was provided with the opportunity to perform a medical accuracy review.

References

  • Davidson JA. The increasing role of primary care physicians in caring for patients with type 2 diabetes mellitus. Mayo Clin Proc. 2010;85(12 Suppl):S3–S4.
  • Shrivastav M, Gibson W Jr, Shrivastav R, et al. Type 2 diabetes management in primary care: the role of retrospective, professional continuous glucose monitoring. Diabetes Spectr. 2018;31(3):279–287.
  • American Diabetes Association. 9. Pharmacologic approaches to glycemic treatment: Standards of Medical Care in Diabetes-2020. Diabetes Care. 2020;43(Suppl 1):S98–S110.
  • Carls G, Huynh J, Tuttle E, et al. Achievement of glycated hemoglobin goals in the US remains unchanged through 2014. Diabetes Ther. 2017;8(4):863–873.
  • American Diabetes Association. 6. Glycemic targets: Standards of Medical Care in Diabetes-2020. Diabetes Care. 2020;43(Suppl 1):S66–S76.
  • Khunti K, Wolden ML, Thorsted BL, et al. Clinical inertia in people with type 2 diabetes: a retrospective cohort study of more than 80,000 people. Diabetes Care. 2013;36(11):3411–3417.
  • Santos Cavaiola T, Kiriakov Y, Reid T. Primary care management of patients with type 2 diabetes: overcoming inertia and advancing therapy with the use of injectables. Clin Ther. 2019;41(2):352–367.
  • Aroda VR. A review of GLP-1 receptor agonists: evolution and advancement, through the lens of randomised controlled trials. Diabetes Obes Metab. 2018;20(Suppl 1):22–33.
  • Chatterjee S, Davies MJ, Khunti K. What have we learnt from “real world” data, observational studies and meta-analyses. Diabetes Obes Metab. 2018;20(Suppl 1):47–58.
  • Levin PA, Nguyen H, Wittbrodt ET, et al. Glucagon-like peptide-1 receptor agonists: a systematic review of comparative effectiveness research. Diabetes Metab Syndr Obes. 2017;10:123–139.
  • Ozempic® (semaglutide) prescribing information [updated Jan 2020; cited 2020 May 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/209637s003lbl.pdf
  • Trulicity® (dulaglutide) prescribing information [updated Feb 2020; cited 2020 May 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125469s033lbl.pdf
  • Victoza® (liraglutide) prescribing information [updated Jun 2019; cited 2020 May 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/022341s031lbl.pdf
  • DeFronzo RA, Triplitt CL, Abdul-Ghani M, et al. Novel agents for the treatment of type 2 diabetes. Diabetes Spectr. 2014;27(2):100–112.
  • DeFronzo RA, Ferrannini E, Groop L, et al. Type 2 diabetes mellitus. Nat Rev Dis Primers. 2015;1:15019.
  • Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007;132(6):2131–2157.
  • Drucker DJ. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab. 2018;27(4):740–756.
  • Drucker DJ, Habener JF, Holst JJ. Discovery, characterization, and clinical development of the glucagon-like peptides. J Clin Invest. 2017;127(12):4217–4227.
  • Holst JJ. The physiology of glucagon-like peptide 1. Physiol Rev. 2007;87(4):1409–1439.
  • Holst JJ, Knop FK, Vilsbøll T, et al. Loss of incretin effect is a specific, important, and early characteristic of type 2 diabetes. Diabetes Care. 2011;34(Suppl 2):S251–S257.
  • Nauck M. Incretin therapies: highlighting common features and differences in the modes of action of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Diabetes Obes Metab. 2016;18(3):203–216.
  • Abdul-Ghani M, DeFronzo RA. Is it time to change the type 2 diabetes treatment paradigm? Yes! GLP-1 RAs should replace metformin in the type 2 diabetes algorithm. Diabetes Care. 2017;40(8):1121–1127.
  • Gentilella R, Pechtner V, Corcos A, et al. Glucagon-like peptide-1 receptor agonists in type 2 diabetes treatment: are they all the same? Diabetes Metab Res Rev. 2019;35(1):e3070.
  • Nauck MA, Meier JJ, Cavender MA, et al. Cardiovascular actions and clinical outcomes with glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Circulation. 2017;136:849–870.
