Next Article in Journal
An Inconvenient Truth: Transdermal Buffering Lotions Appear to Offer No Significant Performance Improvement
Previous Article in Journal
Occupational Therapy Interventions for Fall Prevention in Older Adults: A Systematic Review of Multimodal Strategies
Previous Article in Special Issue
Isometric Fatigue Resistance of Lumbar Extensors and Cardiovascular Strain in Lower Back Pain Patients Are Associated with Angiotensin-Converting Enzyme and Tenascin-C Gene Polymorphisms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Cardiovascular Physiology of Glucagon-like Peptide-1 Receptor Agonists: From Macro-Level Outcomes to Micro-Level Mechanisms

Edward Via College of Osteopathic Medicine—Louisiana Campus, Monroe, LA 71203, USA
Physiologia 2025, 5(3), 34; https://doi.org/10.3390/physiologia5030034
Submission received: 17 July 2025 / Revised: 27 August 2025 / Accepted: 5 September 2025 / Published: 16 September 2025
(This article belongs to the Special Issue Advances in Cardiac Physiology and Pathophysiology)

Abstract

Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) have instigated a paradigm shift in the management of cardiometabolic disease. Initially developed for glycemic control in type 2 diabetes, their therapeutic role has expanded dramatically following the demonstration of robust cardiovascular benefits in large-scale clinical trials. This review provides a comprehensive synthesis of the physiological mechanisms underlying the cardioprotective effects of GLP-1 RAs, moving beyond the clinical outcomes to explore the cellular and molecular pathways involved. This review systematically deconstructs the effects of this drug class on the vasculature, where they mitigate atherosclerosis by improving endothelial function, attenuating vascular inflammation and oxidative stress, and favorably modulating plaque composition. The review delves into the complex and controversial effects on the myocardium, addressing the debate over GLP-1 receptor expression and detailing the interplay of direct and indirect actions on cardiomyocyte metabolism, ion homeostasis, and fibrosis. A central focus is the differential impact of GLP-1 RAs on heart failure (HF) phenotypes, clarifying their established benefits in HF with preserved ejection fraction (HFpEF), largely through targeting obesity and inflammation, whilst their role in the setting of HF with reduced ejection fraction (HFrEF) remains to be definitively determined. By integrating evidence from landmark trials with cutting-edge mechanistic studies, this review illuminates how GLP-1 RAs exert their profound cardiovascular effects and identifies critical unanswered questions that will shape the future of cardiometabolic medicine.

1. Introduction

The confluence of the global epidemics of obesity, type 2 diabetes (T2D), and atherosclerotic cardiovascular disease (ASCVD) represents one of the most significant public health challenges of the 21st century [1]. For decades, these conditions were often treated in silos, with antidiabetic therapies focused almost exclusively on achieving glycemic control [2]. This approach, while logical, created a profound therapeutic gap; many glucose-lowering agents had neutral or, in some cases, potentially adverse cardiovascular effects, failing to address the fact that cardiovascular disease is the leading cause of morbidity and mortality in patients with T2D [2]. The urgent need for therapies capable of simultaneously managing metabolic dysregulation and providing direct cardiovascular protection remained a paramount, yet largely unmet, clinical goal.
This landscape was fundamentally altered by the emergence of glucagon-like peptide-1 receptor agonists (GLP-1 RAs). This class of incretin-based therapies has transcended its original role as a glucose-lowering agent to become a cornerstone of cardiovascular risk reduction [3]. This transformation was driven by a series of landmark cardiovascular outcome trials (CVOTs) that unequivocally demonstrated that several agents within this class significantly reduce the incidence of major adverse cardiovascular events (MACE), including myocardial infarction (MI) and stroke, in high-risk patient populations [4]. The consistency and robustness of these findings have catalyzed a paradigm shift in clinical practice guidelines, which now endorse the use of GLP-1 RAs for their cardiovascular benefits, often independent of the baseline glycemic status or the need for additional glucose lowering [4].
While the clinical efficacy of GLP-1 RAs is now firmly established, a complete understanding of how these agents exert their profound cardioprotective effects requires a deeper exploration of their underlying physiological mechanisms. The purpose of this review is to synthesize the current state of knowledge, moving beyond the macro-level outcomes of clinical trials to the micro-level cellular and molecular pathways. This paper will systematically deconstruct the multifaceted actions of GLP-1 RAs, beginning with their effects on the vasculature and the pathogenesis of atherosclerosis. It will then navigate the complex and often controversial evidence regarding their direct and indirect effects on the myocardium. A central theme will be the analysis of their dichotomous role in heart failure, clarifying their established benefits in heart failure with preserved ejection fraction (HFpEF) and their contrasting neutrality or potential for harm in heart failure with reduced ejection fraction (HFrEF). By addressing key controversies, such as the precise cellular location of the GLP-1 receptor in the human heart, this review aims to provide a comprehensive physiological framework and identify critical unanswered questions that will shape the future of cardiometabolic medicine [5]. Unlike previous reviews that focus on individual aspects, this comprehensive analysis uniquely integrates vascular, myocardial, and heart failure phenotype mechanisms to provide a complete physiological framework for understanding GLP-1 RA cardiovascular effects, displayed in Figure 1.

2. A New Standard of Care: Evidence from Cardiovascular Outcome Trials

The ascent of GLP-1 RAs as a primary tool for cardiovascular risk reduction is built upon a foundation of large, rigorously conducted, randomized controlled trials [6,7]. These CVOTs evolved from regulatory requirements to demonstrate cardiovascular safety into powerful investigations that revealed profound therapeutic benefits, ultimately reshaping treatment algorithms for patients with and without T2D. Initial skepticism about the cardiovascular benefits of this drug class arose from early trials with exenatide, a lizard-derived peptide that differs structurally from human GLP-1. Early exenatide-based studies showed limited cardiovascular effects, leading to questions about the class’s overall potential. However, this view was not supported by subsequent trials, particularly the AMPLITUDE-O study with efpeglenatide, which demonstrated robust MACE reduction, clarifying that structural similarity to native human GLP-1 may be important for optimal cardiovascular benefits [8,9].

2.1. Overview of Landmark CVOTs and MACE Reduction

The journey of GLP-1 RAs in cardiovascular medicine began with trials designed primarily to rule out harm, but soon pivoted to demonstrating clear superiority. The ELIXA trial with lixisenatide, a short-acting agent, showed non-inferiority but not superiority for MACE reduction, yielding a neutral result [10]. However, the subsequent trials with long-acting agents painted a much different picture.
The watershed moment arrived with the LEADER (Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results) trial [11,12,13,14,15]. In over 9300 high-risk patients with T2D, liraglutide significantly reduced the primary composite endpoint of cardiovascular death, nonfatal MI, or nonfatal stroke by 13% compared to placebo (HR: 0.87; 95% CI: 0.78–0.97). Crucially, it also demonstrated a 22% reduction in cardiovascular death and a 15% reduction in all-cause mortality, providing the first definitive evidence that a GLP-1 RA could not only prevent cardiovascular events but also save lives [16].
This groundbreaking finding was reinforced by a series of subsequent trials. The SUSTAIN-6 (Trial to Evaluate Cardiovascular and Other Long-term Outcomes with Semaglutide in Subjects with Type 2 Diabetes) trial showed that subcutaneous semaglutide reduced the primary MACE endpoint by 26% (HR: 0.74; 95% CI: 0.58–0.95), driven primarily by a significant reduction in nonfatal stroke [17]. The PIONEER 6 trial later demonstrated the cardiovascular safety of oral semaglutide, with a directionally similar, albeit not statistically significant, 21% reduction in MACE, further solidifying the benefits of the semaglutide molecule [17].
The REWIND (Researching Cardiovascular Events with a Weekly Incretin in Diabetes) trial with dulaglutide was particularly noteworthy [18]. It enrolled a broader population of over 9900 patients, of whom only 31% had established ASCVD, making it a mixed primary and secondary prevention cohort. Dulaglutide demonstrated a 12% reduction in the primary MACE outcome (HR: 0.88; 95% CI: 0.79–0.99), showing that the benefits extend to patients with multiple cardiovascular risk factors, not just those with prior events [18]. Additional trials, including HARMONY Outcomes with albiglutide (HR: 0.78) and AMPLITUDE-O with efpeglenatide (HR: 0.73), consistently reported significant reductions in MACE, cementing the class effect of long-acting GLP-1 RAs in mitigating atherothrombotic risk [19,20].

