Next Article in Journal
Dental Hygiene Challenges in Children with Autism: Correlation with Parental Stress: A Scoping Review
Previous Article in Journal
Preliminary Studies on Changes in Static Plantar Pressure and Stabilometry in Patients with Ankylosing Spondylitis Undergoing an Exercise Program
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiovascular Protective Properties of GLP-1 Receptor Agonists: More than Just Diabetic and Weight Loss Drugs

by
Richard Le
1,2,
Mau T. Nguyen
2,3,4,
Momina A. Allahwala
2,4,
James P. Psaltis
4,
Chinmay S. Marathe
4,5,
Jessica A. Marathe
2,3,4 and
Peter J. Psaltis
2,3,4,*
1
College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
2
Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
3
Department of Cardiology, Central Adelaide Local Health Network, Adelaide 5000, Australia
4
Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia
5
Department of Endocrinology, Central Adelaide Local Health Network, Adelaide 5000, Australia
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(16), 4674; https://doi.org/10.3390/jcm13164674
Submission received: 27 June 2024 / Revised: 6 August 2024 / Accepted: 7 August 2024 / Published: 9 August 2024
(This article belongs to the Section Pharmacology)

Abstract

:
Owing to their potent glucose-lowering efficacy and substantial weight loss effects, glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are now considered part of the frontline therapeutic options to treat both type 2 diabetes mellitus and nondiabetic overweight/obesity. Stemming from successful demonstration of their cardiometabolic modulation and reduction of major adverse cardiovascular events in clinical outcome trials, GLP-1 RAs have since been validated as agents with compelling cardiovascular protective properties. Studies spanning from the bench to preclinical and large-scale randomised controlled trials have consistently corroborated the cardiovascular benefits of this pharmacological class. Most notably, there is converging evidence that they exert favourable effects on atherosclerotic ischaemic endpoints, with preclinical data indicating that they may do so by directly modifying the burden and composition of atherosclerotic plaques. This narrative review examines the underlying pharmacology and clinical evidence behind the cardiovascular benefits of GLP-1 RAs, with particular focus on atherosclerotic cardiovascular disease. It also delves into the mechanisms that underpin their putative plaque-modifying actions, addresses existing knowledge gaps and therapeutic challenges and looks to future developments in the field, including the use of combination incretin agents for diabetes and weight loss management.

1. Introduction

As the leading cause of morbidity and mortality, cardiovascular disease (CVD) represents a formidable global disease burden [1]. In 2019, it was estimated that of 523 million people affected by all-cause CVD worldwide, there were 197 million prevalent cases of coronary artery disease (CAD), resulting in 9 million deaths [2]. Coronary artery disease, alongside peripheral artery disease (PAD) and ischaemic strokes, is a manifestation of atherosclerosis, a process whereby arteries undergo gradual stiffening and stenosis due to the build-up of inflammatory, fibro-lipidic plaques in their subintimal compartment [3].
As frequently recognised co-morbidities with atherosclerotic cardiovascular disease (ASCVD), type 2 diabetes mellitus (T2DM) and overweight/obesity are rapidly becoming pandemics of dysregulated metabolic health. T2DM currently affects 463 million people globally, a number predicted to increase by 25% in 2030 and 51% in 2045 [4]. Similarly, it has been projected that by 2030, 1.025 billion people will qualify as obese (BMI ≥ 30 kg/m2) [5]. This trajectory portends a myriad of future healthcare challenges. In particular, the burden of CVD will intensify, given that people with diabetes experience a two- to fourfold increased risk of developing ASCVD compared to those without [6] and that CVD accounts for two-thirds of deaths in the overweight/obese population [7]. The inextricable link between these disease states dictates that they cannot be managed in isolation.
Following the Food and Drug Administration’s (FDA) 2008 mandate that new diabetic drugs require a cardiovascular safety assessment, cardiovascular outcome trials (CVOTs) have been conducted to assess all new glucose-lowering pharmacotherapies [8]. In the case of glucagon-like peptide-1 receptor agonists (GLP-1 RAs), they have demonstrated significant reductions in major adverse cardiovascular event (MACE) endpoints, with the observed effects especially strong for reducing ischaemic events related to ASCVD [9]. The cardiovascular benefits of GLP-1 RAs are further supported by considerable preclinical and clinical research, which show that these agents exert favourable modulatory effects on atherosclerosis itself and its risk factors [10]. This narrative review examines the pharmacological properties and cardiovascular benefits of GLP-1 RAs, especially as they pertain to ASCVD. It also explores the spectrum of underlying mechanistic data for their anti-atherosclerotic properties, including those related to, but also independent of, cardiometabolic risk factors.

2. Pharmacology

2.1. GLP-1 Physiology

In 1964, it was found that orally administered glucose elicits a significantly greater insulin response than parenterally administered glucose [11,12]. This phenomenon of exaggerated or amplified insulin response to oral glucose is now known as the ‘incretin effect’ and is substantially reduced in T2DM [13]. The incretin effect was attributed to incretin hormones, particularly glucagon-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) [14,15]. GIP and GLP-1 are secreted from special entero-endocrine ‘K’ and ‘L’ cells, respectively, which are located predominantly in the small intestine and the colon [16].
After being post-translationally processed from proglucagon, GLP-1 is secreted and further converted in humans into its active forms, GLP-1(7–36) amide and GLP-1(7–37) [17]. In health, GLP-1 plasma levels increase fourfold following meals [17]. Postprandial secretion of GLP-1 follows a biphasic pattern: an early phase thought to be neurally mediated and a delayed phase after eating that is in direct response to luminal nutrients, particularly carbohydrates and fats [16,17]. The net result is insulin secretion and inhibition of glucagon secretion from islet β and α cells, respectively [14]. GLP-1 further limits postprandial glucose excursions by decelerating vagally mediated gastric emptying, which slows intestinal glucose absorption [17,18]. These canonical actions contribute to postprandial glucose control.
GLP-1 has been the incretin prioritised in drug discovery programs for T2DM. While GIP loses its insulinotropic action in experimental studies, even at supraphysiological doses, GLP-1 retains this effect under hyperglycaemic conditions [16]. Additionally, GLP-1 has indirect glucagonostatic effects that may assist in counteracting hyperglucagonaemia in T2DM [16,17], which may be mediated through upregulation of somatostatin secretion via somatostatin-2-receptor (SSTR2) [17]. As the insulinotropic effects are glucose-dependent and the counterregulatory response of glucagon to hypoglycaemia remains intact with GLP-1 infusion [19], the risk of hypoglycaemia is low. Hence, incretin-based drug development to control diabetic hyperglycaemia has been biased toward targeting the GLP-1 receptor (GLP-1R).

2.2. Pharmacodynamics of GLP-1 Receptor Agonists

2.2.1. Insulin Secretion

Activation of the GLP-1R results in multiple signalling pathways that increase intracellular calcium, culminating in pancreatic insulin secretion (Figure 1). By imitating the endogenous action of GLP-1, GLP-1 RAs agonise the GLP-1R, a G protein-coupled receptor (GPCR). Once GLP-1 RAs bind, GTP-bound Gαs subunits from the GPCR complexes activate adenylyl cyclase, which converts ATP into cyclic adenosine monophosphate (cAMP). This activates protein kinase A (PKA), which phosphorylates the sulfonylurea receptor-1 (SUR1) subunit of the K+/ATPase channel, leading to early closure of this channel and an amplified physiological Ca2+ influx via voltage-gated calcium channels. Increased insulin secretion can also occur via Gq/phospholipase C (PLC) signalling, where inositol trisphosphate 3 (IP3) binds to its receptor (IP3R) and diacylglycerol (DAG) binds to the ryanodine receptor to stimulate calcium-induced calcium release [17]. In a separate pathway, exchange protein directly activated by cAMP (Epac2) activates ras-proximate-1 (Rap1) [20], which activates PLC and increases downstream IP3/DAG levels, likewise inducing calcium release. Epac2 further enhances insulin exocytosis, as Epac2/Rim2/Piccolo complex binding to the Rab3–insulin interface enables exocytosis of insulin granules [17]. The calcium-permeable transient receptor potential melastatin 2 (TRPM2) channel has also been implicated in GLP-1-related insulin secretion via cAMP and PKA signalling [21]. Desensitisation of the GLP-1R is prevented through regular internalisation for intracellular trafficking and recycling, mediated by Gaq and β-arrestin pathways [22].
In addition to inducing insulin release, GLP-1R agonism can improve function and prevent exhaustion of pancreatic β cells on a transcriptional level by stimulating multiple pro-proliferative and anti-apoptotic pathways, while optimising intracellular metabolism (Figure 1). GLP-1 is thought to enable β islet mitogenesis via cyclin D2/Skp2 upregulation and p21/p27 downregulation, effectively reversing cell cycle arrest of pancreatic cells in hyperglycaemic states [23]. Similarly, GLP-1R agonism induces β cell proliferation and attenuates glucolipotoxicity through AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) signalling [24]. Upregulation of β cell transcription is likewise mediated via a variety of mechanisms including: increased levels of pancreatic and duodenal homeobox 1 (PDX-1) [25]; Wnt/β-catenin-mediated transcription factor 7-like 2 (TCF7L2) expression [17]; calcineurin/nuclear factor of activated T cell (NFAT) signalling [26]; increased insulin-like growth factor-1 receptor (IGF-1R) expression and membrane localisation [27]; increases in ERK1/2 due to GLP-1R-coupled β-arrestin recruitment and activation of Rap1 [17,28] and serine/threonine protein kinase B-raf [29]. Transactivation of epidermal growth factor receptor (EGFR) by GLP-1R occurs via c-Src, which induces phosphoinositide 3-kinase (PI3K) signalling [28] and prevents premature pancreatic cell apoptosis via upregulation of Bcl2 and inhibition of caspase-3 and forkhead box protein 1 (FoxO1) [28], thereby leading to disinhibition of PDX-1 [28]. GLP-1R activation further reduces endoplasmic reticular (ER) stress in β cells, preserving cellular function despite the cellular stresses induced by increased insulin biosynthesis [30] (Figure 1). PKA also activates cAMP response element-binding protein (CREB), which can, in turn, drive β cell growth by stimulating PDX-1 via insulin receptor substrate 2 (Irs2) signalling [17,31]. While CREB regulates the short-term pancreatic effects of GLP-1 RAs, mTOR and hypoxia-inducible factor-1α (HIF-1α) signalling mediate the secondary metabolic reprogramming that alters long-term glucose processing [32]. Collectively, the culmination of these activated pathways allows for enhanced pancreatic β cell viability and function.

2.2.2. Weight Loss and Appetite Suppression

Weight loss is a well-recognised and often beneficial effect of GLP-1 RA therapy. This has been attributed to the modulatory action of central appetite control, which results in caloric restriction and subsequent weight loss [33]. Within the hypothalamus, GLP-1-mediated activation of preproglucagon (Gcg+) neurons in the arcuate nucleus helps regulate appetite, independently of glucose control [34]. Animal studies have shown that GLP-1 can directly activate neurons containing pro-opiomelanocortin/cocaine- and amphetamine-stimulated transcription (POMC+/CART+), resulting in satiety [35]. GLP-1 can concurrently stimulate local inhibitory GABA neurons, which indirectly suppress neurons via agouti-related peptide/neuropeptide Y (AgRP+/NPY+) signalling, thereby reducing feelings of hunger. Control of satiety also occurs when GLP-1R+ neurons in the nucleus tractus solitarius of the hindbrain are stimulated, and this occurs independently of aversion caused by activation of GLP-1R+ neurons in the area postrema [36]. It is thought that vagal innervation is involved in the GLP-1 signalling behind appetite control, given that GLP-1 infusions increase vagal afferent firing and stimulate GLP-1R+ neurons in the nodose ganglion [37]. Furthermore, this theory of vagal dependency is bolstered by the nullification of GLP-1’s anorexic, gastrostatic and glucagonostatic effects in vagotomised patients [37]. There is also evidence that GLP-1 RAs can attenuate hyperstimulation of neural appetite and reward centres (insula, amygdala, putamen and orbitofrontal cortex) through direct GLP-1R agonism [38]. As such, the weight reduction that typically accompanies GLP-1 RA therapy is primarily due to appetite suppression, rather than increased energy expenditure or adipose tissue reduction.

2.2.3. Other Extra-Pancreatic Sites of GLP-1R Expression

The vast range of extra-pancreatic effects exerted by GLP-1 RAs (Figure 2) may be due to the combination of multiple activated intracellular signalling pathways and the breadth of GLP-1R expression across the human body. Although predominantly found in the gastrointestinal and neural tracts, GLP-1R is also present in other tissues, including kidney, skeletal and smooth muscle, liver and adipose [39]. Within the cardiovascular system, it is expressed on cardiomyocytes and vascular smooth muscle and endothelial cells [39], along with circulating inflammatory cells [40]. This may contribute to the cardiovascular benefits achieved clinically with both synthetic GLP-1 and GLP-1 RA administration [41].

2.3. Pharmacokinetics of GLP-1 Receptor Agonists

Native GLP-1 has limited clinical applications due to its labile nature, with rapid degradation by the dipeptidyl peptidase-4 (DPP-4) enzyme contributing to a short plasma half-life of two minutes [17]. GLP-1 RAs were thus designed as synthetic GLP-1 mimetics resistant to DDP-4 degradation, allowing a prolonged insulinotropic response for people with T2DM and thus limiting postprandial glucose excursions. This resistance to proteolytic degradation has been primarily achieved by alterations to the cleavage site of GLP-1, via substitution of alanine with alternative amino acids (Table 1). The endogenous half-life has also been increased through conjugation of the GLP-1 amide backbone with fatty acids, albumin, or human immunoglobulin to reduce the rate of renal elimination [42,43,44]. Therapeutic GLP-1 RAs developed to date have been predominantly administered via subcutaneous injection due to poor oral bioavailability, but the addition of sodium N-[8-(2-hydroxybenzoyl) amino caprylate (SNAC) as an absorption enhancer has circumvented this obstacle and allowed semaglutide to be given as an oral formulation [45] (Table 1).
One method of categorising drugs within the GLP-1 RA class has been according to their plasma half-life. With half-lives of 2–4 h, exenatide and lixisenatide are classed as short-acting agents (Table 1). As a result of their postprandial insulinotropic effects, these agents more closely resemble the effects of endogenous GLP-1. Long-acting GLP-1 RAs, such as liraglutide, dulaglutide and semaglutide in both oral and injectable forms, have half-lives longer than 12 h and primarily lower fasting blood glucose levels.
Due to their inhibition of gastric emptying, GLP-1 RAs may delay the absorption of orally administered medications. However, besides prolonging the Tmax value of other medications, they do not usually affect the pharmacokinetic parameters of other drugs, such as area under curve (AUC) or maximum concentration (Cmax) [46]. Although GLP-1 RAs do not typically cause hypoglycaemic episodes, there is an increased risk of developing hypoglycaemia when used in conjunction with insulin or sulfonylureas, meaning that dose adjustments may be needed [46].
The key adverse effects associated with GLP-1 RAs are gastrointestinal, such as nausea, vomiting, abdominal pain and diarrhoea [47]. These can represent obstacles to patient adherence, with up to 45% of GLP-1 RA users discontinuing therapy within five years [48]. To minimise adverse effects and improve adherence, GLP-1 RAs should be initiated at the lowest dose before gradually being uptitrated [10]. This improves tolerability, as gastrointestinal effects are most severe at initiation and generally wane over time. GLP-1 RAs are generally not recommended in patients with any history of gastrointestinal motility disorders (e.g., gastroparesis), pancreatitis, or pancreatic cancer [49]. There is also caution around using them in people with personal or family history of medullary thyroid carcinoma or multiple endocrine neoplasia type 2 (MEN2) syndrome. This stems from observations of thyroid C-cell hyperplasia in preclinical rodent studies; however, the same effect has not been replicated in large-scale human or primate studies [49]. Among other adverse effects, the SUSTAIN-6 trial of semaglutide also identified a signal for increased risk of diabetic retinopathy complications, but it remains unclear whether this is a class- or agent-specific effect or rather due to rapid HbA1c lowering [50]. The ongoing FOCUS trial (NCT03811561) will investigate this further by monitoring the progression of diabetic retinopathy in patients on semaglutide over a 5-year follow-up period.

3. Cardiovascular Benefits of GLP-1 RAs

As a class, GLP-1 RAs are extremely effective at treating hyperglycaemia and lowering HbA1c levels in people with T2DM compared to other, well-established therapies, with HbA1c level reductions in the range of 0.5–1.5% compared to placebo [51]. In general, long-acting GLP-1 RAs have been demonstrated to be superior to short-acting GLP-1 RAs for glycaemic control, weight loss and tolerability [52]. Furthermore, head-to-head comparisons have shown that semaglutide and liraglutide are the most effective GLP-1 RAs for lowering HbA1c [51].

