Skip to main content
Erschienen in: Advances in Therapy 10/2020

Open Access 04.09.2020 | Review

Oral Semaglutide, A New Option in the Management of Type 2 Diabetes Mellitus: A Narrative Review

verfasst von: Areti Sofogianni, Konstantinos Tziomalos

Erschienen in: Advances in Therapy | Ausgabe 10/2020

Abstract

According to current guidelines, glucagon-like peptide-1 (GLP-1) receptor agonists are the antidiabetic agent of choice in patients with type 2 diabetes mellitus (T2DM) and established cardiovascular disease (CVD) and are also the preferable antidiabetic agent in patients with T2DM without CVD but with indicators of high cardiovascular risk. A limitation in the use of GLP-1 receptor agonists is that they are delivered by subcutaneous injections. In this context, the development of an orally administered formulation of semaglutide offers an additional option in the management of patients with T2DM. In the present review, we discuss the findings of the main trials that evaluated the safety and efficacy of oral semaglutide. Oral semaglutide appears to be more effective in reducing HbA1c levels and body weight than other antidiabetic agents and similarly effective to other GLP-1 receptor agonists. The safety profile of oral semaglutide is also comparable with other members of its class. Even though oral semaglutide did not reduce the incidence of the composite primary endpoint in a randomized controlled trial, a reduction in cardiovascular and all-cause mortality was observed. Therefore, oral semaglutide appears to represent a useful tool in the management of patients with TD2M, particularly those with established CVD or high cardiovascular risk and unwilling to receive injectable GLP-1 receptor agonists.
Hinweise

Digital Features

To view digital features for this article go to https://​doi.​org/​10.​6084/​m9.​figshare.​12800660.
Key Summary Points
Oral semaglutide reduces HbA1c and body weight more than other oral antidiabetic agents
Oral semaglutide induces comparable reductions in HbA1c and body weight with injectable glucagon-like peptide-1 receptor agonists
The safety profile of oral semaglutide is also similar with injectable glucagon-like peptide-1 receptor agonists

Introduction

According to current guidelines, glucagon-like peptide-1 (GLP-1) receptor agonists are the antidiabetic agent of choice in patients with type 2 diabetes mellitus (T2DM) and established cardiovascular disease (CVD) [1]. Moreover, GLP-1 receptor agonists are also the preferable antidiabetic agent in patients with T2DM without CVD but with indicators of high cardiovascular risk, including age ≥ 55 years, coronary, carotid or lower extremity artery stenosis > 50%, left ventricular hypertrophy, estimated glomerular filtration rate (GFR) < 60 ml/min/1.73m2 or albuminuria [1]. Notably, these agents should be considered independently of the baseline HbA1c levels and HbA1c target [1]. Large randomized controlled trials showed that once-daily liraglutide as well as once-weekly semaglutide and dulaglutide reduce cardiovascular morbidity in patients with T2DM and increased cardiovascular risk [24].
A limitation in the use of GLP-1 receptor agonists is that they are delivered by subcutaneous injections. In this context, the development of an orally administered formulation of semaglutide offers an additional option in the management of patients with T2DM. In the present review, we discuss the findings of the main trials that evaluated the safety and efficacy of oral semaglutide as well as the effects of this agent on cardiovascular outcomes and its cost-effectiveness.

Methods

The PubMed database was reviewed for papers published up to June 2020 using the terms “diabetes,” “glucagon-like peptide-1 receptor agonists” and “oral semaglutide.” The references of pertinent articles were also hand-searched for relevant papers. Only studies published in English were considered. This article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors.

