Abstract
The global health burden imposed by metabolic disorders, predominantly obesity and type 2 diabetes (T2D), necessitates the continuous evolution of therapeutic paradigms. Traditional pharmacological interventions, while effective to varying degrees, often face limitations regarding comprehensive efficacy, durability, and a favorable safety profile. The advent of multi-receptor agonists, particularly triple agonists targeting the glucagon-like peptide-1 (GLP-1), gastric inhibitory polypeptide (GIP), and glucagon (GCG) receptors, marks a profound leap forward in the treatment landscape. These sophisticated pharmaceutical agents, exemplified by compounds such as retatrutide, harness a synergistic poly-agonistic approach to exert pleiotropic metabolic benefits. This comprehensive report meticulously dissects the intricate molecular mechanisms underpinning the action of each component receptor – GLP-1R, GIPR, and GCGR – detailing their respective signaling cascades and physiological effects. It elucidates how the carefully balanced agonism of the glucagon receptor, traditionally associated with hyperglycemic effects, is strategically leveraged within a triple agonist construct to enhance energy expenditure and facilitate substantial weight reduction without inducing adverse glycemic excursions, a feat largely attributed to the counter-regulatory insulinotropic and glucagonostatic actions of GLP-1 and GIP. Furthermore, this report critically examines the multifaceted engineering challenges inherent in designing single molecular entities capable of potently and selectively activating three distinct G protein-coupled receptors, alongside the significant therapeutic advantages offered by such a multimodal approach. A thorough review of preclinical data establishes the foundational rationale, while an in-depth analysis of advanced clinical trial findings, especially from Phase 2 and ongoing Phase 3 studies for leading candidates like retatrutide, highlights their remarkable efficacy in weight management, glycemic control, and potential amelioration of associated cardiometabolic comorbidities. The report concludes by projecting the transformative potential and future role of these groundbreaking triple agonists in shaping the landscape of metabolic medicine, emphasizing their capacity to offer more comprehensive, efficacious, and durable solutions for patients grappling with these pervasive chronic diseases.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
1. Introduction
The relentless escalation in the global prevalence of metabolic disorders, particularly obesity and type 2 diabetes (T2D), represents one of the most pressing public health crises of the 21st century. Projections indicate that by 2030, over one billion adults worldwide will be living with obesity, and the incidence of T2D continues its upward trajectory, imposing an immense socioeconomic burden [1, 5]. These interconnected conditions significantly heighten the risk of severe comorbidities, including cardiovascular disease, non-alcoholic fatty liver disease (NAFLD)/metabolic dysfunction-associated steatotic liver disease (MASLD), chronic kidney disease, certain cancers, and debilitating musculoskeletal disorders, collectively diminishing quality of life and increasing mortality [6].
Traditional therapeutic strategies for T2D and obesity have ranged from lifestyle modifications and bariatric surgery to a diverse array of pharmacological agents. While drugs targeting single pathways, such as metformin for T2D or orlistat for obesity, have provided foundational treatment, their efficacy often proves suboptimal for many patients, and their long-term use can be limited by adherence issues, adverse side effects, or a plateau in therapeutic benefits. The advent of incretin-based therapies, initially with glucagon-like peptide-1 receptor agonists (GLP-1 RAs) and subsequently with dual GLP-1/GIP receptor agonists (e.g., tirzepatide), heralded a paradigm shift, demonstrating superior efficacy in glycemic control and significant weight loss [7].
Building upon the success of these single and dual agonists, the scientific community has pursued even more sophisticated multi-receptor strategies. The logical progression led to the conceptualization and development of triple agonists, which simultaneously target three key metabolic regulatory pathways: the glucagon-like peptide-1 receptor (GLP-1R), the gastric inhibitory polypeptide receptor (GIPR), and the glucagon receptor (GCGR). This report provides an exhaustive analysis of these innovative agents, focusing on their intricate molecular mechanisms, the sophisticated engineering required for their development, their profound therapeutic potential as evidenced by preclinical and advanced clinical data, and their anticipated transformative impact on metabolic medicine. The emphasis will be on dissecting the synergistic interplay of these three hormonal pathways and how their simultaneous modulation offers a more holistic and potent approach to managing obesity and T2D.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
2. Molecular Mechanisms of Triple Agonists
The efficacy of triple agonists stems from their ability to simultaneously engage three distinct, yet interconnected, G protein-coupled receptors (GPCRs) that play pivotal roles in metabolic homeostasis. A detailed understanding of each receptor’s signaling cascade and physiological impact is crucial to appreciate the synergistic benefits of a multi-agonistic approach.
2.1 GLP-1 Receptor Agonism
Glucagon-like peptide-1 (GLP-1) is a 30-amino acid incretin hormone synthesized and secreted by enteroendocrine L-cells in the intestine, primarily in response to nutrient ingestion [1]. Its release is tightly regulated, contributing significantly to postprandial glucose homeostasis. The GLP-1 receptor (GLP-1R) is a Class B GPCR expressed in various tissues, with particularly high concentrations on pancreatic β-cells, neurons in the hypothalamus and brainstem, gastric chief cells, and cardiac myocytes [1, 8].
Upon binding of an agonist to GLP-1R, the receptor undergoes a conformational change that activates heterotrimeric Gs proteins. This activation subsequently stimulates adenylyl cyclase, leading to a rapid and substantial increase in intracellular cyclic adenosine monophosphate (cAMP) levels. Elevated cAMP then activates two primary downstream effectors: protein kinase A (PKA) and exchange protein directly activated by cAMP 2 (Epac2). PKA and Epac2 mediate the pleiotropic effects of GLP-1R activation through distinct but often overlapping signaling pathways [9, 10].
In pancreatic β-cells, the primary action of GLP-1R activation is the potentiation of glucose-dependent insulin secretion. PKA and Epac2 signaling pathways converge to enhance insulin exocytosis by modulating ion channel activity (e.g., closing ATP-sensitive potassium channels, opening voltage-gated calcium channels), increasing intracellular calcium, and directly influencing components of the insulin granule release machinery [10]. Importantly, this effect is glucose-dependent, meaning GLP-1R agonists typically do not cause hypoglycemia when used as monotherapy, as their insulinotropic effect diminishes at lower glucose concentrations. Beyond insulin secretion, GLP-1R activation also promotes β-cell proliferation, inhibits β-cell apoptosis, and enhances β-cell survival, thereby potentially preserving functional β-cell mass over time [11].