3.1. Cardiovascular Outcome Trials Data

3.1.1. Composite Adverse Cardiovascular Endpoints

Cardiovascular outcome trials, which have been FDA-mandated since 2008 to identify potential adverse cardiovascular events caused by new anti-diabetic agents [50,53,54,55,56,57,58,59], have highlighted beneficial cardiovascular properties of GLP-1 RAs, albeit with some heterogeneity between different agents (Table 2). All GLP-1 RAs tested have demonstrated noninferiority for cardiovascular endpoints compared to placebo, thus underscoring their cardiovascular safety. Moreover, several have also achieved significant reductions compared to placebo in composite MACEs, usually comprising the endpoints of cardiovascular death, nonfatal myocardial infarction (MI) and nonfatal ischaemic stroke [50,54,56,57,59] (Table 2). Generally, long-acting agents have been associated with greater cardiovascular benefits than short-acting ones (Table 2).
Among the individual trials, the strongest benefit signals were seen with efpeglenatide and subcutaneous semaglutide, which lowered rates of MACEs by 27% (HR 0.73, 95% CI: [0.58, 0.92]) [59] and 26% (HR 0.74, 95% CI: [0.58, 0.95]) [50], respectively (Table 2). Meanwhile, albiglutide, dulaglutide and liraglutide were associated with statistically significant but more modest MACE reductions [54,56,57]. In contrast, the studies for exenatide, lisixenatide and oral semaglutide did not reach statistical significance for superiority compared to placebo [53,55,58]. However, oral semaglutide still reduced cardiovascular and all-cause death on secondary endpoint analysis [58]. The extent of oral semaglutide’s cardiovascular benefit is currently being further investigated with the SOUL trial (NCT03914326) (Table 3).
Further to the individual study results, pooled meta-analysis of over 60,000 patients enrolled in the eight major phase 3 GLP-1 RA trials, of whom 72.4% had established CVD, concluded that GLP-1 RA therapy achieves a relative reduction in MACEs of 14% (HR 0.86, 95% CI [0.80–0.93], p < 0.0001) and all-cause mortality of 12% (HR 0.88, 95% CI [0.82–0.94], p = 0.0001) over a weighted average median follow-up of three years [9]. Secondary analyses of individual MACE components showed that GLP-1 RAs reduced cardiovascular death by 13% (HR 0.87, 95% CI [0.80–0.94], p = 0.001), total MI by 10% (HR 0.90, 95% CI [0.83–0.98], p = 0.020) and total strokes by 17% (HR 0.83, 95% CI [0.76–0.92], p = 0.0002) [9]. A separate meta-analysis has found this MACE benefit is biased towards patients with preestablished cardiovascular disease, with reductions in MACEs of 15% in people with ASCVD (OR 0.85, 95% CI [0.81–0.90], p = 0.00001) compared to 6% in people with multiple cardiovascular risk factors but no ASCVD (OR 0.94, 95% CI [0.83–1.06], p = 0.31) [60].
These benefits position GLP-1 RAs as a suitable addition to the repository of secondary prevention therapies for cardiovascular disease. Another meta-analysis comparing nine diabetic drug classes recently found that in patients with T2DM at increased cardiovascular risk on metformin-based background treatment, GLP-1 RAs and SGLT2 inhibitors stood apart as reducing all-cause mortality and cardiovascular death [61]. Notably, whereas SGLT2 inhibitors reduced heart failure hospitalisation, semaglutide and dulaglutide were associated with lower risk of stroke [61]. It is also important to highlight that the evidence base for GLP-1 RAs in the different trials was achieved on other background cardioprotective therapies. For example, in SUSTAIN-6 64% of participants were also taking aspirin, 21% clopidogrel or ticagrelor, 77% lipid-lowering medication (mostly statins) and 94% anti-hypertensives with 50% on ACE inhibitors [50]. The magnitude of event reduction with GLP-1 RAs, especially semaglutide, also compares favourably with the effectiveness of other drugs commonly used to mitigate cardiovascular risk. By way of reference, a 2011 systematic review of aspirin’s use for primary prevention of MACEs in patients with diabetes demonstrated an overall risk ratio of 0.91 [62]. Meanwhile, statins are recognised to reduce major vascular events by ~21% for every 1.0 mmol/L reduction in LDL-C [63], which is similar to the overall benefit from the ACE inhibitor, ramipril, in the HOPE study where almost 40% of participants had diabetes [64]. Importantly, the effectiveness of GLP-1 RAs for reducing cardiovascular events also sits comfortably alongside other nonglucose-targeting therapies that have been introduced more recently for ASCVD. These include PCSK9 inhibitors (e.g., evolocumab [65]) and anti-inflammatory agents, such as colchicine [66].
When interpreting the results of the GLP-1 RA outcome trials, it is important to note that there was heterogeneity between studies, including differences in baseline cardiovascular risk between trial cohorts. Recruitment in most of the trials was enriched for patients with established CVD to ensure adequate statistical power within short follow-up timeframes, such as in AMPLITUDE-O (efpeglenatide) and SUSTAIN-6 (subcutaneous semaglutide) where >80% had preexisting CVD [50,59]. In contrast, REWIND (dulaglutide) recruited participants with comparatively lower cardiovascular risk, as it had the lowest baseline mean HbA1c of 7.2% and only 31.5% had known CVD [57]. Another characteristic of study design that might confound comparisons between different agents is the temporal difference in excluding patients in the early post-MI period. For instance, LEADER (liraglutide) excluded patients who had suffered a stroke or acute coronary syndrome (ACS) in the two weeks before recruitment [54], whereas SUSTAIN-6 set this exclusion window to 90 days prior [50]. As patients are at highest risk of recurrent ischaemic events in the early post-MI period [67], such differences in exclusion criteria can influence the cardiovascular event rates captured by different studies. For example, the nonbenefit for lixisenatide in the ELIXA trial may be partially attributable to its extremely high-risk cohort, where 100% of participants had established CVD and an ACS within the last 180 days [53]. Hence, it is difficult to draw firm conclusions about the relative merits of different GLP-1 RAs for cardiovascular protection.

3.1.2. Specific Endpoints of Interest

Ischaemic Heart Disease

With respect to individual cardiovascular endpoints, most of the trials have shown benefits in CAD-related outcomes. As described above, meta-analysis of outcome trials has demonstrated a mean relative reduction of 10% in the rates of new and recurrent MI by agents across the GLP-1 RA class [9]. Discounting the trials that did not show benefit for MI, this reduction ranged from 12% for liraglutide in the LEADER trial [54] up to 25% for albiglutide in the HARMONY OUTCOMES trial [56]. Moreover, in the AMPLITUDE-O, HARMONY OUTCOMES and SUSTAIN-6 trials [50,56,59], there were significant reductions in the expanded composite outcome, defined as hospitalisations for unstable angina and urgent coronary revascularisations. Notably, in SUSTAIN-6, subcutaneous semaglutide was associated with 35% reduction in revascularisation procedures of either the coronary or peripheral vessels (HR 0.65, 95% CI: [0.50, 0.86]) [50].

Stroke

GLP-1 RAs have also been linked to significant reductions in cerebrovascular events. By comparison to placebo, dulaglutide (REWIND) and subcutaneous semaglutide (SUSTAIN-6) resulted in relative reductions in nonfatal stroke of 24% and 39%, respectively [50,57]. However, this was not observed in other GLP-1 RA trials. Importantly, meta-analysis has indicated that the salutary effects on stroke outcomes were driven by reductions in ischaemic rather than haemorrhagic stroke [68]. In the primary prevention setting, GLP-1 RAs have been found to reduce the incidence of both nonfatal and total strokes by around 16% [69]. Indeed, GLP-1 RAs and thiazolidinediones are the only two classes of diabetic drugs that have been shown to reduce stroke risk [70].

Heart Failure

As a class, GLP-1 RAs do not reduce heart failure hospitalisations to the same extent as SGLT2 inhibitors, which can reduce heart-failure-related hospitalisations by ~30% [71]. Among the GLP-1 RA outcome trials, only AMPLITUDE-O showed significant reduction in risk of heart-failure-related hospitalisations with a hazard ratio of 0.61 (95% CI [0.38, 0.98]) [59]. However, the recent STEP-HFpEF study found that subcutaneous semaglutide is associated with both symptomatic and functional improvement in patients with nondiabetic obesity (body mass index, BMI ≥ 30 kg/m2) and heart failure with preserved ejection fraction (HFpEF) [72]. After 52 weeks of semaglutide at up to 2.4 mg once weekly, there was significant weight loss, with an estimated different of 10.7 percentage points compared to placebo (p < 0.001). Semaglutide was also associated with improvements in a hierarchical composite endpoint, comprising death, heart failure events, differences in the Kansas City Cardiomyopathy Questionnaire clinical summary score (KCCQ-CSS) and 6 min walk distance (win ratio, 1.72; 95% CI [1.37–2.15]; p < 0.001) [72]. Furthermore, there were also reductions in systolic blood pressure (BP), C-reactive protein (CRP) and N-terminal pro-B-type natriuretic peptide (NT-proBNP) [72]. Whether semaglutide’s effects on these parameters simply reflect the benefits of weight loss, as opposed to bona fide improvement in HFpEF, remains unclear and requires further evaluation.

Peripheral Artery Disease

Given that atherosclerosis also manifests in noncoronary territories, the effect of GLP-1 RAs on the peripheral arterial vasculature is also of major importance. However, in contrast to CAD and stroke, there are much fewer specific data on the effect of GLP-1 RAs on PAD-related outcomes. Each of the major outcome trials (SUSTAIN-6, EXCEL, HARMONY OUTCOMES, REWIND) was underpowered for these events, as the proportion of patients with CAD consistently outnumbered those with PAD [50,55,56,57]. For example, PAD was underrepresented in both LEADER and SUSTAIN-6, affecting only 12.7% and 14.0% of participants at baseline, respectively [73]. Moreover, ELIXA did not feature PAD at all in its inclusion criteria [53]. Furthermore, revascularisation outcome data were also reported as an aggregate of coronary and peripheral interventions, preventing targeted analysis of PAD-related outcomes. As such, future studies with PAD-enriched cohorts and prespecified PAD outcome analyses are required.
Nevertheless, the currently available data do suggest that GLP-1 RAs are safe and associated with a reduction in PAD events [74]. An observational real-world study of two geographically separated Italian cohorts found that GLP-1 RA use decreased rates of PAD (Lombardy cohort: HR 0.72, 95% CI [0.64–0.82]; Apulia cohort: HR 0.80, 95% CI 0.67–0.98) and lower limb complications (Lombardy cohort: HR 0.67, 95% CI [0.56–0.81]; Apulia cohort: HR 0.69, 95% CI [0.51–0.93]) [75]. This was supported by a Danish cohort study of 309,166 patients with diabetes, which similarly showed that GLP-1 RAs were associated with a 50% lower rate of diabetes-associated amputations compared to those not on GLP-1 RA treatment (HR 0.50, 95% CI [0.54–0.74], p < 0.005) [76]. With these signals of safety and possible benefit for PAD-related outcomes, GLP-1 RAs could potentially be adopted as the preferred therapy over other anti-diabetic medications in patients with concurrent T2DM and PAD [77]. This is especially the case given that the CANVAS program identified a twofold increased risk of lower limb amputation with the SGLT2 inhibitor canagliflozin compared to placebo [78]. Indeed, a recent meta-analysis suggested that GLP-1 RAs were associated with significantly lower incidence of lower limb amputations than SGLT2 inhibitors (3.54 ± 3.18 versus 4.72 ± 3.99 events per 1000 patient-years, p = 0.004) [79]. Moving forward, several studies (e.g., STRIDE, STARDUST, LEADPACE) are set to expand upon this area of knowledge by investigating how GLP-1 RAs may affect functional movement and peripheral endothelial function in patients with PAD (Table 3).

3.2. Observational Cardiovascular Data and Real-World Experience

Real-world experience has also been reassuring and congruent with CVOT findings about the safety and benefits of GLP-1 RAs in wider populations of high-cardiovascular-risk patients. Svanström et al. performed a retrospective registry-based cohort analysis of 46,804 patients with T2DM in Denmark and Sweden, who were evenly distributed between the use of liraglutide and DDP-4 inhibitors [80]. Compared to those on DDP-4 inhibitors, patients using liraglutide had a 10% lower risk of MACEs, 22% lower risk of cardiovascular death (unadjusted HR 0.78, 95% CI: [0.68, 0.91]) and 17% lower risk of all-cause death (unadjusted HR 0.83, 95% CI: [0.77, 0.90]) [80]. These results also held with a sensitivity analysis that accounted for HbA1c, smoking status, BP, albuminuria, estimated glomerular filtration rate (eGFR) and BMI [80]. Meanwhile, Trevisan et al. studied 17,868 patients with T2DM and prior MI from the SWEDEHEART registry. Although only used in 2% of the cohort, GLP-1 RAs were associated with 28% lower event risk over a median follow-up of 3 years compared to standard diabetic care (95% CI [0.56, 0.92]) [81]. This was mostly driven by reductions in stroke and re-infarction and was consistent after propensity score matching and across different subgroups. Given the high-fidelity environment of these real-world studies, the results are generalisable to wider populations.
Given the findings of the CVOTs and recent real-world experience, the use of GLP-1 RAs has now become guideline-recommended by the European Society for Cardiology, American Diabetes Association and European Association for the Study of Diabetes in patients with T2DM and concurrent ASCVD or at high risk of ASCVD, such as those with target organ damage or multiple cardiovascular risk factors [82,83]. These agents can be commenced either firstline in drug-naïve patients or secondline in patients already using metformin or other glucose-lowering agents, including insulin.

4. Does Risk Factor Modulation Explain the Cardiovascular Benefit of GLP-1 RAs?

The reductions in cardiovascular outcomes associated with GLP-1 RAs appear to be driven predominantly by reductions in atherosclerotic and/or ischaemic events. However, it is unclear to what extent this is simply mediated by improvements in cardiovascular risk factors, including glycaemia and weight loss, or if additional mechanisms are involved.

4.1. Glycaemic Control

Diabetic hyperglycaemia fosters a pro-inflammatory and oxidative microenvironment that is conducive to the development of ASCVD and its ischaemic complications [84]. While better glycaemic control has long been associated with improved microvascular complications of T2DM, it has been more difficult to demonstrate this relationship for ASCVD (or macrovascular) outcomes [85]. Mediation analyses suggest that glucose lowering may only partially account for the cardiovascular benefits of GLP-1 RA therapy. For example, it has been determined from the LEADER and REWIND trials that HbA1c lowering contributed to 36–41% of cardiovascular event reduction [86,87]. It is also worth noting that in these analyses, changes in other metabolic variables, such as bodyweight, systolic BP and low-density lipoprotein cholesterol (LDL-C), did not reach significance for mediation [86,87]. Other analyses suggest that any MACE reductions associated with HbA1c lowering are predominantly driven by reductions in nonfatal stroke [88].

4.2. Weight Loss

Weight loss can favourably modify cardiometabolic risk factors [89] and reverse the chronic inflammation of an obesogenic microenvironment [90]. Known to be a positive prognostic factor for cardiac outcomes, weight loss also lowers the incidence of clinical endpoints, such as urgent revascularisations, acute ASCVD events and total mortality [91]. Diabetic obesity has also been independently associated with coronary plaque burden, emphasising the need to target this cardiovascular risk factor [92].
Although GLP-1 RAs are one of the most effective agents within the existing pharmacological armamentarium for obesity [93], the degree of weight loss varies between different agents (Table 2). In SUSTAIN-6, 0.5 mg and 1.0 mg doses of subcutaneous semaglutide resulted in mean weight loss of 3.6 kg and 4.9 kg from baseline, compared to 0.7 kg and 0.5 kg with placebo, respectively [50]. For patients with overweight or obesity who did not have T2DM, 2.4 mg of semaglutide in the STEP1 trial led to mean bodyweight reduction of 14.9% (2.4% with placebo), with 86% and 51% of participants achieving ≥5% and ≥15% bodyweight reductions, respectively [94]. Real-world data have shown that high doses of semaglutide (1.7 mg and 2.4 mg) are associated with mean bodyweight percentage reductions of 6% after three months and 12% after six months [95].
It is therefore tempting to causally link the cardiovascular benefits of GLP-1 RA therapy with its associated weight loss. However, weight loss appears to be mostly independent of MACE-lowering effects, as seen in the outcome study data. This is highlighted by similar reductions for primary MACE composite outcomes with dulaglutide and albiglutide, which only yield modest weight loss, and semaglutide and liraglutide, which achieve more substantial weight loss (Table 2). Furthermore, the magnitude of GLP-1 RA-induced weight loss does not fully account for the degree of cardiovascular benefit observed. Bodyweight reductions of 2–5 kg have been shown previously in epidemiological studies to be insufficient to achieve the degree of cardiovascular risk reduction seen in the GLP-1 RA outcome trials. Notably, intensive lifestyle intervention for weight loss in overweight/obese patients did not provide mortality benefit in the LOOK AHEAD trial [96], although post hoc analysis did find that >10% weight loss within the first year of intervention was associated with a 21% reduction in cardiovascular mortality [97]. The Swedish Obese Subjects study supported this, finding that modest weight loss of 5 kg was not enough to alter CVD risk, whereas 10–44 kg weight loss in the setting of bariatric surgery was needed for significant risk factor reduction at 10-year follow-up [98]. A systematic review concurred with these epidemiological observations, concluding that bariatric surgery is superior to GLP-1 RA therapy with regard to weight loss (mean difference −22.68 kg) and BMI reduction (mean difference −8.18 kg/m2) [99]. Retrospective cohort studies have found that due to the magnitude of weight loss, bariatric surgery is superior to GLP-1 RAs for reducing MACEs, although the use of second-generation GLP-1 RAs such as semaglutide was underrepresented in these studies [100,101].
The impetus to prescribe GLP-1 RAs primarily for obesity is continually growing. Although the cardiovascular benefits of using GLP-1 RAs for diabetic patients have been well-documented [9], there was previously a paucity of evidence for using these agents for cardiovascular risk reduction in nondiabetic overweight/obesity. With 17,604 participants aged ≥ 45 years across over 800 sites, the SELECT trial recently showed that once-weekly subcutaneous semaglutide 2.4 mg significantly lowered MACEs by 20% compared to placebo for overweight and obese patients (BMI ≥ 27) with preexisting ASCVD but without diabetes [91] (Table 3). This is in addition to post hoc analysis showing that weight loss was sustained in this cohort for up to four years [102]. An early divergence of the time-to-first-event curves within three months of treatment initiation in SELECT means that the onset of semaglutide’s cardiovascular benefits preceded most of the achieved weight loss or other changes to cardiometabolic risk factors [91]. This is important because it suggests that mechanisms beyond just weight loss are helping to mediate the cardioprotective effects of GLP-1 RAs [91]. Consequently, although semaglutide 2.4 mg (Wegovy) had already been approved in 2021 for chronic weight management in adults with obesity or overweight and at least one weight-related condition, the FDA has now expanded this indication to cardiovascular risk reduction in people with concurrent CVD and overweight/obesity.

4.3. Blood Pressure

All outcome trials also reported reductions in systolic BP [50,53,54,55,56,57,58,59], ranging from an average of 0.65 mmHg with albiglutide [56] to 2.57 mmHg with 1.0 mg subcutaneous semaglutide [50]. Meanwhile, changes in diastolic BP were variable and not significant. A meta-regression analysis has further clarified that reductions in systolic BP are independent of weight loss and HbA1c improvements, raising the point that GLP-1 RAs have an independent anti-hypertensive effect [103].

4.4. Lipids

GLP-1 RAs generally have a positive yet modest effect on blood lipid profiles. In PIONEER 6, oral semaglutide was associated with a reduction of 4–5% in total cholesterol and LDL-C levels and 12% in triglyceride levels from baseline [58]. Similarly, subcutaneous semaglutide in SUSTAIN-6 reduced triglycerides by 7–8% and both total cholesterol and LDL-C by 3% compared to baseline levels [50]. Other trials reported statistically, but not clinically, significant improvements in LDL-C levels [55,57,59]. GLP-1 RAs have also shown potential for use as an adjunctive lipid control measure within real-world settings. For example, liraglutide combined with metformin lowered levels of atherogenic lipoproteins in patients with T2DM and CAD who were already taking statins [104]. Postprandial reductions in remnant cholesterol, an aggregate of very low-density lipoprotein (VLDL) and intermediate-density lipoprotein (IDL), may be mediated by liraglutide-driven reductions of ApoC III [105]. Moreover, detailed lipidomic analysis revealed that liraglutide significantly lowers blood concentrations of multiple lipid species, including ceramides, phosphatidylcholines, phosphatidylethanolamines and triglycerides [106]. Meta-analysis data have also associated GLP-1 RAs with modest lowering of total cholesterol, LDL-C and triglyceride levels, but direct causality between GLP-1 RA-mediated lipid-lowering effects and improved cardiovascular outcomes has not been adequately investigated [107].
The totality of clinical evidence so far suggests that the cardiovascular benefits of GLP-1 RAs cannot be easily attributed to changes in a single risk factor, whether it be glycaemia, weight or other cardiometabolic factors. This has led researchers to explore the effects of GLP-1 RAs on other mechanistic drivers of ASCVD (Table 2).