Pharmacology of Oral Semaglutide

Oral semaglutide is coformulated with the absorption enhancer sodium N-[8-(2-hydroxybenzoyl) aminocaprylate] (SNAC) [5, 6]. SNAC has been shown to be safe [68]. It has been proposed that SNAC increases pH around the tablet, thus protecting semaglutide from degradation by pepsin and by the acidic gastric pH [5]. Moreover, SNAC appears to promote the formation of semaglutide monomers, which are more easily absorbed [5]. Furthermore, SNAC enhances the absorption of semaglutide by inducing membrane fluidization and surface epithelial sloughing in the gastric mucosa [5]. Coadministration of semaglutide with lisinopril, digoxin and warfarin in healthy subjects did not affect the area under the plasma concentration-time curve and the maximum plasma concentration of the latter medications [9]. On the other hand, the plasma concentration-time curve of metformin increased by 32% when coadminstered with semaglutide, whereas its maximum plasma concentration did not change [9]. However, this increase in exposure to metformin does not appear to be clinically relevant [9]. In another study, coadministration of omeprazole did not affect the plasma concentration-time curve and maximum plasma concentration of semaglutide [10]. Food intake reduces the absorption of semaglutide, and therefore patients must wait 30 min before eating after taking semaglutide [8]. Impairment of hepatic or renal function does not appear to affect the pharmacokinetics of semaglutide [11, 12].
The Peptide InnOvatioN for Early DiabEtes Treatment (PIONEER) program compared the safety and efficacy of semaglutide with placebo as well as with other oral and injectable antidiabetic agents (Table 1).
Table 1
Major studies that evaluated the safety and efficacy of oral semaglutide
Study
Ref
n
Follow-up (weeks)
Baseline treatment
Comparator drug
Dosage of semaglutide
Major findings
PIONEER-1
13
703
26
Diet and exercise alone
 
3, 7 or 14 mg/day
Semaglutide reduced HbA1c and weight by 0.7–1.4% and 0.2–2.6 kg more than placebo, respectively
Treatment discontinuation: 2.3–7.4% and 2.2%, respectively
PIONEER-8
18
731
52
Insulin with or without metformin
 
3, 7 or 14 mg/day
Semaglutide reduced HbA1c and weight by 0.5–1.2% and 0.9–3.3 kg more than placebo, respectively
Nausea: 1.6–3.3 times more frequent in the semaglutide arm
PIONEER-7
14
504
52
1–2 oral antidiabetic agents
Sitagliptin 100 mg/day
3, 7 or 14 mg/day
More patients achieved HbA1c levels < 7% in the semaglutide group (63 vs. 28% in the sitagliptin group) and mean weight loss was 2.2 kg greater in the former
PIONEER-3
16
1864
78
Metformin with or without sulfonylurea
Sitagliptin 100 mg/day
3, 7 or 14 mg/day
Semaglutide 7 and 14 mg/day reduced HbA1c and weight more than sitagliptin (by 0.3 and 0.7%, respectively, and by 1.6 and 2.4 kg, respectively)
Treatment discontinuation rates: 15.0, 19.1 and 13.1%, respectively
PIONEER-2
17
822
52
Metformin
Empagliflozin 25 mg/day
14 mg/day
Semaglutide reduced HbA1c and body weight by 0.4% and 0.9 kg more than empaglifozin, respectively
Gastrointestinal adverse events were more common with semaglutide
PIONEER-4
15
711
52
Metformin with or without a SGLT-2 inhibitor
Liraglutide (dose escalated to 1.8 mg/day)
Up to 14 mg/day
Semaglutide reduced HbA1c by 0.2 and 1.2% more than liraglutide and placebo, respectively, and body weight by 1.5 and 4.0 kg more than liraglutide and placebo, respectively
Rates of adverse events were similar in the semaglutide and liraglutide group and lower in the placebo group
PIONEER-5a
19
324
26
Metformin or sulfonylurea, or both, or basal insulin with or without metformin
 
Up to 14 mg/day
Semaglutide reduced HbA1c and weight by 1.1% and 3.7 kg more than placebo, respectively
Treatment discontinuation due to adverse events: 15 and 5%, respectively
PIONEER-9b
20
243
52
 
Liraglutide 0.75 mg/day
14 mg/day
Semaglutide and liraglutide induced similar reductions in HbA1c (1.7 and 1.4%, respectively)
Constipation, which was the most frequent adverse event, occurred in 13 and 19% of patients, respectively
PIONEER-10b
21
458
52
 