  • Nauck MA, Meier JJ. Are all GLP-1 agonists equal in the treatment of type 2 diabetes? Eur J Endocrinol. 2019;181:R211–R234.
  • Ryan D, Acosta A. GLP-1 receptor agonists: nonglycemic clinical effects in weight loss and beyond. Obesity (Silver Spring). 2015;23(6):1119–1129.
  • Byetta® (exenatide) prescribing information [updated Feb 2020; cited 2020 May 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021773s043lbl.pdf
  • Bydureon® (exenatide extended-release) prescribing information [updated Feb 2020; cited 2020 May 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/022200s030lbl.pdf
  • Bydureon BCise® (exenatide extended-release) prescribing information [updated Feb 2020; cited 2020 May 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/209210s011lbl.pdf
  • Adlyxin® (lixisenatide) prescribing information [updated Jan 2019; cited 2020 May 25]. Available from: http://products.sanofi.us/Adlyxin/Adlyxin.pdf
  • Xultophy® (liraglutide + insulin degludec) prescribing information [updated Nov 2019; cited 2020 May 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/208583s014s015lbl.pdf
  • Soliqua® (lixisenatide + insulin glargine) prescribing information [updated Nov 2019; cited 2020 May 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/208673s008s009lbl.pdf
  • Rybelsus® (semaglutide) prescribing information [updated Jan 2020; cited 2020 May 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/213182s000,213051s001lbl.pdf
  • Meier JJ. GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus. Nat Rev Endocrinol. 2012;8(12):728–742.
  • US Food & Drug Administration. FDA approves first oral GLP-1 treatment for type 2 diabetes, 2019 [cited 2020 May 25]. Available from: https://www.fda.gov/news-events/press-announcements/fda-approves-first-oral-glp-1-treatment-type-2-diabetes
  • Clinicaltrials.gov. A heart disease study of semaglutide in patients with type 2 diabetes (SOUL). ClinicalTrials.gov Identifier: NCT03914326 [cited 2020 May 25]. Available from: https://clinicaltrials.gov/ct2/show/NCT03914326
  • Lyseng-Williamson KA. Glucagon-like peptide-1 receptor analogues in type 2 diabetes: their use and differential features. Clin Drug Investig. 2019;39(8):805–819.
  • Lingvay I, Catarig A-M, 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.
  • Ankarfeldt MZ, Adalsteinsson E, Groenwold RH, et al. A systematic literature review on the efficacy-effectiveness gap: comparison of randomized controlled trials and observational studies of glucose-lowering drugs. Clin Epidemiol. 2017;9:41–51.
  • Feher M, Vega-Hernandez G, Mocevic E, et al. Effectiveness of liraglutide and lixisenatide in the treatment of type 2 diabetes: real-world evidence from The Health Improvement Network (THIN) database in the United Kingdom. Diabetes Ther. 2017;8(2):417–431.
  • Martinez L, Penfornis A, Gautier JF, et al. Effectiveness and persistence of liraglutide treatment among patients with type 2 diabetes treated in primary care and specialist settings: a subgroup analysis from the EVIDENCE study, a prospective, 2-year follow-up, observational, post-marketing study. Adv Ther. 2017;34(3):674–685.
  • Mata-Cases M, Franch-Nadal J, Ortega E, et al. Glucagon-like peptide-1 receptor agonists in patients with type 2 diabetes: real-world evidence from a Mediterranean area. Curr Med Res Opin. 2019;35(10):1735–1744.
  • Moreno Obregón F, Miramontes-González JP, Romo Guajardo-Fajardo C, et al. Real-life experience with dulaglutide: analysis of clinical effectiveness to 24 months. Diabetes Res Clin Pract. 2019;158:107916.
  • Nyeland ME, Ploug UJ, Richards A, et al. Evaluation of the effectiveness of liraglutide and sitagliptin in type 2 diabetes: a retrospective study in UK primary care. Int J Clin Pract. 2015;69(3):281–291.
  • Ostawal A, Mocevic E, Kragh N, et al. Clinical effectiveness of liraglutide in type 2 diabetes treatment in the real-world setting: a systematic literature review. Diabetes Ther. 2016;7(3):411–438.
  • Philis-Tsimikas A, Wysham CH, Hardy E, et al. Efficacy and tolerability of exenatide once weekly over 7 years in patients with type 2 diabetes: an open-label extension of the DURATION-1 study. J Diabetes Complications. 2019;33(3):223–230.