5. Evidence of GLP-1 RAs’ Direct Impact on Atherosclerotic Plaques

Numerous preclinical models suggest that the effect of GLP-1 RAs on reducing ischaemic macrovascular events is mediated through direct attenuation of the underlying atherosclerotic substrate. Murine models have particularly contributed to this evidence. Liraglutide and semaglutide both resulted in decreased plaque area and aortic intimal thickening in atherosclerotic models in Apoe−/− and Ldlr−/− mice, with this occurring independently of weight loss and cholesterol-lowering effects [108]. Although both early and late initiation of dulaglutide reduced plaque area in Apoe−/− mice, intervention at early- but not late-stage diabetes was associated with significantly reduced macrophage infiltration into aortic root lesions [109]. Hence, greater attenuation of plaque inflammation may be seen if GLP-1 RAs are administered earlier in the diabetic and atherosclerotic process [109].
In addition to murine research, a study in Ldlr−/− Watanabe heritable hyperlipidaemic (WHHL) rabbits showed that lixisenatide promoted aortic plaque stability, with increased fibrotic tissue and reduced necrotic and calcified areas within plaques, relative to the control group [110]. However, despite these preclinical observations it is interesting that lixisenatide did not reduce MACEs in the ELIXA trial [53]. This discrepancy could potentially be explained by the use of osmotic mini-pumps in the rabbit study, which may have artificially improved the pharmacokinetic efficacy of lixisenatide by circumventing its short half-life [111].
Although there appears to be a positive association between serum GLP-1 levels in patients with CAD and their degree of coronary plaque burden, the causal nature and clinical significance of this finding remain unclear [112]. Several human studies are underway to investigate if GLP-1 RAs directly attenuate plaque formation or at least modulate the composition of plaques to a more passive or stable phenotype (Table 3). The Semaglutide Treatment on Coronary Plaque Progression (STOP) trial (NCT03985384) used serial coronary computed tomography angiography (CCTA) to explore the effect of subcutaneous semaglutide on coronary plaque progression in patients with T2DM who had established ASCVD or at least one additional CVD risk factor [113]. Although unpublished data indicated no significant reduction in plaque volume, there was a signal for a ‘stabilisation’ effect, whereby semaglutide was associated with increased conversion of noncalcified plaque to calcified plaque. The ongoing SAMAS trial (NCT05147896) will examine how oral semaglutide affects parameters of ASCVD, such as arterial stiffness, carotid intima–media thickness and endothelial function, along with atherosclerotic risk factors (e.g., LDL-C, HbA1c, high-sensitivity CRP) [114].

6. Putative Anti-Atherosclerotic Mechanisms

Increasing evidence shows that GLP-1 RAs exert pleiotropic effects on different mediators of atherogenesis, which may help explain their benefits on ASCVD events beyond cardiometabolic risk factor control (Figure 3).

6.1. Correcting Vascular Dysfunction

An intact and functioning endothelium is crucial for maintaining physiological vascular function, with endothelial dysfunction being the first step of the atherogenic cascade. GLP-1 is known to possess vasodilatory effects, due to its direct actions on vascular endothelium. Infusion of GLP-1 has been associated with favourable endothelial function and nitric oxide synthase (NOS)-mediated vasodilation [115]. There is also evidence that it causes vasodilation independent of adenosine [116]. Furthermore, GLP-1 RA-mediated vasodilation can occur in states of low Nox-1/endothelin-1 and normal eNOS/guanylyl cyclase expression [117]. In a mouse study, GLP-1 RA therapy was also found to delay the progression of atherosclerosis through AMP-activated protein kinase (AMPK)-dependent arrest of angiotensin-II-induced vascular smooth muscle cell (VSMC) proliferation [118]. Through targeting AMPK/SIRT1/FoxO3a pathways, healthy vascular function is achieved by maintaining a calponin+SM22α+ VSMC phenotype [119].
With particular focus on diabetic angiopathy, GLP-1 RAs may also attenuate atherosclerotic progression due to the vasoprotective effects of reducing advanced glycosylation end-products (AGEs). In addition to anti-glucolipotoxic effects on the endothelium [120], treatment in Apoe−/− mice inhibits formation of the receptor for advanced glycosylation end-products (RAGE) in the aorta and lowers serum AGE levels [121]. This presents a new opportunity for the treatment of vascular complications associated with diabetes, since the AGE/RAGE interaction is a key component in the progression of diabetic atherosclerosis.

6.2. Targeting Diabetic Dyslipidaemia

Moderating lipid levels is crucial to reducing atherogenesis [122]. GLP-1R agonism modulates different steps of cholesterol homeostasis on both genetic and protein levels. It results in downregulation of lipogenic genes and upregulation of lipolysis in human adipocytes, attenuating a systemic obesogenic state [123]. The GLP-1R is also involved in cholesterol efflux from foam cells mediated by the ATP-binding cassette transporter (ABCA1) [124]. GLP-1 RAs can further downregulate acetyl-CoA acetyltransferase 1 (ACAT1) expression [125] and upregulate signalling between adaptor protein phosphotyrosine interacting with pleckstrin homology (PH) domain and leucine zipper 1 (APPL1) and adiponectin, which suppresses foam cell formation [126]. Whilst the effects of GLP-1 RA administration alone on lipogenesis have not been studied, co-administration of GLP-1 RA with glucagon reduces lipogenesis by lowering expression of β-hydroxy β-methylglutaryl-CoA (HMG-CoA) reductase and sterol regulatory element-binding protein-1c (SBREBP-1C) [127]. Furthermore, GLP-1/glucagon co-agonism improves reverse cholesterol transport and bile acid homeostasis through increased expression of LDLR/ABCA1 and cytochrome P450 family 7 subfamily A member 1 (CYP7A1)/ATP-binding cassette, subfamily B member 11 (ABCB11), respectively [127]. Finally, GLP-1 RAs may have a role in treating nonalcoholic steatohepatitis through lowering hepatic inflammation, steatosis and fibrosis, although the mechanistic basis for this is yet to be determined [128].

6.3. Dampening Inflammation

A chronically dysregulated inflammatory response underpins the progression of atherosclerosis [129]. It is thought that GLP-1 RAs also mediate atheroprotective effects by dampening inflammation systemically, as well as locally in the arterial wall. To this end, they have been shown to downregulate transcriptional expression of genes involved in both inflammation and oxidative stress. In addition to reductions in plasma levels of interferon-γ (IFN-γ) and tumour necrosis factor-α (TNF-α), semaglutide also lowers mRNA levels of other cytokines, such as interleukin-6 (IL-6), chemokine ligand 2 (CCL2) and vascular cell adhesion molecule-1 (VCAM-1), which are involved in leukocyte recruitment and extravasation [108]. Furthermore, GLP-1R agonism can reduce gene transcription of nuclear factor kappa-B (NF-κB) and superoxide dismutase 2 (SOD2) by reversing the hyperglycaemia-induced DNA demethylation [130].
Liraglutide was found to confer resistance to TNF-α- and liposaccharide-induced inflammation and inhibit monocyte recruitment to the vascular endothelium by lowering VCAM-1 and E-selectin expression in cultured human aortic endothelial cells [131]. This was speculated to result from increased calcium/calmodulin-dependent protein kinase I (CaMKI)/CREB levels and induction of calcium/calmodulin-dependent protein kinase kinase-β (CaMKKβ)/AMPK signalling [131]. Furthermore, through the extracellular-signal-regulated kinase 5 (ERK5) pathway, GLP-1 RAs have been shown to increase Krüppel-like factor 2 (KLF2) levels and prevent inhibition of mitogen-activated protein kinase (MAPK), which causes downstream anti-inflammatory effects, including decreased leukocyte adhesion [132]. GLP-1 RAs can additionally guard against hyperglycaemia-induced autoinflammatory damage, inhibiting NLR family pyrin domain-containing 3 (NLRP3) inflammasome formation and thereby conferring anti-pyroptotic effects on cardiomyocytes [133].
There is also evidence for modulatory effects of GLP-1 RAs on macrophage biology. In addition to reducing plaque size and necrotic core area in Apoe−/− Irs2+/− mice, lixisenatide was found to result in a greater proportion of M2-like anti-inflammatory (STAT3+Arginase-1+) macrophages in aortic plaques, with fewer M1-like, pro-inflammatory (STAT1+iNOS+) macrophages [134]. Similarly, liraglutide increased M2-like macrophage populations in Apoe−/− mice and increased aortic plaque expression of the anti-inflammatory mediators IL-10 and Arg-1 [135]. Notably, the oxidative-stress-sensitive channel TRPM2 has also recently been shown to play a role in atherogenesis by promoting macrophage recruitment, foam cell formation and vascular inflammation within atherosclerotic plaques [136,137]. Similarly, it can exacerbate N-methyl-D-aspartate (NMDA) receptor-related excitotoxicity and neuronal death in ischaemic stroke [138]. Given TRPM2’s role in both atherosclerosis and GLP-1-induced insulin secretion as discussed above [21], further research on GLP-1/TRPM2 interactions could enable further pharmacological targeting of macrophage-related inflammation in diabetes and ASCVD.
Despite these potential anti-inflammatory mechanisms, molecular imaging studies have yielded discrepant results about whether GLP-1 RAs can attenuate vascular wall inflammation. Positron emission tomography (PET) imaging of rabbits identified that semaglutide reduced macrophage activation and metabolism in the aortic wall by using [64Cu]Cu-DOTATATE and [18F]FDG radiotracers, respectively [139]. Conversely, the LIRAFLAME trial failed to show a significant effect of liraglutide compared to placebo on vascular inflammation as assessed by [18F]FDG PET after 26 weeks of treatment in people with T2DM [140]. To clarify these discrepant results, further human imaging studies are needed to elucidate the effects of GLP-1 RAs on atherosclerotic plaque burden and composition.

6.4. Reduction in Endoplasmic Reticulum Stress

In addition to reducing endoplasmic reticular (ER) stress in pancreatic β cells in diabetes, GLP-1 RAs can also alleviate the lipotoxic stress that the atherosclerotic milieu chronically places on the ER in macrophages and endothelial cells, thereby helping to mitigate the formation of necrotic cores that are characteristic of unstable plaques [141]. This has been demonstrated through a reduction in levels of inositol-requiring enzyme 1α (IRE1α) and its downstream target c-Jun N-terminal kinase (JNK), which are markers of acute ER stress [142]. Liraglutide was also shown to reverse dextrose- and tunicamycin-induced ER stress in human coronary artery endothelial cells by downregulating the expression of glucose-regulated protein 78 (GRP78)/ATF6 and phosphorylation of protein kinase RNA-like endoplasmic reticulum kinase (PERK)/IRE1α, and this occurred to a greater extent than with SGLT2 inhibition or metformin [143]. In other work, the activation of p38 MAPK by exendin-4 was proposed to protect against lipoapoptosis [144]. Finally, there is also evidence to support that GLP-1R agonism can stabilise plaques not only by limiting inflammation and oxidative stress but also by increasing the expression of sirtuin 6, a DNA repair deacetylase [145]. Taken together, the ability of GLP-1 RAs to limit cellular damage mediated by ER stress is another mechanism by which they may help attenuate atherogenesis and passivate plaques to prevent plaque rupture and atherothrombotic events.

6.5. Regulating Adiponectin and Other Adipokines

Adipokine dysregulation, which presents as hypoadiponectinaemia and hyperleptinaemia, contributes to the insulin resistance seen in T2DM. In fact, low serum adiponectin levels may be an independent predictor for future ASCVD events and a marker of CAD severity [146]. To this end, GLP-1 RAs, such as exendin-4, have been shown to increase adiponectin levels via Sirt1/FoxO1 signalling [147]. Moreover, a meta-analysis of 20 randomised controlled trials revealed that, independent of fat mass changes, liraglutide significantly increased circulating adiponectin levels [148].

6.6. Modification of Epicardial Adipose Tissue

Epicardial adipose tissue (EAT), which is a store of visceral fat located between the myocardium and pericardium, has been implicated in the pathogenesis of ASCVD and has been shown to confer a pro-inflammatory milieu for the neighbouring coronary arteries [149]. With GLP-1R expressed in EAT at much higher levels than in subcutaneous tissue [150], GLP-1 RAs may be able to target EAT for cardiovascular benefit. Dose-dependent reductions in EAT thickness have been observed for both semaglutide and dulaglutide in obese patients with T2DM [151]. This was confirmed via pooled analysis, with a mean reduction of 1.83 mm in EAT thickness following GLP-1 RA use [152]. This presents an exciting and novel therapeutic target for ASCVD prevention and treatment, as reductions in pericoronary EAT have been shown in mechanistic studies to attenuate coronary atherosclerosis [153]. There is ongoing work in this area, including elucidating the modulatory effects of liraglutide on the EAT microenvironment (NCT03260881) (Table 3).

6.7. Remodeling Plaque towards Stability

Plaques that are vulnerable to rupture have a distinctive pathological phenotype, comprising extensive inflammatory infiltration and a necrotic core with an overlying thin fibrous cap [154]. Matrix metalloproteinases (MMPs) degrade the collagen and other extracellular matrix proteins in thin-cap fibroatheromas, resulting in increased vulnerability of the fibrous cap to mechanical forces in the artery and the potential for plaque rupture and thrombosis [154]. Exenatide was shown in a murine model of atherosclerosis to increase the collagen content of plaque and decrease MMP-9/MMP-2 activity, thereby helping to stabilise plaque [155]. This has been proposed to occur via activation of tissue inhibitors of metalloproteinases (TIMPs), which are natural inhibitors of MMPs [156]. In another study, semaglutide reduced the plaque content of CD163+ macrophages [108], which is associated with decreased intraplaque angiogenesis and a lower risk of plaque haemorrhage and rupture [108,157]. The ability of GLP-1 RAs to passivate or stabilise plaque requires further evaluation in human studies but may be a novel and important mechanism by which this class confers a reduced risk of future and recurrent ischaemic events, including both MI and ischaemic stroke.

6.8. Inhibition of Platelet Aggregation and Thrombosis

Platelet aggregation following plaque rupture or erosion causes arterial thrombosis, which restricts tissue perfusion and leads to infarction distal to the occlusion site. In addition to potentially making plaques less vulnerable to rupture, there is also some evidence that GLP-1 RAs may exert anti-thrombotic properties. In human cellular studies, exenatide inhibited the aggregation of platelets induced by thrombin, adenosine diphosphate (ADP) and collagen [158]. Meanwhile, low-dose liraglutide acutely attenuated the aggregation of platelets induced by thromboxane in a small study of nine participants with obesity and prediabetes [159]. Both GLP-1 amides and liraglutide have been demonstrated to impart numerous other effects on platelet biology; these include reduction of arachidonic-induced oxidative stress, increased cGMP/protein kinase G (PKG)/vasodilator-stimulated phosphoprotein (VASP) signalling and reduced PI3K/PKB and MAPK signalling [160]. Furthermore, GLP-1R agonism may also inhibit platelet activation by causing platelets to have higher levels of NO and lower P-selectin and platelet activation complex-1 (PAC-1) [161].
Collectively, there is therefore a large body of mechanistic data to show that GLP-1 RAs exert pleiotropic actions on platelets and immune and vascular cells that would protect against the development and complications of atherosclerosis independently of, and in addition to, their canonical effects on glycaemia, appetite and weight regulation.

7. Future Directions

Although the striking cardiometabolic benefits of GLP-1 RAs in T2DM are well-established, there is evidence that prescribing rates around the world have been suboptimal. Alarmingly, this has also applied to specialist clinic settings for those with T2DM and established CAD, including prior MI [162]. This clinical inertia may stem from insufficient awareness among cardiologists and other physicians regarding the cardioprotective nature of GLP-1 RAs and the mechanistic uncertainty about how they exert their benefits. Paradoxically, as the weight loss properties of GLP-1 RAs have gained widespread attention through traditional and social media platforms, the increasing demand for this off-label indication has created a global shortage, limiting accessibility in many countries. This has prompted critical discussions about prioritising access between different patient subgroups. For many patients with T2DM who have failed other treatments, adequate glycaemic control may be reliant on GLP-1 RA use. It is simultaneously true that with obesity disproportionately affecting lower socioeconomic populations [163], designating weight loss as an off-label indication for GLP-1 RAs could be tantamount to financial gatekeeping, potentially further entrenching health inequities within these populations. Similarly, as we learn more about how GLP-1 RAs protect against atherosclerosis and CVD with trials such as SELECT [91], we will need to consider how management of ASCVD with these agents fits within this wider issue of equitable access and the effects of withdrawal of treatments, which can result in weight regain and reversal of cardiometabolic benefits [164].
Opportunities for intensifying cardiovascular risk reduction in patients with T2DM have also been generated by combining GLP-1 RAs with other therapies, including SGLT2 inhibitors [165]. Based on their potential synergism, a fusion therapy comprising GLP-1 RA with the potent cholesterol-lowering of proprotein convertase subtilisin/kexin type 9 inhibition (PCSK9i) is being developed, especially with an eye to patients who are intolerant or refractory to statins [166]. Recent trial evidence also suggests a beneficial effect of GLP-1 RA use on diabetic renal disease [167], which supports signals observed in the early GLP-1 trials [9].
Beyond GLP-1 RAs, a growing wave of more potent incretin-based treatments now offers the potential for even better glycaemic control and weight loss benefits. Tirzepatide, a novel ‘twincretin’ targeting both GLP-1 and GIP receptors, has demonstrated efficacy for treating T2DM in the SURPASS clinical trial program [168] and overweight/obesity in the SURMOUNT studies [169]. This resulted in FDA approval for T2DM in May 2022 and chronic weight management in late 2023. Although the cardiovascular safety of tirzepatide has been established in short-term trials [170], proof of beneficial effects on hard cardiovascular outcomes is still awaited with dedicated outcome trials ongoing (SURPASS-CVOT, NCT04255433; SURMOUNT-MMO, NCT05556512) (Table 3). Additionally, within the incretin drug pipeline, retatrutide, a unimolecular peptide with tri-agonist activity at the GIP, GLP-1 and glucagon receptors, recently showed promising results in a phase 2 evaluation for both weight loss and glycaemic control [171]. Finally, orforglipron is being developed as the first nonpeptide oral alternative to injectable GLP-1 RAs to help overcome adherence issues of injectable therapies. Early trials have found benefits for both obesity [172] and diabetes [173], expanding oral GLP-1 RA choices. Both retatrutide and orforglipron are now undergoing phase 3 evaluation (Table 3). The increased availability of oral options for GLP-1 RAs may also open up the possibility of their incorporation into fixed-dose polypill combinations [174]. This is particularly important as polypill fixed-dose combinations with statins, ACE inhibitors and aspirin have previously been shown to increase adherence and reduce cardiovascular endpoints by 24–30% compared to standard care [175].