Once-weekly dulaglutide 0.75 mg
14 mg/day
Semaglutide 14 mg/day induced greater reductions in HbA1c (1.7 vs. 1.4%, respectively) and in weight (− 1.6 vs. 1.0 kg, respectively)
Rates of adverse events leading to treatment discontinuation: 6 and 3%, respectively
PIONEER Peptide InnOvatioN for Early DiabEtes Treatment, T2DM type 2 diabetes mellitus, SGLT sodium-glucose co-transporter
aPatients had moderate renal impairment (estimated glomerular filtration rate 30–59 ml/min/1.73 m2
bStudy performed in Japanese patients

Oral Semaglutide Compared with Placebo

In the PIONEER-1 trial, 703 patients with T2DM managed by diet and exercise alone were randomized to receive once-daily oral semaglutide 3, 7 or 14 mg or placebo for 26 weeks [13]. Semaglutide induced dose-dependent reductions in HbA1c (by 0.7, 1.2 and 1.4% more than placebo, respectively) and in body weight (by 0.2, 1.0 and 2.6 kg more than placebo, respectively) [13]. Nausea and diarrhea were the most common adverse events in patients treated with semaglutide and resulted in treatment discontinuation in 2.3, 4.0 and 7.4% of patients, respectively, compared with 2.2% in patients treated with placebo [13].
In the PIONEER-8 study, 731 patients with T2DM uncontrolled on insulin with or without metformin were randomized to receive oral semaglutide 3, 7 or 14 mg/day or placebo for 52 weeks [14]. Semaglutide yielded dose-dependent reductions in HbA1c (0.5, 0.9 and 1.2% more than placebo) and in body weight (0.9, 2.0 and 3.3 kg more than placebo) [14]. However, the incidence of nausea was also dose-dependently higher in patients treated with semaglutide (1.6–3.3 times higher) [14].

Oral Semaglutide Compared with Other Oral Antidiabetic Agents

In the PIONEER-7 study, 504 patients with T2DM uncontrolled on 1–2 oral antidiabetic agents were randomized to receive oral semaglutide with flexible dose adjustments to 3, 7 or 14 mg/day or sitagliptin 100 mg/day for 52 weeks [15]. The proportion of patients who achieved HbA1c levels < 7% was greater in the semaglutide group (63 vs. 28% in the sitagliptin group), and mean weight loss was 2.2 kg greater in the former [15].
In the PIONEER-3 study, 1864 patients with T2DM uncontrolled with metformin with or without sulfonylurea were randomized to receive once-daily oral semaglutide 3, 7 or 14 mg or sitagliptin 100 mg/day for 78 weeks [16]. Semaglutide induced dose-dependent reductions in HbA1c and at the dose of 7 and 14 mg/day was more effective in reducing HbA1c than sitagliptin (difference, 0.3 and 0.7%, respectively) [16]. Semaglutide also induced dose-dependent reductions in body weight that were greater than those of sitagliptin at 78 weeks in all semaglutide doses (difference, 0.8, 1.6 and 2.4 kg with the 3, 7 or 14 mg/day dose, respectively) [16]. Treatment discontinuation rates in the semaglutide 3, 7 or 14 mg and sitagliptin groups were 16.7, 15.0, 19.1 and 13.1%, respectively [16].
In the PIONEER-2 study, 822 patients with T2DM uncontrolled on metformin were randomized to receive oral semaglutide 14 mg/day or empagliflozin 25 mg/day for 52 weeks [17]. Semaglutide reduced HbA1c and body weight more than empaglifozin (mean treatment difference 0.4% and 0.9 kg, respectively) [17]. On the other hand, gastrointestinal adverse events were more common with oral semaglutide [17].

Oral Semaglutide Compared with Injectable Antidiabetic Agents

In the PIONEER-4 study, 711 patients with T2DM uncontrolled despite treatment with metformin (≥ 1500 mg/day or maximum tolerated dose) with or without a sodium-glucose co-transporter-2 inhibitor were randomized to receive once-daily oral semaglutide (dose escalated to 14 mg), once-daily subcutaneous liraglutide (dose escalated to 1.8 mg) or placebo for 52 weeks [18]. Semaglutide reduced HbA1c by 0.2 and 1.2% more than liraglutide and placebo, respectively, and also reduced body weight by 1.5 and 4.0 kg more than liraglutide and placebo, respectively [18]. Rates of adverse events were similar in the semaglutide and liraglutide group and lower in the placebo group [18].