  • Tran S, Retnakaran R, Zinman B, et al. Efficacy of glucagon-like peptide-1 receptor agonists compared to dipeptidyl peptidase-4 inhibitors for the management of type 2 diabetes: a meta-analysis of randomized clinical trials. Diabetes Obes Metab. 2018;20(Suppl 1):68–76.
  • Hussein H, Zaccardi F, Khunti K, et al. Efficacy and tolerability of sodium-glucose co-transporter-2 inhibitors and glucagon-like peptide-1 receptor agonists: a systematic review and network meta-analysis. Diabetes Obes Metab. 2020;22:1035–1046.
  • Castellana M, Cignarelli A, Brescia F, et al. Efficacy and safety of GLP-1 receptor agonists as add-on to SGLT2 inhibitors in type 2 diabetes mellitus: a meta-analysis. Sci Rep. 2019;9(1):19351.
  • Guo M, Gu J, Teng F, et al. The efficacy and safety of combinations of SGLT2 inhibitors and GLP-1 receptor agonists in the treatment of type 2 diabetes or obese adults: a systematic review and meta-analysis. Endocrine. 2020;67(2):294–304.
  • Abd El Aziz MS, Kahle M, Meier JJ, et al. A meta-analysis comparing clinical effects of short- or long-acting GLP-1 receptor agonists versus insulin treatment from head-to-head studies in type 2 diabetic patients. Diabetes Obes Metab. 2017;19(2):216–227.
  • Singh S, Wright EE Jr, Kwan AY, et al. Glucagon-like peptide-1 receptor agonists compared with basal insulins for the treatment of type 2 diabetes mellitus: a systematic review and meta-analysis. Diabetes Obes Metab. 2017;19(2):228–238.
  • Pratley RE, Aroda VR, Lingvay I, et al. Semaglutide versus dulaglutide once weekly in patients with type 2 diabetes (SUSTAIN 7): a randomised, open-label, phase 3b trial. Lancet Diabetes Endocrinol. 2018;6(4):275–286.
  • Ahmann AJ, Capehorn M, Charpentier G, et al. Efficacy and safety of once-weekly semaglutide versus exenatide ER in subjects with type 2 diabetes (SUSTAIN 3): a 56-week, open-label, randomized clinical trial. Diabetes Care. 2018;41(2):258–266.
  • Capehorn MS, Catarig AM, Furberg JK, et al. Efficacy and safety of once-weekly semaglutide 1.0 mg vs once-daily liraglutide 1.2 mg as add-on to 1–3 oral antidiabetic drugs in subjects with type 2 diabetes (SUSTAIN 10). Diabetes Metab. 2020;46(2):100–109.
  • Blevins T, Pullman J, Malloy J, et al. DURATION-5: exenatide once weekly resulted in greater improvements in glycemic control compared with exenatide twice daily in patients with type 2 diabetes. J Clin Endocrinol Metab. 2011;96(5):1301–1310.
  • Witkowski M, Wilkinson L, Webb N, et al. A systematic literature review and network meta-analysis comparing once-weekly semaglutide with other GLP-1 receptor agonists in patients with type 2 diabetes previously receiving 1-2 oral anti-diabetic drugs. Diabetes Ther. 2018;9(3):1149–1167.
  • Maiorino MI, Chiodini P, Bellastella G, et al. The good companions: insulin and glucagon-like peptide-1 receptor agonist in type 2 diabetes. A systematic review and meta-analysis of randomized controlled trials. Diabetes Res Clin Pract. 2019;154:101–115.
  • Castellana M, Cignarelli A, Brescia F, et al. GLP-1 receptor agonist added to insulin versus basal-plus or basal-bolus insulin therapy in type 2 diabetes: a systematic review and meta-analysis. Diabetes Metab Res Rev. 2019;35(1):e3082.
  • Wysham CH, Lin J, Kuritzky L. Safety and efficacy of a glucagon-like peptide-1 receptor agonist added to basal insulin therapy versus basal insulin with or without a rapid-acting insulin in patients with type 2 diabetes: results of a meta-analysis. Postgrad Med. 2017;129(4):436–445.