8. Conclusions

As the field of anti-diabetic treatment continues to evolve rapidly, GLP-1 RAs are gaining attention for their beneficial effects on cardiovascular outcomes and specifically their ability to lower the risk of ischaemic events. A growing body of evidence indicates that beyond improving cardiometabolic health, these drugs may also exert direct anti-atherosclerotic properties via pleiotropic mechanisms that target numerous cellular and molecular mediators implicated in the different stages of plaque formation, destabilisation and thrombosis. Given the established beneficial effects on ASCVD risk reduction, GLP-1 RAs should be considered as the preferred agent for decreasing ASCVD risk in patients with T2DM and overweight/obesity.

Author Contributions

Conceptualisation, R.L. and P.J.P.; writing—original draft preparation, R.L. and P.J.P.; writing—review and editing, R.L., M.T.N., M.A.A., J.P.P., C.S.M., J.A.M. and P.J.P.; visualisation, R.L.; supervision, C.S.M., J.A.M. and P.J.P. All authors have read and agreed to the published version of the manuscript.

Funding

P.J.P. received a L3 Future Leader Fellowship from the National Heart Foundation of Australia (FLF106656).

Acknowledgments

The authors acknowledge the editorial support of Tulin Fisk and Benjamin Lewis from the Lifelong Health Theme, SAHMRI.

Conflicts of Interest

R.L., M.T.N., M.A.A., J.P.P. and J.A.M. have nothing to declare. C.S.M. has received speaking honoraria from Boehringer Ingelheim, Novo Nordisk and Ely Lilly. P.J.P. has received consulting fees from Amgen, Eli Lilly and Esperion and speaker honoraria from Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Merck Schering-Plough, Novartis, Novo Nordisk, Pfizer and Sanofi.