Oral Semaglutide in Special Populations

Studies in special populations yielded similar results. In PIONEER-5, 324 patients with T2DM and moderate renal impairment (estimated GFR 30–59 ml/min/1.73 m2) who were receiving metformin or sulfonylurea, or both, or basal insulin with or without metformin, were randomly assigned to receive oral semaglutide (dose escalated to 14 mg once daily) or placebo for 26 weeks [19]. Oral semaglutide reduced HbA1c by 1.1% and body weight by 3.7 kg more than placebo [19]. However, treatment discontinuation due to adverse events was three times higher with oral semaglutide (15% vs. 5% in the placebo group) [19].
In PIONEER-9, a 52-week, randomized trial in 243 Japanese patients with T2DM, oral semaglutide 14 mg/day induced similar reductions in HbA1c with once-daily liraglutide 0.75 mg (1.7 vs. 1.4 percentage points, respectively) [20]. On the other hand, the incidence of constipation, which was the most frequent adverse event, was lower in the semaglutide group (13 vs. 19% in the liraglutide group) [20].
In PIONEER-10, a 52-week randomized trial in 458 Japanese patients with T2DM, oral semaglutide 14 mg/day induced greater reductions in HbA1c than once-weekly dulaglutide 0.75 mg (1.7 vs. 1.4 percentage points, respectively) and in body weight (− 1.6 vs. 1.0 kg, respectively) [21]. However, the rate of adverse events leading to discontinuation of treatment was two times higher in the semaglutide group (6 vs. 3% in the dulaglutide group) [21].

Meta-Analyses of the Safety and Efficacy of Oral Semaglutide

In a network meta-analysis of 27 randomized controlled studies, oral semaglutide 14 mg/day induced greater reductions in HbA1c than once-weekly dulaglutide 0.75 mg, twice-daily exenatide, once-weekly exenatide 2 mg, liraglutide 1.2 mg and lixisenatide [22]. In addition, oral semaglutide yielded similar reductions in HbA1c with once-weekly semaglutide, once-weekly dulaglutide 1.5 mg and liraglutide 1.8 mg [22]. Moreover, oral semaglutide 14 mg/day induced similar weight loss with once-weekly semaglutide and greater than all other GLP-1 receptor agonists [22]. Oral semaglutide 14 mg/day also induced similar reductions in systolic and diastolic blood pressure with all other GLP-1 receptor agonists except once-weekly semaglutide 1.0 mg, which induced greater reductions in systolic blood pressure (SBP) [22]. The incidence of gastrointestinal adverse events did not differ between the various GLP-1 receptor agonists [22].

Effects of Oral Semaglutide on Cardiovascular Morbidity and Mortality

The effects of oral semaglutide on cardiovascular events was evaluated in the PIONEER-6 trial, a randomized placebo-controlled trial in 3183 patients with T2DM who were ≥ 50 years old and had established CVD or chronic kidney disease or were ≥ 60 years with cardiovascular risk factors only [23]. The trial was designed to rule out an 80% excess cardiovascular risk compared with placebo [noninferiority margin of 1.8 for the upper boundary of the 95% confidence interval (CI) for the hazard ratio (HR) for the primary outcome] [23]. After a median follow-up of 15.9 months, the incidence of the primary outcome (the first occurrence of a major adverse cardiovascular event, i.e., death from cardiovascular causes, nonfatal myocardial infarction or nonfatal stroke) did not differ between the groups assigned to receive semaglutide or placebo (3.8 vs. 4.8%, respectively; HR 0.79, 95% CI 0.57–1.11, p < 0.001 for noninferiority) [23]. Notably, rates of death from cardiovascular causes were lower in patients treated with oral semaglutide (0.9 vs. 1.9% in the placebo group, HR 0.49, 95% CI 0.27–0.92), and rates of death from any cause were also lower in the former (1.4 vs. 2.8% in the placebo group, HR 0.51, 95% CI 0.31–0.84) [23]. Semaglutide reduced HbA1c by 0.7% more than placebo and body weight by 3.4 kg more than placebo [23]. In addition, the reductions in systolic blood pressure (SBP), low-density lipoprotein cholesterol and triglyceride levels were greater in the former [23]. On the other hand, rates of treatment discontinuation were higher in the semaglutide group (11.6 vs. 6.5% in the placebo group), primarily because of nausea, vomiting and diarrhea [23]. In PIONEER-6, when patients with established CVD or established heart failure were analyzed separately, there was no effect of semaglutide on the incidence of major adverse cardiovascular events, i.e., cardiovascular death and nonfatal myocardial infarction or stroke [24]. Similarly, semaglutide had no effect on cardiovascular morbidity in patients without established CVD or heart failure [24]. Interestingly, in a recent network meta-analysis of seven trials in 56,004 patients, oral semaglutide reduced cardiovascular mortality more than exenatide, dulaglutide and lixisenatide and was similarly effective with liraglutide and once-weekly injectable semaglutide [25].