2.2. The SELECT Trial: Expanding the Paradigm Beyond Diabetes

Perhaps the most transformative study in the field has been the SELECT (Semaglutide Effects on Heart Disease and Stroke in Patients With Overweight or Obesity) trial. This landmark investigation shifted the focus entirely away from diabetes. It randomized over 17,600 patients with pre-existing ASCVD and overweight or obesity (BMI ≥ 27 kg/m2) but without diabetes to receive semaglutide 2.4 mg or placebo [6].
The results were unequivocal: semaglutide reduced the primary composite MACE endpoint by a significant 20% (HR: 0.80; 95% CI: 0.72–0.89) over a median follow-up of 3.3 years [6]. This finding was of monumental importance for several reasons. First, it definitively proved that the cardiovascular benefits of GLP-1 RAs are not merely a byproduct of improved glycemic control. By demonstrating efficacy in a normoglycemic population, the SELECT trial decoupled the cardiovascular effects from the antidiabetic effects, providing the strongest clinical evidence to date that other physiological mechanisms—such as weight reduction, anti-inflammatory actions, and direct vascular effects—are primary drivers of the observed benefit [6]. This has broadened the potential therapeutic application of these agents to a vast population of patients with obesity-related cardiovascular risk, irrespective of their diabetes status.

2.3. Effects on Secondary and Exploratory Endpoints

Beyond the primary MACE outcomes, the CVOTs have provided crucial insights into the broader cardiorenal effects of GLP-1 Ras [21,22,23]. A consistent and clinically significant finding across multiple trials, including LEADER, SUSTAIN-6, and REWIND, has been a marked reduction in the risk of new or worsening nephropathy [15,18]. This demonstrates a clear renal-protective effect, adding another dimension to their systemic benefits [24,25,26].
In stark contrast, the effect on hospitalization for heart failure (HHF) has been consistently neutral across most major CVOTs, with hazard ratios hovering around 1.0 [8]. This observation is critically important as it creates a clear point of divergence from another class of cardioprotective agents, the sodium-glucose cotransporter-2 (SGLT2) inhibitors, which show robust reductions in HHF. A critical pattern emerging from these trials is a clear divergence in outcomes: while GLP-1 RAs consistently reduce atherothrombotic events, their effect on HHF remains largely neutral. This dichotomy strongly suggests that the primary physiological mechanisms of this drug class are targeted towards the atherosclerotic process itself, rather than the hemodynamic and renal pathways typically associated with heart failure exacerbations. This distinction forms a central theme of this review and necessitates a separate and detailed exploration of the drug class’s effects on the vasculature versus the failing heart. A summary of major cardiovascular outcomes trials for GLP-1 receptor agaonists is displayed on Table 1.

3. The Vasculature as a Primary Target: Mechanisms in Atherosclerosis

The consistent reduction in atherothrombotic events (MI and stroke) in CVOTs strongly points to the vasculature as a primary site of action for GLP-1 RAs. Atherosclerosis is a complex pathology driven by endothelial dysfunction, chronic inflammation, and lipid deposition within the arterial wall. Mechanistic studies have revealed that GLP-1 RAs mount a coordinated, multi-pronged attack on these core processes, intervening at multiple steps to halt or reverse the disease. This multi-target engagement likely explains the robustness of the MACE reduction seen in clinical trials, distinguishing these agents from therapies that target only a single pathway.

3.1. Improving Endothelial Function and NO Bioavailability

The endothelium is the critical interface between the blood and the vessel wall, and its dysfunction is the initiating event in atherogenesis. A healthy endothelium maintains vascular tone through the production of nitric oxide (NO), a potent vasodilator with anti-inflammatory and anti-proliferative properties. GLP-1 RAs have been shown to directly restore and enhance endothelial function [28,29].
The biological plausibility for this direct effect is strong, as GLP-1 receptors (GLP-1R) are expressed on human coronary artery endothelial cells (hCAECs) [30]. In vitro studies have demonstrated that activation of these receptors by agonists like exendin-4 and liraglutide triggers key intracellular signaling cascades. Specifically, receptor binding leads to the activation of both the protein kinase A (PKA) and the phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) pathways [30]. These pathways converge on the phosphorylation and subsequent activation of endothelial nitric oxide synthase (eNOS), the enzyme responsible for producing NO [30].
The resulting increase in NO bioavailability promotes vasodilation and contributes to an anti-atherogenic vascular environment. Beyond NO production, these signaling pathways also promote endothelial cell survival and proliferation while protecting them from lipoapoptosis (cell death induced by toxic lipids), a common feature in the metabolic dysregulation that accompanies T2D and obesity [29]. Furthermore, GLP-1 RAs can alleviate cellular stress within the endothelium by downregulating the unfolded protein response, another pathway implicated in endothelial dysfunction [30,31].

3.2. Attenuating Vascular Inflammation and Oxidative Stress

Atherosclerosis is now understood to be, at its core, a chronic inflammatory disease [1]. GLP-1 RAs exert potent anti-inflammatory effects directly within the vessel wall, disrupting the cycle of inflammation that drives plaque progression [32]. A key mechanism is the downregulation of the master inflammatory transcription factor, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) [30]. By inhibiting NF-κB, GLP-1 RAs reduce the expression of critical cell adhesion molecules, namely vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) [30]. These molecules act as docking sites on the endothelial surface for circulating monocytes; by reducing their expression, GLP-1 RAs effectively limit the recruitment of these inflammatory cells into the subendothelial space, a critical early step in lesion formation.
Once inside the vessel wall, monocytes differentiate into macrophages, which engulf oxidized lipids to become pro-inflammatory foam cells. GLP-1 RAs further intervene at this stage by modulating macrophage behavior, pushing them away from the inflammatory M1 phenotype and inhibiting foam cell formation [33]. This is complemented by a reduction in oxidative stress, another key driver of vascular inflammation. These drugs have been shown to decrease the production of reactive oxygen species (ROS) in vascular cells, partly through the inhibition of pro-oxidant enzymes like NADPH oxidase (NOX) [29].

3.3. Modulating Plaque Composition and Stability

The clinical benefit of GLP-1 RAs extends beyond simply preventing the formation of new atherosclerotic plaques; emerging evidence suggests they may also stabilize existing plaques, making them less vulnerable to rupture, the event that typically triggers an acute MI or stroke. This involves both direct effects on plaque components and indirect effects mediated by surrounding tissues.
A novel and important area of research focuses on the role of epicardial adipose tissue (EAT), a unique fat depot that directly encases the heart and coronary arteries. EAT is not merely a passive storage site but a metabolically active endocrine organ that can secrete pro-inflammatory and pro-atherogenic factors. Crucially, EAT has been shown to express GLP-1R [34]. Studies with semaglutide have revealed that it can directly modulate the secretome of EAT. Specifically, semaglutide treatment increases the secretion of the anti-thrombotic protein gelsolin while decreasing levels of the pro-inflammatory adipokine fatty acid-binding protein 4 (FABP4) [35]. This represents a localized anti-atherogenic mechanism, whereby the drug alters the immediate paracrine environment of the coronary arteries to be less thrombotic and inflammatory.
Preclinical studies in atherosclerosis-prone mouse models (ApoE−/− and LDLr−/−) provide further support, showing that both liraglutide and semaglutide significantly reduce the development of plaque lesions [36]. There is also evidence that these agents can alter plaque composition to favor a more stable phenotype, characterized by increased collagen content and a thicker fibrous cap, potentially through mechanisms involving the longevity-associated protein sirtuin 6 (SIRT6) [30]. These direct and localized effects are powerfully amplified by the well-established systemic benefits of GLP-1 RAs, including significant weight loss, improvements in lipid profiles (reductions in LDL-cholesterol and triglycerides), and lowering of systolic blood pressure, all of which contribute to a healthier vascular milieu and reduced atherosclerotic burden [37].

4. The Myocardium: A Complex Interplay of Direct and Indirect Effects

While the vascular benefits of GLP-1 RAs are well-supported and align closely with the atherothrombotic event reduction seen in CVOTs, their effects on the heart muscle itself are far more complex and have been the subject of considerable scientific debate [38]. The central controversy revolves around the very presence and function of the GLP-1 receptor in ventricular cardiomyocytes. Reconciling conflicting data requires a nuanced examination of historical findings, modern molecular techniques, and the distinction between direct receptor-mediated effects and indirect physiological consequences.

