Back to Journals » Drug Design, Development and Therapy » Volume 19

A Comprehensive Review on the Pharmacokinetics and Drug−Drug Interactions of Approved GLP-1 Receptor Agonists and a Dual GLP-1/GIP Receptor Agonist

Authors Min JS, Jo SJ, Lee S, Kim DY, Kim DH, Lee CB , Bae SK 

Received 17 November 2024

Accepted for publication 14 April 2025

Published 30 April 2025 Volume 2025:19 Pages 3509—3537

DOI https://doi.org/10.2147/DDDT.S506957

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Prof. Dr. Georgios Panos



Jee Sun Min,1 Seong Jun Jo,1,2 Sangyoung Lee,1 Duk Yeon Kim,1 Da Hyun Kim,1 Chae Bin Lee,3 Soo Kyung Bae1

1College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon, 14662, Republic of Korea; 2Department of Pharmaceutical Sciences, State University of New York, Buffalo, NY, 14214, USA; 3Johns Hopkins Drug Discovery, Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA

Correspondence: Soo Kyung Bae, College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon, 14662, Republic of Korea, Email [email protected]

Abstract: Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are peptide-derived analogs that were initially investigated to treat type 2 diabetes. Recently, a drug targeting the receptors of both GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) (tirzepatide) has been introduced to the market, and its indications have expanded to include treating obesity. Here, we review the pharmacokinetics, pharmacokinetic drug−drug interactions (DDIs), and pharmacokinetic modeling approaches of four currently available GLP-1 RAs (exenatide, liraglutide, dulaglutide, and semaglutide) and tirzepatide. To address the extremely short half-life (2 min) of native human GLP-1, structural modifications have been applied to GLP-1 RAs and a dual GLP-1/GIP RA. These include amino acid sequence substitutions, fatty acid conjugation using a linker, and fusion with albumin or the IgG fragment crystallizable (Fc) region, resulting in minimal metabolism and renal excretion. Due to their diverse structures, the pharmacokinetic profiles vary, and a prolonged half-life may be associated with an increased risk of adverse events. Clinically significant drug-metabolizing enzyme- and transporter-mediated DDIs are yet to be reported. Mechanism-of-action-mediated DDIs are currently limited to those involving delayed gastric emptying, and most studies have found them to be clinically insignificant. However, significant changes in exposure were observed for oral contraceptives and levothyroxine following the administration of tirzepatide and oral semaglutide, respectively, indicating the need for close monitoring in these instances. Thirty models have been developed to predict pharmacokinetics and physiologically based pharmacokinetic modeling can be useful for assessing mechanism-of-action-mediated DDIs. Alterations in the volume of distribution and clearance resulting from other mechanisms of action (eg, reduced fat mass, changes in cytochrome P450 activity, and glomerular filtration rate) are key factors in determining pharmacokinetics. However, the DDIs mediated by these factors remain poorly understood and require further investigation to ensure that GLP-1 RAs can be safely used with concomitant medications.

Keywords: GLP-1, GIP, pharmacokinetics, drug−drug interactions, physiologically based pharmacokinetic model

Introduction

Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are peptide hormone-derived analogs classified as incretin mimetic drugs (Figure 1).1–5 Incretins are gut hormones released into the bloodstream following food consumption that enhances insulin secretion in pancreatic β-cells and help decrease high blood glucose levels.6 Therefore, GLP-1 RAs were first investigated as treatments for type 2 diabetes.7 Compared to other antidiabetic drugs, GLP-1 RAs have the advantage of only being pharmacologically active when blood glucose levels are high.8 This significantly reduces the incidence of hypoglycemia—a frequently reported adverse drug reaction associated with other antidiabetic drugs like sulfonylureas or insulin.9,10 Additionally, GLP-1 RAs suppress glucagon secretion from the α-cells in the pancreas, leading to decreased blood glucose levels.11

Figure 1 Molecular weights and amino acid sequences of human GLP-1 (A), exendin-4 (B), exenatide (C), liraglutide (D), dulaglutide (E), semaglutide (F), human GIP (G), and tirzepatide (H), with substituted amino acids marked in different colors. Created in BioRender. Min, J. (2025) https://BioRender.com/o27r742.

Abbreviations: hGLP-1, human glucagon-like peptide-1; DPP-4, dipeptidyl peptidase-4; IgG4-Fc, immunoglobulin G4 fragment crystallizable; Aib, α-aminoisobutyric acid; OEG, oligoethylene glycol; hGIP, human glucose-dependent insulinotropic polypeptide.

However, a drawback of the native human GLP-1 (Figure 1A) is its extremely short in vivo half-life (2 min) due to fast clearance by dipeptidyl peptidase-4 (DPP-4).1 Thus, numerous studies have used biotechnology to extend the half-life of GLP-1 RAs, with strategies including amino acid sequence substitution, fatty acid conjugation with a linker, fusion of albumin or the IgG fragment crystallizable (Fc) region (Figure 1).5,12,13 These efforts have yielded six GLP-1 RAs−starting with the development of exendin-4 (Figure 1B)-based exenatide (Figure 1C), followed by liraglutide (Figure 1D), albiglutide, dulaglutide (Figure 1E), lixisenatide, and semaglutide (Figure 1F)−that have been approved by the United States Food and Drug Administration (US FDA) to treat type 2 diabetes.14 Among these, albiglutide was pulled from the market in 2017 because of declining sales;15 likewise, lixisenatide was withdrawn from the US market as of January 1, 2023, for commercial reasons, rather than because of any safety or efficacy issues.14

Furthermore, GLP-1 RAs reportedly induce weight loss by reducing appetite and this satiety-promoting effect has recently attracted significant interest.16,17 The appetite-regulating effects of GLP-1 RAs are primarily mediated by both peripheral (vagal) and central nervous system pathways.1,18 Food intake causes the stomach to stretch, activating gastro-mechanoreceptors in the intestinal wall, which then transmit satiety signals via the vagus nerve.19,20 GLP-1 RAs delay gastric emptying and reduce stomach motility in obese patients, contributing to their satiating effect.21 In addition, intracerebro ventricular administration of GLP-1 decreased food intake in rats, suggesting GLP-1 RAs are involved in a central nervous system pathway.22 Eight obese women reported reduced hunger following subcutaneous injections of exenatide and it was related with an enhanced functional connectivity of the nucleus tractus solitarius with the thalamus and hypothalamus.23 Clinical trials also indicated that GLP-1 RA treatment for obese patients presented superior weight loss efficacy compared to other anti-obesity medications.24 Based on these findings, two GLP-1 RAs were approved by the US FDA to treat obesity so far: liraglutide and semaglutide.

Recently, the newly developed drug tirzepatide, which targets both the GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) receptors, was introduced to the market to treat type 2 diabetes and obesity in 2022 and 2023, respectively (Table 1).25,26 GIP (Figure 1G) is another incretin hormone that triggers insulin secretion when a meal is ingested.27 Tirzepatide (Figure 1H) was designed to bind GLP-1/GIP receptors by modifying the native amino acid sequence of GIP.3,28 The combined activation of GLP-1 and GIP receptors more effectively reduces glucose levels and stimulates weight loss compared to the placebo, semaglutide and dulaglutide, or insulin.29–31

Table 1 Information on Currently Available US FDA-Approved GLP-1 RAs and a Dual GLP-1/GIP RA

Several clinical trials and reviews have been conducted to clarify the safety and effectiveness of GLP-1 RAs and a dual GLP-1/GIP RA.32,33 However, given the lack of comprehensive reviews in the existing literature on the pharmacokinetics and drug-drug interactions (DDIs) of GLP-1 RAs and a dual GLP-1/GIP RA, we produced this review to consolidate current knowledge and provide a thorough analysis of key considerations in clinical practice. The main objective of this review is to examine the pharmacokinetics of five peptide-derived incretin mimetic medications—exenatide, liraglutide, dulaglutide, semaglutide, and tirzepatide—including GLP-1 RAs and a dual GLP-1/GIP RA currently used in the market. First, the structures, indications, and doses of GLP-1 RAs and a dual GLP-1/GIP RA are outlined. Second, their pharmacokinetics, efficacy, and relationship between the pharmacokinetics and adverse events are summarized. Third, the pharmacokinetic DDIs of the medications are examined. Fourth, pharmacokinetic modeling approaches and the applications of GLP-1 RAs and a dual GLP-1/GIP RA are described. Finally, future perspectives of these therapies are briefly discussed.

Structures, Indications, and Dosages of GLP-1 RAs and a Dual GLP-1/GIP RA

Exenatide

Exenatide is the first-in-class incretin mimetic drug.34 It is a 39 amino acid (aa)-long synthetic peptide drug (Figure 1C), derived from exendin-4 (Figure 1B), a peptide isolated from the saliva of the lizard Heloderma suspectum.4,35 The immediate-release (IR) formulation of exenatide (Byetta®, Amylin Pharmaceuticals, San Diego, CA, USA) was first developed in 2005 to treat type 2 diabetes (Table 1).34 Initial dosing regimen of Byetta® is 5 μg subcutaneously twice daily and after one month, the dose is increased to 10 μg twice daily based on clinical response (Table 1).36 In 2012, the extended-release (ER) formulation of exenatide (Bydureon®, Amylin Pharmaceuticals) was approved by the US FDA to treat type 2 diabetes mellitus.37 Bydureon Pen® (AstraZeneca, Wilmington, DE, USA) and Bydureon BCise® (AstraZeneca) were developed later, but currently, only the latter, an enhanced injector pen formulation, is prescribed.38 The dosing regimen of Bydureon BCise® is 2 mg once weekly subcutaneously (Table 1).38

Liraglutide

Liraglutide is a 31 aa-long synthetic peptide drug attached to 16 carbon fatty acids (MW: 3751 g/mol) (Figure 1D).39 It was approved by the US FDA in 2014 to treat obesity (Saxenda®, Novo Nordisk, Plainsboro, NJ, USA), after first being approved as a type 2 diabetes medication (Victoza®, Novo Nordisk) in 2010 (Table 1). The available doses of liraglutide are 0.6 mg, 1.2 mg, or 1.8 mg for type 2 diabetes, and 0.6 mg, 1.2 mg, 1.8 mg, 2.4 mg, or 3 mg for obesity, respectively. The initial dosing regimen of liraglutide is 0.6 mg subcutaneously daily, with the dose increased to up to 3 mg for Saxenda® and 1.8 mg for Victoza®, usually after one week at the previous dose.40,41

Dulaglutide

Dulaglutide is a recombinant fusion protein consisting of two identical 31-aa chains, each covalently linked to a single Fc fragment derived from a modified human IgG4 heavy chain (MW: 59,669.8 g/mol) (Figure 1E).42,43 Dulaglutide (Trulicity®, Eli Lilly and Company, Indianapolis, IN, USA) was as a treatment for type 2 diabetes in 2014. It is administered as a subcutaneous injection once weekly, with available doses of 0.75 mg, 1.5 mg, 3 mg, and 4.5 mg (Table 1). The initial dosing regimen of dulaglutide is 0.75 mg subcutaneously weekly, with the dose increased to 4.5 mg, usually one month after the previous dose.43

Semaglutide

Semaglutide is a peptide drug composed of 31 amino acids linked with 18 carbon fatty acids (MW: 4113.6 g/mol) (Figure 1F).44 It was first used to treat type 2 diabetes (Ozempic®, Novo Nordisk) as a subcutaneous injection given once weekly. Subsequently, an oral formulation for once-daily administration (Rybelsus®, Novo Nordisk) was developed in 2019 to treat type 2 diabetes.45 Oral semaglutide is combined with sodium N-(8-[2-hydroxybenzoyl] amino) caprylate (salcaprozate sodium;SNAC), an absorption enhancer that improves semaglutide’s absorption across the gastric mucosa by transcellular mechanisms.46 By 2021, semaglutide (Wegovy®, Novo Nordisk) administered by subcutaneous injection was also approved by the US FDA as an obesity treatment (Table 1).47 The doses of semaglutide available for subcutaneous injection are 0.25 mg, 0.5 mg, 1 mg, or 2 mg for type 2 diabetes, and 0.25 mg, 0.5 mg, 1 mg, 1.7 mg, or 2.4 mg for obesity, respectively. The initial dosing regimen of semaglutide subcutaneous injection is 0.25 mg weekly, with the dose increased up to 2 mg for Ozempic® and 2.4 mg for Wegovy®, respectively, usually after one month at the previous dose.48,49 The available doses of oral semaglutide are 3 mg, 7 mg, and 14 mg (Table 1). The initial dosing regimen of oral semaglutide is 3 mg daily, with the dose increasing to 14 mg daily usually after one month of the previous dose.45

Tirzepatide

Tirzepatide is a 39-aa peptide conjugated with C20 fatty acids (MW: 4813 g/mol) (Figure 1H).3 It was approved by the US FDA in 2023 to treat obesity (Zepbound®, Eli Lilly and Company, Indianapolis, IN, USA) which was initially approved to treat type 2 diabetes in 2022 (Mounjaro®, Eli Lilly and Company) (Table 1). Tirzepatide is available in weekly subcutaneous doses of 2.5 mg, 5 mg, 7.5 mg, 10 mg, 12.5 mg, and 15 mg (Table 1).50,51 The initial dosing regimen of tirzepatide is 2.5 mg weekly, with the dose increased to up to 15 mg weekly usually after one month at the previous dose.50,51

Pharmacokinetics of GLP-1 RAs and a Dual GLP-1/GIP RA

Numerous studies have reported the pharmacokinetics of these drugs, with each study showing varying pharmacokinetic parameters based on differences in the clinical study design, pharmacokinetic parameter calculation approaches, or analytical methods. The pharmacokinetic parameters and profiles discussed in this review were primarily derived from clinical pharmacology and biopharmaceutics reviews and prescribing information released electronically by the US FDA. The pharmacokinetic profiles of GLP-1 RAs and a dual GLP-1/GIP RA vary owing to their diverse structures (Figures 1 and 2).39,52–58 Due to their large molecular weights and high polarity compared to small-molecule drugs, peptide drugs have low membrane permeability, resulting in a relatively small Vd in general.59 Compared to human GLP-1 (Figure 1A), the alanine residue at the second position of the N-terminus in GLP-1 RAs and a dual GLP-1/GIP RA (except for liraglutide) has been substituted to prevent metabolism by DPP-4 (Figure 1). Exenatide (Figure 1C) exhibits relatively fast renal clearance and a short half-life (Figure 2A). The ER formulation of exenatide achieves sustained drug concentrations in plasma over time (Figure 2B). Liraglutide (Figure 1D), dulaglutide (Figure 1E), semaglutide (Figure 1F), and tirzepatide (Figure 1H), which incorporate fatty acid conjugation using a linker or an IgG fragment crystallizable (Fc) region, display minimal renal clearance and prolonged half-lives (Figure 2C–G).60 Detailed information on their pharmacokinetic characteristics is provided below.