Cost-Effectiveness of Oral Semaglutide

In a recent cost-effectiveness analysis, oral semaglutide 14 mg/day was associated with lower cost for achieving HbA1c levels < 6.5% or < 7.0% than dulaglutide, exenatide (once weekly and twice daily), liraglutide and lixisenatide, whereas once-weekly semaglutide 1.0 mg had comparable cost [26]. In a similar study, oral semaglutide 14 mg/day was associated with lower cost for achieving HbA1c levels < 6.5% or < 7.0%, for achieving ≥ 1.0%-point HbA1c reduction and weight loss ≥ 3.0% and for achieving HbA1c levels < 7.0% without hypoglycemia and without weight gain than empagliflozin, sitagliptin and liraglutide [27]. In another recent cost-effectiveness analysis, oral semaglutide 14 mg/day was more cost-effective than empagliflozin and sitagliptin and was also both more effective and less expensive than liraglutide [28].

Oral Versus Injectable Semaglutide

Even though there are no studies that directly compared oral and injectable semaglutide, a network meta-analysis suggested that these two formulations of semaglutide have comparable safety and induce similar reductions in HbA1c and body weight [22]. However, once-weekly injectable semaglutide induces greater reductions in SBP than oral semaglutide [22]. Moreover, in the Trial to Evaluate Cardiovascular and Other Long-term Outcomes with Semaglutide in Subjects with Type 2 Diabetes (SUSTAIN-6), once-weekly semaglutide reduced the incidence of the primary endpoint (the first occurrence of cardiovascular death, nonfatal myocardial infarction, or nonfatal stroke) by 26% compared with placebo in a population with similar size (n = 3297) and characteristics as the PIONEER-6 trial [3]. However, the follow-up period of SUSTAIN-6 was longer than in PIONEER-6 (24 vs. 15.9 months, respectively), and this might have contributed to the different results of the two studies [3, 23]. In addition, once-weekly semaglutide did not reduce cardiovascular or all-cause mortality in contrast to oral semaglutide, which reduced the incidence of both these outcomes [3, 23]. In a network meta-analysis, oral and once-weekly injectable semaglutide yielded similar reductions in cardiovascular mortality [25]. Cost-effectiveness of these two formulations also appears to be comparable [26]. Given the similar safety, efficacy and cost-effectiveness of oral and injectable semaglutide, the choice between the two formulations should be based on patients’ preferences (oral vs. once weekly administration).

Conclusions

Oral semaglutide appears to be more effective in reducing HbA1c levels and body weight than other antidiabetic agents and similarly effective as other GLP-1 receptor agonists. The safety profile of oral semaglutide is also comparable to other members of its class. Even though oral semaglutide did not reduce the incidence of the composite primary endpoint in the PIONEER-6 trial, a reduction in cardiovascular and all-cause mortality was observed. Therefore, oral semaglutide appears to represent a useful tool in the management of patients with TD2M, particularly those with established CVD or high cardiovascular risk and unwilling to receive injectable GLP-1 receptor agonists. Future studies should further evaluate the effects of oral semaglutide on both cardiovascular events and the microvascular complications of T2DM. In addition, studies in specific subgroups, including those with and without established cardiovascular disease as well as in those with diabetic nephropathy, will add new insights into the role of this agent in the management of T2DM.