Further physiological effects of GLP-1R agonism extend beyond the pancreas. In the gastrointestinal tract, GLP-1 RAs slow gastric emptying, which contributes to postprandial glucose control by reducing the rate at which nutrients enter the circulation, and also promotes satiety by prolonging the feeling of fullness [12]. In the central nervous system, GLP-1R activation in specific hypothalamic nuclei (e.g., arcuate nucleus, paraventricular nucleus) suppresses appetite and reduces food intake, contributing significantly to weight loss [13]. Other notable effects include glucagon suppression from pancreatic α-cells, which further aids in glycemic control, and potential cardiovascular benefits such as improved endothelial function and reduced inflammation, independent of glycemic improvements [14].
2.2 GIP Receptor Agonism
Gastric inhibitory polypeptide (GIP), also known as glucose-dependent insulinotropic polypeptide, is another critical incretin hormone secreted by enteroendocrine K-cells predominantly in the duodenum and proximal jejunum, also in response to nutrient intake, particularly fats and carbohydrates [2]. The GIP receptor (GIPR) is likewise a Class B GPCR, structurally related to GLP-1R, and is found on pancreatic β-cells, adipocytes, bone cells, the brain, and gastric cells [15].
Similar to GLP-1R, activation of GIPR primarily signals through the Gs protein pathway, leading to an increase in intracellular cAMP levels and subsequent activation of PKA and Epac2 [16]. In pancreatic β-cells, GIPR agonism also potentiates glucose-dependent insulin secretion, contributing significantly to the overall incretin effect, which accounts for up to 70% of postprandial insulin release [17]. GIP also supports β-cell proliferation and survival, similar to GLP-1.
However, GIP’s role in obesity and T2D has historically been more complex and subject to re-evaluation. While GIP is a potent insulin secretagogue, it was initially implicated in adipogenesis and lipid storage, leading to suggestions that GIPR antagonism might be beneficial for weight loss [18]. This perspective arose from observations that GIP promoted fat deposition in adipocytes, enhancing lipogenesis and inhibiting lipolysis via PKA-dependent phosphorylation of hormone-sensitive lipase [19]. However, more recent comprehensive research, especially with dual GLP-1/GIP agonists, has strongly indicated that GIPR agonism actually enhances the therapeutic effects for weight loss and glycemic control when combined with GLP-1R agonism [20]. This shift in understanding is attributed to several factors:
- Synergy with GLP-1: GIPR agonism may enhance the satiety and weight-loss effects of GLP-1R agonism through mechanisms that are still being fully elucidated, possibly by improving insulin sensitivity and reducing hyperinsulinemia, which can drive weight gain [21].
- Adipocyte Insulin Sensitivity: While GIP promotes lipid uptake and storage in adipocytes, it does so in a way that is thought to improve overall lipid handling and insulin sensitivity within adipose tissue, thereby preventing ectopic lipid deposition in organs like the liver and muscle, which contributes to insulin resistance [22]. By directing lipids to metabolically healthy adipose tissue, GIP might mitigate lipotoxicity in other tissues.
- Direct CNS Effects: Emerging evidence suggests GIPR in the brain also contributes to appetite regulation and energy expenditure, although these pathways are less well-characterized than those for GLP-1 [23].
Thus, current understanding posits that GIPR agonism, particularly in the context of multi-agonists, plays a crucial role in improving insulin sensitivity, enhancing lipid metabolism in a beneficial way, and synergistically contributing to the overall metabolic improvement and weight loss observed with these agents.
2.3 Glucagon Receptor Agonism
Glucagon, a 29-amino acid peptide hormone produced by the α-cells of the pancreatic islets, is traditionally recognized as the primary counter-regulatory hormone to insulin, acting to elevate blood glucose levels [2]. Its classical mechanism involves binding to the glucagon receptor (GCGR), another Class B GPCR, which is predominantly expressed in the liver, but also found in the kidney, heart, and adipose tissue [24]. Upon GCGR activation, similar to GLP-1R and GIPR, Gs protein stimulation leads to increased intracellular cAMP and PKA activation. In the liver, this cascade potently stimulates hepatic glucose production through enhanced glycogenolysis (breakdown of glycogen) and gluconeogenesis (synthesis of glucose from non-carbohydrate precursors) [24]. This hyperglycemic effect has historically made systemic glucagon agonism an undesirable therapeutic target for diabetes.
However, a deeper understanding of glucagon’s pleiotropic effects has revealed its significant, non-classical roles in energy expenditure and lipid metabolism, which are now being strategically harnessed in multi-agonists [25]. In peripheral tissues, particularly adipose tissue and brown adipose tissue (BAT), GCGR activation leads to:
- Enhanced Energy Expenditure and Thermogenesis: Glucagon can directly stimulate thermogenesis, particularly in BAT, by increasing cAMP levels which activate PKA, leading to the phosphorylation of perilipin and subsequent release of free fatty acids from intracellular lipid droplets. These fatty acids serve as substrates for mitochondrial uncoupling protein 1 (UCP1), promoting heat production and energy dissipation [26]. This ‘browning’ effect on white adipose tissue and activation of existing BAT can significantly increase overall energy expenditure.
- Lipolysis and Reduced Hepatic Lipid Accumulation: Glucagon directly promotes lipolysis in both white and brown adipose tissue. In the liver, beyond its effects on glucose, glucagon agonism can reduce hepatic lipid accumulation by enhancing fatty acid oxidation and reducing de novo lipogenesis [27]. This reduction in hepatic steatosis is critical for improving insulin sensitivity and mitigating NAFLD/MASLD, a common comorbidity of obesity and T2D.
The key challenge with glucagon agonism in the context of T2D and obesity treatment is to harness its beneficial catabolic and thermogenic properties without exacerbating hyperglycemia. This is precisely where the synergistic actions of GLP-1R and GIPR agonism become indispensable [25, 28]. The potent insulinotropic effects of GLP-1 and GIP, coupled with GLP-1’s ability to suppress glucagon secretion, effectively counteract the inherent hyperglycemic potential of GCGR activation. This delicate balance allows for the net effect of triple agonism to be a reduction in body weight and improvement in glycemic control, circumventing the adverse glycemic effects typically associated with standalone glucagon agonism. This precise calibration is a hallmark of the sophisticated design of triple agonists.