References

  1. Libby, P. The Changing Landscape of Atherosclerosis. Nature 2021, 592, 524–533. [Google Scholar] [CrossRef] [PubMed]
  2. Roth, G.A.; Mensah, G.A.; Johnson, C.O.; Addolorato, G.; Ammirati, E.; Baddour, L.M.; Barengo, N.C.; Beaton, A.Z.; Benjamin, E.J.; Benziger, C.P.; et al. Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019. J. Am. Coll. Cardiol. 2020, 76, 2982–3021. [Google Scholar] [CrossRef] [PubMed]
  3. Wolf, D.; Ley, K. Immunity and Inflammation in Atherosclerosis. Circ. Res. 2019, 124, 315–327. [Google Scholar] [CrossRef] [PubMed]
  4. Saeedi, P.; Petersohn, I.; Salpea, P.; Malanda, B.; Karuranga, S.; Unwin, N.; Colagiuri, S.; Guariguata, L.; Motala, A.A.; Ogurtsova, K.; et al. Global and Regional Diabetes Prevalence Estimates for 2019 and Projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9th Edition. Diabetes Res. Clin. Pract. 2019, 157, 107843. [Google Scholar] [CrossRef]
  5. Lobstein, T.; Brinsden, H.; Neveux, M. World Obesity Atlas 2022; World Obesity Federation: London, UK, 2022. [Google Scholar]
  6. Bertoluci, M.C.; Rocha, V.Z. Cardiovascular Risk Assessment in Patients with Diabetes. Diabetol. Metab. Syndr. 2017, 9, 25. [Google Scholar] [CrossRef]
  7. The GBD 2015 Obesity Collaborators. Health Effects of Overweight and Obesity in 195 Countries over 25 Years. N. Engl. J. Med. 2017, 377, 13–27. [Google Scholar] [CrossRef] [PubMed]
  8. Hwang, T.; Franklin, J.; Kesselheim, A. Effect of US Food and Drug Administration’s Cardiovascular Safety Guidance on Diabetes Drug Development. Clin. Pharmacol. Ther. 2017, 102, 290–296. [Google Scholar] [CrossRef] [PubMed]
  9. Sattar, N.; Lee, M.M.Y.; Kristensen, S.L.; Branch, K.R.H.; Del Prato, S.; Khurmi, N.S.; Lam, C.S.P.; Lopes, R.D.; McMurray, J.J.V.; 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]
  10. Honigberg, M.C.; Chang, L.-S.; McGuire, D.K.; Plutzky, J.; Aroda, V.R.; Vaduganathan, M. Use of Glucagon-Like Peptide-1 Receptor Agonists in Patients with Type 2 Diabetes and Cardiovascular Disease: A Review. JAMA Cardiol. 2020, 5, 1182. [Google Scholar] [CrossRef]
  11. Mcintyre, N.; Holdsworth, C.D.; Turner, D.S. New Interpretation of Oral Glucose Tolerance. Lancet 1964, 284, 20–21. [Google Scholar] [CrossRef]
  12. Elrick, H.; Stimmler, L.; Hlad, C.J.; Arai, Y. Plasma Insulin Response to Oral and Intravenous Glucose Administration1. J. Clin. Endocrinol. Metab. 1964, 24, 1076–1082. [Google Scholar] [CrossRef]
  13. Nauck, M.A.; Meier, J.J. The Incretin Effect in Healthy Individuals and Those with Type 2 Diabetes: Physiology, Pathophysiology, and Response to Therapeutic Interventions. Lancet Diabetes Endocrinol. 2016, 4, 525–536. [Google Scholar] [CrossRef] [PubMed]
  14. Kreymann, B.; Ghatei, M.A.; Williams, G.; Bloom, S.R. Glucagon-Like Peptide-1 7–36: A Physiological Incretin in Man. Lancet 1987, 330, 1300–1304. [Google Scholar] [CrossRef] [PubMed]
  15. Dupre, J.; Ross, S.A.; Watson, D.; Brown, J.C. Stimulation of Insulin Secretion by Gastric Inhibitory Polypeptide in Man. J. Clin. Endocrinol. Metab. 1973, 37, 826–828. [Google Scholar] [CrossRef] [PubMed]
  16. Nauck, M.A.; Meier, J.J. Incretin Hormones: Their Role in Health and Disease. Diabetes Obes. Metab. 2018, 20, 5–21. [Google Scholar] [CrossRef] [PubMed]
  17. Müller, T.D.; Finan, B.; Bloom, S.R.; D’Alessio, D.; Drucker, D.J.; Flatt, P.R.; Fritsche, A.; Gribble, F.; Grill, H.J.; Habener, J.F.; et al. Glucagon-like Peptide 1 (GLP-1). Mol. Metab. 2019, 30, 72–130. [Google Scholar] [CrossRef] [PubMed]
  18. Wishart, J.M.; Horowitz, M.; Morris, H.A.; Jones, K.L.; Nauck, M.A. Relation between Gastric Emptying of Glucose and Plasma Concentrations of Glucagon-like Peptide-1. Peptides 1998, 19, 1049–1053. [Google Scholar] [CrossRef]
  19. Nauck, M.A.; Heimesaat, M.M.; Behle, K.; Holst, J.J.; Nauck, M.S.; Ritzel, R.; Hüfner, M.; Schmiegel, W.H. Effects of Glucagon-Like Peptide 1 on Counterregulatory Hormone Responses, Cognitive Functions, and Insulin Secretion during Hyperinsulinemic, Stepped Hypoglycemic Clamp Experiments in Healthy Volunteers. J. Clin. Endocrinol. Metab. 2002, 87, 1239–1246. [Google Scholar] [CrossRef] [PubMed]
  20. Leech, C.A.; Chepurny, O.G.; Holz, G.G. Epac2-Dependent Rap1 Activation and the Control of Islet Insulin Secretion by Glucagon-Like Peptide-1. In Vitamins & Hormones; Elsevier: Amsterdam, The Netherlands, 2010; Volume 84, pp. 279–302. ISBN 978-0-12-381517-0. [Google Scholar]
  21. Pang, B.; Kim, S.; Li, D.; Ma, Z.; Sun, B.; Zhang, X.; Wu, Z.; Chen, L. Glucagon-like Peptide-1 Potentiates Glucose-stimulated Insulin Secretion via the Transient Receptor Potential Melastatin 2 Channel. Exp. Ther. Med. 2017, 14, 5219–5227. [Google Scholar] [CrossRef]
  22. Jones, B.; McGlone, E.R.; Fang, Z.; Pickford, P.; Corrêa, I.R.; Oishi, A.; Jockers, R.; Inoue, A.; Kumar, S.; Görlitz, F.; et al. Genetic and Biased Agonist-Mediated Reductions in β-Arrestin Recruitment Prolong cAMP Signaling at Glucagon Family Receptors. J. Biol. Chem. 2021, 296, 100133. [Google Scholar] [CrossRef]
  23. Zhang, Z.; Li, J.; Jiang, X.; Yang, L.; Lei, L.; Cai, D.; Zhang, H.; Chen, H. GLP-1 Ameliorates the Proliferation Activity of INS-1 Cells Inhibited by Intermittent High Glucose Concentrations through the Regulation of Cyclins. Mol. Med. Rep. 2014, 10, 683–688. [Google Scholar] [CrossRef]
  24. Miao, X.-Y.; Gu, Z.-Y.; Liu, P.; Hu, Y.; Li, L.; Gong, Y.-P.; Shu, H.; Liu, Y.; Li, C.-L. The Human Glucagon-like Peptide-1 Analogue Liraglutide Regulates Pancreatic Beta-Cell Proliferation and Apoptosis via an AMPK/mTOR/P70S6K Signaling Pathway. Peptides 2013, 39, 71–79. [Google Scholar] [CrossRef]
  25. Li, Y.; Cao, X.; Li, L.; Brubaker, P.L.; Edlund, H.; Drucker, D.J. β-Cell Pdx1 Expression Is Essential for the Glucoregulatory, Proliferative, and Cytoprotective Actions of Glucagon-Like Peptide-1. Diabetes 2005, 54, 482–491. [Google Scholar] [CrossRef]
  26. Dai, C.; Hang, Y.; Shostak, A.; Poffenberger, G.; Hart, N.; Prasad, N.; Phillips, N.; Levy, S.E.; Greiner, D.L.; Shultz, L.D.; et al. Age-Dependent Human β Cell Proliferation Induced by Glucagon-like Peptide 1 and Calcineurin Signaling. J. Clin. Investig. 2017, 127, 3835–3844. [Google Scholar] [CrossRef]
  27. Cornu, M.; Modi, H.; Kawamori, D.; Kulkarni, R.N.; Joffraud, M.; Thorens, B. Glucagon-like Peptide-1 Increases β-Cell Glucose Competence and Proliferation by Translational Induction of Insulin-like Growth Factor-1 Receptor Expression. J. Biol. Chem. 2010, 285, 10538–10545. [Google Scholar] [CrossRef]
  28. Mayendraraj, A.; Rosenkilde, M.M.; Gasbjerg, L.S. GLP-1 and GIP Receptor Signaling in Beta Cells – A Review of Receptor Interactions and Co-Stimulation. Peptides 2022, 151, 170749. [Google Scholar] [CrossRef]
  29. Trümper, J.; Ross, D.; Jahr, H.; Brendel, M.D.; Göke, R.; Hörsch, D. The Rap–B-Raf Signalling Pathway Is Activated by Glucose and Glucagon-like Peptide-1 in Human Islet Cells. Diabetologia 2005, 48, 1534–1540. [Google Scholar] [CrossRef]
  30. Yusta, B.; Baggio, L.L.; Estall, J.L.; Koehler, J.A.; Holland, D.P.; Li, H.; Pipeleers, D.; Ling, Z.; Drucker, D.J. GLP-1 Receptor Activation Improves β Cell Function and Survival Following Induction of Endoplasmic Reticulum Stress. Cell Metab. 2006, 4, 391–406. [Google Scholar] [CrossRef]
  31. Park, S.; Dong, X.; Fisher, T.L.; Dunn, S.; Omer, A.K.; Weir, G.; White, M.F. Exendin-4 Uses Irs2 Signaling to Mediate Pancreatic β Cell Growth and Function. J. Biol. Chem. 2006, 281, 1159–1168. [Google Scholar] [CrossRef]
  32. Carlessi, R.; Chen, Y.; Rowlands, J.; Cruzat, V.F.; Keane, K.N.; Egan, L.; Mamotte, C.; Stokes, R.; Gunton, J.E.; de Bittencourt, P.I.H.; et al. GLP-1 Receptor Signalling Promotes β-Cell Glucose Metabolism via mTOR-Dependent HIF-1α Activation. Sci. Rep. 2017, 7, 2661. [Google Scholar] [CrossRef]
  33. Aldawsari, M.; Almadani, F.A.; Almuhammadi, N.; Algabsani, S.; Alamro, Y.; Aldhwayan, M. The Efficacy of GLP-1 Analogues on Appetite Parameters, Gastric Emptying, Food Preference and Taste Among Adults with Obesity: Systematic Review of Randomized Controlled Trials. Diabetes Metab. Syndr. Obes. 2023, 16, 575–595. [Google Scholar] [CrossRef] [PubMed]
  34. Singh, I.; Wang, L.; Xia, B.; Liu, J.; Tahiri, A.; El Ouaamari, A.; Wheeler, M.B.; Pang, Z.P. Activation of Arcuate Nucleus Glucagon-like Peptide-1 Receptor-Expressing Neurons Suppresses Food Intake. Cell Biosci. 2022, 12, 178. [Google Scholar] [CrossRef] [PubMed]
  35. Secher, A.; Jelsing, J.; Baquero, A.F.; Hecksher-Sørensen, J.; Cowley, M.A.; Dalbøge, L.S.; Hansen, G.; Grove, K.L.; Pyke, C.; Raun, K.; et al. The Arcuate Nucleus Mediates GLP-1 Receptor Agonist Liraglutide-Dependent Weight Loss. J. Clin. Investig. 2014, 124, 4473–4488. [Google Scholar] [CrossRef] [PubMed]
  36. Huang, K.-P.; Acosta, A.A.; Ghidewon, M.Y.; McKnight, A.D.; Almeida, M.S.; Nyema, N.T.; Hanchak, N.D.; Patel, N.; Gbenou, Y.S.K.; Adriaenssens, A.E.; et al. Dissociable Hindbrain GLP1R Circuits for Satiety and Aversion. Nature 2024, 1–9. [Google Scholar] [CrossRef] [PubMed]
  37. Plamboeck, A.; Veedfald, S.; Deacon, C.F.; Hartmann, B.; Wettergren, A.; Svendsen, L.B.; Meisner, S.; Hovendal, C.; Vilsbøll, T.; Knop, F.K.; et al. The Effect of Exogenous GLP-1 on Food Intake Is Lost in Male Truncally Vagotomized Subjects with Pyloroplasty. Am. J. Physiol. Gastrointest. Liver Physiol. 2013, 304, G1117–G1127. [Google Scholar] [CrossRef] [PubMed]
  38. Van Bloemendaal, L.; IJzerman, R.G.; Ten Kulve, J.S.; Barkhof, F.; Konrad, R.J.; Drent, M.L.; Veltman, D.J.; Diamant, M. GLP-1 Receptor Activation Modulates Appetite- and Reward-Related Brain Areas in Humans. Diabetes 2014, 63, 4186–4196. [Google Scholar] [CrossRef] [PubMed]
  39. Drucker, D.J. Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. Cell Metab. 2018, 27, 740–756. [Google Scholar] [CrossRef]
  40. Drucker, D.J. The Cardiovascular Biology of Glucagon-like Peptide-1. Cell Metab. 2016, 24, 15–30. [Google Scholar] [CrossRef]
  41. Helmstädter, J.; Keppeler, K.; Küster, L.; Münzel, T.; Daiber, A.; Steven, S. Glucagon-like Peptide-1 (GLP-1) Receptor Agonists and Their Cardiovascular Benefits—The Role of the GLP-1 Receptor. Br. J. Pharmacol. 2022, 179, 659–676. [Google Scholar] [CrossRef]
  42. Nauck, M.A.; Meier, J.J. Management of Endocrine Disease: Are All GLP-1 Agonists Equal in the Treatment of Type 2 Diabetes? Eur. J. Endocrinol. 2019, 181, R211–R234. [Google Scholar] [CrossRef]
  43. Tahrani, A.A.; Barnett, A.H.; Bailey, C.J. Pharmacology and Therapeutic Implications of Current Drugs for Type 2 Diabetes Mellitus. Nat. Rev. Endocrinol. 2016, 12, 566–592. [Google Scholar] [CrossRef] [PubMed]
  44. Yoon, K.; Kang, J.; Kwon, S.C.; Trautmann, M.E.; Hompesch, M.; Stewart, J.; Sorli, C.H. Pharmacokinetic and Dose-finding Studies on Efpeglenatide in Patients with Type 2 Diabetes. Diabetes Obes. Metab. 2020, 22, 1292–1301. [Google Scholar] [CrossRef] [PubMed]
  45. Chen, G.; Kang, W.; Li, W.; Chen, S.; Gao, Y. Oral Delivery of Protein and Peptide Drugs: From Non-Specific Formulation Approaches to Intestinal Cell Targeting Strategies. Theranostics 2022, 12, 1419–1439. [Google Scholar] [CrossRef] [PubMed]
  46. Maideen, N.M.P. Pharmacologically Relevant Drug Interactions of Glucagon-like Peptide-1 Receptor Agonists. J. Analytical Pharm. Res. 2019, 8, 51–53. [Google Scholar] [CrossRef]
  47. Filippatos, T.D.; Panagiotopoulou, T.V.; Elisaf, M.S. Adverse Effects of GLP-1 Receptor Agonists. Rev. Diabet. Stud. 2014, 11, 202–230. [Google Scholar] [CrossRef] [PubMed]
  48. Malik, M.E.; Falkentoft, A.C.; Jensen, J.; Zahir, D.; Parveen, S.; Alhakak, A.; Andersson, C.; Petrie, M.C.; Sattar, N.; McMurray, J.J.V.; et al. Discontinuation and Reinitiation of SGLT-2 Inhibitors and GLP-1R Agonists in Patients with Type 2 Diabetes: A Nationwide Study from 2013 to 2021. Lancet Reg. Health Eur. 2023, 29, 100617. [Google Scholar] [CrossRef] [PubMed]
  49. Drab, S.R. Glucagon-Like Peptide-1 Receptor Agonists for Type 2 Diabetes: A Clinical Update of Safety and Efficacy. Curr. Diabetes Rev. 2016, 12, 403–413. [Google Scholar] [CrossRef]
  50. Marso, S.P.; Bain, S.C.; Consoli, A.; Eliaschewitz, F.G.; Jódar, E.; Leiter, L.A.; Lingvay, I.; Rosenstock, J.; Seufert, J.; Warren, M.L.; et al. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N. Engl. J. Med. 2016, 375, 1834–1844. [Google Scholar] [CrossRef]
  51. Xia, L.; Shen, T.; Dong, W.; Su, F.; Wang, J.; Wang, Q.; Niu, S.; Fang, Y. Comparative Efficacy and Safety of 8 GLP-1RAs in Patients with Type 2 Diabetes: A Network Meta-Analysis. Diabetes Res. Clin. Pract. 2021, 177, 108904. [Google Scholar] [CrossRef]
  52. Huthmacher, J.A.; Meier, J.J.; Nauck, M.A. Efficacy and Safety of Short- and Long-Acting Glucagon-Like Peptide 1 Receptor Agonists on a Background of Basal Insulin in Type 2 Diabetes: A Meta-Analysis. Diabetes Care 2020, 43, 2303–2312. [Google Scholar] [CrossRef]
  53. Pfeffer, M.A.; Claggett, B.; Diaz, R.; Dickstein, K.; Gerstein, H.C.; Køber, L.V.; Lawson, F.C.; Ping, L.; Wei, X.; Lewis, E.F.; et al. Lixisenatide in Patients with Type 2 Diabetes and Acute Coronary Syndrome. N. Engl. J. Med. 2015, 373, 2247–2257. [Google Scholar] [CrossRef]
  54. Marso, S.P.; Daniels, G.H.; Brown-Frandsen, K.; Kristensen, P.; Mann, J.F.E.; Nauck, M.A.; Nissen, S.E.; Pocock, S.; Poulter, N.R.; Ravn, L.S.; et al. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2016, 375, 311–322. [Google Scholar] [CrossRef]
  55. Holman, R.R.; Bethel, M.A.; Mentz, R.J.; Thompson, V.P.; Lokhnygina, Y.; Buse, J.B.; Chan, J.C.; Choi, J.; Gustavson, S.M.; Iqbal, N.; et al. Effects of Once-Weekly Exenatide on Cardiovascular Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2017, 377, 1228–1239. [Google Scholar] [CrossRef] [PubMed]
  56. 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]
  57. Gerstein, H.C.; Colhoun, H.M.; Dagenais, G.R.; Diaz, R.; Lakshmanan, M.; Pais, P.; Probstfield, J.; Riesmeyer, J.S.; Riddle, M.C.; Rydén, L.; et al. Dulaglutide and Cardiovascular Outcomes in Type 2 Diabetes (REWIND): A Double-Blind, Randomised Placebo-Controlled Trial. Lancet 2019, 394, 121–130. [Google Scholar] [CrossRef]
  58. Husain, M.; Birkenfeld, A.L.; Donsmark, M.; Dungan, K.; Eliaschewitz, F.G.; Franco, D.R.; Jeppesen, O.K.; Lingvay, I.; Mosenzon, O.; Pedersen, S.D.; et al. Oral Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N. Engl. J. Med. 2019, 381, 841–851. [Google Scholar] [CrossRef]
  59. Gerstein, H.C.; Sattar, N.; Rosenstock, J.; Ramasundarahettige, C.; Pratley, R.; Lopes, R.D.; Lam, C.S.P.; Khurmi, N.S.; Heenan, L.; Del Prato, S.; et al. Cardiovascular and Renal Outcomes with Efpeglenatide in Type 2 Diabetes. N. Engl. J. Med. 2021, 385, 896–907. [Google Scholar] [CrossRef]
  60. Mannucci, E.; Silverii, G.A. Cardiovascular Prevention with Glucose-lowering Drugs in Type 2 Diabetes: An Evidence-based Approach to the Categories of Primary and Secondary Prevention. Diabetes Obes. Metab. 2023, 25, 3435–3443. [Google Scholar] [CrossRef]
  61. Tsapas, A.; Avgerinos, I.; Karagiannis, T.; Malandris, K.; Manolopoulos, A.; Andreadis, P.; Liakos, A.; Matthews, D.R.; Bekiari, E. Comparative Effectiveness of Glucose-Lowering Drugs for Type 2 Diabetes: A Systematic Review and Network Meta-Analysis. Ann. Intern. Med. 2020, 173, 278–286. [Google Scholar] [CrossRef] [PubMed]
  62. Butalia, S.; Leung, A.A.; Ghali, W.A.; Rabi, D.M. Aspirin Effect on the Incidence of Major Adverse Cardiovascular Events in Patients with Diabetes Mellitus: A Systematic Review and Meta-Analysis. Cardiovasc. Diabetol. 2011, 10, 25. [Google Scholar] [CrossRef]
  63. Cholesterol Treatment Trialists’ (Ctt) Collaborators. The Effects of Lowering LDL Cholesterol with Statin Therapy in People at Low Risk of Vascular Disease: Meta-Analysis of Individual Data from 27 Randomised Trials. Lancet 2012, 380, 581–590. [Google Scholar] [CrossRef]
  64. The Heart Outcomes Prevention Evaluation Study Investigators. Effects of an Angiotensin-Converting–Enzyme Inhibitor, Ramipril, on Cardiovascular Events in High-Risk Patients. N. Engl. J. Med. 2000, 342, 145–153. [Google Scholar] [CrossRef]
  65. Sabatine, M.S.; Giugliano, R.P.; Keech, A.C.; Honarpour, N.; Wiviott, S.D.; Murphy, S.A.; Kuder, J.F.; Wang, H.; Liu, T.; Wasserman, S.M.; et al. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N. Engl. J. Med. 2017, 376, 1713–1722. [Google Scholar] [CrossRef]
  66. Tardif, J.-C.; Kouz, S.; Waters, D.D.; Bertrand, O.F.; Diaz, R.; Maggioni, A.P.; Pinto, F.J.; Ibrahim, R.; Gamra, H.; Kiwan, G.S.; et al. Efficacy and Safety of Low-Dose Colchicine after Myocardial Infarction. N. Engl. J. Med. 2019, 381, 2497–2505. [Google Scholar] [CrossRef]
  67. Kalkman, D.N.; Aquino, M.; Claessen, B.E.; Baber, U.; Guedeney, P.; Sorrentino, S.; Vogel, B.; de Winter, R.J.; Sweeny, J.; Kovacic, J.C.; et al. Residual Inflammatory Risk and the Impact on Clinical Outcomes in Patients after Percutaneous Coronary Interventions. Eur. Heart J. 2018, 39, 4101–4108. [Google Scholar] [CrossRef]
  68. Wei, J.; Yang, B.; Wang, R.; Ye, H.; Wang, Y.; Wang, L.; Zhang, X. Risk of Stroke and Retinopathy during GLP-1 Receptor Agonist Cardiovascular Outcome Trials: An Eight RCTs Meta-Analysis. Front. Endocrinol. 2022, 13, 1007980. [Google Scholar] [CrossRef]
  69. Malhotra, K.; Katsanos, A.H.; Lambadiari, V.; Goyal, N.; Palaiodimou, L.; Kosmidou, M.; Krogias, C.; Alexandrov, A.V.; Tsivgoulis, G. GLP-1 Receptor Agonists in Diabetes for Stroke Prevention: A Systematic Review and Meta-Analysis. J. Neurol. 2020, 267, 2117–2122. [Google Scholar] [CrossRef]