Acknowledgements

Funding

No funding or sponsorship was received for this study or publication of this article.

Authorship

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Disclosures

Areti Sofogianni and Konstantinos Tziomalos have nothing to disclose.

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors.

Data Availability

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Buse JB, Wexler DJ, Tsapas A, et al. Update to: management of hyperglycaemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia. 2019;(63):221–8.CrossRef Buse JB, Wexler DJ, Tsapas A, et al. Update to: management of hyperglycaemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia. 2019;(63):221–8.CrossRef
2.
Zurück zum Zitat Marso SP, Daniels GH, Brown-Frandsen K, LEADER Steering Committee, LEADER Trial Investigators, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375:311–22.CrossRef Marso SP, Daniels GH, Brown-Frandsen K, LEADER Steering Committee, LEADER Trial Investigators, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375:311–22.CrossRef
3.
Zurück zum Zitat Marso SP, Bain SC, Consoli A, SUSTAIN-6 Investigators, et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375:1834–44.CrossRef Marso SP, Bain SC, Consoli A, SUSTAIN-6 Investigators, et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375:1834–44.CrossRef
4.
Zurück zum Zitat Gerstein HC, Colhoun HM, Dagenais GR, REWIND Investigators, et al. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomized placebo-controlled trial. Lancet. 2019;394:121–30.CrossRef Gerstein HC, Colhoun HM, Dagenais GR, REWIND Investigators, et al. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomized placebo-controlled trial. Lancet. 2019;394:121–30.CrossRef
5.
Zurück zum Zitat 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:eaar7047.CrossRef 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:eaar7047.CrossRef
6.
Zurück zum Zitat Twarog C, Fattah S, Heade J, Maher S, Fattal E, Brayden DJ. Intestinal permeation enhancers for oral delivery of macromolecules: a comparison between salcaprozate sodium (SNAC) and sodium caprate (C10). Pharmaceutics. 2019;11:78.CrossRef Twarog C, Fattah S, Heade J, Maher S, Fattal E, Brayden DJ. Intestinal permeation enhancers for oral delivery of macromolecules: a comparison between salcaprozate sodium (SNAC) and sodium caprate (C10). Pharmaceutics. 2019;11:78.CrossRef
7.
Zurück zum Zitat Riley MG, Castelli MC, Paehler EA. Subchronic oral toxicity of salcaprozate sodium (SNAC) in Sprague-Dawley and Wistar rats. Int J Toxicol. 2009;28:278–93.CrossRef Riley MG, Castelli MC, Paehler EA. Subchronic oral toxicity of salcaprozate sodium (SNAC) in Sprague-Dawley and Wistar rats. Int J Toxicol. 2009;28:278–93.CrossRef
9.
Zurück zum Zitat Bækdal TA, Borregaard J, Hansen CW, Thomsen M, Anderson TW. Effect of oral semaglutide on the pharmacokinetics of lisinopril, warfarin, digoxin, and metformin in healthy subjects. Clin Pharmacokinet. 2019;58:1193–203.CrossRef Bækdal TA, Borregaard J, Hansen CW, Thomsen M, Anderson TW. Effect of oral semaglutide on the pharmacokinetics of lisinopril, warfarin, digoxin, and metformin in healthy subjects. Clin Pharmacokinet. 2019;58:1193–203.CrossRef
10.
Zurück zum Zitat Bækdal TA, Breitschaft A, Navarria A, Hansen CW. A randomized study investigating the effect of omeprazole on the pharmacokinetics of oral semaglutide. Expert Opin Drug Metab Toxicol. 2018;14:869–77.CrossRef Bækdal TA, Breitschaft A, Navarria A, Hansen CW. A randomized study investigating the effect of omeprazole on the pharmacokinetics of oral semaglutide. Expert Opin Drug Metab Toxicol. 2018;14:869–77.CrossRef
11.