Figure 2 Continued.

Figure 2 Mean plasma concentration-time profiles (AG) and pharmacokinetic parameters (H) of GLP-1 RAs and a dual GLP-1/GIP RA. (A) Mean + standard deviation (SD) plasma concentration-time profile of exenatide IR following a single dose of 10 µg in healthy participants (n = 39). Data obtained from the US FDA clinical pharmacology biopharmaceutics review of Byetta®.52 (B) Mean + standard deviation (SD) plasma concentration-time profile of exenatide ER following a single dose of 2.5 mg in type 2 diabetes participants (n = 14). Adapted from Fineman M, Flanagan S, Taylor K et al. Pharmacokinetics and pharmacodynamics of exenatide extended-release after single and multiple dosing. Clin Pharmacokinet. 2011;50(1):65–74, with permission of Springer Nature.54 Permission conveyed through copyright clearance center, inc. (C) Mean plasma concentration-time profile of liraglutide following 1.8 mg and 3 mg doses at steady state in obese participants (n = 30 and 32, respectively). Data obtained from US FDA clinical pharmacology biopharmaceutics review of liraglutide.39 (D) Mean plasma concentration-time profile of dulaglutide following a single dose of 0.1–12 mg in healthy participants. Data obtained from US FDA clinical pharmacology biopharmaceutics review of dulaglutide.55 (E) Mean + standard deviation (SD) plasma concentration-time profile of oral semaglutide following a single dose of 10 mg in healthy participants (n = 11). Data obtained from US FDA clinical pharmacology biopharmaceutics review of Rybelsus®.57 (F) Mean plasma concentration-time profile of semaglutide following 0.5 mg and 1 mg doses at steady state in healthy participants (n = 8). Data obtained from US FDA clinical pharmacology biopharmaceutics review of semaglutide.56 (G) Mean + standard deviation (SD) plasma concentration-time profile of tirzepatide following a single dose of 0.25–8 mg in healthy participants. Data obtained from US FDA clinical pharmacology biopharmaceutics review of tirzepatide.58 (H) Pharmacokinetic parameters of approved GLP-1 RAs and a dual GLP-1/GIP RA. A power model was used for the statistical analysis of dose proportionality for AUC0-∞ and Cmax. Data points were digitized using GetData Graph Digitizer (version 2.26) from graphical representations. Created in GraphPad Prism 10 (GraphPad Software, San Diego, CA, USA).

Abbreviations: IR, immediate-release; ER, extended-release; SC, subcutaneous; T2DM, type 2 diabetes mellitus; Cmax, maximum plasma concentration; AUC, area under the curve; Tmax, time to reach maximum plasma concentration; t1/2, terminal half-life; Vd, volume of distribution; CL, total body clearance; F, absolute bioavailability; CL/F, apparent clearance; Vd/F, apparent volume of distribution.

Exenatide

Exenatide IR is absorbed rapidly, with maximum plasma concentrations (Cmax) usually attained around 2 h following subcutaneous injection (Figure 2A and H).36,61 However, exenatide ER is encapsulated in microspheres of medical-grade poly-(D,L-lactide-co-glycolide), which ensures the gradual and extended release of the drug (Figure 2B).62 In the first few hours, <1% of the surface-bound exenatide is released. During the subsequent gradual-release phase, which lasted for approximately 10 weeks, exenatide diffused from the deeper parts of the microspheres as the polymer degraded into smaller fragments. This is followed by the final erosion release when the polymer fully hydrolyzes into lactic acid and glycolic acid.54,62 Plasma concentrations of exenatide after ER formulation injection increased in a dose-dependent manner, but not dose-proportionally, across the 2.5–10 mg dose range.54 The mean apparent volume of distribution for exenatide after a single dose administered twice daily is 28.3 L, and it is expected to remain the same for exenatide ER (Figure 2H).38 In nonclinical studies, exenatide was mainly cleared by glomerular filtration, with subsequent proteolytic inactivation occurring in the renal tubules. In humans, exenatide has a dose-independent mean apparent clearance of 9.1 L/h.38

Liraglutide

The pharmacokinetics of liraglutide have been well documented in other reviews.63 In brief, the absolute bioavailability (F) of liraglutide is about 55%; furthermore, it has slow absorption (Tmax: approximately 12 h) (Figure 2C and H).63,64 The volume of distribution (Vd) and CL estimates from a population pharmacokinetic study ranged between 11.0–24.7 L and 0.6–1.2 L/h, respectively, showing consistency across various populations, including healthy individuals and those with type 2 diabetes, as well as dosage levels.63 Its half-life (t1/2) is approximately 13h (11–15h) (Figure 2H). This prolonged half-life (Figure 2C) can be explained by its structure, being composed of human GLP-1 conjugated with 16-carbon fatty acids, which causes noncovalent binding to serum albumin, reducing renal clearance (Figure 1D).65 The metabolism of liraglutide is mainly mediated by DPP-4 and neutral endopeptidase.66 Moreover, unchanged liraglutide was not detected in both urine and feces, implying that it is completely degraded within the body.66

Dulaglutide

Dulaglutide is absorbed slowly, with a median Tmax of 48 h (Figure 2D and H).43 After a single subcutaneous dose of dulaglutide at 0.75 mg or 1.5 mg, the mean absolute bioavailability values (F) were 65% and 47%, respectively.43 Systemic exposure to dulaglutide did not increase proportionally with doses ranging from 0.05 to 8 mg.67 The steady state volume of distribution (Vdss) were 19.2 L and 17.4 L, following the administration of 0.75 mg and 1.5 mg dulaglutide, respectively (Figure 2H).43 Dulaglutide is expected to be primarily metabolized by general protein catabolism, in which it is broken down into its constituent amino acids. The apparent clearance of dulaglutide at steady state was approximately 0.1 L/h for both the 0.75 mg and 1.5 mg once-weekly doses. The half-life (t1/2) of dulaglutide for the 0.75 mg and 1.5 mg once-weekly doses is approximately 5 d, which supports its once-weekly administration (Figure 2D).43

Semaglutide

The absolute bioavailability (F) of subcutaneously administered semaglutide is 89% (Figure 2H).48,49 Despite being co-formulated with an absorption enhancer, the absolute bioavailability of orally administered semaglutide was relatively low, at 0.8%.68 This limited absorption is likely due to semaglutide’s large molecular size.69 Orally administered semaglutide is primarily absorbed in the stomach and is facilitated by an absorption enhancer, with a Tmax of 2–2.5 h (Figure 2E and H).57 Conversely, semaglutide administered subcutaneously is absorbed more slowly, with a Tmax ranging from 36 to 59.8 h. It has a half-life (t1/2) of approximately 7 d, which supports its once-weekly administration (Figure 2F).48 Semaglutide possess a longer fatty acid chain (C-18 fatty acids) (Figure 1F) than liraglutide (C-16 fatty acids) (Figure 1D), which leads to increased albumin binding of the drug.70 In addition, alanine at the 8th position is substituted with 2-amino isobutyric acid in semaglutide, which increases the drug’s resistance to DPP-4 (Figure 1F).71 Such structure differences between liraglutide and semaglutide may result in a longer half-life and less frequent dosing of semaglutide. The apparent clearance and volume of distribution of semaglutide are 0.05 L/h and 12.5 L, respectively (Figure 2H). Semaglutide is metabolized by proteolysis and fatty acid oxidation and 3.1% of unchanged semaglutide is detected in urine.72

Tirzepatide

Tirzepatide has a mean absolute bioavailability (F) of 80% and is absorbed slowly, with a Tmax of 8–72 h in patients with type 2 diabetes (Figure 2G and H).51,73 The systemic exposure to tirzepatide increased dose-proportionally with doses ranging from 0.25 to 15 mg.74 The mean apparent steady-state volume of distribution (Vdss) of tirzepatide in patients with type 2 diabetes is approximately 10.3 L. Metabolism of tirzepatide is primarily mediated by hydrolysis of the peptide and beta-oxidation of the fatty acid (Figure 2H).51 The apparent clearance (CL/F) of tirzepatide is 0.061 L/h, and its half-life (t1/2) is approximately 5 d, which supports once-weekly dosing (Figure 2G).51,75 The metabolites are mainly excreted in urine and feces, while intact tirzepatide is not detected in either.76 The pharmacokinetics of the drugs were found to be similar between healthy individuals and patients with type 2 diabetes.77

Pharmacological Effects and Adverse Events of GLP-1 RAs and a Dual GLP-1/GIP RA

After being absorbed into the bloodstream, both GLP-1 RAs and a dual GLP-1/GIP RA exert their pharmacological effects by interacting with their respective target cell membrane receptors.78 GLP-1 receptors, class B of seven transmembrane G protein-coupled receptors, are expressed in various organs, including the brain, gastrointestinal tract, pancreas, kidneys, lungs, breasts, and thyroid gland.79–81 GLP-1 receptors are also found in blood vessels within specific organs, particularly the kidneys and lungs.79 Recent studies have also reported the presence of GLP-1 receptor mRNA transcripts in both heart chambers and heart muscle cells.82,83 Similarly, GIP receptors are class B-seven-transmembrane G protein-coupled receptors that are also expressed in various organs. However, their binding affinities and specificities differ from those of GLP-1 receptors because of differences in their amino acid sequences.84,85

Their interactions activate various cell signaling pathways, which have been extensively reviewed in previous studies.86,87 Based on this information, numerous clinical trials have been conducted, and the efficacy and adverse events of pivotal clinical trials related to the US FDA approval of GLP-1 RAs and a dual GLP-1/GIP RA for the treatment of type 2 diabetes and obesity are summarized in Table 2.88–96 Among the hitherto developed GLP-1 RAs, semaglutide has shown the highest efficacy with changes in HbA1c (%) and body weight of ‒1.45−1.55 and −14.85%, respectively, while tirzepatide has demonstrated relatively more effective blood glucose-lowering effects (−1.87−2.07) and weight loss (−15−20.9%) (Table 2).

Table 2 Half-Life, Efficacy, and Adverse Events of Approved GLP-1 RAs and a Dual GLP-1/GIP RA

In addition to the glucose-lowering effect mediated by the insulin secretion of pancreatic β-cells, and the central nervous-mediated delayed gastric emptying and satiety effects, some GLP-1 RAs also reportedly have cardioprotective, renoprotective, and hepatoprotective effects.97–102 Evidence indicates that GLP-1 RAs either reduce the risk of major adverse cardiovascular events (MACEs) or have no effect on cardiac safety in patients with type 2 diabetes, based on findings from cardiovascular outcome trials.97,98 Therefore, the indications for liraglutide, dulaglutide, and semaglutide have been expanded to treat patients with type 2 diabetes at risk of MACEs (Table 1). The pharmacological actions mediated by GLP-1 RAs, such as glucose-lowering effects, weight loss, reduced blood pressure, and improved lipid profile, may partially contribute to these cardiovascular outcomes.87,99

Table 2 also summarizes the most frequently reported adverse events from the pivotal clinical trials. GLP-1 RAs and a dual GLP-1/GIP RA administration-associated adverse events are primarily related to the gastrointestinal system, with nausea, vomiting, and diarrhea being the most common issues (Table 2).103 To mitigate these adverse events, the doses of some GLP-1 RAs are carefully titrated (Table 1).104 The administration of GLP-1 RAs to obese patients has also led to significantly higher risks of pancreatitis, bowel obstruction, and gastroparesis compared to those treated with bupropion-naltrexone.105 Increased risk of gallbladder disease was also observed in patients with type 2 diabetes.106 Recent studies have highlighted the association between diabetic retinopathy and GLP-1 RA treatment, indicating the need for further investigation into the underlying mechanisms and more comprehensive evidence.107

For GLP-1 RA, it appears that the incidence of adverse events increased as a drug remains in the body longer (Table 2). For example, exenatide IR, which has a short half-life (t1/2 = 2.4 h), exhibits relatively fewer adverse events, with the percentages of patients experiencing nausea and vomiting being 35.0% and 8.9%, respectively. In contrast, semaglutide, which has a longer half-life (t1/2 = 149–161 h), shows a higher incidence of adverse events (44.2, 31.5, and 24.8% for nausea, diarrhea, and vomiting, respectively) compared to liraglutide (t1/2 = 13 h; 40.2, 20.9, and 20.0% for nausea, diarrhea, and constipation, respectively) (Table 2). The ongoing pharmacovigilance studies also report an increased incidence of gastrointestinal or metabolism and nutrition-related disorders as the half-life of GLP-1 RAs increases.108,109 Traditionally, the relationship between pharmacokinetics and adverse events has been described based on the drug concentration in plasma.110,111 Therefore, the longer the drug concentration is maintained in the body, the more likely it is to have a pronounced and longer-lasting impact on the affected organs.112 However, there is evidence showing that GLP-1 RA-induced delayed gastric emptying experiences rapid tachyphylaxis, attenuated over time, and this effect must be taken into account for long-term use.113 Therefore, findings from real-world evidence studies evaluating the long-term safety of GLP-1 RAs and a dual GLP-1/GIP RA should be carefully considered. There have been emerging reports of an elevated risk of adverse events, such as gastrointestinal and ocular disorders, acute pancreatitis, and thyroid cancer, associated with GLP-1 RA treatment.108,109,114–116 In the future, large-scale data from these pharmacovigilance studies may enable more comprehensive discussions.