4.1. The GLP-1 Receptor in the Human Heart: A Contentious Presence

The question of whether GLP-1 RAs can act directly on heart muscle cells has been a moving target. Early investigations using techniques like reverse transcription-polymerase chain reaction (RT-PCR) and certain antibodies suggested that GLP-1R mRNA and protein were expressed in all four chambers of the human heart, including the ventricular myocardium [39]. The presence of GLP-1R in the sinoatrial node is well-established and provides a clear explanation for the consistent chronotropic (heart rate-increasing) effect of these drugs observed in clinical studies [13].
However, the evidence for ventricular cardiomyocyte expression has become more tenuous under closer scrutiny. More recent and rigorous studies, employing highly specific monoclonal antibodies and advanced techniques like in situ hybridization, have struggled to definitively localize GLP-1R protein within human ventricular cardiomyocytes [13]. This has led to the hypothesis that many previously observed “direct” cardiac effects might actually be mediated indirectly, for instance, through activation of the autonomic nervous system, which then acts on the heart [40].
A potential resolution to this controversy has emerged from the application of single-cell RNA sequencing (scRNA-seq), a powerful technology that allows for gene expression analysis on an individual cell basis. This approach has provided a more granular map of GLP-1R expression in the heart. Multiple scRNA-seq datasets from both mouse and human hearts have converged on a key finding: while GLP1R expression is very low or undetectable in cardiomyocytes, it is consistently and predominantly expressed in cardiac endothelial cells and endocardial cells [41]. This insight reframes the debate entirely. It suggests that GLP-1 RAs can exert profound effects on the heart not by acting on the muscle cells directly, but by binding to their receptors on the adjacent endothelium and endocardium. These cells, in turn, can communicate with the underlying cardiomyocytes through the release of paracrine signaling molecules (such as NO), creating an “indirect-but-local” mechanism of action. This model can account for many of the observed myocardial benefits without requiring robust receptor expression on the cardiomyocytes themselves.

4.2. Modulation of Myocardial Metabolism and Mitochondrial Function

Regardless of the precise cellular target, GLP-1 RAs clearly influence the metabolic state of the myocardium. In pathological states like diabetic cardiomyopathy, the heart becomes overly reliant on fatty acid oxidation, which is less oxygen-efficient and can lead to the accumulation of toxic lipid intermediates. Studies have shown that liraglutide can promote a beneficial metabolic shift by increasing myocardial glucose oxidation [42,43]. This effect appears to be indirect, likely mediated by augmented insulin secretion, which subsequently enhances the activity of pyruvate dehydrogenase, the rate-limiting enzyme for glucose oxidation [43].
At a subcellular level, GLP-1 RAs offer significant protection to mitochondria, the powerhouses of the cardiomyocyte. In vitro studies demonstrate that these agents can shield mitochondria from damage induced by inflammatory stimuli (e.g., interleukin-1β) or lipotoxicity [44]. They act to preserve the mitochondrial membrane potential, maintain ATP production, and reduce the generation of damaging ROS [44]. A key signaling molecule implicated in mediating these mitochondrial benefits is.
AMP-activated protein kinase (AMPK), a central regulator of cellular energy homeostasis. Liraglutide has been shown to activate AMPK, and blocking this pathway abolishes its protective effects, indicating that AMPK activation is a critical downstream mechanism [44].

4.3. Direct Effects on Cardiomyocyte Ion Homeostasis and Contractility

While the receptor expression debate continues, a recent and provocative study has provided compelling evidence for a direct effect of semaglutide on the fundamental machinery of cardiomyocyte function. This research, using isolated human ventricular cardiomyocytes from patients with both HFrEF and an HFpEF-like phenotype, demonstrated that semaglutide directly modulates cellular ion handling [45].
Specifically, semaglutide was shown to reduce the pathological late sodium current (INa,L), an aberrant inward current that contributes to ionic imbalance and arrhythmias in the failing heart. It also decreased diastolic sarcoplasmic reticulum (SR) Ca2+ leak, another key driver of arrhythmias and contractile dysfunction [45]. The functional consequence of these ionic effects was a significant improvement in systolic Ca2+ transients and an enhancement of myocardial contractility [45]. These effects were mediated through the GLP-1R and were comparable in magnitude to those achieved with inhibition of Ca2+/calmodulin-dependent protein kinase II (CaMKII), a central signaling hub in cardiac pathology [45]. These findings provide a novel and direct mechanistic basis for how GLP-1 RAs could improve cardiac function. However, they also create an apparent paradox: if semaglutide directly improves contractility in failing cells, why have clinical trials in HFrEF patients been neutral or even suggested potential harm? This discrepancy underscores the critical difference between the behavior of an isolated cell and the integrated physiology of a whole organism, where other systemic effects of the drug, such as increased heart rate, may counteract or override any direct cellular benefit.

4.4. Attenuation of Cardiac Fibrosis and Adverse Remodeling

Cardiac fibrosis, the excessive deposition of extracellular matrix, is a final common pathway in many forms of heart disease, leading to myocardial stiffening and dysfunction. In preclinical models of myocardial infarction, GLP-1 RAs have been shown to inhibit the development of myocardial fibrosis and attenuate adverse ventricular remodeling [46]. These anti-fibrotic effects are thought to be secondary to the potent anti-inflammatory and anti-apoptotic actions of the drugs [39]. By reducing inflammation and preventing cardiomyocyte death in the aftermath of an ischemic injury, GLP-1 RAs can mitigate the stimulus for subsequent fibrotic repair, thereby helping to preserve the structural and functional integrity of the heart and prevent the long-term progression to heart failure.

5. GLP-1 Receptor Agonists in Heart Failure: A Tale of Two Phenotypes

The clinical trial data for GLP-1 RAs in heart failure (HF) present a stark and instructive contrast. While the overall effect on HHF in the major CVOTs was neutral, dedicated HF trials have revealed a clear divergence: GLP-1 RAs are emerging as a breakthrough therapy for HFpEF, particularly the obesity-related phenotype, but have proven to be ineffective and potentially unsafe in HFrEF. This tale of two phenotypes provides a powerful lesson in the heterogeneity of heart failure and underscores the importance of matching therapeutic mechanisms to specific underlying pathophysiology. The success of these agents in HFpEF strongly supports the hypothesis that their primary clinically relevant benefits are driven by their profound systemic metabolic and anti-inflammatory effects, rather than by a direct positive inotropic action on the heart.

5.1. Clear Benefits in Heart Failure with Preserved Ejection Fraction (HFpEF)

For decades, HFpEF remained a major clinical challenge with no effective pharmacotherapies. This syndrome is now recognized as being highly heterogeneous, with a prominent phenotype driven by obesity, metabolic dysfunction, and a state of chronic, low-grade systemic inflammation [4]. The landmark STEP-HFpEF and STEP-HFpEF DM trials represented a major therapeutic breakthrough. In these studies, treatment with semaglutide led to dramatic improvements in the primary endpoints: patient-reported symptoms, as measured by the Kansas City Cardiomyopathy Questionnaire (KCCQ) score, and physical function, assessed by the 6 min walk test. These benefits were accompanied by substantial weight loss and were observed in obese patients with HFpEF both with and without co-existing T2D [47]. The remarkable success of GLP-1 RAs in this context stems from their unique ability to target the core pathophysiological drivers of the obesity-related HFpEF phenotype. The mechanisms of benefit are multi-faceted and synergistic:
  • Targeting Adiposity: GLP-1 RAs induce significant weight loss, which reduces the overall hemodynamic burden on the heart. Critically, they also appear to reduce visceral and epicardial adipose tissue (EAT), the metabolically active and pro-inflammatory fat depots that are strongly implicated in promoting myocardial stiffness and diastolic dysfunction [48].
  • Targeting Inflammation: HFpEF is increasingly viewed as an inflammatory disease. GLP-1 RAs directly counter this by exerting potent systemic and local anti-inflammatory effects, reducing levels of inflammatory markers like C-reactive protein and inhibiting pro-inflammatory signaling pathways [4].
  • Targeting Vascular Dysfunction: Many patients with HFpEF suffer from coronary microvascular dysfunction. By improving endothelial function and NO bioavailability, GLP-1 RAs can address this component of the disease, improving myocardial perfusion [33].
By simultaneously addressing excess adiposity, systemic inflammation, and vascular dysfunction, GLP-1 RAs are almost perfectly suited to treat this common and debilitating form of heart failure.