Zurück zum Zitat Baekdal TA, Thomsen M, Kupčová V, Hansen CW, Anderson TW. Pharmacokinetics, safety, and tolerability of oral semaglutide in subjects with hepatic impairment. J Clin Pharmacol. 2018;58:1314–23.CrossRef Baekdal TA, Thomsen M, Kupčová V, Hansen CW, Anderson TW. Pharmacokinetics, safety, and tolerability of oral semaglutide in subjects with hepatic impairment. J Clin Pharmacol. 2018;58:1314–23.CrossRef
12.
Zurück zum Zitat Granhall C, Søndergaard FL, Thomsen M, Anderson TW. Pharmacokinetics, safety and tolerability of oral semaglutide in subjects with renal impairment. Clin Pharmacokinet. 2018;57:1571–80.CrossRef Granhall C, Søndergaard FL, Thomsen M, Anderson TW. Pharmacokinetics, safety and tolerability of oral semaglutide in subjects with renal impairment. Clin Pharmacokinet. 2018;57:1571–80.CrossRef
13.
Zurück zum Zitat Aroda VR, Rosenstock J, Terauchi Y, PIONEER 1 Investigators, 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;42:1724–32.CrossRef Aroda VR, Rosenstock J, Terauchi Y, PIONEER 1 Investigators, 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;42:1724–32.CrossRef
14.
Zurück zum Zitat Pratley R, Amod A, Hoff ST, PIONEER 4 investigators, et al. Oral semaglutide versus subcutaneous liraglutide and placebo in type 2 diabetes (PIONEER 4): a randomised, double-blind, phase 3a trial. Lancet. 2019;394:39–50.CrossRef Pratley R, Amod A, Hoff ST, PIONEER 4 investigators, et al. Oral semaglutide versus subcutaneous liraglutide and placebo in type 2 diabetes (PIONEER 4): a randomised, double-blind, phase 3a trial. Lancet. 2019;394:39–50.CrossRef
15.
Zurück zum Zitat Zinman B, Aroda VR, Buse JB, PIONEER 8 Investigators, 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;42:2262–71.CrossRef Zinman B, Aroda VR, Buse JB, PIONEER 8 Investigators, 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;42:2262–71.CrossRef
16.
Zurück zum Zitat Rosenstock J, Allison D, Birkenfeld AL, PIONEER 3 Investigators, 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;321:1466–80.CrossRef Rosenstock J, Allison D, Birkenfeld AL, PIONEER 3 Investigators, 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;321:1466–80.CrossRef
17.
Zurück zum Zitat Rodbard HW, Rosenstock J, Canani LH, PIONEER 2 Investigators, et al. Oral semaglutide versus empagliflozin in patients with type 2 diabetes uncontrolled on metformin: the PIONEER 2 trial. Diabetes Care. 2019;42:2272–81.CrossRef Rodbard HW, Rosenstock J, Canani LH, PIONEER 2 Investigators, et al. Oral semaglutide versus empagliflozin in patients with type 2 diabetes uncontrolled on metformin: the PIONEER 2 trial. Diabetes Care. 2019;42:2272–81.CrossRef
18.
Zurück zum Zitat Pieber TR, Bode B, Mertens A, PIONEER 7 investigators, 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;7:528–39.CrossRef Pieber TR, Bode B, Mertens A, PIONEER 7 investigators, 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;7:528–39.CrossRef
19.
Zurück zum Zitat Mosenzon O, Blicher TM, Rosenlund S, PIONEER 5 Investigators, 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;7:515–27.CrossRef Mosenzon O, Blicher TM, Rosenlund S, PIONEER 5 Investigators, 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;7:515–27.CrossRef
20.
Zurück zum Zitat Yamada Y, Katagiri H, Hamamoto Y, PIONEER 9 investigators, 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;8:377–91.CrossRef Yamada Y, Katagiri H, Hamamoto Y, PIONEER 9 investigators, 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;8:377–91.CrossRef
21.
Zurück zum Zitat Yabe D, Nakamura J, Kaneto H, PIONEER 10 Investigators, 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.CrossRef Yabe D, Nakamura J, Kaneto H, PIONEER 10 Investigators, 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.CrossRef
22.