  70. Benn, M.; Emanuelsson, F.; Tybjærg-Hansen, A.; Nordestgaard, B.G. Impact of High Glucose Levels and Glucose Lowering on Risk of Ischaemic Stroke: A Mendelian Randomisation Study and Meta-Analysis. Diabetologia 2021, 64, 1492–1503. [Google Scholar] [CrossRef]
  71. Zelniker, T.A.; Wiviott, S.D.; Raz, I.; Im, K.; Goodrich, E.L.; Furtado, R.H.M.; Bonaca, M.P.; Mosenzon, O.; Kato, E.T.; Cahn, A.; et al. Comparison of the Effects of Glucagon-Like Peptide Receptor Agonists and Sodium-Glucose Cotransporter 2 Inhibitors for Prevention of Major Adverse Cardiovascular and Renal Outcomes in Type 2 Diabetes Mellitus: Systematic Review and Meta-Analysis of Cardiovascular Outcomes Trials. Circulation 2019, 139, 2022–2031. [Google Scholar] [CrossRef]
  72. Kosiborod, M.N.; Abildstrøm, S.Z.; Borlaug, B.A.; Butler, J.; Rasmussen, S.; Davies, M.; Hovingh, G.K.; Kitzman, D.W.; Lindegaard, M.L.; Møller, D.V.; et al. Semaglutide in Patients with Heart Failure with Preserved Ejection Fraction and Obesity. N. Engl. J. Med. 2023, 389, 1069–1084. [Google Scholar] [CrossRef]
  73. Verma, S.; Al-Omran, M.; Leiter, L.A.; Mazer, C.D.; Rasmussen, S.; Saevereid, H.A.; Sejersten Ripa, M.; Bonaca, M.P. Cardiovascular Efficacy of Liraglutide and Semaglutide in Individuals with Diabetes and Peripheral Artery Disease. Diabetes Obes. Metab. 2022, 24, 1288–1299. [Google Scholar] [CrossRef]
  74. Piechocki, M.; Przewłocki, T.; Pieniążek, P.; Trystuła, M.; Podolec, J.; Kabłak-Ziembicka, A. A Non-Coronary, Peripheral Arterial Atherosclerotic Disease (Carotid, Renal, Lower Limb) in Elderly Patients—A Review PART II—Pharmacological Approach for Management of Elderly Patients with Peripheral Atherosclerotic Lesions Outside Coronary Territory. J. Clin. Med. 2024, 13, 1508. [Google Scholar] [CrossRef]
  75. Baviera, M.; Genovese, S.; Lepore, V.; Colacioppo, P.; Robusto, F.; Tettamanti, M.; D’Ettorre, A.; Avanzini, F.; Fortino, I.; Nicolucci, A.; et al. Lower Risk of Death and Cardiovascular Events in Patients with Diabetes Initiating Glucagon-like Peptide-1 Receptor Agonists or Sodium-glucose Cotransporter-2 Inhibitors: A Real-world Study in Two Italian Cohorts. Diabetes Obes. Metab. 2021, 23, 1484–1495. [Google Scholar] [CrossRef]
  76. Schäfer, Z.; Mathisen, A.; Thomsen, T.R.; Rossing, P.; Kirketerp-Møller, K. Glucagon-like Peptide-1 Treatment Reduces the Risk of Diabetes-Type 2 Related Amputations: A Cohort Study in Denmark. Diabetes Res. Clin. Pract. 2023, 202, 110799. [Google Scholar] [CrossRef]
  77. Liarakos, A.L.; Tentolouris, A.; Kokkinos, A.; Eleftheriadou, I.; Tentolouris, N. Impact of Glucagon-like Peptide 1 Receptor Agonists on Peripheral Arterial Disease in People with Diabetes Mellitus: A Narrative Review. J. Diabetes Complicat. 2023, 37, 108390. [Google Scholar] [CrossRef]
  78. Neal, B.; Perkovic, V.; Mahaffey, K.W.; De Zeeuw, D.; Fulcher, G.; Erondu, N.; Shaw, W.; Law, G.; Desai, M.; Matthews, D.R. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N. Engl. J. Med. 2017, 377, 644–657. [Google Scholar] [CrossRef]
  79. Scheen, A.J. Lower Limb Amputations: Protection with GLP-1 Receptor Agonists Rather than Increased Risk with SGLT2 Inhibitors? Diabetes Metab. 2022, 48, 101325. [Google Scholar] [CrossRef]
  80. Svanström, H.; Ueda, P.; Melbye, M.; Eliasson, B.; Svensson, A.-M.; Franzén, S.; Gudbjörnsdottir, S.; Hveem, K.; Jonasson, C.; Pasternak, B. Use of Liraglutide and Risk of Major Cardiovascular Events: A Register-Based Cohort Study in Denmark and Sweden. Lancet Diabetes Endocrinol. 2019, 7, 106–114. [Google Scholar] [CrossRef]
  81. 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]
  82. Cosentino, F.; Grant, P.J.; Aboyans, V.; Bailey, C.J.; Ceriello, A.; Delgado, V.; Federici, M.; Filippatos, G.; Grobbee, D.E.; Hansen, T.B.; et al. 2019 ESC Guidelines on Diabetes, Pre-Diabetes, and Cardiovascular Diseases Developed in Collaboration with the EASD. Eur. Heart J. 2020, 41, 255–323. [Google Scholar] [CrossRef]
  83. Buse, J.B.; Wexler, D.J.; Tsapas, A.; Rossing, P.; Mingrone, G.; Mathieu, C.; D’Alessio, D.A.; Davies, M.J. 2019 Update to: Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 2020, 43, 487–493. [Google Scholar] [CrossRef]
  84. Butkowski, E.G.; Jelinek, H.F. Hyperglycaemia, Oxidative Stress and Inflammatory Markers. Redox Rep. 2017, 22, 257–264. [Google Scholar] [CrossRef]
  85. Patel, A.; MacMahon, S.; Chalmers, J.; Neal, B.; Billot, L.; Woodward, M.; Marre, M.; Cooper, M.; Glasziou, P.; Grobbee, D.; et al. Intensive Blood Glucose Control and Vascular Outcomes in Patients with Type 2 Diabetes. N. Engl. J. Med. 2008, 358, 2560–2572. [Google Scholar] [CrossRef] [PubMed]
  86. Buse, J.B.; Bain, S.C.; Mann, J.F.E.; Nauck, M.A.; Nissen, S.E.; Pocock, S.; Poulter, N.R.; Pratley, R.E.; Linder, M.; Monk Fries, T.; et al. Cardiovascular Risk Reduction with Liraglutide: An Exploratory Mediation Analysis of the LEADER Trial. Diabetes Care 2020, 43, 1546–1552. [Google Scholar] [CrossRef]
  87. Colhoun, H.M.; Hasenour, C.; Riddle, M.C.; Branch, K.; Konig, M.; Atisso, C.; Lakshmanan, M.; Mody, R.; Gerstein, H.C. Exploring Potential Mediators of the Cardiovascular Benefit of Dulaglutide in REWIND. Diabetes 2020, 69, 194. [Google Scholar] [CrossRef]
  88. Giugliano, D.; Bellastella, G.; Longo, M.; Scappaticcio, L.; Maiorino, M.I.; Chiodini, P.; Esposito, K. Relationship between Improvement of Glycaemic Control and Reduction of Major Cardiovascular Events in 15 Cardiovascular Outcome Trials: A Meta-analysis with Meta-regression. Diabetes Obes. Metab. 2020, 22, 1397–1405. [Google Scholar] [CrossRef]
  89. Wing, R.R.; Lang, W.; Wadden, T.A.; Safford, M.; Knowler, W.C.; Bertoni, A.G.; Hill, J.O.; Brancati, F.L.; Peters, A.; Wagenknecht, L.; et al. Benefits of Modest Weight Loss in Improving Cardiovascular Risk Factors in Overweight and Obese Individuals with Type 2 Diabetes. Diabetes Care 2011, 34, 1481–1486. [Google Scholar] [CrossRef]
  90. Fuster, J.J.; Ouchi, N.; Gokce, N.; Walsh, K. Obesity-Induced Changes in Adipose Tissue Microenvironment and Their Impact on Cardiovascular Disease. Circ. Res. 2016, 118, 1786–1807. [Google Scholar] [CrossRef]
  91. Lincoff, A.M.; Brown-Frandsen, K.; Colhoun, H.M.; Deanfield, J.; Emerson, S.S.; Esbjerg, S.; Hardt-Lindberg, S.; Hovingh, G.K.; Kahn, S.E.; Kushner, R.F.; et al. Semaglutide and Cardiovascular Outcomes in Obesity without Diabetes. N. Engl. J. Med. 2023, 389, 2221–2232. [Google Scholar] [CrossRef]
  92. Kwan, A.C.; May, H.T.; Cater, G.; Sibley, C.T.; Rosen, B.D.; Lima, J.A.C.; Rodriguez, K.; Lappe, D.L.; Muhlestein, J.B.; Anderson, J.L.; et al. Coronary Artery Plaque Volume and Obesity in Patients with Diabetes: The Factor-64 Study. Radiology 2014, 272, 690–699. [Google Scholar] [CrossRef]
  93. Khera, R.; Murad, M.H.; Chandar, A.K.; Dulai, P.S.; Wang, Z.; Prokop, L.J.; Loomba, R.; Camilleri, M.; Singh, S. Association of Pharmacological Treatments for Obesity with Weight Loss and Adverse Events: A Systematic Review and Meta-Analysis. JAMA 2016, 315, 2424–2434. [Google Scholar] [CrossRef] [PubMed]
  94. Wilding, J.P.H.; Batterham, R.L.; Calanna, S.; Davies, M.; Van Gaal, L.F.; Lingvay, I.; McGowan, B.M.; Rosenstock, J.; Tran, M.T.D.; Wadden, T.A.; et al. Once-Weekly Semaglutide in Adults with Overweight or Obesity. N. Engl. J. Med. 2021, 384, 989–1002. [Google Scholar] [CrossRef] [PubMed]
  95. Ghusn, W.; De la Rosa, A.; Sacoto, D.; Cifuentes, L.; Campos, A.; Feris, F.; Hurtado, M.D.; Acosta, A. Weight Loss Outcomes Associated with Semaglutide Treatment for Patients with Overweight or Obesity. JAMA Netw. Open 2022, 5, e2231982. [Google Scholar] [CrossRef] [PubMed]
  96. The Look AHEAD Research Group. Cardiovascular Effects of Intensive Lifestyle Intervention in Type 2 Diabetes. N. Engl. J. Med. 2013, 369, 145–154. [Google Scholar] [CrossRef] [PubMed]
  97. The Look AHEAD Research Group. Association of the Magnitude of Weight Loss and Changes in Physical Fitness with Long-Term Cardiovascular Disease Outcomes in Overweight or Obese People with Type 2 Diabetes: A Post-Hoc Analysis of the Look AHEAD Randomised Clinical Trial. Lancet Diabetes Endocrinol. 2016, 4, 913–921. [Google Scholar] [CrossRef]
  98. Sjöström, C.D.; Lystig, T.; Lindroos, A.K. Impact of Weight Change, Secular Trends and Ageing on Cardiovascular Risk Factors: 10-Year Experiences from the SOS Study. Int. J. Obes. 2011, 35, 1413–1420. [Google Scholar] [CrossRef] [PubMed]
  99. Sarma, S.; Palcu, P. Weight Loss between Glucagon-like Peptide-1 Receptor Agonists and Bariatric Surgery in Adults with Obesity: A Systematic Review and Meta-analysis. Obesity 2022, 30, 2111–2121. [Google Scholar] [CrossRef] [PubMed]
  100. Stenberg, E.; Näslund, E. Major Adverse Cardiovascular Events among Patients with Type-2 Diabetes, a Nationwide Cohort Study Comparing Primary Metabolic and Bariatric Surgery to GLP-1 Receptor Agonist Treatment. Int. J. Obes. 2023, 47, 251–256. [Google Scholar] [CrossRef] [PubMed]
  101. Dicker, D.; Sagy, Y.W.; Ramot, N.; Battat, E.; Greenland, P.; Arbel, R.; Lavie, G.; Reges, O. Bariatric Metabolic Surgery vs Glucagon-Like Peptide-1 Receptor Agonists and Mortality. JAMA Netw. Open 2024, 7, e2415392. [Google Scholar] [CrossRef]
  102. Ryan, D.H.; Lingvay, I.; Deanfield, J.; Kahn, S.E.; Barros, E.; Burguera, B.; Colhoun, H.M.; Cercato, C.; Dicker, D.; Horn, D.B.; et al. Long-Term Weight Loss Effects of Semaglutide in Obesity without Diabetes in the SELECT Trial. Nat. Med. 2024, 30, 2049–2057. [Google Scholar] [CrossRef] [PubMed]
  103. Katout, M.; Zhu, H.; Rutsky, J.; Shah, P.; Brook, R.D.; Zhong, J.; Rajagopalan, S. Effect of GLP-1 Mimetics on Blood Pressure and Relationship to Weight Loss and Glycemia Lowering: Results of a Systematic Meta-Analysis and Meta-Regression. Am. J. Hypertens. 2014, 27, 130–139. [Google Scholar] [CrossRef] [PubMed]
  104. Anholm, C.; Kumarathurai, P.; Pedersen, L.R.; Samkani, A.; Walzem, R.L.; Nielsen, O.W.; Kristiansen, O.P.; Fenger, M.; Madsbad, S.; Sajadieh, A.; et al. Liraglutide in Combination with Metformin May Improve the Atherogenic Lipid Profile and Decrease C-Reactive Protein Level in Statin Treated Obese Patients with Coronary Artery Disease and Newly Diagnosed Type 2 Diabetes: A Randomized Trial. Atherosclerosis 2019, 288, 60–66. [Google Scholar] [CrossRef] [PubMed]
  105. Matikainen, N.; Söderlund, S.; Björnson, E.; Pietiläinen, K.; Hakkarainen, A.; Lundbom, N.; Taskinen, M.-R.; Borén, J. Liraglutide Treatment Improves Postprandial Lipid Metabolism and Cardiometabolic Risk Factors in Humans with Adequately Controlled Type 2 Diabetes: A Single-Centre Randomized Controlled Study. Diabetes Obes. Metab. 2019, 21, 84–94. [Google Scholar] [CrossRef]
  106. Zobel, E.H.; Wretlind, A.; Ripa, R.S.; Rotbain Curovic, V.; von Scholten, B.J.; Suvitaival, T.; Hansen, T.W.; Kjær, A.; Legido-Quigley, C.; Rossing, P. Ceramides and Phospholipids Are Downregulated with Liraglutide Treatment: Results from the LiraFlame Randomized Controlled Trial. BMJ Open Diabetes Res. Care 2021, 9, e002395. [Google Scholar] [CrossRef]
  107. Sun, F.; Wu, S.; Wang, J.; Guo, S.; Chai, S.; Yang, Z.; Li, L.; Zhang, Y.; Ji, L.; Zhan, S. Effect of Glucagon-like Peptide-1 Receptor Agonists on Lipid Profiles Among Type 2 Diabetes: A Systematic Review and Network Meta-Analysis. Clin. Ther. 2015, 37, 225–241. [Google Scholar] [CrossRef]
  108. 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]
  109. Sanada, J.; Obata, A.; Obata, Y.; Fushimi, Y.; Shimoda, M.; Kohara, K.; Nakanishi, S.; Mune, T.; Kaku, K.; Kaneto, H. Dulaglutide Exerts Beneficial Anti Atherosclerotic Effects in ApoE Knockout Mice with Diabetes: The Earlier, the Better. Sci. Rep. 2021, 11, 1425. [Google Scholar] [CrossRef]
  110. Sudo, M.; Li, Y.; Hiro, T.; Takayama, T.; Mitsumata, M.; Shiomi, M.; Sugitani, M.; Matsumoto, T.; Hao, H.; Hirayama, A. Inhibition of Plaque Progression and Promotion of Plaque Stability by Glucagon-like Peptide-1 Receptor Agonist: Serial in Vivo Findings from iMap-IVUS in Watanabe Heritable Hyperlipidemic Rabbits. Atherosclerosis 2017, 265, 283–291. [Google Scholar] [CrossRef] [PubMed]
  111. Ying, Y.-L.; Chen, Y.-C.; Jandeleit-Dahm, K.; Peter, K. GLP-1 Receptor Agonists: An Example of the Challenge for Animal Models to Predict Plaque Instability/Rupture and Cardiovascular Outcomes. Atherosclerosis 2017, 265, 250–252. [Google Scholar] [CrossRef]
  112. Piotrowski, K.; Becker, M.; Zugwurst, J.; Biller-Friedmann, I.; Spoettl, G.; Greif, M.; Leber, A.W.; Becker, A.; Laubender, R.P.; Lebherz, C.; et al. Circulating Concentrations of GLP-1 Are Associated with Coronary Atherosclerosis in Humans. Cardiovasc. Diabetol. 2013, 12, 117. [Google Scholar] [CrossRef]
  113. Hamal, S.; Cherukuri, L.; Shaikh, K.; Kinninger, A.; Doshi, J.; Birudaraju, D.; Budoff, M.J. Effect of Semaglutide on Coronary Atherosclerosis Progression in Patients with Type II Diabetes: Rationale and Design of the Semaglutide Treatment on Coronary Progression Trial. Coron. Artery Dis. 2020, 31, 306–314. [Google Scholar] [CrossRef] [PubMed]
  114. Janić, M.; Rizzo, M.; Cosentino, F.; Pantea Stoian, A.; Lunder, M.; Šabović, M.; Janež, A. Effect of Oral Semaglutide on Cardiovascular Parameters and Their Mechanisms in Patients with Type 2 Diabetes: Rationale and Design of the Semaglutide Anti-Atherosclerotic Mechanisms of Action Study (SAMAS). Diabetes Ther. 2022, 13, 795–810. [Google Scholar] [CrossRef] [PubMed]
  115. Ban, K.; Noyan-Ashraf, M.H.; Hoefer, J.; Bolz, S.-S.; Drucker, D.J.; Husain, M. Cardioprotective and Vasodilatory Actions of Glucagon-Like Peptide 1 Receptor Are Mediated Through Both Glucagon-Like Peptide 1 Receptor–Dependent and –Independent Pathways. Circulation 2008, 117, 2340–2350. [Google Scholar] [CrossRef] [PubMed]
  116. Aetesam-ur-Rahman, M.; Giblett, J.P.; Khialani, B.; Kyranis, S.; Clarke, S.J.; Zhao, T.X.; Braganza, D.M.; Clarke, S.C.; West, N.E.J.; Bennett, M.R.; et al. GLP-1 Vasodilatation in Humans with Coronary Artery Disease Is Not Adenosine Mediated. BMC Cardiovasc. Disord. 2021, 21, 223. [Google Scholar] [CrossRef] [PubMed]
  117. Sukumaran, V.; Tsuchimochi, H.; Sonobe, T.; Waddingham, M.T.; Shirai, M.; Pearson, J.T. Liraglutide Treatment Improves the Coronary Microcirculation in Insulin Resistant Zucker Obese Rats on a High Salt Diet. Cardiovasc. Diabetol. 2020, 19, 24. [Google Scholar] [CrossRef] [PubMed]
  118. Jojima, T.; Uchida, K.; Akimoto, K.; Tomotsune, T.; Yanagi, K.; Iijima, T.; Suzuki, K.; Kasai, K.; Aso, Y. Liraglutide, a GLP-1 Receptor Agonist, Inhibits Vascular Smooth Muscle Cell Proliferation by Enhancing AMP-Activated Protein Kinase and Cell Cycle Regulation, and Delays Atherosclerosis in ApoE Deficient Mice. Atherosclerosis 2017, 261, 44–51. [Google Scholar] [CrossRef] [PubMed]
  119. Liu, Z.; Zhang, M.; Zhou, T.; Shen, Q.; Qin, X. Exendin-4 Promotes the Vascular Smooth Muscle Cell Re-Differentiation through AMPK/SIRT1/FOXO3a Signaling Pathways. Atherosclerosis 2018, 276, 58–66. [Google Scholar] [CrossRef] [PubMed]
  120. Koska, J.; Sands, M.; Burciu, C.; D’Souza, K.M.; Raravikar, K.; Liu, J.; Truran, S.; Franco, D.A.; Schwartz, E.A.; Schwenke, D.C.; et al. Exenatide Protects Against Glucose- and Lipid-Induced Endothelial Dysfunction: Evidence for Direct Vasodilation Effect of GLP-1 Receptor Agonists in Humans. Diabetes 2015, 64, 2624–2635. [Google Scholar] [CrossRef]
  121. Li, P.; Tang, Z.; Wang, L.; Feng, B. Glucagon-like Peptide-1 Analogue Liraglutide Ameliorates Atherogenesis via Inhibiting Advanced Glycation End Product-Induced Receptor for Advanced Glycosylation End Product Expression in Apolipoprotein-E Deficient Mice. Mol. Med. Rep. 2017, 16, 3421–3426. [Google Scholar] [CrossRef]
  122. Ray, K.K.; Ference, B.A.; Séverin, T.; Blom, D.; Nicholls, S.J.; Shiba, M.H.; Almahmeed, W.; Alonso, R.; Daccord, M.; Ezhov, M.; et al. World Heart Federation Cholesterol Roadmap 2022. Glob. Heart 2022, 17, 75. [Google Scholar] [CrossRef]
  123. El Bekay, R.; Coín-Aragüez, L.; Fernández-García, D.; Oliva-Olivera, W.; Bernal-López, R.; Clemente-Postigo, M.; Delgado-Lista, J.; Diaz-Ruiz, A.; Guzman-Ruiz, R.; Vázquez-Martínez, R.; et al. Effects of Glucagon-like Peptide-1 on the Differentiation and Metabolism of Human Adipocytes. Br. J. Pharmacol. 2016, 173, 1820–1834. [Google Scholar] [CrossRef] [PubMed]
  124. Hu, Y.-W.; Yang, J.-Y.; Ma, X.; Chen, Z.-P.; Hu, Y.-R.; Zhao, J.-Y.; Li, S.-F.; Qiu, Y.-R.; Lu, J.-B.; Wang, Y.-C.; et al. A lincRNA-DYNLRB2-2/GPR119/GLP-1R/ABCA1-Dependent Signal Transduction Pathway Is Essential for the Regulation of Cholesterol Homeostasis. J. Lipid Res. 2014, 55, 681–697. [Google Scholar] [CrossRef] [PubMed]
  125. Tashiro, Y.; Sato, K.; Watanabe, T.; Nohtomi, K.; Terasaki, M.; Nagashima, M.; Hirano, T. A Glucagon-like Peptide-1 Analog Liraglutide Suppresses Macrophage Foam Cell Formation and Atherosclerosis. Peptides 2014, 54, 19–26. [Google Scholar] [CrossRef] [PubMed]
  126. Barbieri, M.; Marfella, R.; Esposito, A.; Rizzo, M.R.; Angellotti, E.; Mauro, C.; Siniscalchi, M.; Chirico, F.; Caiazzo, P.; Furbatto, F.; et al. Incretin Treatment and Atherosclerotic Plaque Stability: Role of Adiponectin/APPL1 Signaling Pathway. J. Diabetes Complicat. 2017, 31, 295–303. [Google Scholar] [CrossRef] [PubMed]
  127. Patel, V.; Joharapurkar, A.; Kshirsagar, S.; Sutariya, B.; Patel, M.; Pandey, D.; Patel, H.; Ranvir, R.; Kadam, S.; Patel, D.; et al. Coagonist of GLP-1 and Glucagon Decreases Liver Inflammation and Atherosclerosis in Dyslipidemic Condition. Chem. Biol. Interact. 2018, 282, 13–21. [Google Scholar] [CrossRef]