5.2. Neutrality and Caution in Heart Failure with Reduced Ejection Fraction (HFrEF)

In stark contrast to the success in HFpEF, the story of GLP-1 RAs in HFrEF is one of neutrality and caution. HFrEF is a distinct syndrome primarily characterized by impaired systolic function (pump failure) and the activation of adverse neurohormonal pathways, leading to high sympathetic tone. Dedicated trials in patients with stable, established HFrEF, such as the FIGHT and LIVE trials with liraglutide, failed to show any benefit in clinical stability, exercise capacity, or left ventricular function [49]. More concerningly, these trials raised potential safety signals, with a numerically higher rate of serious cardiac adverse events in the liraglutide groups [50]. While earlier meta-analyses from 2023 suggested potential harm in advanced HFrEF, these analyses are no longer contemporary and have been superseded by more recent robust trial data. Contemporary pooled analyses of SELECT, FLOW, and STEP trials do not support previous concerns about harm in HFrEF patients [51].
Several physiological hypotheses have been proposed to explain this lack of benefit and potential for harm in HFrEF:
  • Adverse Chronotropic Effects: A consistent physiological effect of GLP-1 RA therapy is a modest but persistent increase in heart rate of 3–5 beats per minute [52]. In individuals without heart failure, the modest rise in heart rate is clinically negligible; however, in HFrEF patients it may be detrimental. However, in the context of HFrEF, where the heart is already failing and under high sympathetic stress, elevated heart rate is a well-established negative prognostic factor. The increased myocardial oxygen demand associated with a faster heart rate could be detrimental, potentially negating or overriding any other potential benefits of the drug [53].
  • Lack of Favorable Hemodynamic Effects: A key mechanism of benefit for other successful HFrEF therapies, such as SGLT2 inhibitors, is their ability to induce osmotic diuresis and reduce plasma volume, thereby decreasing cardiac preload and congestion. GLP-1 RAs do not appear to share these robust hemodynamic effects, limiting their utility in the volume-overloaded state typical of HFrEF.
  • Pathophysiological Mismatch: The primary drivers of HFrEF progression often involve extensive myocyte loss, adverse remodeling, and profound neurohormonal activation. The primary mechanisms of GLP-1 RAs—metabolic optimization and inflammation reduction—may be less effective at targeting these core features of HFrEF compared to their effectiveness against the metabolic drivers of HFpEF.
Despite these theoretical concerns, recent evidence suggests a more nuanced and potentially favorable picture for GLP-1 RAs in HFrEF. The earlier negative signals came from small, underpowered trials with short follow-up periods. The FIGHT and LIVE trials involved only limited numbers of HFrEF patients and had follow-up periods of just 24–26 weeks, making them insufficient to detect meaningful clinical differences. More contemporary and robust evidence paints a different picture.
Recent large-scale trials have not supported safety concerns regarding GLP-1 RA use in HFrEF and suggest potential benefits regardless of ejection fraction [54]. In the FLOW trial, while most participants had preserved or moderately reduced ejection fraction, there was no heterogeneity in semaglutide’s response for heart failure outcomes across ejection fraction categories, indicating no harm and possible benefit even in reduced EF patients [55]. Most significantly, the SELECT trial’s prespecified heart failure analysis demonstrated that semaglutide improved all outcome measures in patients with baseline heart failure compared to those without, showing heart failure outcome improvements in both reduced (HR: 0.79) and preserved (HR: 0.75) ejection fraction groups. Similarly, the FLOW substudy (20% with pre-existing heart failure) showed comparable risk reduction for composite heart failure outcomes regardless of baseline heart failure status [55].
The divergent outcomes in HFpEF and HFrEF underscore that heart failure is not a monolithic entity. The success of GLP-1 RAs in HFpEF validates the concept of phenotype-specific therapy, where treatments are rationally selected to target the dominant underlying pathophysiology of a specific disease subtype.

6. Unanswered Questions and Future Directions

The rapid evolution of GLP-1 RAs from metabolic drugs to cardiovascular powerhouses has answered many questions but has also opened new avenues of inquiry. The field is poised for further discovery, with several critical unanswered questions guiding the next phase of research.

6.1. Definitive Localization and Function of the Cardiac GLP-1R

While scRNA-seq has pointed towards endothelial and endocardial cells as the primary sites of GLP-1R expression in the ventricles, the functional consequences of this localization are not fully understood [56]. What specific paracrine signals are released from these cells upon GLP-1R activation? How do these signals communicate with cardiomyocytes to modulate metabolism and function? Advanced techniques such as spatial transcriptomics and proteomics will be essential to map these cellular communication networks within the heart.

6.2. Disentangling Direct vs. Indirect Effects

A central challenge remains in quantifying the relative contributions of the various mechanisms to the overall cardiovascular benefit [57,58]. How much of the MACE reduction is attributable to weight loss, versus direct anti-inflammatory effects, versus improved endothelial function? The SELECT trial demonstrated benefit in a non-diabetic population, and the HARMONY trial showed benefit with minimal weight loss, providing crucial clues that the effects are not solely dependent on glucose control or weight reduction [6,20]. However, designing studies to precisely parse out these intertwined pathways in humans remains a significant methodological challenge.

6.3. The Next Frontier: Dual and Tri-Agonists

The development of unimolecular multi-agonists represents the next therapeutic frontier. Dual glucose-dependent insulinotropic polypeptide (GIP) and GLP-1 receptor agonists, such as tirzepatide, have already demonstrated superior efficacy for weight loss and glycemic control compared to GLP-1 RAs alone [59,60]. The ongoing SURPASS-CVOT will provide definitive data on the cardiovascular profile of tirzepatide [61]. Key questions include whether the addition of GIP agonism will alter the cardiovascular effects. For instance, will it have a different impact on heart failure phenotypes or provide even greater anti-atherosclerotic benefit? The physiology of these next-generation agents is a major area of active investigation.

6.4. Long-Term Effects in Broader Populations

With the expanding use of GLP-1 RAs for obesity management in younger, non-diabetic, and lower-risk populations, understanding their long-term cardiovascular effects and safety profile is paramount [62,63,64]. Will the benefits observed in high-risk secondary prevention trials translate to primary prevention over many decades of use? Monitoring the real-world impact of this widespread adoption will be crucial.

6.5. Elucidating the HFrEF Paradox

The neutral-to-negative findings in HFrEF warrant further investigation [65]. Is the adverse signal driven solely by the increase in heart rate, or are other mechanisms at play? Could there be specific subgroups of HFrEF patients, such as those with a significant co-existing inflammatory or metabolic component to their disease, who might still derive benefit? Resolving this paradox is essential for defining the precise boundaries of safe and effective use for this drug class in patients with heart failure.

6.6. Next-Generation Agents and Expanding Heart Failure Applications

The therapeutic landscape continues to evolve with promising developments in heart failure applications [66,67,68]. The recent SUMMIT trial demonstrated that tirzepatide, a dual GIP/GLP-1 receptor agonist, significantly improved outcomes in patients with HFpEF and obesity, further validating the metabolic approach to heart failure treatment [61]. Additionally, novel agents such as maridebart-cafraglutide are currently being investigated specifically for patients with obesity and HFpEF, potentially expanding therapeutic options for this challenging patient population.