Zurück zum Zitat Nuhoho S, Gupta J, Hansen BB, Fletcher-Louis M, Dang-Tan T, Paine A. Orally administered semaglutide versus GLP-1 RAs in patients with type 2 diabetes previously receiving 1–2 oral antidiabetics: systematic review and network meta-analysis. Diabetes Ther. 2019;10:2183–99.CrossRef Nuhoho S, Gupta J, Hansen BB, Fletcher-Louis M, Dang-Tan T, Paine A. Orally administered semaglutide versus GLP-1 RAs in patients with type 2 diabetes previously receiving 1–2 oral antidiabetics: systematic review and network meta-analysis. Diabetes Ther. 2019;10:2183–99.CrossRef
23.
Zurück zum Zitat Husain M, Birkenfeld AL, Donsmark M, PIONEER 6 Investigators, et al. Oral semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2019;381:841–51.CrossRef Husain M, Birkenfeld AL, Donsmark M, PIONEER 6 Investigators, et al. Oral semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2019;381:841–51.CrossRef
24.
Zurück zum Zitat Husain M, Bain SC, Jeppesen OK, et al. Semaglutide (SUSTAIN and PIONEER) reduces cardiovascular events in type 2 diabetes across varying cardiovascular risk. Diabetes Obes Metab. 2020;22:442–51.CrossRef Husain M, Bain SC, Jeppesen OK, et al. Semaglutide (SUSTAIN and PIONEER) reduces cardiovascular events in type 2 diabetes across varying cardiovascular risk. Diabetes Obes Metab. 2020;22:442–51.CrossRef
25.
Zurück zum Zitat Alfayez OM, Almohammed OA, Alkhezi OS, Almutairi AR, Al Yami MS. Indirect comparison of glucagon like peptide-1 receptor agonists regarding cardiovascular safety and mortality in patients with type 2 diabetes mellitus: network meta-analysis. Cardiovasc Diabetol. 2020;19:96.CrossRef Alfayez OM, Almohammed OA, Alkhezi OS, Almutairi AR, Al Yami MS. Indirect comparison of glucagon like peptide-1 receptor agonists regarding cardiovascular safety and mortality in patients with type 2 diabetes mellitus: network meta-analysis. Cardiovasc Diabetol. 2020;19:96.CrossRef
26.
Zurück zum Zitat Hansen BB, Nuhoho S, Ali SN, et al. Oral semaglutide versus injectable glucagon-like peptide-1 receptor agonists: a cost of control analysis. J Med Econ. 2020;23:650–8.CrossRef Hansen BB, Nuhoho S, Ali SN, et al. Oral semaglutide versus injectable glucagon-like peptide-1 receptor agonists: a cost of control analysis. J Med Econ. 2020;23:650–8.CrossRef
27.
Zurück zum Zitat Hunt B, Hansen BB, Ericsson Å, et al. Evaluation of the cost per patient achieving treatment targets with oral semaglutide: a short-term cost-effectiveness analysis in the United States. Adv Ther. 2019;36:3483–93.CrossRef Hunt B, Hansen BB, Ericsson Å, et al. Evaluation of the cost per patient achieving treatment targets with oral semaglutide: a short-term cost-effectiveness analysis in the United States. Adv Ther. 2019;36:3483–93.CrossRef
28.
Zurück zum Zitat Bain SC, Hansen BB, Malkin SJP, et al. Oral semaglutide versus empagliflozin, sitagliptin and liraglutide in the UK: long-term cost-effectiveness analyses based on the PIONEER clinical trial programme. Diabetes Ther. 2020;11:259–77.CrossRef Bain SC, Hansen BB, Malkin SJP, et al. Oral semaglutide versus empagliflozin, sitagliptin and liraglutide in the UK: long-term cost-effectiveness analyses based on the PIONEER clinical trial programme. Diabetes Ther. 2020;11:259–77.CrossRef
Metadaten
Titel
Oral Semaglutide, A New Option in the Management of Type 2 Diabetes Mellitus: A Narrative Review
verfasst von
Areti Sofogianni
Konstantinos Tziomalos
Publikationsdatum
04.09.2020
Verlag
Springer Healthcare
Erschienen in
Advances in Therapy / Ausgabe 10/2020
Print ISSN: 0741-238X
Elektronische ISSN: 1865-8652
DOI
https://doi.org/10.1007/s12325-020-01478-9

Weitere Artikel der Ausgabe 10/2020

Advances in Therapy 10/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.