Moreover, exenatide (Figure 1C), which shares only 53% amino acid sequence homology with human GLP-1 (Figure 1A), has been reported to be associated with increased antibody development and exhibited a broad spectrum of adverse events.117–119 Therefore, relationship between amino acid sequence of GLP-1 RAs and a dual GLP-1/GIP RA and the incidence of adverse events also need to be considered.

Pharmacokinetic DDIs of Peptide Drugs

Recently, the US FDA has released draft guidance addressing clinical considerations for peptide drugs.120 The guidance defines a “peptide” as a molecule consisting of 40 or fewer amino acids. It recommends conducting in vitro studies related to cytochrome P450s (CYPs) and transporters if hepatic and/or biliary excretion constitutes >20% of the drug’s total elimination or if the drug’s primary target organ is the liver. Additionally, if the mechanism of action of the peptide drug can alter the pharmacokinetics of other co-administered drugs, it is recommended that the sponsor evaluate the peptide drug as a perpetrator.

Peptide drugs are mainly eliminated by protease enzymes, while liver uptake is minimal. This allows them to bypass metabolism by hepatic enzymes such as CYPs or UDP-glucuronosyltransferases (UGTs).121–123 Consequently, drug-metabolizing enzyme-mediated DDIs, typically involving competition for the same CYP binding site with another drug, are uncommon in peptide drug-related DDIs.124 Moreover, many peptide drugs are not known to be substrates or inhibitors of transporters at present, indicating that transporter-mediated biliary or renal excretion may not be a major pathway for their elimination.124 Therefore, it was speculated that peptide drugs are unlikely to be involved in the occurrence of transporter-mediated DDIs.122,124

However, increasing evidence is reporting the involvement of peptide drugs in the occurrence of CYP or transporter-mediated DDIs.123 The native glucagon peptide stimulates cAMP production, and elevated cAMP levels in rat hepatocytes indirectly reduce CYP2C11 expression.125 Moreover, research indicates that growth hormone enhances CYP3A4 expression in human hepatocytes, while synthetic somatostatin analogs like octreotide, which inhibit endogenous growth hormone, may decrease CYP3A4 activity.126 Co-administration of warfarin (a substrate of CYP1A2, CYP2C9, and CYP3A4) and octreotide led to high INR levels in humans, which may be attributed to the increased warfarin exposure caused by octreotide’s inhibitory effect on CYP3A4.127 Additionally, cyclosporine is an 11 aa-long lipophilic cyclic polypeptide used as an immunosuppressant.128 Cyclosporine has been identified as a substrate for CYP3A4, P-glycoprotein (P-gp), and organic anion transporting polypeptides (OATPs), while also acting as a potent inhibitor of CYP3A4, multidrug resistance-associated protein 2 (MRP2), OATPs, P-gp, and breast cancer resistance protein (BCRP).129131 Cyclosporine was found to inhibit CYP3A4 with a Ki of 1.42 µM in human liver microsomes, and systemic exposure of atorvastatin increased 15-fold in humans with the co-administration of cyclosporine.132,133

A recent in vitro DDI study of ATSP-7041, a stapled α-helical peptide being developed as an anticancer agent, revealed minimal CYP inhibition. However, uptake transporter assays indicated that ATSP-7041 acts as both a substrate for OATPs and a potent inhibitor of OATP1B1 (IC50: 0.81 µM), suggesting the potential for clinically relevant DDIs. Furthermore, ATSP-7041 inhibited the P-gp and BCRP-mediated efflux of the substrate.134 In addition, a clinically significant DDI was reported in a Phase I study of ALRN-6924, an analog of ATSP-7041 and an inhibitor of OATP1B3.135 A patient co-administered with telmisartan (an OATP1B3 substrate) presented with hypotension, which may have been caused by increased exposure of telmisartan mediated by ATSP-7041.135,136 Based on these findings, CYP- and transporter-mediated DDIs involving peptide drugs are not negligible and should be carefully evaluated.

Pharmacokinetic DDIs of GLP-1 RAs and a Dual GLP-1/GIP RA

Drug-Metabolizing Enzyme-and Transporter-Mediated DDIs

GLP-1 RAs and a dual GLP-1/GIP RA are now approved for the treatment of type 2 diabetes and obesity, with patients frequently managing coexisting metabolic diseases that require additional medications.137 Consequently, using multiple medications can result in DDIs, often leading to adverse drug reactions, making it crucial to assess the DDI potential.138 To date, there are limited data on clinically significant metabolic enzyme- or transporter-mediated DDIs involving GLP-1 RAs and a dual GLP-1/GIP RA. In vitro DDI studies detailed in clinical pharmacology and biopharmaceutics reviews were not included for exenatide and dulaglutide, but were included for liraglutide, semaglutide, and tirzepatide (Table 3).39,56–58,139 All the investigated GLP-1 RAs and a dual GLP-1/GIP RA exhibited minimal inhibition or induction of CYP enzymes. However, semaglutide and tirzepatide inhibited OATP1B1/3 activity, despite their relatively large molecular size, in the absence of bovine serum albumin (BSA). In addition, P-gp- and BCRP-mediated efflux was inhibited by tirzepatide. However, plasma concentrations of semaglutide at steady state are estimated to be about 100 times lower than the IC50 values for OATP1B1/3 (3.50 μM and 2.95 μM, respectively).56 Moreover, when BSA was added, the inhibitory effect disappeared or was diminished, probably due to the binding of the fatty acid residues of GLP-1 RAs and a dual GLP-1/GIP RA to BSA.70

Table 3 Overview of in vitro Drug-Metabolizing Enzyme and Transporter-Mediated DDI Studies by Approved GLP-1 RAs and a Dual GLP-1/GIP RA

In addition, a clinical DDI study was conducted to investigate the inhibitory effect of the absorption enhancer for oral semaglutide (SNAC) on BCRP, OAT1/3, and OATP1B1 based on in vitro data.140 However, the administration of SNAC alone did not alter the plasma concentrations of furosemide (an OAT1 and OAT3 substrate) or rosuvastatin (a BCRP and OATP1B1 substrate). Therefore, the sponsors concluded that clinically significant DDIs mediated by direct inhibition of CYP enzymes or transporters are unlikely to occur with the administration of GLP-1 RAs and a dual GLP-1/GIP RA.

Mechanisms of Action-Mediated DDIs

Furthermore, assessing DDIs for GLP-1 RAs and a dual GLP-1/GIP RA requires additional considerations, including the possible occurrence of DDIs mediated by mechanisms of action that remain poorly understood (Figure 3).120 Long-acting GLP-1 RAs remain in the body for an extended period (eg, with a mean residence time [MRT] of 224 hours at a steady state after administering 14 mg of oral semaglutide).141 This prolonged duration may be driven by albumin binding to the fatty acid residues of GLP-1 RAs and their ability to escape protease metabolism due to amino acid substitutions (Figure 1).141 They remain in the body, interacting with GLP-1 receptors in various organs, which could lead to DDIs through mechanisms of action such as slowing gastric emptying, reducing fat mass and inflammation, and increasing glomerular filtration rate (GFR) and renal plasma flow, consequently altering the pharmacokinetics of the victim drug (Figure 3).124,142 Investigations into mechanism of action-mediated pharmacokinetic DDIs are currently limited to those mediated by the slowing of gastric emptying, with examples summarized in other reviews.124,143

Figure 3 Flowchart illustrating the process of pharmacokinetic DDIs mediated by the mechanisms of action of approved GLP-1 RAs and a dual GLP-1/GIP RA; 1) GLP-1 RAs or a dual GLP-1/GIP RA (perpetrator) administration triggers receptor activation in GLP-1 receptor-expressing organs, leading to drug efficacy → 2) Physiological changes in the human body due to the drug effect → 3) Pharmacokinetic changes of small-molecule drugs (victim) influenced by these physiological changes. Created in BioRender. Min, J. (2025) https://BioRender.com/g44y018.

Notes: aPerpetrator, the drug that influences the pharmacokinetics of another drug; bVictim, the drug altered by the DDIs; cMechanism of action-mediated pharmacokinetic changes requiring additional validation through follow-up studies.

The pharmacokinetic DDI are commonly expressed in terms of “victim drug” and “perpetrator drug”. The perpetrator drug is the drug which affects the pharmacokinetics of the other drug; the victim drug is the drug affected by the DDI.144 GLP-1 RAs or a dual GLP-1/GIP RA as perpetrator drugs in combination with a victim drug, a number of clinical trials have been conducted to evaluate DDI potentials (Table 4 and Table 5). The regulatory guidance for DDI studies emphasizes that “perpetrators” must attain systemic exposure consistent with the highest therapeutic dose administered under steady-state conditions.144,145 Therefore, the effect of GLP-1 RAs or a dual GLP-1/GIP RA at steady-state conditions on the victim drug was generally investigated in the studied DDIs. Currently, an exact standard for determining pharmacokinetic changes caused by DDIs that may have a clinically significant impact has not been established. However, regulatory guidance states that when the ratio of the area under the concentration-time curve (AUC) of the victim drug with and without the perpetrator drug falls outside the range of 0.8–1.25, there is a potential risk of a clinically significant impact, necessitating further investigation.146 The rationale for selecting the victim drugs was based on their being commonly used or a narrow therapeutic index, with different Biopharmaceutics Classification System (BCS) classifications.124 The list of victim drugs that have been investigated to confirm the magnitude of DDIs caused by delayed gastric emptying is as follows: acetaminophen, metoprolol and oral contraceptives (BCS Class I); statins and griseofulvin (BCS Class II); levothyroxine, lisinopril, metformin, and sitagliptin (BCS Class III); digoxin and furosemide (BCS Class IV).124,140,143,147–163

Table 4 Changes in Oral Contraceptive Pharmacokinetics without and with Treatment with Approved GLP-1 RAs or a Dual GLP-1/GIP RA

Table 5 Changes in Narrow Therapeutic Index Drug Pharmacokinetics without and with Treatment with Approved GLP-1 RAs or a Dual GLP-1/GIP RA

In general, the administration of GLP-1 RAs delayed the absorption of the tested drugs by decreasing Cmax and/or delaying tmax. However, the AUC of the victim drugs were not altered by GLP-1 RA administration in most cases, suggesting that clinically significant DDIs are unlikely to occur.124 Conversely, both the Cmax and AUC of statins decreased by 28–70% and 0–40%, respectively, after GLP-1 RA subcutaneous administration.140,147,153,155,159 However, statins are known to exhibit high interindividual variability in terms of pharmacokinetics, and GLP-1 RA administration did not affect the lipid-lowering effect of statins in clinical studies, indicating minimal clinical significance.147,164 Moreover, the AUC of metformin increased by 32%, while the Cmax of metformin remained unchanged following oral semaglutide administration.158 In addition, the Cmax and AUC of furosemide were decreased by 34% and increased by 28%, respectively.140 Due to metformin and furosemide’s wide therapeutic index, the risk of clinically significant DDIs is low.140,158

Administration of SNAC, the absorption enhancer for oral semaglutide, did not alone alter exposure to lisinopril, warfarin, digoxin, or metformin. The AUC and Cmax ratios with or without SNAC administration remained within the 0.8–1.25 range.158

DDIs with Oral Contraceptives

Some GLP-1 RAs and a dual GLP-1/GIP RA altered the exposure of oral contraceptives, implying the potential for clinically significant DDIs (Table 4).55,57,148,155,157,161,163 About 25% of women between the ages of 15 and 44 use oral contraceptives, highlighting the importance of studying their potential DDIs.165,166 The Cmax and AUC values of ethinylestradiol and levonorgestrel were not affected by the administration of 14 mg oral or 1 mg subcutaneous semaglutide, indicating that it is not clinically relevant (Table 4).56,157 Notable changes in the Cmax ratios of oral contraceptives after the administration of 10 µg exenatide twice daily seem to be caused by their administration 30 min after exenatide, when the delayed gastric emptying effect was at its peak. Therefore, it was concluded that clinically significant pharmacokinetic changes are unlikely to occur.148,167 Similarly, alterations in the pharmacokinetics after 1.8 mg liraglutide and 1.5 mg dulaglutide administration have also been shown to have minimal clinical significance.161,168

The most significant pharmacokinetic alterations were reported following the administration of a single 5 mg dose of tirzepatide, with the Cmax and AUC values of ethinylestradiol decreasing by 59% and 21%, respectively, and those of norelgestromin by 55% and 22%, respectively (Table 4).161 However, the pharmacokinetic /pharmacodynamic relationship of oral contraceptives is not yet fully defined; therefore, additional studies are required to explore how changes in exposure influence their effectiveness.166 Moreover, this implies that tirzepatide more strongly affects the absorption of oral contraceptives and that this increased impact may be driven by rapid dose escalation.161 Unlike other pharmacodynamic effects, such as lowering blood glucose or reducing appetite, the delay in gastric emptying shows tachyphylaxis, with the effect gradually decreasing over time.169 Therefore, the investigators have suggested switching to a non-oral contraceptive option or adding a barrier method of contraception for 4 weeks after tirzepatide administration is initiated or if the dose is substantially increased.50,51

DDIs with Narrow Therapeutic Index Drugs

Other clinical studies on DDIs have investigated the delayed gastric emptying effect of GLP-1 RA on the pharmacokinetics of drugs with narrow therapeutic index (Table 5). It was concluded that the Cmax and AUC changes of digoxin after GLP-1 RA administration may not be clinically significant.55,56,58,150,153 Likewise, the pharmacokinetics of warfarin were not altered by semaglutide administration.158,159 The AUC of S- and R-warfarin was not affected by dulaglutide and tirzepatide administration.55,58 However, postmarketing data on exenatide indicate an increase in INR, which is sometimes associated with bleeding events, highlighting the need for close INR monitoring after GLP-1 RA treatment.36

Furthermore, DDIs between levothyroxine and oral semaglutide, caused by delayed gastric emptying, have been explored (Table 5).160 Levothyroxine, a synthetic form of the thyroid hormone thyroxine, is an orally administered narrow therapeutic index drug used to treat hypothyroidism.170 It has been reported that gastrointestinal motility may affect its bioavailability, and reduced absorption is a leading contributor to refractory hypothyroidism.171,172 Therefore, it is recommended to administer levothyroxine at least 1 h before meals.173 Based on this information, the influence of gastric emptying effect by oral semaglutide administration on pharmacokinetics of levothyroxine was investigated.160 After multiple oral doses of 14 mg oral semaglutide, the Tmax of levothyroxine was delayed by 2 h, with a 12% decrease in the Cmax and a 33% increase in the AUC, indicating potential clinical relevance (Table 5). Therefore, monitoring thyroid parameters is advised when oral semaglutide is co-administered.45,160

In summary, most of the studied drugs did not show clinically significant DDIs related to delayed gastric emptying caused by GLP-1 RAs or a dual GLP-1/GIP RA administration. However, oral contraceptives and levothyroxine, showed notable changes in AUC following the administration of tirzepatide and oral semaglutide, respectively, implying a potential risk of a clinically significant DDIs (Table 4 and Table 5). This indicates the need for close monitoring when GLP-1 RAs and a dual GLP-1/GIP RA are co-administered, especially when starting treatment or increasing the dose. For a more accurate assessment, large-scale pharmacokinetic/pharmacodynamic (PK/PD) studies need to be conducted. Delayed gastric emptying may undergo tachyphylaxis, potentially reducing the impact of DDIs with repeated dosing.169,174 Moreover, delayed absorption of the concomitant drug due to delayed gastric emptying can be minimized by administering the drug 1 h before GLP-1 RAs or a dual GLP-1/GIP RA administration.168 However, the possibility of DDIs related to delayed gastric emptying cannot be excluded,175 so investigators recommend monitoring when oral drugs are co-administered with GLP-1 RAs and a dual GLP-1/GIP RA.