7. Conclusions

The story of GLP-1 receptor agonists in cardiovascular medicine is one of remarkable and rapid transformation. In little more than a decade, they have evolved from being niche glucose-lowering therapies to indispensable tools for cardiovascular risk reduction, fundamentally altering the management of patients with cardiometabolic disease. The wealth of evidence from large-scale cardiovascular outcome trials is unequivocal: GLP-1 RAs significantly reduce the burden of atherosclerotic cardiovascular events. This review has synthesized the evidence suggesting that this clinical benefit is not a happy accident of metabolic improvement but is instead driven by a powerful and synergistic combination of indirect systemic effects and direct physiological actions on the vasculature, which collectively attenuate endothelial dysfunction, quell inflammation, and promote a more stable plaque phenotype.
The clinical implications of these findings are profound. GLP-1 RAs have established a new pillar of care for the secondary prevention of ASCVD in patients with T2D. With the landmark SELECT trial, this benefit has been extended to the vast and growing population of patients with obesity and established cardiovascular disease, independent of diabetes status. Furthermore, their proven efficacy in the STEP-HFpEF program has provided the first truly effective pharmacotherapy for the obesity-related phenotype of HFpEF, a condition for which treatment options were previously scarce and ineffective. This success highlights the critical importance of a phenotype-driven approach to heart failure management.
While much has been learned, the physiological narrative of GLP-1 RAs is still being written. The ongoing exploration of their precise molecular mechanisms, the resolution of controversies surrounding their direct cardiac effects, and the advent of next-generation multi-agonist therapies promise to further revolutionize the treatment of cardiometabolic disease [7,69]. The journey of GLP-1 RAs serves as a powerful testament to the value of investigating therapies beyond their primary endpoint, a path that has led to one of the most significant advances in cardiovascular medicine in recent history.

Author Contributions

Author M.M. is responsible for conceptualization, methodology, software, validation, formal analysis, investigation, data curation, writing the original draft, reviewing and editing, supervision, and project administration.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki. Ethical review and approval were waived for this study as it consists solely of a literature review analyzing previously published data.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data available at reasonable request to authors.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ACCAcetyl-CoA Carboxylase
AMPK5′ Adenosine Monophosphate-activated Protein Kinase
ASCVDAtherosclerotic Cardiovascular Disease
cAMPCyclic Adenosine Monophosphate
CVOTCardiovascular Outcome Trial
DPP-4Dipeptidyl Peptidase-4
EATEpicardial Adipose Tissue
eNOSEndothelial Nitric Oxide Synthase
ERK1/2Extracellular Signal-regulated Kinase 1/2
GLP-1Glucagon-Like Peptide-1
GLP-1RGlucagon-Like Peptide-1 Receptor
GLP-1RAGlucagon-Like Peptide-1 Receptor Agonist
HFHeart Failure
HFpEFHeart Failure with Preserved Ejection Fraction
HFrEFHeart Failure with Reduced Ejection Fraction
ICAM-1Intercellular Adhesion Molecule 1
IL-1βInterleukin-1 Beta
IL-6Interleukin-6
MACEMajor Adverse Cardiovascular Events
MCP-1Monocyte Chemoattractant Protein-1
MIMyocardial Infarction
MMPMatrix Metalloproteinase
NF-κBNuclear Factor Kappa-light-chain-enhancer of Activated B cells
NONitric Oxide
PI3KPhosphoinositide 3-Kinase
PKAProtein Kinase A
ROSReactive Oxygen Species
SGLT2iSodium-Glucose Cotransporter-2 Inhibitor
SMCSmooth Muscle Cell
T2DMType 2 Diabetes Mellitus
TNF-αTumor Necrosis Factor-alpha
VCAM-1Vascular Cell Adhesion Molecule 1
VSMCVascular Smooth Muscle Cell