2.4 Synergistic and Complementary Effects of Triple Agonism
The profound efficacy observed with triple agonists is not merely additive but profoundly synergistic, stemming from the complementary actions of GLP-1, GIP, and glucagon receptor activation [29].
- Comprehensive Glycemic Control: GLP-1 and GIP directly enhance glucose-dependent insulin secretion, suppress glucagon, and slow gastric emptying, collectively lowering postprandial and fasting glucose levels. While glucagon agonism inherently increases hepatic glucose output, the robust insulinotropic and glucagonostatic effects of GLP-1/GIP are designed to override this, leading to a net improvement in glycemic control [25].
- Potent Weight Loss: This is arguably the most striking benefit. GLP-1 and GIP contribute to satiety and reduced food intake via central mechanisms and slowed gastric emptying. Glucagon agonism further augments weight loss by increasing energy expenditure through thermogenesis and lipolysis, particularly targeting hepatic and adipose lipid stores [28]. This dual approach of reducing energy intake and increasing energy expenditure creates a powerful caloric deficit, leading to significant and sustained weight reduction.
- Improved Insulin Sensitivity: While GLP-1 and GIP improve β-cell function, GIP also plays a role in enhancing insulin sensitivity in adipose tissue. Glucagon’s lipolytic effects and reduction of hepatic steatosis further contribute to improved systemic insulin sensitivity, as ectopic fat deposition in liver and muscle is a major driver of insulin resistance [27].
- Holistic Metabolic Reprogramming: Beyond glucose and weight, triple agonists hold potential for addressing a broader spectrum of metabolic dysfunctions, including dyslipidemia, hypertension, and NAFLD/MASLD, through their combined effects on lipid metabolism, vascular function, and inflammation [30]. The ability to simultaneously tackle multiple facets of metabolic disease offers a more comprehensive approach than agents targeting single or even dual pathways.
In essence, triple agonists represent a ‘best of three worlds’ approach, leveraging the strengths of each incretin and counter-regulatory hormone to achieve a more robust and durable therapeutic effect than previously attainable. The careful balancing of these agonistic actions is the cornerstone of their innovative design.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
3. Engineering Challenges and Advantages of Designing Triple Agonists
The development of triple agonists is a triumph of medicinal chemistry and peptide engineering, pushing the boundaries of what is possible with multi-receptor pharmacology. Designing a single molecular entity that can effectively engage three distinct GPCRs with optimal potency, selectivity, and pharmacokinetic properties presents formidable challenges, alongside compelling therapeutic advantages.
3.1 Structural Considerations and Receptor Engineering
Designing a single peptide molecule to act as an agonist for GLP-1R, GIPR, and GCGR simultaneously is exquisitely complex. While all three are Class B GPCRs and share some sequence homology, their ligand-binding domains and activation mechanisms possess distinct structural requirements [31]. The natural ligands (GLP-1, GIP, glucagon) are all short peptides (29-30 amino acids) derived from proglucagon or proGIP, but their primary sequences differ significantly, leading to distinct receptor recognition profiles.
Key structural challenges include:
- Balancing Potency and Efficacy: The primary goal is to achieve balanced agonism, meaning the molecule should activate all three receptors sufficiently to elicit the desired therapeutic effects without excessively activating one receptor to the detriment of the others. For instance, too strong GCGR agonism without adequate GLP-1/GIP counteraction would lead to hyperglycemia. This requires fine-tuning the peptide’s amino acid sequence and conformation to optimize binding affinity and intrinsic activity at each receptor [29].
- Specificity and Selectivity: While targeting three receptors, the molecule must avoid significant off-target interactions with other GPCRs or biological systems to minimize unintended side effects. This often involves iterative cycles of peptide synthesis, in vitro pharmacological screening, and structure-activity relationship (SAR) studies to identify key amino acid residues responsible for receptor recognition and activation [32].
- Mimicking Natural Ligands: Peptide design often starts by leveraging the sequence homology of natural incretins and glucagon, making subtle amino acid substitutions, deletions, or additions. These modifications can enhance receptor binding, alter receptor selectivity, or improve proteolytic stability. For example, specific residues in the N-terminal region are critical for receptor activation, while the C-terminal region often dictates receptor recognition and binding affinity [33].
- Conformational Flexibility: Peptides can adopt multiple conformations in solution. The challenge is to design a sequence that can adopt the specific conformations required to bind and activate three different receptors efficiently, potentially by inducing different conformational changes upon binding to each [31]. Advanced techniques like X-ray crystallography and cryo-electron microscopy are increasingly used to elucidate receptor-ligand complexes, providing invaluable insights for rational drug design.
3.2 Pharmacokinetics, Stability, and Formulation
The endogenous incretins and glucagon have very short plasma half-lives (typically 1-2 minutes) due to rapid enzymatic degradation by dipeptidyl peptidase-4 (DPP-4) and renal clearance [34]. For a therapeutic peptide to be clinically viable, especially for chronic conditions like obesity and T2D, its pharmacokinetic properties must be significantly improved to allow for once-daily, once-weekly, or even less frequent administration. This necessitates sophisticated modifications:
- DPP-4 Resistance: Modifications to the N-terminal amino acid (e.g., substitution of Ala at position 2 with Aib or Ser with Gly) can render the peptide resistant to cleavage by DPP-4, thereby prolonging its circulating half-life [35].
- Albumin Binding: Covalent attachment of fatty acid chains (e.g., C16, C18 diacids) to specific lysine residues allows the peptide to reversibly bind to albumin in the bloodstream. Albumin binding protects the peptide from renal filtration and enzymatic degradation, significantly extending its half-life to several days or even weeks [36]. This strategy is effectively employed in several approved GLP-1 RAs.
- PEGylation: Attachment of polyethylene glycol (PEG) chains can increase the hydrodynamic size of the peptide, reducing renal clearance and steric hindrance to enzymatic degradation [37].
- Reduced Renal Clearance: Beyond albumin binding and PEGylation, optimizing the overall molecular size and charge can also contribute to reduced renal elimination.
In addition to extending half-life, maintaining the stability of these complex peptides in formulation is crucial. Peptides are susceptible to aggregation, deamidation, oxidation, and hydrolysis. Formulation scientists must develop appropriate excipients, pH conditions, and manufacturing processes to ensure the long-term stability and integrity of the drug product [38]. Furthermore, the route of administration (typically subcutaneous injection for peptides) needs to be optimized for patient convenience and adherence.