The Developed Pharmacokinetic Models for GLP-1 RAs and a Dual GLP-1/GIP RA

Pharmacokinetic models of GLP-1 RAs and a dual GLP-1/GIP RA have been developed as clinical support tools, enabling the prediction of their pharmacokinetic behavior across different clinical scenarios (Table 6). The developed pharmacokinetic models of GLP-1 RAs and a dual GLP-1/GIP RA were searched in the PubMed electronic database. The search terms were “exenatide”, “liraglutide”, “dulaglutide”, “semaglutide”, or “tirzepatide”, along with “pharmacokinetic model”. A total of 26 articles were identified as of October 8, 2024. Clinical pharmacology and biopharmaceutics reviews released electronically by the US FDA were also examined for information on the pharmacokinetic models of GLP-1 RAs and a dual GLP-1/GIP RA.39,52,55–58,139,176 Thirty pharmacokinetic models were developed to predict the pharmacokinetics of GLP-1 RAs and a dual GLP-1/GIP RA. Among the GLP-1 RAs and a dual GLP-1/GIP RA, exenatide has the most developed pharmacokinetic model (9 of 30),61,177–184 followed by semaglutide (8 of 30),68,162,182,185–189 liraglutide and dulaglutide (each 5 of 30),156,177,182,189–194 and tirzepatide (3 of 30).58,73,195 The developed pharmacokinetic models were used either solely to predict pharmacokinetics (18 of 30) or integrated with pharmacodynamic models to evaluate responses (12 of 30). Most of the developed pharmacokinetic models were population pharmacokinetic models utilizing nonlinear mixed effect modeling (22 of 30). The models have been used to predict pharmacokinetics in special populations, including those with hepatic or renal impairment, as well as in disease states such as type 2 diabetes and obesity. Additionally, pharmacokinetic differences were predicted in subjects based on factors such as age, body weight, sex, race, ethnicity, and injection site, with most GLP-1 RAs and a dual GLP-1/GIP RA exhibiting only minor variations.

Table 6 The Developed Pharmacokinetic (PK) and Pharmacokinetic/Pharmacodynamic (PK/PD) Models of Approved GLP-1 RAs and a Dual GLP-1/GIP RA

Physiologically based pharmacokinetic (PBPK) models have been developed for exenatide (1 of 30), dulaglutide (1 of 30), semaglutide (1 of 30), and tirzepatide (2 of 30). The developed models were used to investigate the initial dose for pediatric patients (2 of 30), the potential for DDIs caused by delayed gastric emptying (2 of 30), and the pharmacokinetics of once-monthly dosing (1 of 30) (Table 6).

Delayed gastric emptying-mediated DDIs have been investigated using both PBPK models (2 of 30) and population pharmacokinetic models (1 of 30). For example, increased gastric emptying time due to the mechanism of action of dulaglutide was described in the developed PBPK model by modulating the default gastric MRT.156 Administering 1.5 mg of dulaglutide reportedly decreased gastric emptying by roughly threefold, with the initial MRT (0.27 h) increasing to 0.84 h.196 The refined model successfully described the pharmacokinetic profiles of the affected drug, atorvastatin acid and lactone, after dulaglutide administration in SimcypTM (Certara UK Limited, Sheffield, United Kingdom).156 It appears that clinically significant DDIs caused by delayed gastric emptying affecting the pharmacokinetics of the victim drugs are unlikely to occur.58,156,162

Future Perspectives

GLP-1 RAs Currently Under Development

Based on previous findings, dosages that enable longer durations and less frequent administrations have been investigated. For example, efpeglenatide, a long-acting GLP-1 RA and an exendin analog linked to an IgG4 Fc fragment, has been explored for once-monthly administration to treat type 2 diabetes.197,198 Additionally, efforts have been made to develop oral formulations with improved bioavailability (F). Orforglipron, a nonpeptide GLP-1 RA administered once daily, showed dose-dependent efficacy on weight loss in a Phase II trial.199

Currently, investigations into GLP-1 RAs and similar drugs are underway to obtain approval for additional indications based on their diverse pharmacological effects. Liraglutide and semaglutide have beneficial effects on non-alcoholic steatohepatitis.200,201 Additionally, incretin mimetic drugs targeting multiple receptors for greater efficacy, including glucagon and GIP receptors, are under development.202 A Phase 2a study of efocipegtrutide (HM15211), a new long-acting triple GLP-1/GIP/glucagon RA is currently being conducted to assess its use to treat non-alcoholic steatohepatitis.203 Survodutide, a novel dual GLP-1/ glucagon RA, significantly reduced body weight and improved metabolic dysfunction-associated steatohepatitis compared to the placebo in phase II trials.204 Mazdutide, a dual GLP-1/glucagon RA, and retatrutide, a triple GLP-1/GIP/glucagon RA, also have both shown significant weight loss effects in Phase 2 trials.205,206

Among the drugs discussed, including US FDA-approved GLP-1 RAs, tirzepatide was reported to achieve the most significant reduction in blood glucose levels. This weight loss effect was most potent in the CagriSema (cagrilintide and semaglutide combination therapy)-treated group, followed by tirzepatide and retatrutide.97 These new GLP-1 RAs, including dual and triple agonists, are now in Phase 3 trials, and the clinical use of more potent GLP-1 RAs with additional indications is expected.

Drug-Metabolizing Enzyme- and Transporter- Mediated DDIs of GLP-1 RAs and a Dual GLP-1/GIP RA

To date, no clinically relevant drug-metabolizing enzyme- or transporter-mediated DDIs involving GLP-1 RAs or a dual GLP-1/GIP RA have been reported (Table 3). However, some transporters, including OATP1B1/3 and OAT3, demonstrated inhibition following treatment with GLP-1 RAs and a dual GLP-1/GIP RA in vitro, suggesting that the assessment of transporter-mediated DDI by GLP-1 RAs may be necessary (Table 3). Moreover, glucagon treatment in human hepatocytes leads to the downregulation of CYP gene expression, and the underlying mechanism may be driven by the activation of glucagon receptors expressed in the liver.125,207,208 Drugs targeting both GLP-1 and glucagon receptors are currently under development, and it is vital to examine their indirect effects on CYP gene expression. Currently, in vitro studies on peptide-mediated DDIs, which would provide better in vitro to in vivo extrapolation, remain poorly established.207 A recent study introduced new experiment designs using HepatoPac® (Hepregen Corporation, Medford, MA, USA), spheroids or liver-on-a-chip models to assess the modulation of CYPs and transporter gene expression by peptides, but further large-scale experiments are still needed.209

Pharmacokinetic DDIs Caused by Mechanisms of Actions Following GLP-1 RAs or a Dual GLP-1/GIP RA Treatment

GLP-1 RAs are currently attracting significant attention because of their remarkable weight loss-promoting effects. From a pharmacokinetic perspective, besides increased gastric emptying, obesity has been suggested to modulate CYP gene expression, and increased fat mass may result in an increased Vd of lipophilic drugs.210,211 The weight loss effects from GLP-1 RA treatment may reverse these changes and alter the pharmacokinetics of co-administered lipophilic drugs by decreasing the Vd (Figure 3). For example, the Vd of diazepam, lorazepam, and nitrazepam decreased by 3.2-, 1.7-, and 2.1-fold, respectively, in non-obese participants, and their CL and t1/2 were altered, indicating the need for dose adjustment.212

Furthermore, the anti-inflammatory properties of GLP-1 RAs constitute another notable effect. They can reduce levels of inflammatory biomarkers, such as C-reactive protein and inflammatory cytokines (eg, tumor necrosis factor-alpha and interleukin-6).100,101 This positively impacts various pathophysiological conditions, including type 2 diabetes, obesity, and non-alcoholic fatty liver disease.100–102 In addition, inflammatory cytokines are reportedly involved in CYP gene expression, with the reduced cytokine levels-mediated by GLP-1 RA administration may alter CYP gene expression, potentially leading to changes in the pharmacokinetics of co-administered drugs (Figure 3).213

Finally, cardiovascular outcome trials have demonstrated that GLP-1 RAs offer beneficial effects on kidney function in type 2 diabetes and decrease the risk of major adverse kidney events in patients with type 2 diabetes and acute kidney disease.99,214 These renoprotective effects are expected to be driven by both direct and indirect mechanisms including reducing albuminuria, reducing inflammation, natriuresis and diuresis induction, and antioxidative effects.215 Moreover, GLP-1 RA treatment reportedly increased renal plasma flow and GFR in rats, leading to natriuresis and diuresis.216 It is known that changes in GFR can affect the elimination of many drugs including narrow therapeutic index drugs (aminoglycosides, lithium, digoxin), diuretics, and nonsteroidal anti-inflammatory drugs.217–221 Therefore, pharmacokinetic changes in drugs mediated by increased GFR and renal plasma flow following GLP-1 RA administration may occur, and the impact of its mechanism of action need to be investigated in the future. Moreover, as these mechanisms of action may occur concurrently, it is important to investigate alterations in drug exposure when multiple mechanisms are considered simultaneously.

Utilization of PBPK Modeling Approaches to Assess DDIs Caused by Mechanisms of Actions

Evaluating the mechanism of action (eg, delayed gastric emptying, reduced fat mass and inflammation, and changes in GFR)-mediated DDIs of GLP-1 RAs and a dual GLP-1/GIP RA is often challenging to control for and clinical studies are costly. Therefore, modeling approaches can be valuable for examining the extent of pharmacokinetic DDIs driven by these mechanisms. Among various modeling techniques, PBPK modeling appears to be particularly promising. Notably, the US FDA submission documents for tirzepatide include predictions of DDIs mediated by delayed gastric emptying using PBPK modeling, rather than conducting clinical DDI studies for certain drugs (Table 6).58 PBPK modeling is a mathematical approach that uses differential equations to predict pharmacokinetics in humans and other species. It has become a valuable tool in various stages of drug development, including first-in-human dose calculation, predicting DDI potential, and assessing pharmacokinetic profiles in special populations such as pediatrics and patients with renal or hepatic impairment.222,223 In particular, it is widely used to predict potential pharmacokinetic DDIs of small molecules.224 Physiological properties of the body, including organ mass and volume, blood flow rate, plasma protein levels, and the abundance of enzyme and transporter expression, are mathematically described within the model.225,226 Therefore, the mechanism of action of GLP-1 RAs and a dual GLP-1/GIP RA can be characterized by modulating physiological properties within the model, enabling the prediction of altered pharmacokinetics for the affected drug. To effectively develop a PBPK model for studying mechanism-of-action-mediated DDIs, it is essential to carefully assess the extent to which these mechanisms impact pharmacokinetics. After the validation process, the developed PBPK model can be used to assess the impact of the mechanism of action on the pharmacokinetics of various victim drugs.58,156

Conclusion

In this review, pharmacokinetics, pharmacokinetic drug−drug interactions (DDIs), and pharmacokinetic modeling approaches of four currently available GLP-1 RAs (exenatide, liraglutide, dulaglutide, and semaglutide) and a dual GLP-1/GIP RA (tirzepatide), are discussed. Substitutions in amino acid sequences, fatty acid conjugation using a linker, and fusion with albumin or the IgG fragment crystallizable (Fc) region in GLP-1 RAs and a dual GLP-1/GIP RA have minimized metabolism and renal excretion. The pharmacokinetic profiles vary due to their diverse structures and prolonged half-life appears to be associated with an increased risk of adverse events. To date, there have been no reports of pharmacokinetic DDIs involving metabolic enzymes or transporters. However, there is the potential for DDIs of GLP-1 RAs to occur through their mechanisms of action. Among the possible mechanisms underlying DDIs, those related to delayed gastric emptying have been most extensively studied. The pharmacokinetics of most victim drugs were not affected by GLP-1 RAs. However, clinically significant DDIs were observed with oral contraceptives after tirzepatide administration and with levothyroxine after oral semaglutide administration. Therefore, close monitoring is recommended when oral drugs are co-administered with GLP-1 RAs or a dual GLP-1/GIP RA. Drug-metabolizing enzyme- and transporter-mediated DDIs involving GLP-1 RAs and a dual GLP-1/GIP RA require careful evaluation. Thirty pharmacokinetic models were developed to predict the pharmacokinetics of GLP-1 RAs and a dual GLP-1/GIP RA. Among these models, PBPK modeling can be useful for assessing DDIs caused by different mechanisms of action. Alterations in Vd resulting from reduced fat mass and alterations in clearance resulting from changes in CYP activity and GFR are key factors in determining pharmacokinetics. These mechanisms may accelerate the pharmacokinetic changes in the affected drug and lead to clinically relevant DDIs, particularly in drugs with a narrow therapeutic window. However, the DDIs mediated by these factors remain poorly understood and will require further investigation to ensure the safe use of GLP-1 RAs with concomitant medications.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

This study was supported by a grant of Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare (HF20C0002), The Basic Science Research Program through the National Research Foundation of Korea, funded by The Ministry of Education (2018R1A6A1A03025108), and the Research Fund of The Catholic University of Korea (2023).