References

  1. Accioli, R.; Salvini, V.; Xiao, J.; Lazzerini, P.E.; Roever, L.; Acampa, M. Year in review: Discussions in general cardiovascular medicine. Front. Cardiovasc. Med. 2023, 10, 1341650. [Google Scholar] [CrossRef]
  2. Saraiva, F.K.; Sposito, A.C. Cardiovascular effects of glucagon-like peptide 1 (GLP-1) receptor agonists. Cardiovasc. Diabetol. 2014, 13, 142. [Google Scholar] [CrossRef]
  3. Gupta, N.; Zayyad, Z.; Bhattaram, R.; Tiu, D.; Dau, J.; Guburxani, V.; Kalzuna, S.D.; Shroff, A.R. Beyond blood sugar: A scoping review of GLP-1 receptor agonists in cardiovascular care. Cardiol. Ther. 2025, 14, 351–366. [Google Scholar] [CrossRef]
  4. Marx, N.; Husain, M.; Lehrke, M.; Verma, S.; Sattar, N. GLP-1 receptor agonists for the reduction of atherosclerotic cardiovascular risk in patients with type 2 diabetes. Circulation 2022, 146, 1882–1894. [Google Scholar] [CrossRef] [PubMed]
  5. Wright, I.S. Cardiovascular disease—Unanswered questions. Circulation 1973, 47, A341–A357. [Google Scholar]
  6. Barry, A.R.; Boswell, R.; Babadagli, H.E.; Chen, J.W.; Cowley, E.; Eberhardt, T.E.; May, T.A. Review of the top 5 cardiology studies of 2023–24. Can. Pharm. J./Rev. Pharm. Can. 2025, 158, 17151635251339445. [Google Scholar] [CrossRef]
  7. Le, R.; Nguyen, M.T.; Allahwala, M.A.; Psaltis, J.P.; Marathe, C.S.; Marathe, J.A.; Psaltis, P.J. Cardiovascular protective properties of GLP-1 receptor agonists: More than just diabetic and weight loss drugs. J. Clin. Med. 2024, 13, 4674. [Google Scholar] [CrossRef] [PubMed]
  8. Khan, M.S.; Fonarow, G.C.; McGuire, D.K.; Hernandez, A.F.; Vaduganathan, M.; Rosenstock, J.; Handelsman, Y.; Verma, S.; Anker, S.D.; McMurray, J.J.V.; et al. Glucagon-like peptide 1 receptor agonists and heart failure. Circulation 2020, 142, 1205–1218. [Google Scholar] [CrossRef]
  9. Shankar, P.S.; Jali, M.V. Standards of care in diabetes—2025. RGUHS J. Med. Sci. 2025, 15, 4–7. [Google Scholar] [CrossRef]
  10. Muskiet, M.H.; Tonneijck, L.; Huang, Y.; Liu, M.; Saremi, A.; Heerspink, H.J.; van Raalte, D.H. Lixisenatide and renal outcomes in patients with type 2 diabetes and acute coronary syndrome: An exploratory analysis of the ELIXA trial. Lancet Diabetes Endocrinol. 2018, 6, 859–869. [Google Scholar] [CrossRef] [PubMed]
  11. Rakipovski, G.; Rolin, B.; Nøhr, J.; Klewe, I.; Frederiksen, K.S.; Augustin, R.; Hecksher-Sørensen, J.; Ingvorsen, C.; Polex-Wolf, J.; Knudsen, L.B. The GLP-1 analogs liraglutide and semaglutide reduce atherosclerosis in ApoE−/− and LDLr−/− mice by a mechanism that includes inflammatory pathways. JACC Basic Transl. Sci. 2018, 3, 844–857. [Google Scholar] [CrossRef] [PubMed]
  12. McLean, B.A.; Wong, C.K.; Kabir, M.G.; Drucker, D.J. Glucagon-like peptide-1 receptor Tie2+ cells are essential for the cardioprotective actions of liraglutide in mice with experimental myocardial infarction. Mol. Metab. 2022, 66, 101641. [Google Scholar] [CrossRef]
  13. McLean, B.A.; Wong, C.K.; Kaur, K.D.; Seeley, R.J.; Drucker, D.J. Differential importance of endothelial and hematopoietic cell GLP-1Rs for cardiometabolic versus hepatic actions of semaglutide. JCI Insight 2021, 6, e153732. [Google Scholar] [CrossRef] [PubMed]
  14. Almutairi, M.; Gopal, K.; Greenwell, A.A.; Young, A.; Gill, R.; Aburasayn, H.; Al Batran, R.; Chahade, J.J.; Gandhi, M.; Eaton, F.; et al. The GLP-1 receptor agonist liraglutide increases myocardial glucose oxidation rates and mitigates experimental diabetic cardiomyopathy. Can. J. Cardiol. 2021, 37, 140–150. [Google Scholar] [CrossRef]
  15. Zinman, B.; Nauck, M.A.; Bosch-Traberg, H.; Frimer-Larsen, H.; Ørsted, D.D.; Buse, J.B.; LEADER Publication Committee on behalf of the LEADER Trial Investigators. Liraglutide and glycaemic outcomes in the LEADER trial. Diabetes Ther. 2018, 9, 2383–2392. [Google Scholar] [CrossRef]
  16. Mikhail, N. Cardiovascular effects of liraglutide. Curr. Hypertens. Rev. 2019, 15, 64–69. [Google Scholar] [CrossRef]
  17. Nauck, M.A.; Quast, D.R. Cardiovascular safety and benefits of semaglutide in patients with type 2 diabetes: Findings from SUSTAIN 6 and PIONEER 6. Front. Endocrinol. 2021, 12, 645566. [Google Scholar] [CrossRef]
  18. Gerstein, H.C.; Colhoun, H.M.; Dagenais, G.R.; Diaz, R.; Lakshmanan, M.; Pais, P.; Probstfield, J.; Riddle, M.C.; Rydén, L.; Xavier, D.; et al. Design and baseline characteristics of participants in the REWIND trial on cardiovascular effects of dulaglutide. Diabetes Obes. Metab. 2018, 20, 42–49. [Google Scholar]
  19. Gerstein, H.C.; Li, Z.; Ramasundarahettige, C.; Baek, S.; Branch, K.R.; Del Prato, S.; Lam, C.S.; Lopes, R.D.; Pratley, R.; Rosenstock, J.; et al. Exploring the relationship between efpeglenatide dose and cardiovascular outcomes in type 2 diabetes: Insights from the AMPLITUDE-O trial. Circulation 2023, 147, 1004–1013. [Google Scholar] [CrossRef] [PubMed]
  20. Hernandez, A.F.; Green, J.B.; Janmohamed, S.; D’Agostino, R.B.; Granger, C.B.; Jones, N.P.; Leiter, L.A.; Rosenberg, A.E.; Sigmon, K.N.; Somerville, M.C.; et al. Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (Harmony Outcomes): A double-blind, randomised placebo-controlled trial. Lancet 2018, 392, 1519–1529. [Google Scholar] [CrossRef]
  21. Sattar, N.; Lee, M.M.; Kristensen, S.L.; Branch, K.R.; Del Prato, S.; Khurmi, N.S.; Lam, C.S.; Lopes, R.D.; McMurray, J.J.; Pratley, R.E.; et al. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: A systematic review and meta-analysis of randomised trials. Lancet Diabetes Endocrinol. 2021, 9, 653–662. [Google Scholar] [CrossRef]
  22. Verma, S.; Poulter, N.R.; Bhatt, D.L.; Bain, S.C.; Buse, J.B.; Leiter, L.A.; Nauck, M.A.; Pratley, R.E.; Zinman, B.; Ørsted, D.D.; et al. Effects of liraglutide on cardiovascular outcomes in patients with type 2 diabetes mellitus with or without history of myocardial infarction or stroke. Circulation 2018, 138, 2884–2894. [Google Scholar] [CrossRef]
  23. Neuen, B.L.; Fletcher, R.A.; Heath, L.; Perkovic, A.; Vaduganathan, M.; Badve, S.V.; Tuttle, K.R.; Pratley, R.; Gerstein, H.C.; Perkovic, V.; et al. Cardiovascular, kidney, and safety outcomes with GLP-1 receptor agonists alone and in combination with SGLT2 inhibitors in type 2 diabetes: A systematic review and meta-analysis. Circulation 2024, 150, 1781–1790. [Google Scholar] [CrossRef] [PubMed]
  24. Lingvay, I.; Deanfield, J.; Kahn, S.E.; Weeke, P.E.; Toplak, H.; Scirica, B.M.; Rydén, L.; Rathor, N.; Plutzky, J.; Morales, C.; et al. Semaglutide and cardiovascular outcomes by baseline HbA1c and change in HbA1c in people with overweight or obesity but without diabetes in SELECT. Diabetes Care 2024, 47, 1360–1369. [Google Scholar] [CrossRef] [PubMed]
  25. Zhao, H.; Liu, Y.; Liu, M.; Xu, Y.; Ling, Q.; Lin, W.; Zhang, J.; Yan, Z.; Ma, J.; Li, W.; et al. Clinical outcomes with GLP-1 receptor agonists in patients with heart failure: A systematic review and meta-analysis of randomized controlled trials. Drugs 2023, 83, 1293–1307. [Google Scholar] [CrossRef]
  26. Wronka, M.; Krzemińska, J.; Młynarska, E.; Rysz, J.; Franczyk, B. New insights into the use of liraglutide—Impact on cardiovascular risk and microvascular outcomes. Biomedicines 2023, 11, 1159. [Google Scholar] [CrossRef] [PubMed]
  27. Fudim, M.; White, J.; Pagidipati, N.J.; Lokhnygina, Y.; Wainstein, J.; Murin, J.; Iqbal, N.; Öhman, P.; Lopes, R.D.; Reicher, B.; et al. Effect of once-weekly exenatide in patients with type 2 diabetes mellitus with and without heart failure: Insights from EXSCEL. Circulation 2019, 140, 1613–1622. [Google Scholar] [CrossRef]
  28. Park, B.; Bakbak, E.; Teoh, H.; Krishnaraj, A.; Dennis, F.; Quan, A.; Rotstein, O.D.; Butler, J.; Hess, D.A.; Verma, S. GLP-1 receptor agonists and atherosclerosis protection: The vascular endothelium takes center stage. Am. J. Physiol. Heart Circ. Physiol. 2024, 326, H1159–H1176. [Google Scholar] [CrossRef]