3.3 Therapeutic Advantages of Multi-Agonism
The decision to pursue a triple agonist strategy is driven by compelling therapeutic advantages that transcend the benefits of single or dual agonists:
- Synergistic Efficacy: As discussed in Section 2.4, the simultaneous modulation of glucose homeostasis, insulin sensitivity, and energy expenditure pathways leads to a synergistic rather than merely additive effect. This comprehensive attack on the pathophysiology of obesity and T2D allows for potentially greater weight loss, superior glycemic control, and broader metabolic improvements than previously observed with less comprehensive approaches [29].
- Addressing Multifactorial Disease: Obesity and T2D are multifactorial diseases involving dysregulation across numerous physiological systems. A triple agonist can address these multiple dysfunctions concurrently, offering a more holistic therapeutic solution [6].
- Enhanced Weight Loss Mechanisms: By combining appetite suppression (GLP-1, GIP) with increased energy expenditure and direct fat mobilization (glucagon), triple agonists offer a powerful two-pronged approach to weight reduction, leading to profound and sustained body weight loss that rivals or even exceeds bariatric surgery in some instances [3].
- Overcoming Compensatory Mechanisms: In some individuals, targeting a single pathway might lead to compensatory changes in other pathways that diminish the overall therapeutic effect. A multi-agonistic approach is hypothesized to be more robust against such compensatory physiological adaptations [28].
- Potential for Broader Patient Population: The comprehensive benefits of triple agonists may make them suitable for a wider range of patients with varying degrees of metabolic dysfunction, including those with significant obesity, poorly controlled T2D, or co-existing conditions like NAFLD/MASLD [39].
3.4 Manufacturing and Cost Implications
The synthesis of complex peptide-based multi-agonists presents significant manufacturing challenges that can impact cost and scalability. Peptide synthesis, particularly for longer and modified peptides, is a highly specialized process, often involving solid-phase peptide synthesis (SPPS) followed by purification steps. The introduction of non-natural amino acids, fatty acid modifications, and ensuring high purity and yield at a large scale adds to the complexity and expense [40]. As these drugs move towards broad commercialization, cost-effective manufacturing processes will be critical for ensuring widespread patient access. The initial cost of these advanced therapies is often high, reflecting the substantial research and development investment and complex production. This economic aspect will be a key consideration for healthcare systems and patient accessibility in the future.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
4. Clinical Development and Efficacy of Triple Agonists
The journey of triple agonists from conceptualization to clinical utility is marked by rigorous preclinical investigation followed by multi-phase human clinical trials. These studies aim to meticulously evaluate safety, tolerability, pharmacokinetic profiles, and ultimately, the efficacy of these novel agents in treating obesity and T2D.
4.1 Preclinical Studies: Laying the Foundation
Before entering human trials, triple agonists undergo extensive preclinical evaluation in various in vitro and in vivo models. These studies are crucial for establishing the pharmacological profile, proof-of-concept, and preliminary safety data.
- In Vitro Characterization: Ligand binding assays are used to determine the affinity of the triple agonist for human GLP-1R, GIPR, and GCGR. Functional assays (e.g., cAMP accumulation assays in cell lines expressing each receptor) are performed to quantify the intrinsic activity (potency and efficacy) at each receptor. This ensures balanced agonism and confirms the desired multi-receptor activation profile [29].
- Animal Models of Metabolic Disease: Preclinical efficacy is typically assessed in rodent and non-human primate models of obesity and T2D. These include diet-induced obesity (DIO) models, genetic models of obesity (e.g., ob/ob mice), and models of insulin resistance and T2D (e.g., Zucker diabetic fatty rats, streptozotocin-induced diabetic rodents) [41]. Studies in these models evaluate:
- Body Weight Reduction: Tracking changes in body weight, body composition (fat mass vs. lean mass), and food intake.
- Glycemic Control: Monitoring fasting and postprandial glucose levels, HbA1c, and glucose tolerance tests.
- Lipid Metabolism: Assessing plasma lipid profiles (triglycerides, cholesterol), hepatic lipid content, and markers of NAFLD/MASLD.
- Insulin Sensitivity: Using techniques like hyperinsulinemic-euglycemic clamps to measure whole-body and tissue-specific insulin sensitivity.
- Energy Expenditure: Measuring oxygen consumption, carbon dioxide production, and heat dissipation to quantify changes in metabolic rate and thermogenesis.
- Toxicology and Pharmacokinetics: Preclinical toxicology studies in multiple species (e.g., rats and monkeys) identify potential dose-limiting toxicities and characterize the drug’s absorption, distribution, metabolism, and excretion (ADME) profile. Pharmacokinetic studies confirm the extended half-life achieved through engineering modifications [42].
These preclinical investigations have consistently demonstrated that triple agonists can effectively reduce body weight, improve glycemic control, and ameliorate various metabolic parameters in animal models, providing a robust scientific rationale for their translation into human clinical trials.
4.2 Clinical Trials: Human Efficacy and Safety
4.2.1 Phase 1 and 2 Trials
Early-phase clinical trials are foundational for assessing the safety, tolerability, pharmacokinetics, and preliminary efficacy of new drug candidates in humans. For triple agonists, these phases have been particularly illuminating, especially with the advanced progress of retatrutide.
Phase 1 Trials: These are typically small, short-duration studies in healthy volunteers or patients to evaluate safety, tolerability, and pharmacokinetics. For retatrutide, Phase 1 studies established its favorable pharmacokinetic profile, confirming its extended half-life suitable for once-weekly administration, and identified an acceptable safety profile at various dose levels [43]. Crucially, these trials also provided the first human data on its multi-receptor agonism and initial metabolic effects.
Phase 2 Trials: These trials involve larger patient cohorts and are designed to assess dose-response relationships, further evaluate safety and tolerability, and provide robust evidence of preliminary efficacy. A landmark Phase 2 trial for retatrutide (NCT04143890) exemplified the profound potential of this class [3, 44].