Disclosure

The authors report no conflicts of interest in this work.

References

1. Crane J, McGowan B. The GLP-1 agonist, liraglutide, as a pharmacotherapy for obesity. Ther Adv Chronic Dis. 2016;7(2):92–107. doi:10.1177/2040622315620180

2. Lucey M, Ashik T, Marzook A, et al. Acylation of the incretin peptide exendin-4 directly impacts glucagon-like peptide-1 receptor signaling and trafficking. Mol Pharmacol. 2021;100(4):319–334. doi:10.1124/molpharm.121.000270

3. Gallwitz B. Clinical perspectives on the use of the GIP/GLP-1 receptor agonist tirzepatide for the treatment of type-2 diabetes and obesity. Front Endocrinol. 2022;13:1004044. doi:10.3389/fendo.2022.1004044

4. Doyle ME, Theodorakis MJ, Holloway HW, Bernier M, Greig NH, Egan JM. The importance of the nine-amino acid C-terminal sequence of exendin-4 for binding to the GLP-1 receptor and for biological activity. Regul Pept. 2003;114(2–3):153–158. doi:10.1016/s0167-0115(03)00120-4

5. Yu M, Benjamin MM, Srinivasan S, et al. Battle of GLP-1 delivery technologies. Adv Drug Deliv Rev. 2018;130:113–130. doi:10.1016/j.addr.2018.07.009

6. Faillie JL, Yin H, OHY Y, et al. Incretin-based drugs and risk of intestinal obstruction among patients with type 2 diabetes. Clin Pharmacol Ther. 2022;111(1):272–282. doi:10.1002/cpt.2430

7. Parks M, Rosebraugh C. Weighing risks and benefits of liraglutide--the FDA’s review of a new antidiabetic therapy. N Engl J Med. 2010;362(9):774–777. doi:10.1056/NEJMp1001578

8. Meloni AR, DeYoung MB, Lowe C, Parkes DG. GLP-1 receptor activated insulin secretion from pancreatic beta-cells: mechanism and glucose dependence. Diabetes Obes Metab. 2013;15(1):15–27. doi:10.1111/j.1463-1326.2012.01663.x

9. Cryer PE, Davis SN, Shamoon H. Hypoglycemia in diabetes. Diabetes Care. 2003;26(6):1902–1912. doi:10.2337/diacare.26.6.1902

10. Schopman JE, Simon AC, Hoefnagel SJ, Hoekstra JB, Scholten RJ, Holleman F. The incidence of mild and severe hypoglycaemia in patients with type 2 diabetes mellitus treated with sulfonylureas: a systematic review and meta-analysis. Diabetes Metab Res Rev. 2014;30(1):11–22. doi:10.1002/dmrr.2470

11. Zhang Y, Parajuli KR, Fava GE, et al. GLP-1 receptor in pancreatic alpha-cells regulates glucagon secretion in a glucose-dependent bidirectional manner. Diabetes. 2019;68(1):34–44. doi:10.2337/db18-0317

12. Tan H, Su W, Zhang W, Wang P, Sattler M, Zou P. Recent advances in half-life extension strategies for therapeutic peptides and proteins. Curr Pharm Des. 2018;24(41):4932–4946. doi:10.2174/1381612825666190206105232

13. Scheen AJ. New therapeutic approaches in type 2 diabetes. Acta Clin Belg. 2008;63(6):402–407. doi:10.1179/acb.2008.083

14. Latif W, Lambrinos KJ, Patel P, Rodriguez R. Compare and Contrast the Glucagon-Like Peptide-1 Receptor Agonists (GLP1RAs). StatPearls; 2024.

15. Chudleigh RA, Platts J, Bain SC. Comparative effectiveness of long-acting GLP-1 receptor agonists in type 2 diabetes: a short review on the emerging data. Diabetes Metab Syndr Obes. 2020;13:433–438. doi:10.2147/DMSO.S193693

16. Flint A, Raben A, Astrup A, Holst JJ. Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans. J Clin Invest. 1998;101(3):515–520. doi:10.1172/JCI990

17. Friedrichsen M, Breitschaft A, Tadayon S, Wizert A, Skovgaard D. The effect of semaglutide 2.4 mg once weekly on energy intake, appetite, control of eating, and gastric emptying in adults with obesity. Diabetes Obes Metab. 2021;23(3):754–762. doi:10.1111/dom.14280

18. Alruwaili H, Dehestani B, le Roux CW. Clinical impact of liraglutide as a treatment of obesity. Clin Pharmacol. 2021;13:53–60. doi:10.2147/CPAA.S276085

19. Cork SC. The role of the vagus nerve in appetite control: implications for the pathogenesis of obesity. J Neuroendocrinol. 2018;30(11):e12643. doi:10.1111/jne.12643

20. Shah M, Vella A. Effects of GLP-1 on appetite and weight. Rev Endocr Metab Disord. 2014;15(3):181–187. doi:10.1007/s11154-014-9289-5

21. Nauck MA, Niedereichholz U, Ettler R, et al. Glucagon-like peptide 1 inhibition of gastric emptying outweighs its insulinotropic effects in healthy humans. Am J Physiol. 1997;273(5):E981–8. doi:10.1152/ajpendo.1997.273.5.E981

22. Turton MD, O’Shea D, Gunn I, et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature. 1996;379(6560):69–72. doi:10.1038/379069a0

23. Coveleskie K, Kilpatrick LA, Gupta A, et al. The effect of the GLP-1 analogue Exenatide on functional connectivity within an NTS-based network in women with and without obesity. Obes Sci Pract. 2017;3(4):434–445. doi:10.1002/osp4.124

24. Wang JY, Wang QW, Yang XY, et al. GLP-1 receptor agonists for the treatment of obesity: role as a promising approach. Front Endocrinol. 2023;14:1085799. doi:10.3389/fendo.2023.1085799

25. Nauck MA, D’Alessio DA. Tirzepatide, a dual GIP/GLP-1 receptor co-agonist for the treatment of type 2 diabetes with unmatched effectiveness regrading glycaemic control and body weight reduction. Cardiovasc Diabetol. 2022;21(1):169. doi:10.1186/s12933-022-01604-7

26. Abbasi J. FDA Green-Lights Tirzepatide, Marketed as Zepbound, for chronic weight management. JAMA. 2023;330(22):2143–2144. doi:10.1001/jama.2023.24539

27. Seino Y, Fukushima M, Yabe D. GIP and GLP-1, the two incretin hormones: similarities and differences. J Diabetes Investig. 2010;1(1–2):8–23. doi:10.1111/j.2040-1124.2010.00022.x

28. Scheen AJ. Dual GIP/GLP-1 receptor agonists: new advances for treating type-2 diabetes. Ann Endocrinol. 2023;84(2):316–321. doi:10.1016/j.ando.2022.12.423

29. Frias JP, Davies MJ, Rosenstock J, et al. Tirzepatide versus semaglutide once weekly in patients with type 2 diabetes. N Engl J Med. 2021;385(6):503–515. doi:10.1056/NEJMoa2107519

30. Frias JP, Nauck MA, Van J, et al. Efficacy and tolerability of tirzepatide, a dual glucose-dependent insulinotropic peptide and glucagon-like peptide-1 receptor agonist in patients with type 2 diabetes: a 12-week, randomized, double-blind, placebo-controlled study to evaluate different dose-escalation regimens. Diabetes Obes Metab. 2020;22(6):938–946. doi:10.1111/dom.13979

31. Frias JP, Nauck MA, Van J, et al. Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet. 2018;392(10160):2180–2193. doi:10.1016/S0140-6736(18)32260-8

32. Trujillo JM, Nuffer W, Smith BA. GLP-1 receptor agonists: an updated review of head-to-head clinical studies. Ther Adv Endocrinol Metab. 2021;12:2042018821997320. doi:10.1177/2042018821997320

33. Nauck MA, Quast DR, Wefers J, Meier JJ. GLP-1 receptor agonists in the treatment of type 2 diabetes - state-of-the-art. Mol Metab. 2021;46:101102. doi:10.1016/j.molmet.2020.101102

34. Davidson MB, Bate G, Kirkpatrick P. Exenatide. Nat Rev Drug Discov. 2005;4(9):713–714. doi:10.1038/nrd1828

35. Gentilella R, Bianchi C, Rossi A, Rotella CM. Exenatide: a review from pharmacology to clinical practice. Diabetes Obes Metab. 2009;11(6):544–556. doi:10.1111/j.1463-1326.2008.01018.x

36. AstraZeneca Pharmaceuticals LP. Byetta (exenatide) injection, for subcutaneous use: prescribing information. Wilmington, DE: AstraZeneca Pharmaceuticals LP; 2022. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/021773s048lbl.pdf. Accessed September 24, 2024.

37. AstraZeneca Pharmaceuticals LP. Bydureon (exenatide extended-release) for injectable suspension: prescribing information. Wilmington, DE: AstraZeneca Pharmaceuticals LP; 2022. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/022200s034lbl.pdf. Accessed September 24, 2024.

38. AstraZeneca Pharmaceuticals LP. Bydureon BCise (exenatide extended-release) injection, for subcutaneous use: prescribing information. Wilmington, DE: AstraZeneca Pharmaceuticals LP; 2022. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/209210s023lbl.pdf. Accessed September 24, 2024.

39. United State Food and Drug Administration. Clinical pharmacology biopharmaceutics review: liraglutide. 2013. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/206321Orig1s000ClinPharmR.pdf. Accessed June 24, 2024.

40. Novo Nordisk. Saxenda (liraglutide) injection, for subcutaneous use: prescribing information. Bagsvaerd, Denmark: Novo Nordisk; 2023. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/206321s016lbl.pdf. Accessed September 24, 2024.

41. Novo Nordisk. Victoza (liraglutide) injection, for subcutaneous use: prescribing information. Bagsværd, Denmark: Novo Nordisk; 2023. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/022341s039lbl.pdf. Accessed September 24, 2024.

42. Glaesner W, Vick AM, Millican R, et al. Engineering and characterization of the long-acting glucagon-like peptide-1 analogue LY2189265, an Fc fusion protein. Diabetes Metab Res Rev. 2010;26(4):287–296. doi:10.1002/dmrr.1080

43. Eli Lilly and Company. Trulicity (dulaglutide) injection, for subcutaneous use: prescribing information. Indianapolis, IN: Eli Lilly and Company; 2022. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/125469s051lbl.pdf. Accessed September 24, 2024.

44. Mahapatra MK, Karuppasamy M, Sahoo BM. Semaglutide, a glucagon like peptide-1 receptor agonist with cardiovascular benefits for management of type 2 diabetes. Rev Endocr Metab Disord. 2022;23(3):521–539. doi:10.1007/s11154-021-09699-1

45. Novo Nordisk. Rybelsus (semaglutide) tablets, for oral use: prescribing information. Bagsværd, Denmark: Novo Nordisk; 2024. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/213051s018lbl.pdf. Accessed September 24, 2024.

46. Buckley ST, Baekdal TA, Vegge A, et al. Transcellular stomach absorption of a derivatized glucagon-like peptide-1 receptor agonist. Sci Transl Med. 2018;10(467). doi:10.1126/scitranslmed.aar7047

47. Singh G, Krauthamer M, Bjalme-Evans M. Wegovy (semaglutide): a new weight loss drug for chronic weight management. J Investig Med. 2022;70(1):5–13. doi:10.1136/jim-2021-001952

48. Novo Nordisk. Wegovy (semaglutide) injection, for subcutaneous use: prescribing information. Bagsværd, Denmark: Novo Nordisk; 2024. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/215256s011lbl.pdf. Accessed September 24, 2024.

49. Novo Nordisk. Ozempic (semaglutide) injection, for subcutaneous use: prescribing information. Bagsværd, Denmark: Novo Nordisk; 2023. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/209637s020s021lbl.pdf. Accessed September 24, 2024.

50. Eli Lilly and Company. Zepbound (tirzepatide) injection, for subcutaneous use: prescribing information. Indianapolis, IN: Eli Lilly and Company; 2024. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/217806s003lbl.pdf. Accessed September 24, 2024.

51. Eli Lilly and Company. Mounjaro (tirzepatide) injection, for subcutaneous use: prescribing information. Indianapolis, IN: Eli Lilly and Company; 2023. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/215866Orig1s002s006lbl.pdf. Accessed September 24, 2024.

52. United State Food and Drug Administration. Clinical pharmacology biopharmaceutics review: Byetta®. 2003. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2005/021773_Byetta_biopharmr.PDF. Accessed June 24, 2024.

53. Linnebjerg H, Kothare PA, Park S, et al. Effect of renal impairment on the pharmacokinetics of exenatide. Br J Clin Pharmacol. 2007;64(3):317–327. doi:10.1111/j.1365-2125.2007.02890.x

54. Fineman M, Flanagan S, Taylor K, et al. Pharmacokinetics and pharmacodynamics of exenatide extended-release after single and multiple dosing. Clin Pharmacokinet. 2011;50(1):65–74. doi:10.2165/11585880-000000000-00000

55. United State Food and Drug Administration. Clinical pharmacology biopharmaceutics review: Dulaglutide. 2014. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/125469Orig1s000ClinPharmR.pdf. Accessed June 24, 2024.

56. United State Food and Drug Administration. Clinical pharmacology biopharmaceutics review: Ozempic®. 2016. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2017/209637Orig1s000ClinPharmR.pdf. Accessed June 24, 2024.

57. United State Food and Drug Administration. Clinical pharmacology biopharmaceutics review: Rybelsus®. 2019. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2019/213051Orig1s000ClinPharmR.pdf. Accessed June 24, 2024.