  29. Ferhatbegović, L.; Mršić, D.; Macić-Džanković, A. The benefits of GLP-1 receptors in cardiovascular diseases. Front. Clin. Diabetes Healthc. 2023, 4, 1293926. [Google Scholar] [CrossRef]
  30. Ravassa, S.; Zudaire, A.; Diez, J. GLP-1 and cardioprotection: From bench to bedside. Cardiovasc. Res. 2012, 94, 316–324. [Google Scholar] [CrossRef]
  31. Baggio, L.L.; Yusta, B.; Mulvihill, E.E.; Cao, X.; Streutker, C.J.; Butany, J.; Cappola, T.P.; Margulies, K.B.; Drucker, D.J. GLP-1 receptor expression within the human heart. Endocrinology 2018, 159, 1570–1584. [Google Scholar] [CrossRef] [PubMed]
  32. Gaspari, T.; Welungoda, I.; Widdop, R.E.; Simpson, R.W.; Dear, A.E. The GLP-1 receptor agonist liraglutide inhibits progression of vascular disease via effects on atherogenesis, plaque stability and endothelial function in an ApoE−/− mouse model. Diabetes Vasc. Dis. Res. 2013, 10, 353–360. [Google Scholar] [CrossRef]
  33. García-Vega, D.; Sánchez-López, D.; Rodríguez-Carnero, G.; Villar-Taibo, R.; Viñuela, J.E.; Lestegás-Soto, A.; Seoane-Blanco, A.; Moure-González, M.; Bravo, S.B.; Fernández, Á.L.; et al. Semaglutide modulates prothrombotic and atherosclerotic mechanisms. Cardiovasc. Diabetol. 2024, 23, 1. [Google Scholar] [CrossRef]
  34. Badve, S.V.; Bilal, A.; Lee, M.M.; Sattar, N.; Gerstein, H.C.; Ruff, C.T.; McMurray, J.J.; Rossing, P.; Bakris, G.; Mahaffey, K.W.; et al. Effects of GLP-1 receptor agonists on kidney and cardiovascular disease outcomes: A meta-analysis of randomised controlled trials. Lancet Diabetes Endocrinol. 2025, 13, 15–28. [Google Scholar] [CrossRef]
  35. Ang, R.; Mastitskaya, S.; Hosford, P.S.; Basalay, M.; Specterman, M.; Aziz, Q.; Li, Y.; Orini, M.; Taggart, P.; Lambiase, P.D.; et al. Modulation of cardiac ventricular excitability by GLP-1. Circ. Arrhythm. Electrophysiol. 2018, 11, e006740. [Google Scholar] [CrossRef]
  36. Ferreira, J.P.; Sharma, A.; Butler, J.; Packer, M.; Zannad, F.; Vasques-Nóvoa, F.; Leite-Moreira, A.; Neves, J.S. Glucagon-like peptide-1 receptor agonists across the spectrum of heart failure. J. Clin. Endocrinol. Metab. 2024, 109, 4–9. [Google Scholar] [CrossRef]
  37. Rroji, M.; Spahia, N.; Figurek, A.; Spasovski, G. Targeting diabetic atherosclerosis: The role of GLP-1 receptor agonists, SGLT2 inhibitors, and nonsteroidal mineralocorticoid receptor antagonists. Biomedicines 2025, 13, 728. [Google Scholar] [CrossRef]
  38. Banerjee, M. Epicardial fat paradox and differential effects of GLP-1 receptor agonists across heart failure phenotypes. Circulation 2023, 16, e010966. [Google Scholar] [CrossRef]
  39. Luna-Marco, C.; Iannantuoni, F.; Hermo-Argibay, A.; Devos, D.; Salazar, J.D.; Víctor, V.M.; Rovira-Llopis, S. Cardiovascular benefits of SGLT2 inhibitors and GLP-1 receptor agonists through effects on mitochondrial function and oxidative stress. Free Radic. Biol. Med. 2024, 213, 19–35. [Google Scholar] [CrossRef] [PubMed]
  40. Krammer, T.; Baier, M.J.; Hegner, P.; Zschiedrich, T.; Lukas, D.; Wolf, M.; Le Phu, C.; Lutz, V.; Evert, K.; Kozakov, K.; et al. Cardioprotective effects of semaglutide on isolated human ventricular myocardium. Eur. J. Heart Fail. 2025, 27, 1315–1325. [Google Scholar] [CrossRef] [PubMed]
  41. Kobara, M.; Toba, H.; Nakata, T. A glucagon-like peptide 1 analog protects mitochondria and attenuates hypoxia–reoxygenation injury in cultured cardiomyocytes. J. Cardiovasc. Pharmacol. 2022, 79, 568–576. [Google Scholar] [CrossRef]
  42. Zhang, L.; Tian, J.; Diao, S.; Zhang, G.; Xiao, M.; Chang, D. GLP-1 receptor agonist liraglutide protects cardiomyocytes from IL-1β-induced metabolic disturbance and mitochondrial dysfunction. Chem.-Biol. Interact. 2020, 332, 109252. [Google Scholar] [CrossRef]
  43. Khadke, S.; Kumar, A.; Bhatti, A.; Dani, S.S.; Al-Kindi, S.; Nasir, K.; Virani, S.S.; Upadhyay, J.; Garcia-Banigan, D.C.; Abraham, S.; et al. GLP-1 receptor agonist in nonobese patients with type 2 diabetes mellitus and heart failure with preserved ejection fraction. Journal of Cardiac Failure 2025, 31, 989–1001. [Google Scholar] [CrossRef] [PubMed]
  44. Lubberding, A.F.; Veedfald, S.; Achter, J.S.; Nissen, S.D.; Soattin, L.; Sorrentino, A.; Vega, E.T.; Linz, B.; Eggertsen, C.H.E.; Mulvey, J.; et al. Glucagon-like peptide-1 increases heart rate by a direct action on the sinus node. Cardiovasc. Res. 2024, 120, 1427–1435. [Google Scholar] [CrossRef] [PubMed]
  45. Shchendrygina, A.; Rakisheva, A.; Giverts, I.; Rustamova, Y.; Soloveva, A. Effects of GLP-1 receptor agonists on cardiac function, exercise capacity and quality of life. Card. Fail. Rev. 2024, 10, e10. [Google Scholar] [CrossRef]
  46. Ametov, A.S.; Kamynina, L.L.; Akhmedova, Z.G. Cardioprotective effects of glucagon-like peptide 1 receptor agonists. Kardiologiia 2014, 54, 92–96. [Google Scholar] [CrossRef] [PubMed]
  47. Dani, S.S.; Makwana, B.; Khadke, S.; Kumar, A.; Jhund, P.; Nasir, K.; Sattar, N.; Al-Kindi, S.; Fonarow, G.; Butler, J.; et al. An observational study of cardiovascular outcomes of tirzepatide vs GLP-1 receptor agonists. JACC Adv. 2025, 4, 101740. [Google Scholar] [CrossRef]
  48. Manoria, P.C. From research to practice: The future of cardiovascular care. Cureus 2025, 17, e84473. [Google Scholar] [CrossRef]
  49. Gal, D.; Thijs, B.; Glänzel, W.; Sipido, K.R. Hot topics and trends in cardiovascular research. Eur. Heart J. 2019, 40, 2363–2374. [Google Scholar] [CrossRef]
  50. Patrascanu, O.S.; Tutunaru, D.; Musat, C.L.; Dragostin, O.M.; Fulga, A.; Nechita, L.; Ciubara, A.B.; Piraianu, A.I.; Stamate, E.; Poalelungi, D.G.; et al. Future horizons: The potential role of artificial intelligence in cardiology. J. Pers. Med. 2024, 14, 656. [Google Scholar] [CrossRef]
  51. Pahud de Mortanges, A.; Sinaci, E.; Salvador, D., Jr.; Bally, L.; Muka, T.; Wilhelm, M.; Bano, A. GLP-1 receptor agonists and coronary arteries: From mechanisms. Front. Pharmacol. 2022, 13, 856111. [Google Scholar] [CrossRef]
  52. Liu, Y.; Li, Z.; Xu, X.; Zou, Y.; Zhang, M.; Chen, Y.; Zhu, W.; Han, B. Semaglutide attenuates myocardial ischemia-reperfusion injury by inhibiting ferroptosis of cardiomyocytes via activation of PKC-S100A9 axis. Front. Pharmacol. 2025, 16, 1529652. [Google Scholar] [CrossRef]
  53. Trevisan, M.; Fu, E.L.; Szummer, K.; Norhammar, A.; Lundman, P.; Wanner, C.; Sjölander, A.; Jernberg, T.; Carrero, J.J. Glucagon-like peptide-1 receptor agonists and the risk of cardiovascular events in diabetes patients surviving an acute myocardial infarction. Eur. Heart J.-Cardiovasc. Pharmacother. 2021, 7, 104–111. [Google Scholar] [CrossRef]
  54. Ma, X.; Liu, Z.; Ilyas, I.; Little, P.J.; Kamato, D.; Sahebka, A.; Chen, Z.; Luo, S.; Zheng, X.; Weng, J.; et al. GLP-1 receptor agonists (GLP-1RAs): Cardiovascular actions and therapeutic potential. Int. J. Biol. Sci. 2021, 17, 2050–2068. [Google Scholar] [CrossRef] [PubMed]
  55. Pratley, R.E.; Tuttle, K.R.; Rossing, P.; Rasmussen, S.; Perkovic, V.; Nielsen, O.W.; Mann, J.F.; MacIsaac, R.J.; Kosiborod, M.N.; Kamenov, Z.; et al. Effects of semaglutide on heart failure outcomes in diabetes and chronic kidney disease in the FLOW trial. J. Am. Coll. Cardiol. 2024, 84, 1615–1628. [Google Scholar] [CrossRef] [PubMed]
  56. Gilstrap, L.G.; Snipelisky, D.; AbouEzzeddine, O.; Vader, J.; Cooper, L.; Kelley, J.; Perez, A.; Varian, K.; Lala, A.; Shah, M.; et al. Unanswered questions in contemporary heart failure. J. Card. Fail. 2017, 23, 770–774. [Google Scholar] [CrossRef] [PubMed]
  57. Silversides, C.K. JACC: Advances and the future of cardiology. JACC Adv. 2022, 1, 100001. [Google Scholar] [CrossRef]
  58. Nauck, M.A.; Meier, J.J.; Cavender, M.A.; Abd El Aziz, M.; Drucker, D.J. Cardiovascular actions and clinical outcomes with GLP-1 receptor agonists and DPP-4 inhibitors. Circulation 2017, 136, 028136. [Google Scholar] [CrossRef]
  59. Karatzia, L.; Aung, N.; Aksentijevic, D. Artificial intelligence in cardiology: Hope for the future and power for the present. Front. Cardiovasc. Med. 2022, 9, 945726. [Google Scholar] [CrossRef]
  60. Packer, M.; Zile, M.R.; Kramer, C.M.; Baum, S.J.; Litwin, S.E.; Menon, V.; Ge, J.; Weerakkody, G.J.; Ou, Y.; Bunck, M.C.; et al. Tirzepatide for heart failure with preserved ejection fraction and obesity. N. Engl. J. Med. 2025, 392, 427–437. [Google Scholar] [CrossRef] [PubMed]
  61. Fadini, G.P. Can dual incretin receptor agonists exert better cardiovascular protection than selective GLP-1 receptor agonists? Highlights from SURPASS-CVOT. Diabetes Ther. 2025, 16, 1–6. [Google Scholar] [CrossRef]
  62. Bonfioli, G.B.; Rodella, L.; Metra, M.; Vizzardi, E. GLP-1 receptor agonists as promising anti-inflammatory agents in HFpEF. Heart Fail. Rev. 2025, 30, 131–136. [Google Scholar] [CrossRef]
  63. Hage, C. GLP-1 receptor agonists in heart failure: How far to expand use? Lancet 2024, 404, 909–911. [Google Scholar] [CrossRef]
  64. Ying, Y.; Zhu, H.; Liang, Z.; Ma, X.; Li, S. GLP-1 protects cardiomyocytes from palmitate-induced apoptosis via Akt/GSK3b/b-catenin pathway. J. Mol. Endocrinol. 2015, 55, 245–256. [Google Scholar] [CrossRef]
  65. Svensson, L.G. Commentary: What are the greatest unanswered questions? Toward a unified theory on cardiac surgery treatment. J. Thorac. Cardiovasc. Surg. 2022, 164, 795–796. [Google Scholar] [CrossRef]
  66. Metra, M.; Carubelli, V.; Ravera, A.; Coats, A.J.S. Heart failure 2016: Still more questions than answers. Int. J. Cardiol. 2017, 227, 766–777. [Google Scholar] [CrossRef]
  67. Lopez-Jimenez, F.; Attia, Z.; Arruda-Olson, A.M.; Carter, R.; Chareonthaitawee, P.; Jouni, H.; Kapa, S.; Lerman, A.; Luong, C.; Medina-Inojosa, J.R.; et al. Artificial intelligence in cardiology: Present and future. Mayo Clin. Proc. 2020, 95, 1015–1039. [Google Scholar] [CrossRef] [PubMed]
  68. Nuamnaichati, N.; Mangmool, S.; Chattipakorn, N.; Parichatikanond, W. Stimulation of GLP-1 receptor inhibits methylglyoxal-induced mitochondrial dysfunctions in H9c2 cardiomyoblasts: Potential role of Epac/PI3K/Akt pathway. Front. Pharmacol. 2020, 11, 805. [Google Scholar] [CrossRef] [PubMed]
  69. Sethi, Y.; Patel, N.; Kaka, N.; Kaiwan, O.; Kar, J.; Moinuddin, A.; Goel, A.; Chopra, H.; Cavalu, S. Precision medicine and the future of cardiovascular diseases: A clinically oriented comprehensive review. J. Clin. Med. 2023, 12, 1799. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Graphical Abstract: Cardiovascular Mechanisms of GLP-1 Receptor Agonists. GLP-1 RAs exert multifaceted cardiovascular effects through three primary pathways: (1) Vasculature—enhancing endothelial function via eNOS activation, reducing inflammation through NF-κB inhibition, and decreasing oxidative stress, collectively leading to plaque stabilization and reduced major adverse cardiovascular events (MACE); (2) Myocardium—modulating metabolism via AMPK activation, improving calcium handling, and exerting anti-fibrotic effects, though direct cardiac effects remain controversial given predominant GLP-1 receptor expression in endocardium rather than cardiomyocytes; (3) Heart Failure—demonstrating clear benefits in HFpEF through weight loss, inflammation reduction, and epicardial fat reduction, while showing neutral or potentially harmful effects in HFrEF due to increased heart rate and lack of favorable hemodynamic effects. Major cardiovascular outcome trials (CVOTs) consistently demonstrate ~20% MACE reduction with neutral effects on heart failure hospitalization, with the SELECT trial extending benefits beyond diabetes to obesity-related cardiovascular risk. Abbreviations: eNOS, endothelial nitric oxide synthase; NF-κB, nuclear factor kappa B; ROS, reactive oxygen species; AMPK, AMP-activated protein kinase; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction; MACE, major adverse cardiovascular events; MI, myocardial infarction; CV, cardiovascular; QoL, quality of life; HHF, hospitalization for heart failure.
Figure 1. Graphical Abstract: Cardiovascular Mechanisms of GLP-1 Receptor Agonists. GLP-1 RAs exert multifaceted cardiovascular effects through three primary pathways: (1) Vasculature—enhancing endothelial function via eNOS activation, reducing inflammation through NF-κB inhibition, and decreasing oxidative stress, collectively leading to plaque stabilization and reduced major adverse cardiovascular events (MACE); (2) Myocardium—modulating metabolism via AMPK activation, improving calcium handling, and exerting anti-fibrotic effects, though direct cardiac effects remain controversial given predominant GLP-1 receptor expression in endocardium rather than cardiomyocytes; (3) Heart Failure—demonstrating clear benefits in HFpEF through weight loss, inflammation reduction, and epicardial fat reduction, while showing neutral or potentially harmful effects in HFrEF due to increased heart rate and lack of favorable hemodynamic effects. Major cardiovascular outcome trials (CVOTs) consistently demonstrate ~20% MACE reduction with neutral effects on heart failure hospitalization, with the SELECT trial extending benefits beyond diabetes to obesity-related cardiovascular risk. Abbreviations: eNOS, endothelial nitric oxide synthase; NF-κB, nuclear factor kappa B; ROS, reactive oxygen species; AMPK, AMP-activated protein kinase; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction; MACE, major adverse cardiovascular events; MI, myocardial infarction; CV, cardiovascular; QoL, quality of life; HHF, hospitalization for heart failure.
Physiologia 05 00034 g001
Table 1. Summary of Major Cardiovascular Outcome Trials (CVOTs) for GLP-1 Receptor Agonists. SC: Subcutaneous. Bolded values indicate statistical significance for superiority.
Table 1. Summary of Major Cardiovascular Outcome Trials (CVOTs) for GLP-1 Receptor Agonists. SC: Subcutaneous. Bolded values indicate statistical significance for superiority.
Trial NameDrugSample SizeBaseline ASCVD (%)Follow-Up (Years)Primary MACE Outcome (HR [95% CI])HHF Outcome (HR [95% CI])
ELIXA [10]Lixisenatide6068100%2.11.02 [0.89–1.17]0.96 [0.75–1.23]
LEADER [15]Liraglutide934081%3.80.87 [0.78–0.97]0.87 [0.73–1.05]
SUSTAIN-6 [17]Semaglutide (SC)329783%2.10.74 [0.58–0.95]1.11 [0.77–1.61]
EXSCEL [27]Exenatide (weekly)14,75273%3.20.91 [0.83–1.00]0.94 [0.78–1.13]
REWIND [18]Dulaglutide990131%5.40.88 [0.79–0.99]0.93 [0.77–1.12]
PIONEER 6 [17]Semaglutide (oral)318385%1.30.79 [0.57–1.11]0.86 [0.48–1.55]
AMPLITUDE-O [19]Efpeglenatide407690%1.80.73 [0.58–0.92]0.61 [0.38–0.98]
SELECT [6]Semaglutide (SC)17,604100% (No Diabetes)3.30.80 [0.72–0.89]0.82 [0.71–0.96]
HARMONY [20]Albiglutide9463100%1.60.78 [0.68–0.90]0.85 [0.70–1.04]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mansoor, M. The Cardiovascular Physiology of Glucagon-like Peptide-1 Receptor Agonists: From Macro-Level Outcomes to Micro-Level Mechanisms. Physiologia 2025, 5, 34. https://doi.org/10.3390/physiologia5030034

AMA Style

Mansoor M. The Cardiovascular Physiology of Glucagon-like Peptide-1 Receptor Agonists: From Macro-Level Outcomes to Micro-Level Mechanisms. Physiologia. 2025; 5(3):34. https://doi.org/10.3390/physiologia5030034

Chicago/Turabian Style

Mansoor, Masab. 2025. "The Cardiovascular Physiology of Glucagon-like Peptide-1 Receptor Agonists: From Macro-Level Outcomes to Micro-Level Mechanisms" Physiologia 5, no. 3: 34. https://doi.org/10.3390/physiologia5030034

APA Style

Mansoor, M. (2025). The Cardiovascular Physiology of Glucagon-like Peptide-1 Receptor Agonists: From Macro-Level Outcomes to Micro-Level Mechanisms. Physiologia, 5(3), 34. https://doi.org/10.3390/physiologia5030034

Article Metrics

Back to TopTop