  • US Food & Drug Administration. Guidance document: diabetes mellitus – evaluating cardiovascular risk in new antidiabetic therapies to treat type 2 diabetes, 2008 [cited 2020 May 25]. Available from: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/diabetes-mellitus-evaluating-cardiovascular-risk-new-antidiabetic-therapies-treat-type-2-diabetes
  • Pfeffer MA, Claggett B, Diaz R, et al. Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med. 2015;373(23):2247–2257.
  • Gerstein HC, Colhoun HM, Dagenais GR, et al. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet. 2019;394(10193):121–130.
  • Holman RR, Bethel MA, Mentz RJ, et al. Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2017;377(13):1228–1239.
  • Marso SP, Daniels GH, Brown-Frandsen K, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375(4):311–322.
  • Marso SP, Bain SC, Consoli A, et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375(19):1834–1844.
  • Hinnen D, Kruger DF. Cardiovascular risks in type 2 diabetes and the interpretation of cardiovascular outcome trials. Diabetes Metab Syndr Obes. 2019;12:447–455.
  • Reid TS. Practical use of glucagon-like peptide-1 receptor agonist therapy in primary care. Clin Diabetes. 2013;31(4):148–157.
  • Milne N. How to use GLP-1 receptor agonist therapy safely and effectively. Diabet Prim Care. 2019;21:45–46.
  • Abd El Aziz M, Cahyadi O, Meier JJ, et al. Incretin-based glucose-lowering medications and the risk of acute pancreatitis and malignancies: a meta-analysis based on cardiovascular outcomes trials. Diabetes Obes Metab. 2020;22:699–704.
  • Prasad-Reddy L, Isaacs D. A clinical review of GLP-1 receptor agonists: efficacy and safety in diabetes and beyond. Drugs Context. 2015;4:212283.
  • Azoulay L, Filion KB, Platt RW, et al. Incretin based drugs and the risk of pancreatic cancer: international multicentre cohort study. BMJ. 2016;352:i581.
  • Boniol M, Franchi M, Bota M, et al. Incretin-based therapies and the short-term risk of pancreatic cancer: results from two retrospective cohort studies. Diabetes Care. 2018;41(2):286–292.
  • Pinto LC, Falcetta MR, Rados DV, et al. Glucagon-like peptide-1 receptor agonists and pancreatic cancer: a meta-analysis with trial sequential analysis. Sci Rep. 2019;9(1):2375.
  • Byetta® (exenatide) Summary of Product Characteristics [updated Mar 2020; cited 2020 May 25]. Available from: https://www.ema.europa.eu/en/documents/product-information/byetta-epar-product-information_en.pdf
  • Bydureon® (exenatide extended-release) Summary of Product Characteristics [updated Feb 2020; cited 2020 May 25]. Available from: https://www.ema.europa.eu/en/documents/product-information/bydureon-epar-product-information_en.pdf
  • Lyxumia® (lixisenatide) Summary of Product Characteristics [updated Mar 2020; cited 2020 May 25]. Available from: https://www.ema.europa.eu/en/documents/product-information/lyxumia-epar-product-information_en.pdf
  • Ozempic® (semaglutide) Summary of Product Characteristics [updated Apr 2020; cited 2020 May 25]. Available from: https://www.ema.europa.eu/en/documents/product-information/ozempic-epar-product-information_en.pdf
  • Trulicity® (dulaglutide) Summary of Product Characteristics [updated Nov 2019; cited 2020 May 25]. Available from: https://www.ema.europa.eu/en/documents/product-information/trulicity-epar-product-information_en.pdf
  • Victoza® (liraglutide) Summary of Product Characteristics [updated Nov 2019; cited 2020 May 25]. Available from: https://www.ema.europa.eu/en/documents/product-information/victoza-epar-product-information_en.pdf
  • Brunton SA, Mosenzon O, Wright EE. Integrating oral semaglutide into clinical practice in primary care: for whom, when, and how? Postgrad Med. 2020;132(S2):48–60.
  • Bain SC, Klufas MA, Ho A, et al. Worsening of diabetic retinopathy with rapid improvement in systemic glucose control: a review. Diabetes Obes Metab. 2019;21(3):454–466.
  • Avgerinos I, Karagiannis T, Malandris K, et al. Glucagon-like peptide-1 receptor agonists and microvascular outcomes in type 2 diabetes: a systematic review and meta-analysis. Diabetes Obes Metab. 2019;21(1):188–193.