58. United State Food and Drug Administration. Clinical pharmacology biopharmaceutics review: Mounjaro®. 2021. Available from: https://www.accessdata.fda.gov. Accessed June 24, 2024.

59. Collins L, Costello RA. Glucagon-Like Peptide-1 Receptor Agonists. StatPearls; 2024.

60. Gentilella R, Pechtner V, Corcos A, Consoli A. Glucagon-like peptide-1 receptor agonists in type 2 diabetes treatment: are they all the same? Diabetes Metab Res Rev. 2019;35(1):e3070. doi:10.1002/dmrr.3070

61. Cirincione B, Mager DE. Population pharmacokinetics of exenatide. Br J Clin Pharmacol. 2017;83(3):517–526. doi:10.1111/bcp.13135

62. DeYoung MB, MacConell L, Sarin V, Trautmann M, Herbert P. Encapsulation of exenatide in poly-(D,L-lactide-co-glycolide) microspheres produced an investigational long-acting once-weekly formulation for type 2 diabetes. Diabetes Technol Ther. 2011;13(11):1145–1154. doi:10.1089/dia.2011.0050

63. Jacobsen LV, Flint A, Olsen AK, Ingwersen SH. Liraglutide in type 2 diabetes mellitus: clinical pharmacokinetics and pharmacodynamics. Clin Pharmacokinet. 2016;55(6):657–672. doi:10.1007/s40262-015-0343-6

64. Elbrond B, Jakobsen G, Larsen S, et al. Pharmacokinetics, pharmacodynamics, safety, and tolerability of a single-dose of NN2211, a long-acting glucagon-like peptide 1 derivative, in healthy male subjects. Diabetes Care. 2002;25(8):1398–1404. doi:10.2337/diacare.25.8.1398

65. Plum A, Jensen LB, Kristensen JB. In vitro protein binding of liraglutide in human plasma determined by reiterated stepwise equilibrium dialysis. J Pharm Sci. 2013;102(8):2882–2888. doi:10.1002/jps.23648

66. Malm-Erjefalt M, Bjornsdottir I, Vanggaard J, et al. Metabolism and excretion of the once-daily human glucagon-like peptide-1 analog liraglutide in healthy male subjects and its in vitro degradation by dipeptidyl peptidase IV and neutral endopeptidase. Drug Metab Dispos. 2010;38(11):1944–1953. doi:10.1124/dmd.110.034066

67. Burness CB, Scott LJ. Dulaglutide: a review in type 2 diabetes. BioDrugs. 2015;29(6):407–418. doi:10.1007/s40259-015-0143-4

68. Overgaard RV, Navarria A, Ingwersen SH, Baekdal TA, Kildemoes RJ. Clinical pharmacokinetics of oral semaglutide: analyses of data from clinical pharmacology trials. Clin Pharmacokinet. 2021;60(10):1335–1348. doi:10.1007/s40262-021-01025-x

69. Mahato RI, Narang AS, Thoma L, Miller DD. Emerging trends in oral delivery of peptide and protein drugs. Crit Rev Ther Drug Carrier Syst. 2003;20(2–3):153–214. doi:10.1615/critrevtherdrugcarriersyst.v20.i23.30

70. Lau J, Bloch P, Schaffer L, et al. Discovery of the once-weekly Glucagon-Like Peptide-1 (GLP-1) analogue semaglutide. J Med Chem. 2015;58(18):7370–7380. doi:10.1021/acs.jmedchem.5b00726

71. Deacon CF, Knudsen LB, Madsen K, Wiberg FC, Jacobsen O, Holst JJ. Dipeptidyl peptidase IV resistant analogues of glucagon-like peptide-1 which have extended metabolic stability and improved biological activity. Diabetologia. 1998;41(3):271–278. doi:10.1007/s001250050903

72. Jensen L, Helleberg H, Roffel A, et al. Absorption, metabolism and excretion of the GLP-1 analogue semaglutide in humans and nonclinical species. Eur J Pharm Sci. 2017;104:31–41. doi:10.1016/j.ejps.2017.03.020

73. Schneck K, Urva S. Population pharmacokinetics of the GIP/GLP receptor agonist tirzepatide. CPT Pharmacometrics Syst Pharmacol. 2024;13(3):494–503. doi:10.1002/psp4.13099

74. Coskun T, Sloop KW, Loghin C, et al. LY3298176, a novel dual GIP and GLP-1 receptor agonist for the treatment of type 2 diabetes mellitus: from discovery to clinical proof of concept. Mol Metab. 2018;18:3–14. doi:10.1016/j.molmet.2018.09.009

75. Furihata K, Mimura H, Urva S, Oura T, Ohwaki K, Imaoka T. A Phase 1 multiple-ascending dose study of tirzepatide in Japanese participants with type 2 diabetes. Diabetes Obes Metab. 2022;24(2):239–246. doi:10.1111/dom.14572

76. Syed YY. Tirzepatide: first Approval. Drugs. 2022;82(11):1213–1220. doi:10.1007/s40265-022-01746-8

77. Al-Horani RA, Chedid M. Tirzepatide: a new generation therapeutic for diabetes type 2. Endocr Metab Immune Disord Drug Targets. 2023;23(8):1046–1050. doi:10.2174/1871530322666221004151212

78. Mayo KE, Miller LJ, Bataille D, et al. International Union of Pharmacology. XXXV. The glucagon receptor family. Pharmacol Rev. 2003;55(1):167–194. doi:10.1124/pr.55.1.6

79. Korner M, Stockli M, Waser B, Reubi JC. GLP-1 receptor expression in human tumors and human normal tissues: potential for in vivo targeting. J Nucl Med. 2007;48(5):736–743. doi:10.2967/jnumed.106.038679

80. Oh YS, Jun HS. Effects of glucagon-like peptide-1 on oxidative stress and Nrf2 signaling. Int J Mol Sci. 2017;19(1). doi:10.3390/ijms19010026

81. Song G, Yang D, Wang Y, et al. Human GLP-1 receptor transmembrane domain structure in complex with allosteric modulators. Nature. 2017;546(7657):312–315. doi:10.1038/nature22378

82. Baggio LL, Yusta B, Mulvihill EE, et al. GLP-1 receptor expression within the human heart. Endocrinology. 2018;159(4):1570–1584. doi:10.1210/en.2018-00004

83. Wallner M, Kolesnik E, Ablasser K, et al. Exenatide exerts a PKA-dependent positive inotropic effect in human atrial myocardium: GLP-1R mediated effects in human myocardium. J Mol Cell Cardiol. 2015;89(Pt B):365–375. doi:10.1016/j.yjmcc.2015.09.018

84. Smit FX, van der Velden WJC, Kizilkaya HS, et al. Investigating GIPR (ant)agonism: a structural analysis of GIP and its receptor. Structure. 2021;29(7):679–693e6. doi:10.1016/j.str.2021.04.001

85. Usdin TB, Mezey E, Button DC, Brownstein MJ, Bonner TI. Gastric inhibitory polypeptide receptor, a member of the secretin-vasoactive intestinal peptide receptor family, is widely distributed in peripheral organs and the brain. Endocrinology. 1993;133(6):2861–2870. doi:10.1210/endo.133.6.8243312

86. Drucker DJ. Mechanisms of action and therapeutic application of Glucagon-like peptide-1. Cell Metab. 2018;27(4):740–756. doi:10.1016/j.cmet.2018.03.001

87. Cornell S. A review of GLP-1 receptor agonists in type 2 diabetes: a focus on the mechanism of action of once-weekly agents. J Clin Pharm Ther. 2020;45(Suppl 1):17–27. doi:10.1111/jcpt.13230

88. 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. doi:10.1210/jc.2010-2081

89. Buse JB, Rosenstock J, Sesti G, et al. Liraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6). Lancet. 2009;374(9683):39–47. doi:10.1016/S0140-6736(09)60659-0

90. Pi-Sunyer X, Astrup A, Fujioka K, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med. 2015;373(1):11–22. doi:10.1056/NEJMoa1411892

91. Dungan KM, Povedano ST, Forst T, et al. Once-weekly dulaglutide versus once-daily liraglutide in metformin-treated patients with type 2 diabetes (AWARD-6): a randomised, open-label, phase 3, non-inferiority trial. Lancet. 2014;384(9951):1349–1357. doi:10.1016/S0140-6736(14)60976-4

92. 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. doi:10.1016/S2213-8587(17)30013-X

93. 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. doi:10.2337/dc19-0749

94. Wilding JPH, Batterham RL, Calanna S, et al. Once-weekly semaglutide in adults with overweight or obesity. N Engl J Med. 2021;384(11):989–1002. doi:10.1056/NEJMoa2032183

95. Rosenstock J, Wysham C, Frias JP, et al. Efficacy and safety of a novel dual GIP and GLP-1 receptor agonist tirzepatide in patients with type 2 diabetes (SURPASS-1): a double-blind, randomised, phase 3 trial. Lancet. 2021;398(10295):143–155. doi:10.1016/S0140-6736(21)01324-6

96. Jastreboff AM, Aronne LJ, Ahmad NN, et al. Tirzepatide once weekly for the treatment of obesity. N Engl J Med. 2022;387(3):205–216. doi:10.1056/NEJMoa2206038

97. 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. doi:10.1056/NEJMoa1603827

98. 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. doi:10.1056/NEJMoa1612917

99. Mosterd CM, Bjornstad P, van Raalte DH. Nephroprotective effects of GLP-1 receptor agonists: where do we stand? J Nephrol. 2020;33(5):965–975. doi:10.1007/s40620-020-00738-9

100. Bendotti G, Montefusco L, Lunati ME, et al. The anti-inflammatory and immunological properties of GLP-1 Receptor Agonists. Pharmacol Res. 2022;182:106320. doi:10.1016/j.phrs.2022.106320

101. Mehdi SF, Pusapati S, Anwar MS, et al. Glucagon-like peptide-1: a multi-faceted anti-inflammatory agent. Front Immunol. 2023;14:1148209. doi:10.3389/fimmu.2023.1148209

102. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–867. doi:10.1038/nature05485

103. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006;368(9548):1696–1705. doi:10.1016/S0140-6736(06)69705-5

104. Klein KR, Clemmensen KKB, Fong E, Olsen S, Abrahamsen T, Lingvay I. Occurrence of gastrointestinal adverse events upon GLP-1 receptor agonist initiation with concomitant metformin use: a post hoc analysis of LEADER, STEP 2, SUSTAIN-6, and PIONEER 6. Diabetes Care. 2024;47(2):280–284. doi:10.2337/dc23-1791

105. Sodhi M, Rezaeianzadeh R, Kezouh A, Etminan M. Risk of gastrointestinal adverse events associated with glucagon-like peptide-1 receptor agonists for weight loss. JAMA. 2023;330(18):1795–1797. doi:10.1001/jama.2023.19574

106. Rubino DM, Greenway FL, Khalid U, et al. Effect of weekly subcutaneous semaglutide vs daily liraglutide on body weight in adults with overweight or obesity without diabetes: the STEP 8 randomized clinical trial. JAMA. 2022;327(2):138–150. doi:10.1001/jama.2021.23619

107. Kapoor I, Sarvepalli SM, D’Alessio D, Grewal DS, Hadziahmetovic M. GLP-1 receptor agonists and diabetic retinopathy: a meta-analysis of randomized clinical trials. Surv Ophthalmol. 2023;68(6):1071–1083. doi:10.1016/j.survophthal.2023.07.002

108. Liu L, Chen J, Wang L, Chen C, Chen L. Association between different GLP-1 receptor agonists and gastrointestinal adverse reactions: a real-world disproportionality study based on FDA adverse event reporting system database. Front Endocrinol. 2022;13:1043789. doi:10.3389/fendo.2022.1043789

109. He L, Li Q, Yang Y, et al. Pharmacovigilance study of GLP-1 receptor agonists for metabolic and nutritional adverse events. Front Pharmacol. 2024;15:1416985. doi:10.3389/fphar.2024.1416985

110. Walker DK. The use of pharmacokinetic and pharmacodynamic data in the assessment of drug safety in early drug development. Br J Clin Pharmacol. 2004;58(6):601–608. doi:10.1111/j.1365-2125.2004.02194.x

111. United State Food and Drug Administration, Center for Drug Evaluation and Research. Guidance for industry: exposure-response relationship-study design, data analysis, and regulatory applications. 2003.

112. Zhou Y, Chen M, Liu L, Chen Z. Difference in gastrointestinal risk associated with use of GLP-1 receptor agonists: a real-world pharmacovigilance study. Diabetes Metab Syndr Obes. 2022;15:155–163. doi:10.2147/DMSO.S348025

113. Nauck MA, Kemmeries G, Holst JJ, Meier JJ. Rapid tachyphylaxis of the glucagon-like peptide 1-induced deceleration of gastric emptying in humans. Diabetes. 2011;60(5):1561–1565. doi:10.2337/db10-0474

114. Luo ZY, Li X, Chen CT, et al. Ocular adverse events associated with GLP-1 receptor agonists: a real-world study based on the FAERS database and network pharmacology. Expert Opin Drug Saf. 2024:1–10. doi:10.1080/14740338.2024.2419989

115. Guo H, Guo Q, Li Z, Wang Z. Association between different GLP-1 receptor agonists and acute pancreatitis: case series and real-world pharmacovigilance analysis. Front Pharmacol. 2024;15:1461398. doi:10.3389/fphar.2024.1461398

116. Makunts T, Joulfayan H, Abagyan R. Thyroid hyperplasia and neoplasm adverse events associated with GLP-1 receptor agonists in FDA adverse event reporting system. medRxiv. 2023. doi:10.1101/2023.11.19.23298750

117. Wu T, Zhang Y, Shi Y, et al. Safety of glucagon-like peptide-1 receptor agonists: a real-world study based on the US FDA adverse event reporting system database. Clin Drug Investig. 2022;42(11):965–975. doi:10.1007/s40261-022-01202-1

118. Holst JJ. From the incretin concept and the discovery of GLP-1 to today’s diabetes therapy. Front Endocrinol. 2019;10:260. doi:10.3389/fendo.2019.00260

119. Garber AJ. Long-acting glucagon-like peptide 1 receptor agonists: a review of their efficacy and tolerability. Diabetes Care. 2011;34(Suppl 2):S279–84. doi:10.2337/dc11-s231

120. United State Food and Drug Administration, center for drug evaluation and research. Guidance for Industry: clinical Pharmacology considerations for peptide drug products. 2023. Available from: https://www.fda.gov/media/171901/download. Accessed March 3, 2024.