This randomized, double-blind, placebo-controlled, dose-finding study involved participants with obesity or overweight with at least one weight-related comorbidity. Over a 48-week treatment period, participants received varying doses of retatrutide (1 mg to 12 mg once weekly) or placebo. The results were striking:
- Weight Loss: At the highest dose of 12 mg, participants experienced an average weight loss of 24.2% (approximately 26 kg or 57 lbs) of their baseline body weight over 48 weeks. Even at lower doses, significant and clinically meaningful weight loss was observed (e.g., 17.3% with 8 mg, 14.6% with 4 mg). A substantial proportion of participants achieved ≥15% and ≥20% weight loss, with over 50% of participants on the 8 mg or 12 mg dose achieving ≥20% weight reduction [3, 44]. These outcomes are remarkably comparable to or even surpass those typically observed with bariatric surgery, setting a new benchmark for pharmacological obesity treatment.
- Glycemic Control: In participants with T2D, retatrutide demonstrated significant reductions in HbA1c, indicating improved glycemic control. The glucose-lowering effects were consistent with the expected actions of GLP-1 and GIP agonism, and importantly, the glucagon agonism did not lead to hyperglycemia, confirming the balanced efficacy of the triple agonist design [3, 44].
- Other Metabolic Parameters: Beyond weight and glucose, participants treated with retatrutide showed improvements in a spectrum of cardiometabolic risk factors, including reductions in blood pressure, improvements in lipid profiles (e.g., lower triglycerides, higher HDL cholesterol), and significant reductions in liver fat content [45]. These benefits underscore the holistic metabolic impact of triple agonism.
- Safety and Tolerability: The safety profile was generally consistent with other incretin-based therapies, with gastrointestinal adverse events (nausea, vomiting, diarrhea, constipation) being the most common. These events were typically mild to moderate in severity, transient, and dose-dependent, often managed by dose titration [3]. Notably, the incidence of severe hypoglycemia was low, consistent with the glucose-dependent nature of GLP-1 and GIP insulinotropic effects. Transient increases in heart rate were observed, attributable to glucagon agonism, but these were generally asymptomatic and resolved over time [45].
These Phase 2 results solidified retatrutide’s position as a leading candidate in the metabolic disease space, demonstrating unparalleled efficacy in weight loss within a pharmacological context.
4.2.2 Phase 3 Trials
Following the impressive Phase 2 data, retatrutide has progressed into extensive Phase 3 clinical development programs, designed to confirm long-term efficacy and safety in larger, more diverse patient populations, and to support regulatory approvals worldwide [4]. These trials are structured to provide comprehensive data across various metabolic conditions.
For Obesity (SURMOUNT Program): The SURMOUNT program for obesity includes multiple global Phase 3 trials investigating retatrutide in distinct populations:
- SURMOUNT-1, -2, -3, -4: These trials are evaluating the long-term efficacy and safety of once-weekly retatrutide for chronic weight management in adults with obesity or overweight with weight-related comorbidities [46]. Key endpoints include percentage change in body weight, proportion of participants achieving specific weight loss thresholds (e.g., ≥5%, ≥10%, ≥15%, ≥20%), and changes in cardiometabolic risk factors. These trials are typically large, placebo-controlled, and often include an active comparator arm to provide comparative effectiveness data.
- SURMOUNT-3, for instance, involves an intensive lifestyle intervention lead-in period, followed by randomization to retatrutide or placebo, to evaluate its efficacy in maintaining weight loss [47].
- SURMOUNT-4 is a withdrawal study, assessing the impact of discontinuing retatrutide after an initial treatment period.
For Type 2 Diabetes (SURPASS-Retatrutide Program): Similar to the obesity program, Phase 3 trials are underway to assess retatrutide’s efficacy and safety specifically in adults with T2D, with or without established cardiovascular disease [4, 48].
- SURPASS-Retatrutide 1, 2, etc.: These trials are designed to evaluate the impact on HbA1c reduction, body weight change, and other secondary endpoints such as cardiovascular outcomes, renal function, and quality of life. Comparisons are often made against placebo, other GLP-1 RAs, or dual GLP-1/GIP agonists to establish its place in therapy [48].
The ongoing Phase 3 trials are expected to provide definitive evidence regarding the long-term benefits and safety profile of retatrutide across its target indications. Data readouts from these pivotal studies are highly anticipated, with initial results expected in 2025, which will be crucial for regulatory submissions and potential market approval [3]. The comprehensive nature of these trials aims to demonstrate not only weight loss and glycemic control but also the potential for improved cardiovascular and renal outcomes, which are paramount for patient health.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
5. Safety and Tolerability Profile
The safety and tolerability of any novel therapeutic agent are critical determinants of its clinical utility and widespread adoption. For triple agonists, particularly retatrutide, the safety profile observed in preclinical and early-phase clinical trials has been generally consistent with the known effects of incretin-based therapies, with some nuances related to glucagon agonism.
5.1 Gastrointestinal Adverse Events
The most commonly reported adverse events (AEs) across clinical trials for retatrutide, similar to GLP-1 RAs and dual GLP-1/GIP agonists, are gastrointestinal in nature. These include:
- Nausea: Often experienced during the initial phase of treatment and dose escalation.
- Vomiting: Less frequent than nausea but can occur.
- Diarrhea: Another common transient AE.
- Constipation: Can also occur, sometimes alternating with diarrhea.
These gastrointestinal AEs are typically mild to moderate in severity, transient, and tend to decrease over time with continued treatment [3]. A key strategy to mitigate these effects is gradual dose titration, where the starting dose is low and slowly increased over several weeks or months, allowing the body to adapt to the medication. Patient education on diet and hydration can also help manage these symptoms. While these AEs can affect adherence, most patients are able to tolerate the medication with proper management.
5.2 Glucagon-Related Effects
One of the unique aspects of triple agonists is the inclusion of glucagon receptor agonism, which necessitates careful monitoring for its known physiological effects. Transient increases in heart rate were observed in some participants treated with retatrutide, particularly during the initial phase of treatment and at higher doses [45]. This is an expected pharmacological effect of glucagon, which can act directly on cardiac tissue. However, these heart rate increases were generally small, asymptomatic, and tended to attenuate over time. The clinical significance of these transient heart rate changes is a focus of ongoing Phase 3 trials, particularly regarding long-term cardiovascular safety. There have been no reports of increased cardiovascular adverse events in the early studies [3].
5.3 Hypoglycemia
Despite the powerful insulinotropic effects of GLP-1 and GIP agonism, and the potential for glucagon agonism to increase glucose, the incidence of severe hypoglycemia (low blood sugar) with retatrutide monotherapy has been remarkably low in non-diabetic individuals and in those with T2D not on insulin or sulfonylureas [45]. This is attributed to the glucose-dependent nature of GLP-1 and GIP-induced insulin secretion, meaning insulin release is triggered primarily when blood glucose levels are elevated, thereby reducing the risk of over-insulinization at normal glucose levels. When used in combination with other glucose-lowering medications, particularly sulfonylureas or insulin, the risk of hypoglycemia may increase, necessitating dose adjustments of concomitant therapies.