  • Vilsbøll T, Bain SC, Leiter LA, et al. Semaglutide, reduction in glycated haemoglobin and the risk of diabetic retinopathy. Diabetes Obes Metab. 2018;20(4):889–897.
  • Aroda VR, Ahmann A, Cariou B, et al. Comparative efficacy, safety, and cardiovascular outcomes with once-weekly subcutaneous semaglutide in the treatment of type 2 diabetes: insights from the SUSTAIN 1-7 trials. Diabetes Metab. 2019;45(5):409–418.
  • Ahrén B, Masmiquel L, Kumar H, et al. Efficacy and safety of once-weekly semaglutide versus once-daily sitagliptin as an add-on to metformin, thiazolidinediones, or both, in patients with type 2 diabetes (SUSTAIN 2): a 56-week, double-blind, phase 3a, randomised trial. Lancet Diabetes Endocrinol. 2017;5(5):341–354.
  • Aroda VR, Bain SC, Cariou B, et al. Efficacy and safety of once-weekly semaglutide versus once-daily insulin glargine as add-on to metformin (with or without sulfonylureas) in insulin-naive patients with type 2 diabetes (SUSTAIN 4): a randomised, open-label, parallel-group, multicentre, multinational, phase 3a trial. Lancet Diabetes Endocrinol. 2017;5(5):355–366.
  • Rodbard HW, Lingvay I, Reed J, et al. Semaglutide added to basal insulin in type 2 diabetes (SUSTAIN 5): a randomized, controlled trial. J Clin Endocrinol Metab. 2018;103(6):2291–2301.
  • Sorli C, Harashima SI, Tsoukas GM, et al. Efficacy and safety of once-weekly semaglutide monotherapy versus placebo in patients with type 2 diabetes (SUSTAIN 1): a double-blind, randomised, placebo-controlled, parallel-group, multinational, multicentre phase 3a trial. Lancet Diabetes Endocrinol. 2017;5(4):251–260.
  • 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.
  • American Diabetes Association. 11. Microvascular complications and foot care: Standards of Medical Care in Diabetes-2020. Diabetes Care. 2020;43(Suppl 1):S135–S151.
  • Jorsal A, Kistorp C, Holmager P, et al. Effect of liraglutide, a glucagon-like peptide-1 analogue, on left ventricular function in stable chronic heart failure patients with and without diabetes (LIVE)-a multicentre, double-blind, randomised, placebo-controlled trial. Eur J Heart Fail. 2017;19(1):69–77.
  • Tougaard RS, Jorsal A, Tarnow L, et al. Heart rate increases in liraglutide treated chronic heart failure patients: association with clinical parameters and adverse events [published online ahead of print, 2020 Apr 15]. Scand Cardiovasc J. 2020;1–6.
  • Kristensen SL, Rørth R, Jhund PS, et al. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet Diabetes Endocrinol. 2019;7(10):776–785.
  • Buysman EK, Liu F, Hammer M, et al. Impact of medication adherence and persistence on clinical and economic outcomes in patients with type 2 diabetes treated with liraglutide: a retrospective cohort study. Adv Ther. 2015;32(4):341–355.
  • Mody R, Grabner M, Yu M, et al. Real-world effectiveness, adherence and persistence among patients with type 2 diabetes mellitus initiating dulaglutide treatment. Curr Med Res Opin. 2018;34(6):995–1003.
  • Nguyen H, Dufour R, Caldwell-Tarr A. Glucagon-like peptide-1 receptor agonist (GLP-1RA) therapy adherence for patients with type 2 diabetes in a Medicare population. Adv Ther. 2017;34(3):658–673.
  • Giorgino F, Penfornis A, Pechtner V, et al. Adherence to antihyperglycemic medications and glucagon-like peptide 1-receptor agonists in type 2 diabetes: clinical consequences and strategies for improvement. Patient Prefer Adherence. 2018;12:707–719.
  • Alatorre C, Fernández Landó L, Yu M, et al. Treatment patterns in patients with type 2 diabetes mellitus treated with glucagon-like peptide-1 receptor agonists: higher adherence and persistence with dulaglutide compared with once-weekly exenatide and liraglutide. Diabetes Obes Metab. 2017;19(7):953–961.
  • Johnston SS, Nguyen H, Felber E, et al. Retrospective study of adherence to glucagon-like peptide-1 receptor agonist therapy in patients with type 2 diabetes mellitus in the United States. Adv Ther. 2014;31(11):1119–1133.