121. Glucagon-Like Peptide-1 (GLP-1) Analogues. LiverTox: clinical and research information on drug-induced liver injury. 2012.

122. Neumiller JJ. Clinical pharmacology of incretin therapies for type 2 diabetes mellitus: implications for treatment. Clin Ther. 2011;33(5):528–576. doi:10.1016/j.clinthera.2011.04.024

123. Jyrkas J, Tolonen A. Hepatic in vitro metabolism of peptides; Comparison of human liver S9, hepatocytes and Upcyte hepatocytes with cyclosporine A, leuprorelin, desmopressin and cetrorelix as model compounds. J Pharm Biomed Anal. 2021;196:113921. doi:10.1016/j.jpba.2021.113921

124. Calvarysky B, Dotan I, Shepshelovich D, Leader A, Cohen TD. Drug-drug interactions between glucagon-like peptide 1 receptor agonists and oral medications: a systematic review. Drug Saf. 2024;47(5):439–451. doi:10.1007/s40264-023-01392-3

125. Iber H, Li-Masters T, Chen Q, Yu S, Morgan ET. Regulation of hepatic cytochrome P450 2C11 via cAMP: implications for down-regulation in diabetes, fasting, and inflammation. J Pharmacol Exp Ther. 2001;297(1):174–180.

126. Liddle C, Goodwin BJ, George J, Tapner M, Farrell GC. Separate and interactive regulation of cytochrome P450 3A4 by triiodothyronine, dexamethasone, and growth hormone in cultured hepatocytes. J Clin Endocrinol Metab. 1998;83(7):2411–2416. doi:10.1210/jcem.83.7.4877

127. Yilmaz B, Kemal Y, Teker F. Be careful before prescribing warfarin and octreotide together: a new drug-drug interaction report. Hippokratia. 2014;18(4):377.

128. Stevenson W, Chauhan SK, Dana R. Dry eye disease: an immune-mediated ocular surface disorder. Arch Ophthalmol. 2012;130(1):90–100. doi:10.1001/archophthalmol.2011.364

129. Lechner C, Reichel V, Moenning U, Reichel A, Fricker G. Development of a fluorescence-based assay for drug interactions with human Multidrug Resistance Related Protein (MRP2; ABCC2) in MDCKII-MRP2 membrane vesicles. Eur J Pharm Biopharm. 2010;75(2):284–290. doi:10.1016/j.ejpb.2010.03.008

130. Shitara Y. Clinical importance of OATP1B1 and OATP1B3 in drug-drug interactions. Drug Metab Pharmacokinet. 2011;26(3):220–227. doi:10.2133/dmpk.DMPK-10-RV-094

131. Qadir M, O’Loughlin KL, Fricke SM, et al. Cyclosporin A is a broad-spectrum multidrug resistance modulator. Clin Cancer Res. 2005;11(6):2320–2326. doi:10.1158/1078-0432.CCR-04-1725

132. Niwa T, Yamamoto S, Saito M, Shiraga T, Takagi A. Effect of cyclosporine and tacrolimus on cytochrome p450 activities in human liver microsomes. Yakugaku Zasshi. 2007;127(1):209–216. doi:10.1248/yakushi.127.209

133. Lemahieu WP, Hermann M, Asberg A, et al. Combined therapy with atorvastatin and calcineurin inhibitors: no interactions with tacrolimus. Am J Transplant. 2005;5(9):2236–2243. doi:10.1111/j.1600-6143.2005.01005.x

134. Ishikawa R, Saito K, Misawa T, Demizu Y, Saito Y. Identification of the stapled alpha-helical peptide ATSP-7041 as a substrate and strong inhibitor of OATP1B1 in vitro. Biomolecules. 2023;13(6). doi:10.3390/biom13061002

135. Saleh MN, Patel MR, Bauer TM, et al. Phase 1 trial of ALRN-6924, a dual inhibitor of MDMX and MDM2, in patients with solid tumors and lymphomas bearing wild-type TP53. Clin Cancer Res. 2021;27(19):5236–5247. doi:10.1158/1078-0432.CCR-21-0715

136. Ishiguro N, Maeda K, Kishimoto W, et al. Predominant contribution of OATP1B3 to the hepatic uptake of telmisartan, an angiotensin II receptor antagonist, in humans. Drug Metab Dispos. 2006;34(7):1109–1115. doi:10.1124/dmd.105.009175

137. Indu R, Adhikari A, Maisnam I, Basak P, Sur TK, Das AK. Polypharmacy and comorbidity status in the treatment of type 2 diabetic patients attending a tertiary care hospital: an observational and questionnaire-based study. Perspect Clin Res. 2018;9(3):139–144. doi:10.4103/picr.PICR_81_17

138. Hamilton RA, Briceland LL, Andritz MH. Frequency of hospitalization after exposure to known drug-drug interactions in a Medicaid population. Pharmacotherapy. 1998;18(5):1112–1120.

139. United State Food and Drug Administration. Clinical pharmacology biopharmaceutics review: Wegovy®. 2020. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2021/215256Orig1s000ClinPharmR.pdf. Accessed June 24, 2024.

140. Jordy AB, Albayaty M, Breitschaft A, et al. Effect of oral semaglutide on the pharmacokinetics of levonorgestrel and ethinylestradiol in healthy postmenopausal women and furosemide and rosuvastatin in healthy subjects. Clin Pharmacokinet. 2021;60(9):1171–1185. doi:10.1007/s40262-020-00976-x

141. Xie P, Abildlund MT, Baekdal TA, et al. A phase 1, randomized, double-blind, placebo-controlled trial investigating the pharmacokinetics, pharmacodynamics, safety and tolerability of oral semaglutide in healthy Chinese subjects. Diabetes Obes Metab. 2024;26(8):3068–3077. doi:10.1111/dom.15624

142. Maselli DB, Camilleri M. Effects of GLP-1 and its analogs on gastric physiology in diabetes mellitus and obesity. Adv Exp Med Biol. 2021;1307:171–192. doi:10.1007/5584_2020_496

143. Hurren KM, Pinelli NR. Drug-drug interactions with glucagon-like peptide-1 receptor agonists. Ann Pharmacother. 2012;46(5):710–717. doi:10.1345/aph.1Q583

144. European Medicines Agency. Guideline on the investigation of drug interactions. 2012. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-investigation-drug-interactions-revision-1_en.pdf. Accessed Feb 2, 2025.

145. United State Food and Drug Administration, center for drug evaluation and research. Guidance for Industry: drug interaction studies–study design, data analysis, implications for dosing, and labeling recommendations. 2012. Available from: https://www.fda.gov/media/171901/download. Accessed March 3, 2024.

146. United State Food and Drug Administration, Center for Drug Evaluation and Research. M12 drug interaction studies guidance for industry. 2024. Available from: https://www.fda.gov/media/161199/download. Accessed March 10, 2025.

147. Kothare PA, Linnebjerg H, Skrivanek Z, et al. Exenatide effects on statin pharmacokinetics and lipid response. Int J Clin Pharmacol Ther. 2007;45(2):114–120. doi:10.5414/cpp45114

148. Kothare PA, Seger ME, Northrup J, Mace K, Mitchell MI, Linnebjerg H. Effect of exenatide on the pharmacokinetics of a combination oral contraceptive in healthy women: an open-label, randomised, crossover trial. BMC Clin Pharmacol. 2012;12:8. doi:10.1186/1472-6904-12-8

149. Kothare PA, Soon DK, Linnebjerg H, et al. Effect of exenatide on the steady-state pharmacokinetics of digoxin. J Clin Pharmacol. 2005;45(9):1032–1037. doi:10.1177/0091270005278806

150. Blase E, Taylor K, Gao HY, Wintle M, Fineman M. Pharmacokinetics of an oral drug (Acetaminophen) administered at various times in relation to subcutaneous injection of exenatide (exendin-4) in healthy subjects. J Clin Pharmacol. 2005;45(5):570–577. doi:10.1177/0091270004274432

151. Soon D, Kothare PA, Linnebjerg H, et al. Effect of exenatide on the pharmacokinetics and pharmacodynamics of warfarin in healthy Asian men. J Clin Pharmacol. 2006;46(10):1179–1187. doi:10.1177/0091270006291622

152. Linnebjerg H, Kothare P, Park S, Mace K, Mitchell M. The effect of exenatide on lisinopril pharmacodynamics and pharmacokinetics in patients with hypertension. Int J Clin Pharmacol Ther. 2009;47(11):651–658. doi:10.5414/cpp47651

153. Malm-Erjefalt M, Ekblom M, Vouis J, Zdravkovic M, Lennernas H. Effect on the gastrointestinal absorption of drugs from different classes in the biopharmaceutics classification system, when treating with liraglutide. Mol Pharm. 2015;12(11):4166–4173. doi:10.1021/acs.molpharmaceut.5b00278

154. Kapitza C, Zdravkovic M, Hindsberger C, Flint A. The effect of the once-daily human glucagon-like peptide 1 analog liraglutide on the pharmacokinetics of Acetaminophen. Adv Ther. 2011;28(8):650–660. doi:10.1007/s12325-011-0044-y

155. de la Pena A, Cui X, Geiser J, Loghin C. Dose adjustment is recommended for digoxin, warfarin, atorvastatin or a combination oral contraceptive when coadministered with dulaglutide. Clin Pharmacokinet. 2017;56(11):1415–1427. doi:10.1007/s40262-017-0531-7

156. Morse BL, Alberts JJ, Posada MM, et al. Physiologically-based pharmacokinetic modeling of atorvastatin incorporating delayed gastric emptying and acid-to-lactone conversion. CPT Pharmacometrics Syst Pharmacol. 2019;8(9):664–675. doi:10.1002/psp4.12447

157. Kapitza C, Nosek L, Jensen L, Hartvig H, Jensen CB, Flint A. Semaglutide, a once-weekly human GLP-1 analog, does not reduce the bioavailability of the combined oral contraceptive, ethinylestradiol/levonorgestrel. J Clin Pharmacol. 2015;55(5):497–504. doi:10.1002/jcph.443

158. Baekdal 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(9):1193–1203. doi:10.1007/s40262-019-00756-2

159. Hausner H, Derving Karsbol J, Holst AG, et al. Effect of semaglutide on the pharmacokinetics of metformin, warfarin, atorvastatin and digoxin in healthy subjects. Clin Pharmacokinet. 2017;56(11):1391–1401. doi:10.1007/s40262-017-0532-6

160. Hauge C, Breitschaft A, Hartoft-Nielsen ML, Jensen S, Baekdal TA. Effect of oral semaglutide on the pharmacokinetics of thyroxine after dosing of levothyroxine and the influence of co-administered tablets on the pharmacokinetics of oral semaglutide in healthy subjects: an open-label, one-sequence crossover, single-center, multiple-dose, two-part trial. Expert Opin Drug Metab Toxicol. 2021;17(9):1139–1148. doi:10.1080/17425255.2021.1955856

161. Skelley JW, Swearengin K, York AL, Glover LH. The impact of tirzepatide and glucagon-like peptide 1 receptor agonists on oral hormonal contraception. J Am Pharm Assoc. 2024;64(1):204–211e4. doi:10.1016/j.japh.2023.10.037

162. Langeskov EK, Kristensen K. Population pharmacokinetic of paracetamol and atorvastatin with co-administration of semaglutide. Pharmacol Res Perspect. 2022;10(4):e00962. doi:10.1002/prp2.962

163. Jacobsen LV, Vouis J, Hindsberger C, Zdravkovic M. Treatment with liraglutide--a once-daily GLP-1 analog--does not reduce the bioavailability of ethinyl estradiol/levonorgestrel taken as an oral combination contraceptive drug. J Clin Pharmacol. 2011;51(12):1696–1703. doi:10.1177/0091270010389471

164. Shitara Y, Sugiyama Y. Pharmacokinetic and pharmacodynamic alterations of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors: drug-drug interactions and interindividual differences in transporter and metabolic enzyme functions. Pharmacol Ther. 2006;112(1):71–105. doi:10.1016/j.pharmthera.2006.03.003

165. Cooper DB, Patel P. Oral Contraceptive Pills. StatPearls; 2024.

166. Li L, Yang X, Tran D, Seo SK, Lu Y. Combined oral contraceptives as victims of drug interactions. Drug Metab Dispos. 2023;51(6):718–732. doi:10.1124/dmd.122.000854

167. Sun H, Sivasubramanian R, Vaidya S, Barve A, Jarugula V. Drug-drug interaction studies with oral contraceptives: pharmacokinetic/pharmacodynamic and study design considerations. J Clin Pharmacol. 2020;60(Suppl 2):S49–S62. doi:10.1002/jcph.1765

168. Maideen NMP. Pharmacologically relevant drug interactions of Glucagon-like peptide-1 receptor agonists. J Anal Pharm Res. 2019;8(2):51–53.