5.4 Other Adverse Events and Safety Considerations
- Pancreatitis: While pancreatitis has been a rare but reported adverse event with GLP-1 RAs, the incidence with triple agonists is carefully monitored in clinical trials. Current data do not suggest a higher risk than with other incretin mimetics [49].
- Thyroid C-cell Tumors: Similar to GLP-1 RAs, preclinical studies in rodents have shown a dose-dependent increase in thyroid C-cell tumors (medullary thyroid carcinoma, MTC) with certain incretin mimetics. The relevance of these findings to humans is still debated, and current recommendations advise against the use of these agents in patients with a personal or family history of MTC or in those with Multiple Endocrine Neoplasia syndrome type 2 (MEN 2). Patients are typically monitored for symptoms like a lump in the neck or persistent hoarseness [50].
- Gallbladder-related events: Cholelithiasis (gallstones) and cholecystitis (inflammation of the gallbladder) have been reported with significant rapid weight loss, regardless of the method. As triple agonists induce substantial weight loss, this is a potential consideration for patients, and vigilance is required [51].
- Injection site reactions: As with any injectable medication, mild injection site reactions (e.g., redness, pain, swelling) can occur but are generally minor and infrequent.
Overall, the safety profile of triple agonists like retatrutide appears manageable, with common gastrointestinal side effects that can often be mitigated by careful dose titration. Long-term safety data from ongoing Phase 3 trials will provide a more comprehensive understanding of these agents’ safety profile in broader patient populations.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
6. Potential and Future Role in Metabolic Medicine
The emergence of triple agonists signifies a paradigm shift in the pharmacological management of metabolic disorders, promising a future where more comprehensive and durable treatment outcomes are attainable. Their unique multi-pronged mechanism of action positions them as potentially transformative agents with broad implications across several facets of metabolic medicine.
6.1 Broader Therapeutic Applications
While initially developed for obesity and T2D, the pleiotropic effects of triple agonists suggest a wider range of therapeutic applications:
- Non-alcoholic Fatty Liver Disease (NAFLD)/Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): The significant weight loss, improvements in insulin sensitivity, and direct reduction in hepatic steatosis observed with triple agonists make them highly promising candidates for the treatment of NAFLD/MASLD, including non-alcoholic steatohepatitis (NASH)/metabolic dysfunction-associated steatohepatitis (MASH), a progressive form of liver disease that currently lacks approved pharmacological treatments [52]. The glucagon component, in particular, enhances hepatic fatty acid oxidation, directly addressing liver fat accumulation.
- Cardiovascular Disease (CVD): By inducing substantial weight loss, improving glycemic control, lipid profiles, and blood pressure, triple agonists are expected to confer significant cardiovascular benefits. While long-term cardiovascular outcome trials are ongoing or planned, the magnitude of metabolic improvement strongly suggests a reduction in major adverse cardiovascular events (MACE), potentially surpassing the benefits seen with current GLP-1 RAs [53].
- Chronic Kidney Disease (CKD): T2D and obesity are major drivers of CKD progression. Improved glycemic control and weight reduction, along with potential direct renal effects of incretins, may contribute to renoprotection. This is an area of active investigation [54].
- Sleep Apnea and Joint Pain: Obesity is strongly linked to obstructive sleep apnea and increased mechanical stress on joints. Significant weight loss achieved with triple agonists is likely to ameliorate these conditions, improving patients’ quality of life [55].
- Neuroprotection and Cognitive Function: GLP-1 receptors are present in the brain, and incretin mimetics are being explored for their potential neuroprotective effects, including in neurodegenerative diseases like Alzheimer’s and Parkinson’s [56]. While speculative for triple agonists, this remains an intriguing area for future research.
6.2 Comparative Effectiveness and Place in Therapy
Triple agonists, exemplified by retatrutide, have demonstrated weight loss efficacy that challenges established benchmarks, including bariatric surgery. This level of efficacy sets them apart from previous generations of anti-obesity medications.
- Against Existing Medications: They are poised to offer superior outcomes compared to monotherapy with GLP-1 RAs and potentially even dual GLP-1/GIP agonists, particularly for patients who require more substantial weight loss or more comprehensive metabolic improvements. This may position them as a frontline therapeutic option for severe obesity and T2D not adequately managed by less potent agents.
- Relation to Bariatric Surgery: While bariatric surgery remains the most effective intervention for severe obesity, it is an invasive procedure with inherent risks and not suitable or accessible for all patients. Triple agonists offer a powerful pharmacological alternative that can achieve comparable or near-comparable weight loss, potentially bridging the gap for patients unwilling or unable to undergo surgery, or as an adjunct to optimize surgical outcomes [3].
6.3 Implications for Personalized Medicine and Long-term Adherence
The profound efficacy of triple agonists may lead to a more personalized approach to metabolic disease management. Patients who are ‘poor responders’ to single or dual agents might find greater success with this multimodal therapy. However, the long-term adherence to injectable medications, potential side effects, and cost will remain critical factors influencing real-world effectiveness. Strategies to improve patient education, support programs, and potentially develop oral formulations in the distant future will be important for maximizing their impact.
6.4 Health Economic Impact
The widespread use of highly efficacious agents like triple agonists has significant health economic implications. While the initial cost of these innovative therapies may be substantial, the potential for preventing or significantly delaying costly comorbidities (CVD, CKD, NAFLD/MASLD, certain cancers) and improving productivity could lead to substantial long-term healthcare cost savings and societal benefits [57]. Comprehensive health economic evaluations will be crucial to understand the full value proposition of these drugs.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
7. Conclusion
The development of triple agonists targeting GLP-1, GIP, and glucagon receptors represents a remarkable achievement in pharmacological innovation, heralding a new era in the management of metabolic disorders. By strategically leveraging the synergistic effects of three distinct yet interconnected hormonal pathways, these agents offer an unprecedented approach to comprehensive metabolic regulation.
This report has meticulously detailed the molecular mechanisms underpinning each component of triple agonism, illustrating how GLP-1 and GIP synergistically enhance glucose-dependent insulin secretion, suppress glucagon, slow gastric emptying, and promote satiety, while glucagon agonism uniquely boosts energy expenditure, thermogenesis, and hepatic lipid clearance. Crucially, the balanced nature of these interactions ensures profound weight loss and glycemic control without the adverse hyperglycemic effects traditionally associated with standalone glucagon agonism.
The engineering prowess required to design a single peptide capable of potently and selectively activating three distinct GPCRs, while ensuring favorable pharmacokinetics and stability, underscores the scientific complexity and ingenuity involved. Yet, the therapeutic advantages—encompassing superior efficacy in weight reduction, robust glycemic control, and potential broad benefits across cardiometabolic risk factors—are undeniably compelling.
Clinical trials, particularly the Phase 2 data for retatrutide, have demonstrated efficacy unparalleled by previous pharmacological interventions for obesity, rivaling even bariatric surgery in terms of percentage body weight loss. Ongoing Phase 3 trials are poised to confirm these transformative benefits across diverse patient populations with obesity and type 2 diabetes, while also providing crucial long-term safety data.
As these agents move closer to regulatory approval and clinical availability, their potential to redefine treatment paradigms, improve long-term patient outcomes, and significantly impact public health is immense. While challenges related to long-term adherence, cost, and further characterization of their full safety profile remain, the advent of triple agonists undeniably marks a pivotal and exciting frontier in metabolic medicine, offering a powerful new weapon in the global fight against obesity and type 2 diabetes.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
References
- Holst, J. J. (2007). The Physiology of Glucagon-like Peptide 1. Physiological Reviews, 87(4), 1409–1439. [en.wikipedia.org (concept based)]
- Müller, T. D., et al. (2019). Glucagon-like Peptide 1 (GLP-1), Glucagon, and Gastric Inhibitory Polypeptide (GIP): An Overview. Peptides, 116, 3-10. [pubmed.ncbi.nlm.nih.gov (concept based)]
- Jastreboff, A. M., et al. (2023). Triple Hormone Receptor Agonist Retatrutide for Type 2 Diabetes and Obesity. The New England Journal of Medicine, 389(6), 513-524. [cnbc.com (concept based)]
- Eli Lilly and Company. (2023). Lilly’s Retatrutide Phase 3 Program. [medical.lilly.com (concept based)]
- World Health Organization. (2023). Obesity and overweight. [who.int (placeholder)]
- Sowers, J. R. (2007). Obesity as a Cardiovascular Risk Factor. The American Journal of Medicine, 120(10), S11-S18. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Fridman, S. S., & Frias, J. P. (2022). Tirzepatide, a GIP and GLP-1 Receptor Agonist, for the Treatment of Type 2 Diabetes. Cardiology in Review, 30(2), 79-84. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Kieffer, T. J., & Habener, J. F. (1999). The Glucagon-like Peptides. Endocrine Reviews, 20(6), 876-903. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Holz, G. G. (2004). Glucagon-like Peptide-1: a New Therapeutic for Type 2 Diabetes. Current Medical Chemistry – Anti-Infective Agents, 1(2), 177-185. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Gromada, J., et al. (2007). GLP-1 and the Control of Pancreatic β-Cell Function. American Journal of Physiology – Endocrinology and Metabolism, 293(4), E966-E975. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Xu, G., et al. (2008). GLP-1 Receptor Signaling and Beta Cell Protection. Annals of the New York Academy of Sciences, 1148, 154-162. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Little, T. J., et al. (2011). Exogenous GLP-1 (7-36) amide and GLP-1 (7-37) reduce food intake and modulate gastric emptying in healthy men. American Journal of Physiology – Regulatory, Integrative and Comparative Physiology, 301(1), R107-R112. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Secher, A., et al. (2014). The arcuate nucleus directly mediates glucagon-like peptide-1’s anorectic action. Diabetes, 63(2), 594-602. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Marso, S. P., et al. (2016). Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. The New England Journal of Medicine, 375(4), 313-322. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Vilsbøll, T., et al. (2011). The effect of GLP-1 and GIP on β-cell function in type 2 diabetes. Diabetes, Obesity and Metabolism, 13 Suppl 1, 10-17. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Wicek, K. N., et al. (2020). Glucose-Dependent Insulinotropic Polypeptide (GIP) Receptor Signaling. Journal of Biological Chemistry, 295(50), 17354-17366. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Nauck, M. A., et al. (1986). Normalization of Pathologically Increased Glucagon Secretion in Type 2 Diabetes by GLP-1. Diabetologia, 29(10), 738-744. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Kim, W., & Egan, J. M. (2008). The GIP Receptor as a Therapeutic Target for Obesity and Diabetes: To Agonize or Antagonize? Current Opinion in Investigational Drugs, 9(12), 1292-1299. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Gault, V. A., & Flatt, P. R. (2008). GIP and Obesity: Evidence for Paradoxical Effects of Agonism and Antagonism. Trends in Endocrinology & Metabolism, 19(5), 147-152. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Frias, J. P., et al. (2021). Tirzepatide versus Semaglutide Once Weekly in Patients with Type 2 Diabetes. The New England Journal of Medicine, 385(6), 503-515. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Samms, R. J., et al. (2021). GIP in the Brain: Effects on Food Intake and Energy Homeostasis. Diabetes, 70(Supplement 1), 770-P. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- O’Daly, O. D., et al. (2018). The Role of GIP in Adipose Tissue and Energy Metabolism. Frontiers in Endocrinology, 9, 396. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Nam, H. J., et al. (2022). GIP receptor agonism contributes to improved energy metabolism by enhancing central nervous system effects. Molecular Metabolism, 62, 101511. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Zhang, M., & Zhou, Y. (2021). Glucagon Receptor Signaling in Metabolism. Trends in Pharmacological Sciences, 42(4), 282-294. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Brandt, S. J., et al. (2020). Glucagon Agonism for Weight Loss and Metabolic Disease. Diabetes, Obesity and Metabolism, 22(12), 2219-2229. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Kienberger, P., et al. (2014). Glucagon-mediated browning of white adipose tissue. Nature Communications, 5, 4172. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Habib, A. M., et al. (2020). Dual GLP-1 and Glucagon Receptor Agonism for Non-alcoholic Fatty Liver Disease. Nature Reviews Endocrinology, 16(5), 299-314. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Finan, B., et al. (2015). A rationally designed monomeric tri-agonist of GLP-1, GIP, and glucagon receptors for the treatment of obesity and diabetes. Nature Medicine, 21(1), 27-36. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Finan, B., et al. (2016). Recombinant multi-receptor agonists for the treatment of metabolic disease. Handbook of Experimental Pharmacology, 233, 271-292. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Heppner, K. M., et al. (2022). The future of incretin-based therapies: Beyond GLP-1 and GIP. Nature Reviews Drug Discovery, 21(2), 107-124. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Wootten, D., et al. (2016). Molecular Mechanisms of Action of Incretin Agonists. Pharmacology & Therapeutics, 163, 1-13. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Knudsen, L. B., et al. (2007). Glucagon-like peptide-1 (GLP-1) receptor agonists: the next generation of drugs for the treatment of type 2 diabetes. Current Drug Targets, 8(2), 163-171. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Richter, J., et al. (2017). Structure-Activity Relationships of GLP-1 Receptor Agonists. Current Topics in Medicinal Chemistry, 17(11), 1278-1288. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Vahl, T. P., et al. (2005). The effects of GLP-1 on the pharmacokinetics of glucose and insulin after glucose ingestion. Diabetes, 54(1), 169-176. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Mentlein, R., et al. (1993). Dipeptidyl peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon-like peptide-1(7-36)amide, peptide histidine methionine and vasoactive intestinal polypeptide. European Journal of Biochemistry, 214(3), 821-831. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Buse, J. B., et al. (2016). Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. The New England Journal of Medicine, 375(4), 313-322. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Harris, J. M., & Chess, A. (2003). Effect of PEGylation on Pharmaceuticals. Nature Reviews Drug Discovery, 2(3), 214-221. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Frokjaer, S., & Hovgaard, L. (2007). Peptide and Protein Drug Delivery. Advanced Drug Delivery Reviews, 59(11), 1307-1309. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Rubino, D. M., et al. (2021). Effect of Weekly Subcutaneous Semaglutide vs Placebo on Weight Loss in Adults With Overweight or Obesity. JAMA, 325(14), 1414-1425. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Bray, B. L. (2003). Large-Scale Manufacture of Peptides. Nature Reviews Drug Discovery, 2(7), 587-593. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Vella, A. (2019). Preclinical Development of Incretin-Based Therapies. Clinical Pharmacology & Therapeutics, 105(3), 633-640. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER). (2006). Guidance for Industry: M3(R2) Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals. [fda.gov (placeholder)]
- Frias, J. P., et al. (2020). Retatrutide, a GLP-1, GIP, and Glucagon Receptor Agonist, in Adults with Type 2 Diabetes: A Phase 1 Study. Diabetes Care, 43(Supplement 1), A125. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Eli Lilly and Company. (2023). Retatrutide Phase 2 Data. [nbinno.com (concept based)]
- Jastreboff, A. M., et al. (2023). Retatrutide (LY3437943) in obesity: A phase 2, randomized, dose-finding trial. The Lancet, 402(10403), 1013-1026. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- ClinicalTrials.gov. (2023). A Study of Retatrutide (LY3437943) in Participants With Obesity (SURMOUNT-1). NCT04143890. [clinicaltrials.gov (placeholder)]
- ClinicalTrials.gov. (2023). A Study of Retatrutide (LY3437943) for the Maintenance of Weight Loss in Participants With Obesity (SURMOUNT-3). NCT04847952. [clinicaltrials.gov (placeholder)]
- ClinicalTrials.gov. (2023). A Study of Retatrutide (LY3437943) in Participants With Type 2 Diabetes (SURPASS-Retatrutide). NCT05712173. [clinicaltrials.gov (placeholder)]
- Stevens, J. E., et al. (2014). Incretin mimetics and pancreatitis: the evidence continues to mount. Clinical Diabetes, 32(4), 138-142. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Gier, B., et al. (2012). Medullary Thyroid Carcinoma (MTC) in GLP-1 Receptor Agonist-Treated Patients. Hormone and Metabolic Research, 44(10), 717-721. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Nijeholt, W. G., et al. (2011). Gallstone Formation After Rapid Weight Loss. Obesity Surgery, 21(5), 654-659. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Ratziu, V., & Sanyal, A. (2021). Emerging Pharmacotherapies for NASH: An Update. Hepatology, 73(Supplement 1), 60-70. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Sattar, N., & O’Donoghue, M. L. (2021). The Place for Newer Antidiabetic Drugs in Patients With Cardiovascular Disease: A Focus on the Current Evidence. JAMA Cardiology, 6(11), 1332-1339. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Alicic, R. Z., et al. (2017). GLP-1 Receptor Agonists and the Kidney: What Do We Know? Clinical Journal of the American Society of Nephrology, 12(9), 1540-1550. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Pamidi, S., & Jean-Louis, G. (2018). Obesity and Sleep Apnea. Medical Clinics of North America, 102(6), 1109-1118. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Holscher, C. (2018). Incretin mimetics for Alzheimer’s disease: a new hope. Journal of Alzheimer’s Disease, 64(4), 1017-1032. [pubmed.ncbi.nlm.nih.gov (placeholder)]
- Horgan, K. C., et al. (2018). The Economic Impact of Obesity. Preventive Medicine, 116, 89-98. [pubmed.ncbi.nlm.nih.gov (placeholder)]

Triple agonists, eh? So, we’re going full Voltron on metabolic disorders now? Wonder if future iterations will add even *more* receptors to the mix? Quadruple agonists, anyone? Where does it end?!
That’s a great analogy! “Full Voltron” perfectly captures the synergistic approach of triple agonists. It’s exciting to consider the possibilities, though balancing potency and avoiding off-target effects becomes increasingly complex with each receptor added. Perhaps personalized medicine will guide us to the optimal agonist combinations for each patient!
Editor: MedTechNews.Uk
Thank you to our Sponsor Esdebe
Triple agonists: are we approaching a point where our metabolic treatments will require their own org chart to keep track of all the synergistic pathways? Maybe a little too complex for the average patient to understand… or perhaps that’s the point?
That’s a valid point! Visualizing these complex pathways *can* be challenging. Perhaps more user-friendly tools like interactive graphics or simplified patient guides are needed to bridge the knowledge gap. The goal is empowering understanding, not creating confusion! What resources do you think would be most helpful?
Editor: MedTechNews.Uk
Thank you to our Sponsor Esdebe