  128. Yabut, J.M.; Drucker, D.J. Glucagon-like Peptide-1 Receptor-Based Therapeutics for Metabolic Liver Disease. Endocr. Rev. 2023, 44, 14–32. [Google Scholar] [CrossRef]
  129. Nguyen, M.; Fernando, S.; Schwarz, N.; Tan, J.; Bursill, C.; Psaltis, P. Inflammation as a Therapeutic Target in Atherosclerosis. J. Clin. Med. 2019, 8, 1109. [Google Scholar] [CrossRef]
  130. Scisciola, L.; Rizzo, M.R.; Cataldo, V.; Fontanella, R.A.; Balestrieri, M.L.; D’Onofrio, N.; Marfella, R.; Paolisso, G.; Barbieri, M. Incretin Drugs Effect on Epigenetic Machinery: New Potential Therapeutic Implications in Preventing Vascular Diabetic Complications. FASEB J. 2020, 34, 16489–16503. [Google Scholar] [CrossRef]
  131. Krasner, N.M.; Ido, Y.; Ruderman, N.B.; Cacicedo, J.M. Glucagon-Like Peptide-1 (GLP-1) Analog Liraglutide Inhibits Endothelial Cell Inflammation through a Calcium and AMPK Dependent Mechanism. PLoS ONE 2014, 9, e97554. [Google Scholar] [CrossRef]
  132. Yue, W.; Li, Y.; Ou, D.; Yang, Q. The GLP-1 Receptor Agonist Liraglutide Protects against Oxidized LDL-Induced Endothelial Inflammation and Dysfunction via KLF2. IUBMB Life 2019, 71, 1347–1354. [Google Scholar] [CrossRef]
  133. Wei, H.; Bu, R.; Yang, Q.; Jia, J.; Li, T.; Wang, Q.; Chen, Y. Exendin-4 Protects against Hyperglycemia-Induced Cardiomyocyte Pyroptosis via the AMPK-TXNIP Pathway. J. Diabetes Res. 2019, 2019, 8905917. [Google Scholar] [CrossRef] [PubMed]
  134. Vinué, Á.; Navarro, J.; Herrero-Cervera, A.; García-Cubas, M.; Andrés-Blasco, I.; Martínez-Hervás, S.; Real, J.T.; Ascaso, J.F.; González-Navarro, H. The GLP-1 Analogue Lixisenatide Decreases Atherosclerosis in Insulin-Resistant Mice by Modulating Macrophage Phenotype. Diabetologia 2017, 60, 1801–1812. [Google Scholar] [CrossRef] [PubMed]
  135. Bruen, R.; Curley, S.; Kajani, S.; Crean, D.; O’Reilly, M.E.; Lucitt, M.B.; Godson, C.G.; McGillicuddy, F.C.; Belton, O. Liraglutide Dictates Macrophage Phenotype in Apolipoprotein E Null Mice during Early Atherosclerosis. Cardiovasc. Diabetol. 2017, 16, 143. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, Y.; Ying, F.; Tian, X.; Lei, Z.; Li, X.; Lo, C.-Y.; Li, J.; Jiang, L.; Yao, X. TRPM2 Promotes Atherosclerotic Progression in a Mouse Model of Atherosclerosis. Cells 2022, 11, 1423. [Google Scholar] [CrossRef] [PubMed]
  137. Zong, P.; Feng, J.; Yue, Z.; Yu, A.S.; Vacher, J.; Jellison, E.R.; Miller, B.; Mori, Y.; Yue, L. TRPM2 Deficiency in Mice Protects against Atherosclerosis by Inhibiting TRPM2–CD36 Inflammatory Axis in Macrophages. Nat. Cardiovasc. Res. 2022, 1, 344–360. [Google Scholar] [CrossRef] [PubMed]
  138. Zong, P.; Feng, J.; Yue, Z.; Li, Y.; Wu, G.; Sun, B.; He, Y.; Miller, B.; Yu, A.S.; Su, Z.; et al. Functional Coupling of TRPM2 and Extrasynaptic NMDARs Exacerbates Excitotoxicity in Ischemic Brain Injury. Neuron 2022, 110, 1944–1958. [Google Scholar] [CrossRef] [PubMed]
  139. Jensen, J.K.; Binderup, T.; Grandjean, C.E.; Bentsen, S.; Ripa, R.S.; Kjaer, A. Semaglutide Reduces Vascular Inflammation Investigated by PET in a Rabbit Model of Advanced Atherosclerosis. Atherosclerosis 2022, 352, 88–95. [Google Scholar] [CrossRef]
  140. Ripa, R.S.; Zobel, E.H.; von Scholten, B.J.; Jensen, J.K.; Binderup, T.; Diaz, L.J.; Curovic, V.R.; Hansen, T.W.; Rossing, P.; Kjaer, A. Effect of Liraglutide on Arterial Inflammation Assessed as [18F]FDG Uptake in Patients With Type 2 Diabetes: A Randomized, Double-Blind, Placebo-Controlled Trial. Circ. Cardiovasc. Imaging 2021, 14, e012174. [Google Scholar] [CrossRef]
  141. Ni, L.; Yang, L.; Lin, Y. Recent Progress of Endoplasmic Reticulum Stress in the Mechanism of Atherosclerosis. Front. Cardiovasc. Med. 2024, 11, 1413441. [Google Scholar] [CrossRef]
  142. Bretón-Romero, R.; Weisbrod, R.M.; Feng, B.; Holbrook, M.; Ko, D.; Stathos, M.M.; Zhang, J.; Fetterman, J.L.; Hamburg, N.M. Liraglutide Treatment Reduces Endothelial Endoplasmic Reticulum Stress and Insulin Resistance in Patients with Diabetes Mellitus. J. Am. Heart Assoc. 2018, 7, e009379. [Google Scholar] [CrossRef] [PubMed]
  143. Kapadia, P.; Bikkina, P.; Landicho, M.A.; Parekh, S.; Haas, M.J.; Mooradian, A.D. Effect of Anti-Hyperglycemic Drugs on Endoplasmic Reticulum (ER) Stress in Human Coronary Artery Endothelial Cells. Eur. J. Pharmacol. 2021, 907, 174249. [Google Scholar] [CrossRef]
  144. Erdogdu, Ö.; Eriksson, L.; Xu, H.; Sjöholm, Å.; Zhang, Q.; Nyström, T. Exendin-4 Protects Endothelial Cells from Lipoapoptosis by PKA, PI3K, eNOS, P38 MAPK, and JNK Pathways. J. Mol. Endocrinol. 2013, 50, 229–241. [Google Scholar] [CrossRef]
  145. Balestrieri, M.L.; Rizzo, M.R.; Barbieri, M.; Paolisso, P.; D’Onofrio, N.; Giovane, A.; Siniscalchi, M.; Minicucci, F.; Sardu, C.; D’Andrea, D.; et al. Sirtuin 6 Expression and Inflammatory Activity in Diabetic Atherosclerotic Plaques: Effects of Incretin Treatment. Diabetes 2015, 64, 1395–1406. [Google Scholar] [CrossRef]
  146. Zhao, S.; Kusminski, C.M.; Scherer, P.E. Adiponectin, Leptin and Cardiovascular Disorders. Circ. Res. 2021, 128, 136–149. [Google Scholar] [CrossRef]
  147. Wang, A.; Li, T.; An, P.; Yan, W.; Zheng, H.; Wang, B.; Mu, Y. Exendin-4 Upregulates Adiponectin Level in Adipocytes via Sirt1/Foxo-1 Signaling Pathway. PLoS ONE 2017, 12, e0169469. [Google Scholar] [CrossRef]
  148. Simental-Mendía, L.E.; Sánchez-García, A.; Linden-Torres, E.; Simental-Mendía, M. Impact of Glucagon-like Peptide-1 Receptor Agonists on Adiponectin Concentrations: A Meta-analysis of Randomized Controlled Trials. Br. J. Clin. Pharmacol. 2021, 87, 4140–4149. [Google Scholar] [CrossRef]
  149. McAninch, E.A.; Fonseca, T.L.; Poggioli, R.; Panos, A.L.; Salerno, T.A.; Deng, Y.; Li, Y.; Bianco, A.C.; Iacobellis, G. Epicardial Adipose Tissue Has a Unique Transcriptome Modified in Severe Coronary Artery Disease: Epicardial Fat Transcriptome in CAD. Obesity 2015, 23, 1267–1278. [Google Scholar] [CrossRef]
  150. Iacobellis, G.; Camarena, V.; Sant, D.; Wang, G. Human Epicardial Fat Expresses Glucagon-Like Peptide 1 and 2 Receptors Genes. Horm. Metab. Res. 2017, 49, 625–630. [Google Scholar] [CrossRef] [PubMed]
  151. Iacobellis, G.; Villasante Fricke, A.C. Effects of Semaglutide Versus Dulaglutide on Epicardial Fat Thickness in Subjects with Type 2 Diabetes and Obesity. J. Endocr. Soc. 2020, 4, bvz042. [Google Scholar] [CrossRef] [PubMed]
  152. Berg, G.; Barchuk, M.; Lobo, M.; Nogueira, J.P. Effect of Glucagon-like Peptide-1 (GLP-1) Analogues on Epicardial Adipose Tissue: A Meta-Analysis. Diabetes Metab. Syndr. 2022, 16, 102562. [Google Scholar] [CrossRef] [PubMed]
  153. McKenney, M.L.; Schultz, K.A.; Boyd, J.H.; Byrd, J.P.; Alloosh, M.; Teague, S.D.; Arce-Esquivel, A.A.; Fain, J.N.; Laughlin, M.H.; Sacks, H.S.; et al. Epicardial Adipose Excision Slows the Progression of Porcine Coronary Atherosclerosis. J. Cardiothorac. Surg. 2014, 9, 2. [Google Scholar] [CrossRef]
  154. Gaba, P.; Gersh, B.J.; Muller, J.; Narula, J.; Stone, G.W. Evolving Concepts of the Vulnerable Atherosclerotic Plaque and the Vulnerable Patient: Implications for Patient Care and Future Research. Nat. Rev. Cardiol. 2023, 20, 181–196. [Google Scholar] [CrossRef]
  155. Yang, G.; Lei, Y.; Inoue, A.; Piao, L.; Hu, L.; Jiang, H.; Sasaki, T.; Wu, H.; Xu, W.; Yu, C.; et al. Exenatide Mitigated Diet-Induced Vascular Aging and Atherosclerotic Plaque Growth in ApoE-Deficient Mice under Chronic Stress. Atherosclerosis 2017, 264, 1–10. [Google Scholar] [CrossRef]
  156. Garczorz, W.; Gallego-Colon, E.; Kosowska, A.; Kłych-Ratuszny, A.; Woźniak, M.; Marcol, W.; Niesner, K.J.; Francuz, T. Exenatide Exhibits Anti-Inflammatory Properties and Modulates Endothelial Response to Tumor Necrosis Factor α-Mediated Activation. Cardiovasc. Ther. 2018, 36, e12317. [Google Scholar] [CrossRef]
  157. Guo, L.; Akahori, H.; Harari, E.; Smith, S.L.; Polavarapu, R.; Karmali, V.; Otsuka, F.; Gannon, R.L.; Braumann, R.E.; Dickinson, M.H.; et al. CD163+ Macrophages Promote Angiogenesis and Vascular Permeability Accompanied by Inflammation in Atherosclerosis. J. Clin. Investig. 2018, 128, 1106–1124. [Google Scholar] [CrossRef]
  158. Cameron-Vendrig, A.; Reheman, A.; Siraj, M.A.; Xu, X.R.; Wang, Y.; Lei, X.; Afroze, T.; Shikatani, E.; El-Mounayri, O.; Noyan, H.; et al. Glucagon-Like Peptide 1 Receptor Activation Attenuates Platelet Aggregation and Thrombosis. Diabetes 2016, 65, 1714–1723. [Google Scholar] [CrossRef]
  159. Cahill, K.N.; Amin, T.; Boutaud, O.; Printz, R.; Newcomb, D.C.; Foer, D.; Hodson, D.J.; Broichhagen, J.; Beckman, J.A.; Yu, C.; et al. Glucagon-Like Peptide-1 Receptor Regulates Thromboxane-Induced Human Platelet Activation. JACC Basic. Transl. Sci. 2022, 7, 713–715. [Google Scholar] [CrossRef]
  160. Barale, C.; Buracco, S.; Cavalot, F.; Frascaroli, C.; Guerrasio, A.; Russo, I. Glucagon-like Peptide 1-Related Peptides Increase Nitric Oxide Effects to Reduce Platelet Activation. Thromb. Haemost. 2017, 117, 1115–1128. [Google Scholar] [CrossRef]
  161. Zhang, Y.; Chen, R.; Jia, Y.; Chen, M.; Shuai, Z. Effects of Exenatide on Coagulation and Platelet Aggregation in Patients with Type 2 Diabetes. Drug Des. Dev. Ther. 2021, 15, 3027–3040. [Google Scholar] [CrossRef]
  162. Fadini, G.P.; Frison, V.; Simioni, N.; Lapolla, A.; Gatti, A.; Bossi, A.C.; Del Buono, A.; Fornengo, P.; Gottardo, L.; Laudato, M.; et al. Changes in the Prescription of Glucose-Lowering Medications in Patients with Type 2 Diabetes Mellitus After a Cardiovascular Event: A Call to Action From the DATAFILE Study. J. Am. Heart Assoc. 2019, 8, e012244. [Google Scholar] [CrossRef]
  163. Volaco, A.; Cavalcanti, A.M.; Filho, R.P.; Precoma, D.B. Socioeconomic Status: The Missing Link Between Obesity and Diabetes Mellitus? Curr. Diabetes Rev. 2018, 14, 321–326. [Google Scholar] [CrossRef]
  164. Wilding, J.P.H.; Batterham, R.L.; Davies, M.; Van Gaal, L.F.; Kandler, K.; Konakli, K.; Lingvay, I.; McGowan, B.M.; Oral, T.K.; Rosenstock, J.; et al. Weight Regain and Cardiometabolic Effects after Withdrawal of Semaglutide: The STEP 1 Trial Extension. Diabetes Obes. Metab. 2022, 24, 1553–1564. [Google Scholar] [CrossRef]
  165. Castellana, M.; Cignarelli, A.; Brescia, F.; Perrini, S.; Natalicchio, A.; Laviola, L.; Giorgino, F. Efficacy and Safety of GLP-1 Receptor Agonists as Add-on to SGLT2 Inhibitors in Type 2 Diabetes Mellitus: A Meta-Analysis. Sci. Rep. 2019, 9, 19351. [Google Scholar] [CrossRef]
  166. Chodorge, M.; Celeste, A.J.; Grimsby, J.; Konkar, A.; Davidsson, P.; Fairman, D.; Jenkinson, L.; Naylor, J.; White, N.; Seaman, J.C.; et al. Engineering of a GLP-1 Analogue Peptide/Anti-PCSK9 Antibody Fusion for Type 2 Diabetes Treatment. Sci. Rep. 2018, 8, 17545. [Google Scholar] [CrossRef]
  167. Perkovic, V.; Tuttle, K.R.; Rossing, P.; Mahaffey, K.W.; Mann, J.F.E.; Bakris, G.; Baeres, F.M.M.; Idorn, T.; Bosch-Traberg, H.; Lausvig, N.L.; et al. Effects of Semaglutide on Chronic Kidney Disease in Patients with Type 2 Diabetes. N. Engl. J. Med. 2024, 391, 109–121. [Google Scholar] [CrossRef]
  168. Frías, J.P.; Davies, M.J.; Rosenstock, J.; Pérez Manghi, F.C.; Fernández Landó, L.; Bergman, B.K.; Liu, B.; Cui, X.; Brown, K. Tirzepatide versus Semaglutide Once Weekly in Patients with Type 2 Diabetes. N. Engl. J. Med. 2021, 385, 503–515. [Google Scholar] [CrossRef]
  169. Jastreboff, A.M.; Aronne, L.J.; Ahmad, N.N.; Wharton, S.; Connery, L.; Alves, B.; Kiyosue, A.; Zhang, S.; Liu, B.; Bunck, M.C.; et al. Tirzepatide Once Weekly for the Treatment of Obesity. N. Engl. J. Med. 2022, 387, 205–216. [Google Scholar] [CrossRef]
  170. Sattar, N.; McGuire, D.K.; Pavo, I.; Weerakkody, G.J.; Nishiyama, H.; Wiese, R.J.; Zoungas, S. Tirzepatide Cardiovascular Event Risk Assessment: A Pre-Specified Meta-Analysis. Nat. Med. 2022, 28, 591–598. [Google Scholar] [CrossRef]
  171. Rosenstock, J.; Frias, J.; Jastreboff, A.M.; Du, Y.; Lou, J.; Gurbuz, S.; Thomas, M.K.; Hartman, M.L.; Haupt, A.; Milicevic, Z.; et al. Retatrutide, a GIP, GLP-1 and Glucagon Receptor Agonist, for People with Type 2 Diabetes: A Randomised, Double-Blind, Placebo and Active-Controlled, Parallel-Group, Phase 2 Trial Conducted in the USA. Lancet 2023, 402, 529–544. [Google Scholar] [CrossRef]
  172. Wharton, S.; Blevins, T.; Connery, L.; Rosenstock, J.; Raha, S.; Liu, R.; Ma, X.; Mather, K.J.; Haupt, A.; Robins, D.; et al. Daily Oral GLP-1 Receptor Agonist Orforglipron for Adults with Obesity. N. Engl. J. Med. 2023, 389, 877–888. [Google Scholar] [CrossRef]
  173. Frias, J.P.; Hsia, S.; Eyde, S.; Liu, R.; Ma, X.; Konig, M.; Kazda, C.; Mather, K.J.; Haupt, A.; Pratt, E.; et al. Efficacy and Safety of Oral Orforglipron in Patients with Type 2 Diabetes: A Multicentre, Randomised, Dose-Response, Phase 2 Study. Lancet 2023, 402, 472–483. [Google Scholar] [CrossRef] [PubMed]
  174. Espinosa, E.V.P.; Matute, E.M.; Sosa Guzmán, D.M.; Khasawneh, F.T. The Polypill: A New Alternative in the Prevention and Treatment of Cardiovascular Disease. J. Clin. Med. 2024, 13, 3179. [Google Scholar] [CrossRef] [PubMed]
  175. Castellano, J.M.; Pocock, S.J.; Bhatt, D.L.; Quesada, A.J.; Owen, R.; Fernandez-Ortiz, A.; Sanchez, P.L.; Marin Ortuño, F.; Vazquez Rodriguez, J.M.; Domingo-Fernández, A.; et al. Polypill Strategy in Secondary Cardiovascular Prevention. N. Engl. J. Med. 2022, 387, 967–977. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Intracellular signalling pathways induced by GLP-1 RA binding to GLP-1R in pancreatic β cells. These result in attenuation of endoplasmic reticulum (ER) stress (pink), inhibition of apoptosis (purple), increased insulin exocytosis (green), β cell proliferation (yellow) and improved glucose handling and homeostasis (white). AC: adenylyl cyclase, AKT/PKB: protein kinase B, AMPK: adenosine monophosphate-activated protein kinase, ATF4: activating transcription factor 4, ATP: adenosine triphosphate, B-raf: serine/threonine–protein kinase B-raf, Ca2+: calcium ions, cAMP: cyclic adenosine monophosphate, CREB: cAMP-response element binding protein, c-Src: tyrosine–protein kinase Src, DAG: diacylglycerol, dephosph-eIF2α: dephosphorylated eukaryotic initiation factor 2α, EGFR: epidermal growth factor receptor, Epac2: exchange protein activated by cAMP 2, ERK1/2: extracellular signal-regulated kinase 1/2, FOXO1: forkhead box protein O1, GLP-1R: glucagon-like peptide 1 receptor, GLP-1 RA: glucagon-like peptide 1 receptor agonist, HIF-α: hypoxia-inducible factor 1-α, IGF1R: insulin-like growth factor 1 receptor, IGF2: insulin-like growth factor 2, IP3: inositol 1,4,5-trisphosphate, IP3R: inositol 1,4,5-trisphosphate receptor, Irs2: insulin receptor substrate 2, mTOR: mammalian target of rapamycin, PDX-1: pancreatic and duodenal homeobox 1, PI3K: phosphoinositide 3-kinase, PIP2: phosphatidylinositol (4,5)-diphosphate, PLC: phospholipase C, PKA: protein kinase A, Rap1: Ras-proximate-1, RyR: ryanodine receptor, SUR1: sulfonylurea receptor 1, TCF7L2: transcription factor 7 like 2, VGCC: voltage-gated calcium channel, Wnt: wingless-related integration site. Created with BioRender.com.
Figure 1. Intracellular signalling pathways induced by GLP-1 RA binding to GLP-1R in pancreatic β cells. These result in attenuation of endoplasmic reticulum (ER) stress (pink), inhibition of apoptosis (purple), increased insulin exocytosis (green), β cell proliferation (yellow) and improved glucose handling and homeostasis (white). AC: adenylyl cyclase, AKT/PKB: protein kinase B, AMPK: adenosine monophosphate-activated protein kinase, ATF4: activating transcription factor 4, ATP: adenosine triphosphate, B-raf: serine/threonine–protein kinase B-raf, Ca2+: calcium ions, cAMP: cyclic adenosine monophosphate, CREB: cAMP-response element binding protein, c-Src: tyrosine–protein kinase Src, DAG: diacylglycerol, dephosph-eIF2α: dephosphorylated eukaryotic initiation factor 2α, EGFR: epidermal growth factor receptor, Epac2: exchange protein activated by cAMP 2, ERK1/2: extracellular signal-regulated kinase 1/2, FOXO1: forkhead box protein O1, GLP-1R: glucagon-like peptide 1 receptor, GLP-1 RA: glucagon-like peptide 1 receptor agonist, HIF-α: hypoxia-inducible factor 1-α, IGF1R: insulin-like growth factor 1 receptor, IGF2: insulin-like growth factor 2, IP3: inositol 1,4,5-trisphosphate, IP3R: inositol 1,4,5-trisphosphate receptor, Irs2: insulin receptor substrate 2, mTOR: mammalian target of rapamycin, PDX-1: pancreatic and duodenal homeobox 1, PI3K: phosphoinositide 3-kinase, PIP2: phosphatidylinositol (4,5)-diphosphate, PLC: phospholipase C, PKA: protein kinase A, Rap1: Ras-proximate-1, RyR: ryanodine receptor, SUR1: sulfonylurea receptor 1, TCF7L2: transcription factor 7 like 2, VGCC: voltage-gated calcium channel, Wnt: wingless-related integration site. Created with BioRender.com.
Jcm 13 04674 g001
Figure 2. Summary of the cardiometabolic benefits of GLP-1 RA therapy. Green upward arrow: increases; Red downward arrow, decreases. Created with BioRender.com.
Figure 2. Summary of the cardiometabolic benefits of GLP-1 RA therapy. Green upward arrow: increases; Red downward arrow, decreases. Created with BioRender.com.
Jcm 13 04674 g002