169. Urva S, Coskun T, Loghin C, et al. The novel dual glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1 (GLP-1) receptor agonist tirzepatide transiently delays gastric emptying similarly to selective long-acting GLP-1 receptor agonists. Diabetes Obes Metab. 2020;22(10):1886–1891. doi:10.1111/dom.14110

170. Wartofsky L. Levothyroxine: therapeutic use and regulatory issues related to bioequivalence. Expert Opin Pharmacother. 2002;3(6):727–732. doi:10.1517/14656566.3.6.727

171. Benvenga S, Bartolone L, Squadrito S, Lo Giudice F, Trimarchi F. Delayed intestinal absorption of levothyroxine. Thyroid. 1995;5(4):249–253. doi:10.1089/thy.1995.5.249

172. Virili C, Antonelli A, Santaguida MG, Benvenga S, Centanni M. Gastrointestinal Malabsorption of Thyroxine. Endocr Rev. 2019;40(1):118–136. doi:10.1210/er.2018-00168

173. Jonklaas J, Bianco AC, Bauer AJ, et al. Guidelines for the treatment of hypothyroidism: prepared by the American thyroid association task force on thyroid hormone replacement. Thyroid. 2014;24(12):1670–1751. doi:10.1089/thy.2014.0028

174. De Block C, Bailey C, Wysham C, Hemmingway A, Allen SE, Peleshok J. Tirzepatide for the treatment of adults with type 2 diabetes: an endocrine perspective. Diabetes Obes Metab. 2023;25(1):3–17. doi:10.1111/dom.14831

175. Raven LM, Stoita A, Feller RB, Brown C, Greenfield JR. Delayed gastric emptying with perioperative use of glucagon-like peptide-1 receptor agonists. Am J Med. 2023;136(12):e233–e234. doi:10.1016/j.amjmed.2023.07.016

176. United State Food and Drug Administration. Clinical pharmacology biopharmaceutics review: exenatide extended-release. 2017. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2005/021773_Byetta_biopharmr.PDF. Accessed June 24, 2024.

177. Watson E, Jonker DM, Jacobsen LV, Ingwersen SH. Population pharmacokinetics of liraglutide, a once-daily human glucagon-like peptide-1 analog, in healthy volunteers and subjects with type 2 diabetes, and comparison to twice-daily exenatide. J Clin Pharmacol. 2010;50(8):886–894. doi:10.1177/0091270009354996

178. Chen T, Mager DE, Kagan L. Interspecies modeling and prediction of human exenatide pharmacokinetics. Pharm Res. 2013;30(3):751–760. doi:10.1007/s11095-012-0917-z

179. Voronova V, Zhudenkov K, Penland RC, Boulton DW, Helmlinger G, Peskov K. Exenatide effects on gastric emptying rate and the glucose rate of appearance in plasma: a quantitative assessment using an integrative systems pharmacology model. Diabetes Obes Metab. 2018;20(8):2034–2038. doi:10.1111/dom.13326

180. Elkeeb R, Eid T, Yu J, Nguyen H, Atef E. Can a monthly exenatide extended release regimen provide a therapeutic and cost benefit? Biopharm Drug Dispos. 2021;42(6):245–251. doi:10.1002/bdd.2279

181. Cirincione B, Edwards J, Mager DE. Population pharmacokinetics of an extended-release formulation of exenatide following single- and multiple-dose administration. AAPS J. 2017;19(2):487–496. doi:10.1208/s12248-016-9975-1

182. Overgaard RV, Lindberg SO, Thielke D. Impact on HbA1c and body weight of switching from other GLP-1 receptor agonists to semaglutide: a model-based approach. Diabetes Obes Metab. 2019;21(1):43–51. doi:10.1111/dom.13479

183. Li H, Xu J, Fan X. Target-mediated pharmacokinetic/pharmacodynamic model based meta-analysis and dosing regimen optimization of a long-acting release formulation of exenatide in patients with type 2 diabetes mellitus. J Pharmacol Sci. 2015;127(2):170–180. doi:10.1016/j.jphs.2014.12.004

184. Cirincione B, LaCreta F, Sager P, Mager DE. Model-based evaluation of exenatide effects on the QT interval in healthy subjects following continuous IV infusion. J Clin Pharmacol. 2017;57(8):956–965. doi:10.1002/jcph.882

185. Machado TR, Honorio T, Souza Domingos TF, et al. Physiologically based pharmacokinetic modelling of semaglutide in children and adolescents with healthy and obese body weights. Br J Clin Pharmacol. 2023;89(10):3175–3194. doi:10.1111/bcp.15816

186. Petri KCC, Ingwersen SH, Flint A, Zacho J, Overgaard RV. Exposure-response analysis for evaluation of semaglutide dose levels in type 2 diabetes. Diabetes Obes Metab. 2018;20(9):2238–2245. doi:10.1111/dom.13358

187. Overgaard RV, Delff PH, Petri KCC, Anderson TW, Flint A, Ingwersen SH. Population pharmacokinetics of semaglutide for type 2 diabetes. Diabetes Ther. 2019;10(2):649–662. doi:10.1007/s13300-019-0581-y

188. Strathe A, Horn DB, Larsen MS, et al. A model-based approach to predict individual weight loss with semaglutide in people with overweight or obesity. Diabetes Obes Metab. 2023;25(11):3171–3180. doi:10.1111/dom.15211

189. Tham LS, Pantalone KM, Dungan K, et al. A model-based simulation of glycaemic control and body weight when switching from semaglutide to 3.0- and 4.5-mg doses of once-weekly dulaglutide. Diabetes Obes Metab. 2022;24(2):302–311. doi:10.1111/dom.14582

190. Petri KC, Jacobsen LV, Klein DJ. Comparable liraglutide pharmacokinetics in pediatric and adult populations with type 2 diabetes: a population pharmacokinetic analysis. Clin Pharmacokinet. 2015;54(6):663–670. doi:10.1007/s40262-014-0229-z

191. Overgaard RV, Petri KC, Jacobsen LV, Jensen CB. Liraglutide 3.0 mg for weight management: a population pharmacokinetic analysis. Clin Pharmacokinet. 2016;55(11):1413–1422. doi:10.1007/s40262-016-0410-7

192. Papathanasiou T, Strathe A, Agerso H, Lund TM, Overgaard RV. Impact of dose-escalation schemes and drug discontinuation on weight loss outcomes with liraglutide 3.0 mg: a model-based approach. Diabetes Obes Metab. 2020;22(6):969–977. doi:10.1111/dom.13985

193. Geiser JS, Heathman MA, Cui X, et al. Clinical pharmacokinetics of dulaglutide in patients with type 2 diabetes: analyses of data from clinical trials. Clin Pharmacokinet. 2016;55(5):625–634. doi:10.1007/s40262-015-0338-3

194. Tang CC, Lim J, Loo LS, Jung H, Konig M, Tham LS. Practical applications of a nausea and vomiting model in the clinical development of additional doses of dulaglutide. J Clin Pharmacol. 2024;64(2):215–226. doi:10.1002/jcph.2373

195. Guan R, Li X, Ma G. Prediction of pediatric dose of tirzepatide from the reference adult dose using physiologically based pharmacokinetic modelling. Front Pharmacol. 2023;14:1326373. doi:10.3389/fphar.2023.1326373

196. Tham LSSK, Geiser JS, Posada MM, Dickinson GL. Integration of population exposure-response and physiological based pharmacokinetic modeling approaches to evaluate gastric-emptying induced drug interaction risks for dulaglutide. 2018.

197. Yoon KH, Kang J, Kwon SC, et al. Pharmacokinetic and dose-finding studies on efpeglenatide in patients with type 2 diabetes. Diabetes Obes Metab. 2020;22(8):1292–1301. doi:10.1111/dom.14032

198. Sharma D, Verma S, Vaidya S, Kalia K, Tiwari V. Recent updates on GLP-1 agonists: current advancements & challenges. Biomed Pharmacother. 2018;108:952–962. doi:10.1016/j.biopha.2018.08.088

199. Wharton S, Blevins T, Connery L, et al. Daily oral GLP-1 receptor agonist orforglipron for adults with obesity. N Engl J Med. 2023;389(10):877–888. doi:10.1056/NEJMoa2302392

200. Newsome PN, Buchholtz K, Cusi K, et al. A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. N Engl J Med. 2021;384(12):1113–1124. doi:10.1056/NEJMoa2028395

201. Armstrong MJ, Barton D, Gaunt P, et al. Liraglutide efficacy and action in non-alcoholic steatohepatitis (LEAN): study protocol for a phase II multicentre, double-blinded, randomised, controlled trial. BMJ Open. 2013;3(11):e003995. doi:10.1136/bmjopen-2013-003995

202. Knerr PJ, Mowery SA, Douros JD, et al. Next generation GLP-1/GIP/glucagon triple agonists normalize body weight in obese mice. Mol Metab. 2022;63:101533. doi:10.1016/j.molmet.2022.101533

203. Abdelmalek MF, Suzuki A, Sanchez W, et al. A phase 2, adaptive randomized, double-blind, placebo-controlled, multicenter, 52-week study of HM15211 in patients with biopsy-confirmed non-alcoholic steatohepatitis - Study design and rationale of HM-TRIA-201 study. Contemp Clin Trials. 2023;130:107176. doi:10.1016/j.cct.2023.107176

204. Sanyal AJ, Bedossa P, Fraessdorf M, et al. A Phase 2 Randomized Trial of Survodutide in MASH and Fibrosis. N Engl J Med. 2024;391(4):311–319. doi:10.1056/NEJMoa2401755

205. Jastreboff AM, Kaplan LM, Frias JP, et al. Triple-hormone-receptor agonist retatrutide for obesity—a phase 2 trial. N Engl J Med. 2023;389(6):514–526. doi:10.1056/NEJMoa2301972

206. Ji L, Jiang H, Cheng Z, et al. A phase 2 randomised controlled trial of mazdutide in Chinese overweight adults or adults with obesity. Nat Commun. 2023;14(1):8289. doi:10.1038/s41467-023-44067-4

207. Sall C, Alifrangis L, Dahl K, Friedrichsen MH, Nygard SB, Kristensen K. In vitro CYP450 enzyme down-regulation by GLP-1/glucagon co-agonist does not translate to observed drug-drug interactions in the clinic. Drug Metab Dispos. 2022. doi:10.1124/dmd.122.000865

208. Simonsen L, Lau J, Kruse T, et al. Preclinical evaluation of a protracted GLP-1/glucagon receptor co-agonist: translational difficulties and pitfalls. PLoS One. 2022;17(3):e0264974. doi:10.1371/journal.pone.0264974

209. Norgaard RA, Bhatt DK, Jarvinen E, et al. Evaluating drug-drug interaction risk associated with peptide analogues using advanced in vitro systems. Drug Metab Dispos. 2023. doi:10.1124/dmd.123.001441

210. Gouju J, Legeay S. Pharmacokinetics of obese adults: not only an increase in weight. Biomed Pharmacother. 2023;166:115281. doi:10.1016/j.biopha.2023.115281

211. Kvitne KE, Robertsen I, Skovlund E, et al. Short- and long-term effects of body weight loss following calorie restriction and gastric bypass on CYP3A-activity - a non-randomized three-armed controlled trial. Clin Transl Sci. 2022;15(1):221–233. doi:10.1111/cts.13142

212. Cheymol G. Clinical pharmacokinetics of drugs in obesity. An update. Clin Pharmacokinet. 1993;25(2):103–114. doi:10.2165/00003088-199325020-00003

213. Aitken AE, Morgan ET. Gene-specific effects of inflammatory cytokines on cytochrome P450 2C, 2B6 and 3A4 mRNA levels in human hepatocytes. Drug Metab Dispos. 2007;35(9):1687–1693. doi:10.1124/dmd.107.015511

214. Pan HC, Chen JY, Chen HY, et al. GLP-1 receptor agonists’ impact on cardio-renal outcomes and mortality in T2D with acute kidney disease. Nat Commun. 2024;15(1):5912. doi:10.1038/s41467-024-50199-y

215. Greco EV, Russo G, Giandalia A, Viazzi F, Pontremoli R, De Cosmo S. GLP-1 receptor agonists and kidney protection. Medicina. 2019;55(6). doi:10.3390/medicina55060233

216. Crajoinas RO, Oricchio FT, Pessoa TD, et al. Mechanisms mediating the diuretic and natriuretic actions of the incretin hormone glucagon-like peptide-1. Am J Physiol Renal Physiol. 2011;301(2):F355–63. doi:10.1152/ajprenal.00729.2010

217. Lumholtz B, Kampmann TJ, Siersbaek-Nielsen K, Hansen JM. Dose regimen of kanamycin and gentamicin. Acta Med Scand. 1974;196(6):521–524. doi:10.1111/j.0954-6820.1974.tb01054.x

218. Triggs EJ, Johnson JM, Learoyd B. Absorption and disposition of ampicillin in the elderly. Eur J Clin Pharmacol. 1980;18(2):195–198. doi:10.1007/BF00561590

219. Somogyi A, Hewson D, Muirhead M, Bochner F. Amiloride disposition in geriatric patients: importance of renal function. Br J Clin Pharmacol. 1990;29(1):1–8. doi:10.1111/j.1365-2125.1990.tb03595.x

220. Portnoi VA. Digitalis delirium in elderly patients. J Clin Pharmacol. 1979;19(11–12):747–750. doi:10.1002/j.1552-4604.1979.tb01646.x

221. Oberbauer R, Krivanek P, Turnheim K. Pharmacokinetics of indomethacin in the elderly. Clin Pharmacokinet. 1993;24(5):428–434. doi:10.2165/00003088-199324050-00007

222. Sager JE, Yu J, Ragueneau-Majlessi I, Isoherranen N. Physiologically Based Pharmacokinetic (PBPK) modeling and simulation approaches: a systematic review of published models, applications, and model verification. Drug Metab Dispos. 2015;43(11):1823–1837. doi:10.1124/dmd.115.065920

223. Jones HM, Chen Y, Gibson C, et al. Physiologically based pharmacokinetic modeling in drug discovery and development: a pharmaceutical industry perspective. Clin Pharmacol Ther. 2015;97(3):247–262. doi:10.1002/cpt.37

224. Min JS, Bae SK. Prediction of drug-drug interaction potential using physiologically based pharmacokinetic modeling. Arch Pharm Res. 2017;40(12):1356–1379. doi:10.1007/s12272-017-0976-0

225. Jamei M, Dickinson GL, Rostami-Hodjegan A. A framework for assessing inter-individual variability in pharmacokinetics using virtual human populations and integrating general knowledge of physical chemistry, biology, anatomy, physiology and genetics: a tale of ‘bottom-up’ vs ‘top-down’ recognition of covariates. Drug Metab Pharmacokinet. 2009;24(1):53–75. doi:10.2133/dmpk.24.53

226. Rowland M, Peck C, Tucker G. Physiologically-based pharmacokinetics in drug development and regulatory science. Annu Rev Pharmacol Toxicol. 2011;51:45–73. doi:10.1146/annurev-pharmtox-010510-100540

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 4.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.