  • Lemstra M, Nwankwo C, Bird Y, et al. Primary nonadherence to chronic disease medications: a meta-analysis. Patient Prefer Adherence. 2018;12:721–731.
  • Hamersky CM, Fridman M, Gamble CL, et al. Injectable antihyperglycemics: a systematic review and critical analysis of the literature on adherence, persistence, and health outcomes. Diabetes Ther. 2019;10(3):865–890.
  • Holman RR, Paul SK, Bethel MA, et al. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med. 2008;359(15):1577–1589.
  • Laiteerapong N, Ham SA, Gao Y, et al. The legacy effect in type 2 diabetes: impact of early glycemic control on future complications (The Diabetes & Aging Study). Diabetes Care. 2019;42(3):416–426.
  • Paul SK, Klein K, Thorsted BL, et al. Delay in treatment intensification increases the risks of cardiovascular events in patients with type 2 diabetes. Cardiovasc Diabetol. 2015;14:100.
  • Thieu VT, Robinson S, Kennedy-Martin T, et al. Patient preferences for glucagon-like peptide 1 receptor-agonist treatment attributes. Patient Prefer Adherence. 2019;13:561–576.
  • Polonsky WH, Henry RR. Poor medication adherence in type 2 diabetes: recognizing the scope of the problem and its key contributors. Patient Prefer Adherence. 2016;10:1299–1307.
  • Okemah J, Peng J, Quiñones M. Addressing clinical inertia in type 2 diabetes mellitus: a review. Adv Ther. 2018;35:1735–1745.
  • Bui V, Neumiller JJ. Oral semaglutide. Clin Diabetes. 2018;36(4):327–329.
  • Buckley ST, Bækdal TA, Vegge A, et al. Transcellular stomach absorption of a derivatized glucagon-like peptide-1 receptor agonist. Sci Transl Med. 2018;10(467):eaar7047.
  • Renukuntla J, Vadlapudi AD, Patel A, et al. Approaches for enhancing oral bioavailability of peptides and proteins. Int J Pharm. 2013;447:75–93.
  • Granhall C, Donsmark M, Blicher TM, et al. Safety and pharmacokinetics of single and multiple ascending doses of the novel oral human GLP-1 analogue, oral semaglutide, in healthy subjects and subjects with type 2 diabetes. Clin Pharmacokinet. 2019;58(6):781–791.
  • Novo Nordisk. Rybelsus® approved in Japan for the treatment of type 2 diabetes, 2020 [cited 2020 Jul 7]. Available from: https://www.novonordisk.com/media/news-details.2325697.html
  • Rybelsus® product monograph including patient medication information; date of initial approval: March 30, 2020 [cited 2020 Jun 24]. Available from: https://www.novonordisk.ca/content/dam/Canada/AFFILIATE/www-novonordisk-ca/OurProducts/PDF/Rybelsus-PM-EN-monograph.pdf
  • Rybelsus® Summary of Product Characteristics [updated Jun 2020; cited 2020 Jun 24]. Available from: https://www.ema.europa.eu/en/documents/product-information/rybelsus-epar-product-information_en.pdf
  • Rybelsus® Swiss public assessment report; decision date: 24 March 2020 [cited 2020 Jun 24]. Available from: https://www.swissmedic.ch/dam/swissmedic/de/dokumente/zulassung/swisspar/67446-rybelsus-01-swisspar-20200430.pdf.download.pdf/SwissPAR_Rybelsus_final.pdf
  • Aroda VR, Rosenstock J, Terauchi Y, et al. 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.
  • Rodbard HW, Rosenstock J, Canani LH, et al. 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. 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.
  • Pratley R, Amod A, Hoff ST, et al. 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.
  • Mosenzon O, Blicher TM, Rosenlund S, et al. 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.
  • Husain M, Birkenfeld AL, Donsmark M, et al. Oral semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2019;381(9):841–851.
  • Pieber TR, Bode B, Mertens A, et al. 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.
  • Zinman B, Aroda VR, Buse JB, et al. 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.
  • Yabe D, Nakamura J, Kaneto H, et al. 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;8:392–406.
  • Yamada Y, Katagiri H, Hamamoto Y, et al. 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;8:377–391.