Figure 3. Putative Molecular Mechanisms for Anti-Atherogenic Effects of GLP-1 RAs. ABCA1: ATP-binding cassette transporter A1, ANG-II: angiotensin-II, AMPK: AMP-activated protein kinase, ANP: atrial natriuretic peptide, Arg-1: arginase-1, ATF6: cyclic AMP-dependent transcription factor-6, CCL2: C–C motif chemokine ligand 2, CCR7: C-C chemokine receptor type 7, CD62p: P-selectin, CD163+ Mac: cluster of differentiation 163+ macrophage, cGMP: cyclic guanine monophosphate, CRP: C-reactive protein, eNOS: endothelial nitric oxide synthase, FOXO3a: forkhead transcription factor O subfamily member 3a, HTN: hypertension, iNOS: inducible nitric oxide synthase, IFN-γ: interferon-γ, IL-1β: interleukin-1β, IL-6: interleukin-6, IL-10: interleukin-10, IRE1α: inositol-requiring enzyme 1, JNK: c-Jun N-terminal kinase, KLF2: Krüppel-like factor 2, LDL-C: low-density lipoprotein cholesterol, MAPK: mitogen-activated protein kinase, Mφ: macrophage, M1: M1 macrophage, M2: M2 macrophage, MMP: matrix metalloproteinase, MCP-1: monocyte chemoattractant protein-1, NLRP3: nucleotide-binding domain (NOD)-like receptor protein 3, NO: nitric oxide, PERK: protein kinase R-like ER kinase, PKG: protein kinase G, RAGE: receptor for advanced glycation end-products, ROS: reactive oxygen species, SIRT1: sirtuin 1, STAT1: signal transducer and activator of transcription 1, STAT3: signal transducer and activator of transcription 3, TAG: triacylglyceride, TIMP: tissue inhibitor of metalloproteinases, TNF-α: tumour necrosis factor-α, VASP: vasodilator-stimulated phosphoprotein, VCAM1: vascular cell adhesion protein 1, VSMC: vascular smooth muscle cell. Green upward arrow, increases. Red downward arrow, reduces. Red cross, inhibits. Created with BioRender.com.
Figure 3. Putative Molecular Mechanisms for Anti-Atherogenic Effects of GLP-1 RAs. ABCA1: ATP-binding cassette transporter A1, ANG-II: angiotensin-II, AMPK: AMP-activated protein kinase, ANP: atrial natriuretic peptide, Arg-1: arginase-1, ATF6: cyclic AMP-dependent transcription factor-6, CCL2: C–C motif chemokine ligand 2, CCR7: C-C chemokine receptor type 7, CD62p: P-selectin, CD163+ Mac: cluster of differentiation 163+ macrophage, cGMP: cyclic guanine monophosphate, CRP: C-reactive protein, eNOS: endothelial nitric oxide synthase, FOXO3a: forkhead transcription factor O subfamily member 3a, HTN: hypertension, iNOS: inducible nitric oxide synthase, IFN-γ: interferon-γ, IL-1β: interleukin-1β, IL-6: interleukin-6, IL-10: interleukin-10, IRE1α: inositol-requiring enzyme 1, JNK: c-Jun N-terminal kinase, KLF2: Krüppel-like factor 2, LDL-C: low-density lipoprotein cholesterol, MAPK: mitogen-activated protein kinase, Mφ: macrophage, M1: M1 macrophage, M2: M2 macrophage, MMP: matrix metalloproteinase, MCP-1: monocyte chemoattractant protein-1, NLRP3: nucleotide-binding domain (NOD)-like receptor protein 3, NO: nitric oxide, PERK: protein kinase R-like ER kinase, PKG: protein kinase G, RAGE: receptor for advanced glycation end-products, ROS: reactive oxygen species, SIRT1: sirtuin 1, STAT1: signal transducer and activator of transcription 1, STAT3: signal transducer and activator of transcription 3, TAG: triacylglyceride, TIMP: tissue inhibitor of metalloproteinases, TNF-α: tumour necrosis factor-α, VASP: vasodilator-stimulated phosphoprotein, VCAM1: vascular cell adhesion protein 1, VSMC: vascular smooth muscle cell. Green upward arrow, increases. Red downward arrow, reduces. Red cross, inhibits. Created with BioRender.com.
Jcm 13 04674 g003
Table 1. Chemical and pharmacokinetic properties of GLP-1 RAs. Adapted from Nauck and Meier, 2019; Tahrani, Barnett and Bailey, 2016; Yoon et al., 2020 [42,43,44].
Table 1. Chemical and pharmacokinetic properties of GLP-1 RAs. Adapted from Nauck and Meier, 2019; Tahrani, Barnett and Bailey, 2016; Yoon et al., 2020 [42,43,44].
Agent Name Brand Name Molecular Structure Molecular Weight (g/mol) Administration Dosing Half-Life (t1/2) Tmax
Exenatide Byetta®Modelled on exendin-4 from lizard Heloderma suspectum 39 a.a. peptide amide4186.6S/C twice daily.
1 h before meals, >6 h apart
Uptitration recommended
5 µg
10 µg
3–4 h2 h
Exenatide Extended-Release Bydureon Bcise®Modelled on exendin-4
39 a.a. synthetic peptide, suspension of microspheres in a MCT vehicle
4186.6S/C once weekly
Independent of meals
2 mg3–4 h §6 w
Lixisenatide † Adlyxinc®Modelled on exendin-4
44 a.a., amidated at the C-terminal a.a.
Contains poly-lysine tail
4858.6S/C once daily
1 h before first meal of day
Uptitration recommended
10 µg
20 µg
2–4 h1–2 h
Liraglutide Victoza®Modelled on mammalian GLP-1
Substitution of arginine for lysine at position 34. Attachment of a C-16 fatty acid (palmitic acid) at position 26
3751.3S/C once daily
Independent of meals
Uptitration recommended
0.6 mg
1.2 mg
1.8 mg
10–15 h9–12 h
Semaglutide (S/C) Ozempic®Modelled on mammalian GLP-1
Albumin binding, with a hydrophilic spacer and a C18 fatty di-acid
4113.6S/C once weekly
Independent of meals
Uptitration recommended
0.5 mg
1 mg
2 mg
1 w1–3 d
Semaglutide (Oral) Rybelsus®Modelled on mammalian GLP-1
Albumin binding, with a hydrophilic spacer and a C18 fatty di-acid
SNAC for enhanced stomach absorption
4113.6Oral once daily
>30 min before first meal of day
Uptitration recommended
7 mg
14 mg
1 w1 h
Dulaglutide Trulicity®Modelled on mammalian GLP-1. Fusion of 2 disulfide-linked GLP-1 chains to the Fc region of human IgG459,669.8S/C once weekly
Independent of meals
0.75 mg
1.5 mg
4 d12–72 h
Albiglutide Tanzeum®Modelled on mammalian GLP-1
2 chains of GLP-1 bound to albumin
72,070.4S/C once weekly
Independent of meals
Uptitration recommended
30 mg
50 mg
6–7 d3–5 d
Efpeglenatide †† FDA approval pendingExendin-based
Conjugation of exendin chain to IgG4 Fc fragment
NAS/C once weekly
Dosing time/regime not yet standardised
4 mg
6 mg
135–180 h72–144 h
§ Not formally assessed. However, given that this drug works by having exenatide bound to microspheres, it technically has the same half-life as the short-acting version, and this results in significant interindividual variability in absorption. † Lixisenatide has been discontinued by Sanofi in the UK and the USA. †† Since FDA approval is pending for efpeglenatide, standardised dosing and its brand name are not available. Abbreviations: a.a.: amino acid, d: days, h: hours, MCT: medium-triglyceride, S/C: subcutaneous, SNAC: sodium N-[8-(2-hydroxybenzoyl) amino caprylate, Tmax: time to peak drug concentration, w: weeks.
Table 2. Review of Cardiovascular Outcome Trials (CVOTs) for GLP-1 Receptor Agonists.
Table 2. Review of Cardiovascular Outcome Trials (CVOTs) for GLP-1 Receptor Agonists.
Trial ELIXA LEADER SUSTAIN-6 EXSCEL Harmony Outcomes REWIND PIONEER 6 AMPLITUDE-O
Reference Pfeffer et al. (2015) [53]Marso et al. (2016) [54]Marso et al. (2016) [50]Holman et al. (2017) [55]Hernandez et al. (2018) [56]Gerstein et al. (2019) [57]Husain et al. (2019) [58]Gerstein et al. (2021) [59]
Agent Name LixisenatideLiraglutideSemaglutide (S/C)Exenatide AlbiglutideDulaglutideSemaglutide (Oral)Efpeglenatide
Active Intervention Daily S/C lixisenatide
(10–20 μg)
Daily S/C liraglutide
(1.8 mg)
Weekly semaglutide S/C (0.5 or 1.0 mg)Weekly S/C ER exenatide
(2 mg)
Weekly S/C albiglutide
(30–50 mg)
Weekly S/C dulaglutide
(1.5 mg)
Daily oral semaglutide
(14 mg)
Weekly S/C efpeglenatide
(4 or 6 mg)
Participant Number 60689340329714,7529463990131834076
Patient Cohort Characteristics T2DM + ACS in last 180 d
Age ≥ 30
T2DM + high CV risk (age ≥ 50 with prior CAD, stroke, PVD, HF, CKD; or age ≥ 60 with CV RFs)T2DM + high CV risk (prior CAD, stroke, PVD, HF, CKD; or age ≥ 60 with CV RFs)T2DM +/− prior CVD
(70% had prior CVD)
T2DM + age ≥ 40 + prior CVD, stroke, PADT2DM + age ≥ 50 + CVD or CV RFsHigh CV risk (with established CVD or CKD; or age ≥ 60 with CV RFs)T2DM + age ≥ 18 + CVD; or
T2DM + age ≥ 50 + CV RFs + CKD
Median Follow-Up 2.1 years 3.8 years2 years3.2 years1.5 years5.4 years1.3 years1.8 years
Changes in HbA1c−0.27%−0.40%−1.1% (0.5 mg)
−1.4% (1.0 mg)
−0.7%−0.63% (8 m)
−0.52% (16 m)
−0.61%−0.7%−1.24%
Definition of Primary Endpoint CV death, MI, stroke or hospitalisation for unstable anginaFirst occurrence CV death, nonfatal MI or strokeFirst occurrence CV death, nonfatal MI or strokeFirst occurrence CV death, nonfatal MI or strokeFirst occurrence CV death, nonfatal MI or strokeFirst occurrence CV death, nonfatal MI or strokeFirst occurrence CV death, nonfatal MI or strokeFirst occurrence death from CV or undetermined causes, nonfatal MI or stroke
MACE Result (HR) 1.02 (95% CI [0.89, 1.17], p = 0.81)0.87 (95% CI [0.78, 0.97], p = 0.01)0.74 (95% CI [0.58, 0.95], p = 0.02)0.91 (95% CI [0.83, 1.00], p = 0.06)0.78 (95% CI [0.68–0.90], p = 0.0006)0.88 (95% CI [0.79–0.99], p = 0.026)0.79 (95% CI [0.57, 1.11], p = 0.001)0.73 (95% CI [0.58–0.92], p = 0.007)
All-Cause Death (HR) 0.94 (95% CI [0.78–1.13])0.85 (95% CI [0.74–0.97])1.05 (95% CI [0.74–1.50])0.86 (95% CI [0.77–0.97])0.95 (95% CI [0.79–1.16])0.90 (95% CI [0.80–1.01])0.51 (95% CI [0.31–0.84])0.78 (95% CI [0.58–1.06])
Cardiovascular Death (HR) 0.98 (95% CI [0.78–1.22])0.78 (95% CI [0.66–0.93])0.98 (95% CI [0.65–1.48])0.88 (95% CI [0.76–1.02])0.93 (95% CI [0.73–1.19])0.91 (95% CI [0.78–1.06])0.49 (95% CI [0.27–0.92])0.72 (95% CI [0.50–1.03])
Nonfatal Myocardial Infarction (HR) 1.03 (95% CI [0.87–1.22]) *0.88 (95% CI [0.75–1.03])0.74 (95% CI [0.51–1.08])0.97 (95% CI [0.85–1.10]) *0.75 (95% CI [0.61–0.90]) *0.96 (95% CI [0.79–1.16])1.18 (95% CI [0.73–1.90])0.78 (95% CI [0.55–1.10])
Nonfatal Stroke (HR) 1.12 (95% CI [0.79–1.58]) **0.89 (95% CI [0.72–1.11])0.61 (95% CI [0.38–0.99])0.85 (95% CI [0.70–1.03]) **0.86 (95% CI [0.66–1.14]) **0.76 (95% CI [0.61–0.95])0.74 (95% CI [0.35–1.57])0.80 (95% CI [0.48–1.31])
Hospitalisation for Heart Failure (HR) 0.96 (95% CI [0.75–1.23])0.87 (95% CI [0.73–1.05])1.11 (95% CI [0.77–1.61])0.94 (95% CI [0.78–1.13])0.85 (95% CI [0.70–1.04]) ***0.93 (95% CI [0.77–1.12])0.86 (95% CI [0.49–1.55])0.61 (95% CI [0.38–0.98])
Effect on Heart Rate +0.4 bpm+3.0 bpm+2.1 bpm
(0.5 mg)
+2.4 bpm
(1.0 mg)
+2.51 bpm+1.3 bpm+1.87 bpm+4 bpm+3.9 bpm
Effect on Weight from Baseline −0.6 kg−2.3 kg−3.6 kg (0.5 mg)
−4.9 kg (1.0 mg)
−1.27kg−0.66 kg (8 m)
−0.83 kg (16 m)
−1.46 kg−4.2kg−2.6 kg
Effect on Systolic Blood Pressure −0.8 mmHg−1.2 mmHg−3.4 mmHg
(0.5 mg)
−5.4 mmHg
(1.0 mg)
−1.57 mmHg−0.65 mmHg
(8 m)
−0.67 mmHg
(16 m)
−1.70 mmHg−2.6 mmHg−1.5 mmHg
Overall Effect on CVD No benefitMACE benefit + reduced all-cause and CV mortalityMACE benefit + reduced strokeNo MACE benefit but reduced all-cause mortalityMACE benefit + reduced MIMACE benefit + reduced strokeNo MACE benefit but reduced all-cause and CV mortalityMACE benefit + reduced HF hospitalisation
* This was reported as an aggregate of fatal and nonfatal myocardial infarction. ** This was reported as an aggregate of fatal and nonfatal stroke. *** This was reported as a composite of death from cardiovascular causes and hospital admission for heart failure. Trial abbreviations: AMPLITUDE-O: Effect of Efpeglenatide on Cardiovascular Outcomes; ELIXA: Evaluation of Lixisenatide in Acute Coronary Syndrome; EXSCEL: Exenatide Study of Cardiovascular Event Lowering; Harmony Outcomes: Effect of Albiglutide, When Added to Standard Blood Glucose Lowering Therapies, on Major Cardiovascular Events in Subjects With Type 2 Diabetes Mellitus; LEADER: Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results; PIONEER 6: Peptide Innovation for Early Diabetes Treatment 6; REWIND: Researching cardiovascular Events with a Weekly INcretin in Diabetes; SUSTAIN-6: Trial to Evaluate Cardiovascular and Other Long-term Outcomes with Semaglutide in Subjects with Type 2 Diabetes. Other abbreviations: ACS: acute coronary syndrome; bpm: beats per minute; CAD: coronary artery disease; CI: confidence interval; CKD: chronic kidney disease; CV: cardiovascular; CVD: cardiovascular disease; ER: extended release; HbA1c: glycated hemoglobin; HF: heart failure; HR: hazard ratio; MACE: major adverse cardiovascular event; MI: myocardial infarction; m: months; S/C: subcutaneous; T2DM: type 2 diabetes mellitus; PAD: peripheral arterial disease; PVD: peripheral vascular disease; RFs: risk factors.
Table 3. Active, In-Progress and Upcoming GLP-1 RA/incretin clinical trials registered on www.clinicaltrials.gov.
Table 3. Active, In-Progress and Upcoming GLP-1 RA/incretin clinical trials registered on www.clinicaltrials.gov.
Trial Name ClinicalTrials.gov ID Purpose Phase Trial Status Estimated Completion Expected Sample Size Inclusion Criteria Active Intervention Primary Outcome Measures
LEADPACE NCT04146155Liraglutide in diabetic patients with PADPhase 4UnknownDecember 2021200Age > 40; T2DM; HbA1c: 7.5–14%; PADLiraglutideInitial and absolute claudication distance and assessment of limb ischaemia
STARDUST NCT04881110Liraglutide on peripheral arterial perfusionPhase 4UnknownJune 202250T2DM; HbA1c 6.5–8%; PADLiraglutide Transcutaneous oxygen pressure (mmHg) on anterior and posterior tibial arteries
STOP NCT03985384Semaglutide on coronary plaque progression and composition using serial CTCAPhase 4Completed, awaiting publicationDecember 2022140Age ≥ 40; T2DM; HbA1c ≥ 7.0%; prior ASCVD or ≥1 CV RFSemaglutide S/CChange in noncalcified plaque volume on CTCA
SAMAS NCT05147896Anti-atherosclerotic mechanisms of oral semaglutidePhase 4Active, recruitingDecember 2023100T2DM; HbA1c ≤ 8.5%
BMI ≥ 30
Semaglutide oralcIMT;
endothelial function; arterial stiffness
Effects of Liraglutide on Epicardial Fat Pro-Inflammatory Genes in Type 2 Diabetes and Coronary Artery Disease NCT03260881Liraglutide on the inflammatory environment of EATPhase 4Active, recruitingDecember 202340T2DM; HbA1c ≤ 9%; BMI ≥ 27 and/or WC ≥ 102 cm (men) or 88 cm (women);
stable CAD requiring CABG
Liraglutide for minimum of 4–12 w prior to CABG EAT inflammation as measured by mRNA and protein expression of tumour necrosis factor (TNF)-alpha and interleukin (IL)-6 from blood sample
STRIDE NCT04560998Semaglutide on walking distance in PAD + T2DMPhase 3Active, not recruitingJuly 2024800T2DM; PADSemaglutide S/C Change in maximum walking distance
SOUL NCT03914326Oral semaglutide in T2DM with ASCVD and/or CKDPhase 3Active, not recruitingJuly 20249642Age ≥ 50; T2DM; HbA1c 6.5–10.0%; CAD, CeVD, PAD or CKDSemaglutide oralTime to first CV death, nonfatal MI or stroke
SURPASS-CVOT NCT04255433Tirzepatide vs. dulaglutide in T2DM and high CV riskPhase 3Active, not recruitingOctober 202413,299T2DM; HbA1c 7.0–10.5%;
ASCVD; BMI ≥ 25
Tirzepatide S/C Time to first CV death, MI or stroke
SURMOUNT-MMO NCT05556512Tirzepatide vs. placebo in nondiabetic overweight/obesity with high ASCVD risk Phase 3 Active, recruitingOctober 202715,000Age ≥ 40; BMI ≥ 27; prior ASCVD or high CV riskTirzepatide S/C Time to first all-cause death, nonfatal MI, nonfatal stroke, coronary revascularisation or heart failure events
TRIUMPH-3 NCT05882045Retatrutide vs. placebo in obesity with prior CVDPhase 3Active, recruitingFebruary 20261800BMI ≥ 35.0;
prior MI, stroke or PAD
Retatrutide S/C Percent change from baseline in body weight
ACHIEVE-4 NCT05803421Orforglipron vs. insulin glargine in T2DM and overweight/obesity and high CV riskPhase 3Active, recruitingDecember 20252620T2DM; HbA1c 7.0–10.5%; BMI ≥ 25; high CV riskOrforglipron oralTime to first CV death, MI, stroke or hospitalisation for unstable angina
Trial abbreviations: LEADPACE: Liraglutide Efficacy and Action on Type 2 Diabetes With Peripheral Atherosclerotic intErmittent Claudication: a Prospective, 24-week, Multicenter, Randomized, Controlled Clinical Study; STARDUST: Effects of the Glucagon Like-peptide 1 (GLP-1) Receptor Agonist Liraglutide on Lower Limb Perfusion in People With Type 2 Diabetes and Peripheral Artery Disease: a Randomized Controlled Trial; STOP: Semaglutide Treatment on Coronary Plaque Progression Trial; SAMAS: Semaglutide Anti-Atherosclerotic Mechanisms of Action Study in Type 2 Diabetes Patients; STRIDE: Effects of Semaglutide on Functional Capacity in Patients With Type 2 Diabetes and Peripheral Arterial Disease; SOUL: Semaglutide Cardiovascular Outcomes Trial in Patients With Type 2 Diabetes; SURPASS-CVOT: The Effect of Tirzepatide Versus Dulaglutide on Major Adverse Cardiovascular Events in Patients With Type 2 Diabetes; SURMOUNT-MMO: A Phase 3, Randomized, Double-blind, Placebo-Controlled Study to Investigate the Effect of Tirzepatide on the Reduction of Morbidity and Mortality in Adults With Obesity; TRIUMPH-3: A Study of Retatrutide (LY3437943) in Participants With Obesity and Cardiovascular Disease; ACHIEVE-4: A Study of Daily Oral Orforglipron (LY3502970) Compared With Insulin Glargine in Participants With Type 2 Diabetes and Obesity or Overweight at Increased Cardiovascular Risk. Other abbreviations: ASCVD: atherosclerotic cardiovascular disease; BMI: body mass index; CAD: coronary artery disease; CABG: coronary artery bypass graft; CeVD: cerebrovascular disease; cIMT: carotid intima–media thickness; CKD: chronic kidney disease; CTCA: computed tomography coronary angiogram; CV: cardiovascular; CVD: cardiovascular disease; EAT: epicardial adipose tissue; HbA1c: glycated hemoglobin; MI: myocardial infarction; PAD: peripheral artery disease; RF: risk factor; S/C: subcutaneous; T2DM: type 2 diabetes mellitus; WC: waist circumference.
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

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. https://doi.org/10.3390/jcm13164674

AMA Style

Le R, Nguyen MT, Allahwala MA, Psaltis JP, Marathe CS, Marathe JA, Psaltis PJ. Cardiovascular Protective Properties of GLP-1 Receptor Agonists: More than Just Diabetic and Weight Loss Drugs. Journal of Clinical Medicine. 2024; 13(16):4674. https://doi.org/10.3390/jcm13164674

Chicago/Turabian Style

Le, Richard, Mau T. Nguyen, Momina A. Allahwala, James P. Psaltis, Chinmay S. Marathe, Jessica A. Marathe, and Peter J. Psaltis. 2024. "Cardiovascular Protective Properties of GLP-1 Receptor Agonists: More than Just Diabetic and Weight Loss Drugs" Journal of Clinical Medicine 13, no. 16: 4674. https://doi.org/10.3390/jcm13164674

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop