IMR Press / FBL / Volume 28 / Issue 11 / DOI: 10.31083/j.fbl2811315
Open Access Review
Potential Role of GLP-1 Based Therapeutics in Coronary Artery Disease
Show Less
1 Department of Cardiology, Fengcheng People’s Hospital, Fengcheng Hospital Affiliated to Yichun University, 331100 Fengcheng, Jiangxi, China
2 Prevention & Healthcare Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 030032 Taiyuan, Shanxi, China
3 Health Commission of Shanxi Province, 030032 Taiyuan, Shanxi, China
4 Department of Cardiology, The Third Clinical Medical College of Shanxi Medical University, 030032 Taiyuan, Shanxi, China
*Correspondence: 825283252@qq.com (Wenbo Li); 13485385229@163.com (Lihui Zhang)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2023, 28(11), 315; https://doi.org/10.31083/j.fbl2811315
Submitted: 21 May 2023 | Revised: 30 July 2023 | Accepted: 17 August 2023 | Published: 29 November 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Glucagon-like peptide-1 (GLP-1), an incretin hormone primarily secreted by intestinal L cells, regulates glucose metabolism by increasing insulin synthesis and secretion, decreasing plasma glucagon levels, reducing food intake, and slowing gastric emptying. This has led to the development of GLP-1 receptor (GLP-1R) agonists as a treatment for diabetes and obesity. In addition to being present in beta cells, GLP-1R has also been identified in blood vessels and the heart, suggesting that GLP-1R agonists may have an impact on cardiovascular health. There is now substantial evidence supporting GLP-1’s protective effects on the cardiovascular system. This review summarizes the current research on GLP-1-based therapy for coronary artery disease (CAD) by examining its protective effects against inflammation and ischemia/reperfusion injury and analyzing clinical trials on GLP-1-based therapies for CAD. Although results from various studies were inconsistent, the challenge of transitioning GLP-1-based therapies from the laboratory to the clinical setting remains. Further well-designed and high-quality studies are necessary to determine the efficacy and safety of GLP-1 for patients with CAD.

Keywords
glucagon-like peptide-1
GLP-1 receptor agonists
coronary artery disease
acute/chronic coronary syndromes
ischemia/reperfusion injury
inflammation
1. Introduction

The close relationship between coronary artery disease (CAD) and type 2 diabetes mellitus (T2DM) has been established by current research. Patients with T2DM are more likely to experience CAD earlier in life and in a more severe and widespread manner compared to non-diabetic patients [1]. The progression of arteriosclerosis in this population occurs earlier and at a faster rate compared to non-diabetic individuals [2]. The significant public health impact caused by ischemic heart disease is projected to rise as the population ages and the incidence of comorbidities such as obesity and diabetes increases. According to projections, the global prevalence of diabetes among adults aged 20–79 years is expected to surge in the coming decades, reaching 783.2 million by 2045 [3].

Two significant trials have been reported: DCCT (the Diabetes Control and Complications Trial) [4] in type 1 diabetes (T1DM) and ADVANCE (Action in Diabetes and Vascular Disease: Preterax and Diamicron MR Controlled Evaluation) [5] in T2DM. The results from both trials showed that improved glycemic control reduces microvascular disease and can effectively lower long-term cardiovascular complications. The United Kingdom Prospective Diabetes Study [6] suggested that intensive glycemic control can significantly reduce the occurrence of diabetes-related events. However, the impacts on cardiovascular events and mortality were not substantial. Another study revealed that the intensive-therapy regimen resulted in an unexpected increase in mortality [7]. As a result, the Food and Drug Administration (FDA) established guidelines that any anti-hyperglycemic prescription medications must demonstrate the safety of their effects on the heart and blood vessels before they can be marketed. This policy was revised and strengthened in March 2020 [8].

Fortunately, several new pharmacological agents, such as glucagon-like peptide-1 (GLP-1) receptor agonists, dipeptidyl peptidase-4 (DPP-4) inhibitors, and sodium-glucose co-transporter 2 (SGLT2) inhibitors have obtained acceptance among medical practitioners and patients due to their reliable cardiovascular profiles as demonstrated in cardiovascular outcome studies (CVOTs) [9, 10]. Despite this, controversies surrounding their risk of cardiovascular events persist. In this review, we aim to summarize and evaluate the available evidence from pre-clinical studies and clinical trials regarding the effects and underlying mechanisms of GLP-1-based therapies on CAD.

2. The Properties of GLP-1 and GLP-1 Receptor Agonists

GLP-1 was first discovered as a hormone that stimulates insulin production and is secreted by intestinal endocrine L-cells. GLP-1 is released within minutes of consuming food and helps to speed up the metabolism of nutrients. One of the defining characteristics of GLP-1 is its extremely brief half-life of approximately 2 minutes, making it highly susceptible to decomposition by DPP-4. The predominant biologically active variant of GLP-1, known as GLP-1 (7-36), initiates signaling cascades through the GLP-1 receptor (GLP-1R). Yet, it undergoes rapid conversion into GLP-1 (9-36) [11]. The GLP-1R is a protein consisting of 463 amino acids belonging to the Class B family of 7-transmembrane-spanning receptors [12]. GLP-1 and GLP-1 receptor agonists exert multiple physiological effects, including increased insulin production and release, decreased glucagon secretion, delayed gastric emptying, and increased satiety by acting on the GLP-1R.

The initial approval of the first GLP-1R agonist for the management of T2DM in 2005 was attributed to the therapeutic impact of GLP-1 on glucose metabolism [13]. Since then, the FDA has approved various GLP-1R agonist products with diverse structures and pharmacokinetics (as shown in Table 1). These drugs are commonly recommended as combination therapy in addition to metformin for patients who have not achieved their glycemic goals, particularly those who need to lose weight and reduce the risk of hypoglycemia [14]. The expression of GLP-1R is not limited to pancreatic islet cells but is also observed in multiple other anatomical locations, including the lung, brain, kidney, stomach, and liver. Considerable evidence suggests that the expression of GLP-1R has been further localized in cardiomyocytes, microvascular endothelium, the endocardium, the coronary arteries, and the smooth muscle cells [15].

Table 1.Comparison of GLP-1-receptor agonist products approved by the FDA for treatment of T2DM.
Generic Proprietary Dosing regimen Half-life Sponsor Year approved by FDA
Exenatide Byetta Subcutaneous injection twice-daily 2.5 h AstraZeneca 2005
Liraglutide Victoza Subcutaneous injection once-daily 12 h Novo Nordisk 2010
Exenatide extended-release (ER) Bydureon Subcutaneous injection once-weekly n/a AstraZeneca 2012
Albiglutide* Tanzeum Subcutaneous injection once-weekly 120 h GlaxoSmithKline 2014
Dulaglutide Trulicity Subcutaneous injection once-weekly 90 h Eli Lilly 2014
Liraglutide Saxenda Subcutaneous injection once-daily 12 h Novo Nordisk 2014
Lixisenatide Adlyxin Subcutaneous injection once-daily 3~4 h Sanofi 2016
Semaglutide Ozempic Subcutaneous injection once-weekly 160 h Novo Nordisk 2018
Oral semaglutide Rybelsus Oral once-daily 160 h Novo Nordisk 2018
Semaglutide Wegovy Subcutaneous injection once-weekly 160 h Novo Nordisk 2021

*Albiglutide was discontinued in 2017 due to a decrease in sales. GLP-1, Glucagon-like peptide-1; FDA, Food and Drug Administration; T2DM, type 2 diabetes mellitus. n/a: not applicable.

While GLP-1R agonism is primarily recognized for its insulin-stimulating and weight-reducing properties, studies have also demonstrated its several positive effects on the cardiovascular system in animal models [16]. Importantly, the improvement in cardiovascular function brought about by GLP-1 is not solely dependent on its ability to enhance energy metabolism and reduce body weight. GLP-1 may enhance cardiovascular function through direct action on GLP-1R and indirect mechanisms that are unrelated to its mode of action in the heart.

3. Pre-Clinical Evidence for the Anti-Inflammation Effects of GLP-1

Inflammation, especially in those with diabetes, plays an important role in the development of atherosclerosis. Systemic inflammation can speed up the advancement of the disease and heighten the susceptibility to other CAD, such as acute coronary syndromes (ACS) and myocardial infarction (MI). Proinflammatory stimuli can cause atherosclerosis, which is marked by elevated levels of Tumor Necrosis Factor-α (TNF-α), Interleukin 6 (IL-6), C-Reactive Protein (CRP), or endotoxin [17]. Anti-inflammatory drugs were postulated to have beneficial effects on the natural progression of CAD by slowing down plaque expansion, reducing the risk of acute plaque changes, and improving the outcome of MI events [18].

Both endogenous GLP-1 and GLP-1R agonists can diminish cardiovascular inflammation via direct and indirect means. GLP-1 infusion decreases the elevation in microvascular permeability during inflammation caused by lipopolysaccharide (LPS), and the positive impact may be due to GLP-1R/cAMP pathways [19]. Liraglutide has a protective effect against TNFα-induced inflammation in human aortic endothelial cells by inhibiting nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, Protein kinase C (PKC-α), Nuclear factor-kappaB (NF-κB) signaling [20].

Sirtuin 6 (SIRT6), a histone deacetylase, has been recognized as a promising therapeutic target for the treatment of inflammatory vascular diseases due to its effects on NF-κB in HUVECs [21]. Plaques in individuals with diabetes have more inflammation and higher collagen content, along with increased SIRT6 expression, compared to those in non-diabetic individuals. GLP-1-based therapy can reduce the inflammation in diabetic plaques by regulating SIRT6/NF-κB signaling [21].

Liraglutide reduces the level of E-selectin and vascular cell adhesion molecule (VCAM-1) in response to endotoxin stimulation. This inhibitory effect was nullified by inhibiting calmodulin-dependent protein kinase kinase-β (CAMKKβ). In addition, knocking down 5 adenosine monophosphate-activated protein kinase (AMPK) abolished the liraglutide-stimulated phosphorylation of AMPK and its anti-inflammatory effect. This finding indicates that the activation of CaMKKβ and AMPK is correlated with the anti-inflammatory effect of liraglutide in endothelial cells [22]. A one-week administration of liraglutide had an anti-inflammatory effect in C57BL/6 mice with obesity produced by a high-fat diet, independent of changes in body weight. These obese mice exhibited a decrease in the level of phosphorylated AMP-activated protein kinase (pAMPK), along with cardiac insulin resistance. Additionally, there was an increase in the expression of TNF-α and NF-κB [18]. Our previous research also showed that recombination of GLP-1 and liraglutide can suppress the expression of matrix metalloproteinase 1 (MMP1) by inhibiting the ERK1/2-NF-κB signaling pathway, reversing vascular remodeling [23]. Additionally, studies have demonstrated that the administration of exenatide elicits a strong and rapid anti-inflammatory action, regardless of weight loss, and this effect becomes evident within the first two hours of treatment [24]. These findings suggest that the direct anti-inflammatory properties of GLP-1 may be separate from its hypoglycemic effects.

Intriguingly, in a mouse model of atherosclerosis, it has been demonstrated that GLP-1 (9-37) and (28-37), which are breakdown products of GLP-1 (7-37), have the potential to decrease inflammation within plaques and enhance their stability, despite their inability to control glucose metabolism [25]. Future studies are required to clarify the exact impact of GLP-1-based treatment on atherosclerosis and coronary plaque stability. It is speculated that the mechanism may be associated with the indirect control of weight loss, glucose levels, blood pressure, inhibition of inflammation, and platelet activation [26]. However, a more detailed analysis of these elements falls outside the scope of this review.

4. GLP-1-Based Therapies and Coronary Blood Flow

Abnormal coronary blood flow is believed to contribute to ischemic heart disease. Insufficient coronary blood flow leads to fibrosis and necrosis of the heart muscle. Reviving the balance between oxygen consumption and demand is critical to preserving heart muscle in the face of obstructed coronary arteries [27]. While coronary intervention is an effective solution, finding effective drugs is also important.

Previous research has demonstrated that GLP-1R agonists can increase coronary flow in aerobically perfused mouse hearts through GLP-1R-dependent and GLP-1R-independent mechanisms [28]. The administration of GLP-1 through infusion has been observed to result in an augmentation in microvascular blood volume and flow velocity. This effect is believed to be facilitated by a process that relies on nitric oxide, as evidenced by the utilization of contrast-enhanced ultrasound imaging [29]. Additionally, another study has demonstrated that GLP-1 administration leads to an augmentation of coronary blood flow in conscious dogs with dilated cardiomyopathy produced by rapid pacing [30].

In a post-resuscitation swine model, continuous GLP-1 infusion increased coronary flow reserve, as estimated by intracoronary Doppler flow measurements [31]. Another study found that the administration of human GLP-1 by intravenous infusion following cardiac arrest and resuscitation resulted in enhanced coronary microvascular function via reducing oxidative stress [32]. Our team has also reported that GLP-1 exhibits a dosage-dependent inhibition of thromboxane receptor agonist-induced contraction in rat coronary artery rings, and this effect may be due to stimulation of potassium-ATP currents in the vascular endothelium [33]. Additionally, the presence of the relaxant effect of GLP-1 was not detected following the removal of the endothelial layer, indicating the potential involvement of the vascular endothelium. Putative mechanisms may include stimulation of the potassium-ATP currents [33]. A study on a rat model of metabolic syndrome found that chronic administration of liraglutide improved the capacity for nitric oxide-mediated dilation in coronary vessel internal diameter. These improvements were observed irrespective of any changes in body mass or control of blood glucose levels [34].

Clinical studies, however, have produced mixed results. A small, short-term clinical study revealed that infusion of GLP-1 resulted in a notable enhancement of resting myocardial blood flow (MBF) by about 24% in patients with T2DM but without heart failure or CAD, as seen by quantitative positron emission tomography (PET) [35]. Comparable findings were found in healthy participants using semi-quantitative contrast-enhanced echocardiography [36]. Conversely, a study of patients with non-diabetic chronic heart failure found that liraglutide administration for 24 weeks did not affect myocardial blood flow despite reducing weight, HbA1c, and 2-hour glucose values compared to placebo [37].

Although the relationship between GLP-1 and its receptor agonists and the nature of coronary arteries has been studied, the exact mechanisms linking GLP-1 to improved coronary blood flow are still unclear. One possible explanation is that GLP-1 may indirectly improve myocardial capillaries through the inhibition of myocardial hypertrophy [38, 39]. Currently, large randomized controlled trials investigating the influence of GLP-1 on coronary blood flow in CAD patients are lacking, and the available data is derived from small-sample studies with varying inclusion criteria. Additional studies are warranted to examine the role of GLP-1-based therapies in addressing issues such as coronary spasm, slow flow, and no flow, which are increasingly becoming angiographic phenomena in the treatment of CAD [40].

5. GLP-1-Based Therapies and Ischemia-Reperfusion Injury

Ischemia-reperfusion (I/R) injury is the main pathological manifestation of CAD that causes myocardial damage. Currently, there is no effective medication to defend the cardiovascular system against I/R. Rapid increases in intracellular cAMP and calcium levels brought on by GLP-1 activation trigger PKA activation and phosphorylation of cyclic AMP response element binding protein (CREB). GLP-1R agonists can modulate the activity of CREB by stimulating cytoplasmic-to-nuclear translocation of target of rapamycin (TOR) complex-2 (TORC2) [16]. Moreover, GLP-1 receptor activation also activates PKC, extracellular regulated kinase 1/2 (ERK1/2), AMPK, and phosphoinositide 3-kinase (PI3K) [41]. These pro-survival intracellular signaling pathways and proteins protect against I/R injury.

The impact of GLP-1-based therapy on ischemia-reperfusion (I/R)-induced cardiac damage has been largely demonstrated in animal models [28, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52]. However, these studies have exhibited variable findings, with some reporting positive outcomes, while others have shown neutral or negative effects (as shown in Table 2, Ref. [28, 42, 44, 45, 47, 48, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59]). For example, Kavianipour et al. [53] showed that the administration of recombinant GLP-1 did not limit infarct size during I/R when utilizing a porcine heart model. Similarly, in other studies using porcine ischemia-reperfusion models induced by balloon occlusion, liraglutide also exerts neutral properties on infarct size [54, 55]. Timmers et al. [56] demonstrated that exenatide treatment reduced myocardial infarct size in Dalland Landrace pig I/R models. The variation in the consequences of GLP-1 on I/R injury may also be attributed to variations in dosing times, whether it was given before or after the injury or the duration and methods used to artificially obstruct coronary blood flow.

Table 2.The effect of GLP-1 on Ischemia-reperfusion (I/R) injury and its underlying mechanisms.
Authors Agent Subjects Results or potential mechanisms Ref
Ban et al. GLP-1 (7-36), GLP-1 (9-36) Mice Both GLP-1 (7-36) and GLP-1 (9-36) reduced ischemic damage after I/R and increased cGMP release in wild-type and Glp1r(-/-) mice [28]
Bose et al. GLP-1 Rat Activation of cAMP and PI3K [42]
Bose et al. GLP-1 Rat Activation of mTOR/p70s6 [44]
Huisamen et al. GLP-1 (7-36) Rat Activation of AMPK and PKB/Akt [45]
Noyan-Ashraf et al. Liraglutide Mice Activation of PKB, GSK3beta, PPARbeta/delta, Nrf-2, and HO-1 [47]
Sonne et al. Exendin-4, GLP-1 (9-36) Rat Exendin-4 protect against I/R injury by activating GLP-1R [52]
Eid et al. Exendin-4 Rat Activation of SIRT1, SIRT3 and AMPK [51]
Bao et al. Albiglutide Rat Increasing both glucose and lactate oxidation [48]
Wohlfart et al. Lixisenatide Rat Independent of GLP-1R [50]
Kavianipour et al. GLP-1 Pig GLP-1 does not limit infarct size [53]
Kristensen et al. liraglutide Pig Liraglutide has a neutral effect on myocardial infarct size [54]
Ekström et al. liraglutide Pig Liraglutide does not reduce infarct size [55]
Timmers et al. Exenatide Pig Exenatide reduces infarct size by increasing the expression of PKB and Bcl-2 [56]
Siraj et al. GLP-1 (28-36) hcaECs, hcaSMCs Activation of MTPα and sAC [57]
Ban et al. Exendin-4, GLP-1 (9-36) Rat, CMs cAMP and phosphorylation of ERK1/2 and the phosphoinositide 3-kinase target protein kinase B/Akt [58]
Besch et al. Exenatide Patients admitted for CABG Exenatide does not exert any additional cardioprotective effect compared to insulin in patients undergoing scheduled CABG surgery [59]

Abbreviations: AMPK, AMP-activated protein kinase; Bcl-2, B-cell lymphoma 2; CMs, cardiomyocytes; cAMP, cyclic adenosine 3,5-monophosphate; CABG, coronary artery bypass graft; GLP-1-Tf, GLP-1 to human transferrin; GSK3beta, Glycogen synthase kinase-3; HO-1, heme oxygenase-1; hcaECs, human coronary artery endothelial cells; hcaSMCs, human coronary artery smooth muscle cells; sAC, soluble adenylyl cyclase; SIRT6, sirtuin 6; mTOR, mammalian target of rapamycin; MTPα, mitochondrial trifunctional protein-α; Nrf-2, NF-E2-related factor 2; PI3K, phosphoinositide 3-kinase; PKB, Protein kinase B; PPARbeta/delta, Peroxisome proliferator-activated receptor beta/delta; p70s6, 70-kDa ribosomal protein S6 kinase; sAC, soluble adenylyl cyclase.

Moreover, the protective effects of GLP-1 seem to be independent of its receptors [28]. In the C57BL/6J mice I/R model, pre-treatment with GLP-1 (28-36) reduces myocardial infarct size through the suppression of inhibiting mitochondrial trifunctional protein-α, and the protective property was preserved in Glp1r-/- mice [57]. Ban and colleagues [58] revealed that the protective effects of exendin-4 were abolished in cardiomyocytes treated with exendin (9–39), as well as in cardiomyocytes isolated from GLP-1r-/- mice. Also, GLP-1 (9-36), a GLP-1 metabolite made by enzymatic cleavage that was thought to be biologically inactive at first, reduced the size of an infarct after I/R in a rat model and protected cardiomyocytes from GLP-1r-/- mice. In a clinical trial conducted by Besch et al. [59], the researchers did not observe additional cardioprotective effects on I/R injury with exenatide versus insulin in patients undergoing coronary artery bypass grafting (CABG) surgery.

The disparities between the results mentioned above can be attributed to several factors. Firstly, different dosing phases may have led to varying results. Several studies have suggested that early drug administration exhibits the greatest protection against myocardial I/R injury. Secondly, differences in results may be due to inadequate doses. Clinical studies of drugs often use off-label doses, which presents challenges in establishing comparability between clinical and pre-clinical studies. Finally, other interfering or confounding factors may have overshadowed the beneficial impact of GLP-1-based therapy. For instance, the aforementioned randomized controlled trial utilized insulin in both the control and study groups, and it is well-established that insulin protects against I/R injury [60, 61]. Therefore, more research is needed to evaluate the potential benefits of various GLP-1R agonists in the context of I/R damage.

6. Clinical Data on Coronary Artery Disease from GLP-1-Based Therapies.
6.1 Chronic Coronary Syndromes

Evidence on the impact of GLP-1RAs on outcomes in chronic coronary syndromes (CCS) is still minimal (as shown in Table 3, Ref. [62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79]), particularly from clinical trials. Research has demonstrated that GLP-1 protects against ischemic dysfunction as evaluated by left ventricular (LV) conductance catheter in patients with CAD [62, 63]. Read et al. [64] did a study that backs up this claim. They found that GLP-1 therapy protects the heart against ischemic LV dysfunction in patients with CAD who are waiting for revascularization. Furthermore, the benefits were found to be sustained even during a hyperglycemic hyperinsulinemic clamp (HHC), indicating that they are independent of glycemic control [65].

Table 3.Overview of studies reporting on GLP-1-based therapies for CAD.
Authors Year Subjects N Agent Result Ref
Read et al. 2011 Patients with normal LV function and single-vessel coronary disease within the LAD artery undergoing elective PCI 20 GLP-1 (7-36) GLP-1 improved recovery of LV systolic and diastolic function at 30 minutes after balloon occlusion compared with control [62]
McCormick et al. 2015 Patients with preserved LV function and single-vessel coronary disease within the LAD artery undergoing elective PCI 20 GLP-1 (7-36) Pre-treatment with GLP-1 (7-36) protects the heart against ischemic LV dysfunction and improves the recovery of function during reperfusion [63]
Read et al. 2012 Patients with CAD and good LV function awaiting revascularization 14 GLP-1 Global LV function was greater at peak stress during GLP-1 infusion compared with control [64]
McCormick et al. 2015 Patients with obstructive CAD (at least one proximal stenosis >70% and diabetes 10 GLP-1 (7-36) GLP-1 improved both global and regional myocardial performance at peak stress and at 30-min recovery [65]
Kumarathurai et al. 2016 Patient with T2D and stable CAD 41 Liraglutide Liraglutide did not improve LV ejection fraction when compared to placebo [66]
Kumarathurai et al. 2021 Patients with CAD and newly diagnosed T2DM 40 Liraglutide Liraglutide did not improve diastolic function parameters in subjects with T2DM, CAD, and preserved LVEF [67]
Myat et al. 2021 Patients with chronic stable angina 22 Liraglutide Liraglutide did not improve exercise tolerance or hemodynamics compared with saline placebo during serial treadmill testing [68]
Suhrs et al. 2019 Female overweight patients with CMD and angina symptoms 29 Liraglutide Liraglutide did not alleviate angina and improve coronary microvascular function [69]
Chen et al. 2016 Patients with STEMI undergoing PCI 210 Liraglutide Liraglutide lowers the prevalence of no-reflow as compared with the control group, and the incidence of MACE is no different [70]
Lønborg et al. 2012 Patients with STEMI and TIMI flow 0/1 148 Exenatide Exenatide reduces infarct size only when the duration of ischemia before primary PCI is less than 132 mins [71]
Nikolaidis et al. 2004 Patient with AMI and LVEF <40% after successful reperfusion 21 GLP-1 (7-36) GLP-1 significantly improved LVEF (from 29 ± 2% to 39 ± 2%, p < 0.01) compared with control subjects [73]
Chen et al. 2016 Patient with STEMI undergoing PPCI 77 Liraglutide Liraglutide increases salvage index and reduces infarct size [74]
Chen et al. 2015 Patient with STEMI undergoing PPCI 85 Liraglutide Liraglutide increases LVEF at 3 months [75]
Chen et al. 2016 Patient with NSTEMI 90 Liraglutide Liraglutide increases LVEF at 3 months [76]
Lønborg et al. 2012 Patients with STEMI and TIMI flow 0/1 172 Exenatide Exenatide increases salvage index [72]
Woo et al. 2013 Patients with STEMI and TIMI flow 0 116 Exenatide Exenatide reduces infarct size, and biomarkers rise [77]
Roos et al. 2016 Patients with STEMI and TIMI flow 0/1. T2DM was excluded. 91 Exenatide No difference was found in infarct size [78]
Pfeffer et al. 2015 Patients with T2DM and recent ACS 6068 Lixisenatide No significant between-group differences in the rate of hospitalization for heart failure or the rate of death [79]

Abbreviation: ACS, acute coronary syndrome; AMI, acute myocardial infarction; CAD, coronary artery disease; CMD, Coronary microvascular dysfunction; LAD, Left anterior descending artery; LV, left ventricular; LVEF, left ventricular ejection fraction; NSTEMI, non-ST- elevation myocardial infarction; PCI, percutaneous coronary intervention; PPCI, primary percutaneous coronary intervention; STEMI, ST-elevation myocardial infarction; T2DM, Type 2 diabetes mellitus; TIMI, thrombolysis in myocardial infarction.

In a randomized controlled trial (RCT) with patients who had stable CAD, preserved LVEF, and T2DM, giving liraglutide for 12 weeks failed to show a significant improvement in left ventricular ejection fraction (LVEF) [66]. However, no decline in cardiac function was observed, suggesting that liraglutide appears to be a safe therapeutic alternative when used in combination with metformin therapy in these patients. In another RCT conducted by the same research team, it was concluded that liraglutide did not have any impact on diastolic function [67]. In the LIONESS study [80], patients with chronic stable angina were given liraglutide for 3 plus 3 weeks to see how it affected exercise hemodynamics during exercise stress testing. Results showed that liraglutide had no effect on any improvements in parameters of hemodynamic performance during exercise or the magnitude of ST-segment depression seen at peak exercise [68]. Another open-label study that enrolled women with coronary microvascular dysfunction in which participants were treated with liraglutide for 3 months showed liraglutide failed to produce any improvements in symptoms or coronary flow velocity reserve (CFVR) [69].

6.2 Acute Coronary Syndromes

Despite having a cardioprotective effect, the impact of GLP-1 based therapies on ACS has not been as strong as anticipated from pre-clinical studies. However, recent research indicates that GLP-1R agonists have a favorable cardiovascular safety profile for patients with T2DM. A prospective study reported that pre-treatment with liraglutide demonstrated a significant association with a reduced occurrence of no-reflow in patients with ST-elevation myocardial infarction (STEMI) as compared to placebo (5% vs. 15%, p = 0.01) [70]. However, the primary endpoint of the study was to determine the prevalence of angiographic no-reflow as assessed by subjective TIMI flow. Other objective assessment techniques might provide a clearer understanding of microvascular injury. Following a 3-month period of observation, there was no significant difference in the occurrence of major adverse cardiovascular events (MACE) between the liraglutide group and the control group (8% vs. 15%, p = 0.12) [70].

Studies indicate that administering cardioprotective medications early during reperfusion provides optimal protection from I/R injury [81]. In a randomized trial conducted by Lønborg et al. [71, 72], the administration of exenatide during the period of ischemia and continuing for 6 hours reduced final infarct size among patients with STEMI and a brief ischemic duration (132 minutes). However, patients with a prolonged ischemic delay (>132 minutes) did not exhibit any cardioprotective effect. Nikolaidis et al. [73] demonstrated that administering recombinant GLP-1 by continuous infusion for 72 hours after successful reperfusion improved LV function in patients who had experienced acute myocardial infarction (AMI).

Another clinical trial conducted to evaluate the role of liraglutide on reperfusion injury in patients with STEMI who underwent primary PCI found that administering liraglutide before intervention and maintaining it for one week resulted in an increased salvage index and improved infarct size after 3 months. Similarly, there was no difference noted in the incidence of MACE during the half-year follow-up period [74]. Moreover, one week of subcutaneous liraglutide treatment in both STEMI and non-STEMI patients revealed an enhancement in LVEF during a subsequent 3-month follow-up period [75, 76]. Two studies conducted in a population of patients with STEMI who underwent primary PCI suggested that adjunctive exenatide therapy could also limit infarct size and increase salvage index [72, 77]. The findings of these abovementioned studies differ from those of a similar randomized clinical trial examining exenatide, with no apparent difference in final infarct size (18.8 ± 13.2 vs. 18.8 ± 11.3% of left ventricular mass, p = 0.965) [78].

The ELIXA trial (The Evaluation of Lixisenatide in Acute Coronary Syndrome) [82] provided high certainty evidence that treatment with lixisenatide has a neutral cardiovascular profile for patients with T2DM who recently experienced ACS. The agent had no substantial impact on the rate of MACE or any other adverse events, including hypoglycemia, pancreatitis, pancreatic neoplasms, or allergic reactions. Although there were only 2 years of follow-up in this clinical setting, enough cardiovascular events were observed to rule out a significant non-glycemic cardiovascular benefit [79]. Clinical evidence also suggests that GLP-1-based treatments have a protective impact on cardiovascular outcomes in diabetics with either STEMI or non-STEMI [83, 84]. A meta-analysis including six RCTs suggested that GLP-1R agonists improved LVEF by 2.46 [95% CI: 0.23–4.70%] and reduced infarct size in both grams and as a percentage of the area at risk [WMD –5.29, 95% CI: –10.39 to –0.19; WMD –0.08, 95% CI: –0.12 to –0.04, respectively] when compared to placebo [85].

7. Cardiovascular Outcomes of Liraglutide and Semaglutide

In the LEADER trial (Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes), liraglutide 1.8 mg (Victoza) reduced the risk of the primary outcome (cardiac death, myocardial infarction, or stroke) by 13% in diabetics with high cardiovascular risk compared to placebo [86]. Based on these significant cardiovascular benefits, in 2017, the European Medicines Agency incorporated the outcomes derived from the LEADER trial into the prescription label for the 3 mg dosage of liraglutide (Saxenda) [87]. Although some studies suggest GLP-1 receptor agonists demonstrate a dose-response relationship in HbA1c reduction [88] and weight loss [89], whether higher doses can further reduce cardiovascular events needs further investigation.

Results from the SUSTAIN 6 trial (Semaglutide Unabated Sustainability in Treatment of Type 2 Diabetes) demonstrated that injectable semaglutide, when compared to placebo, resulted in a 26% relative and a 2.3% absolute reduction in the risk of the 3-points MACE composite endpoint in diabetics with CAD or at high atherosclerotic cardiovascular disease (ASCVD) risk [90]. In the PIONEER 6 trial (Peptide InnOvatioN for Early diabEtes tReatment), treatment with oral semaglutide in diabetics with high cardiovascular risk reduced the occurrence of MACE by 21% and significantly reduced cardiovascular death by 53% compared to placebo [91]. With the FDA approval of Wegovy (semaglutide) injection for chronic weight management in obese or overweight adults in 2021 [92], semaglutide appears to have the trend of becoming one of the most widely used GLP-1 receptor agonists. Another ongoing SOUL (Semaglutide cardiOvascular oUtcomes triaL) trial enrolled 9650 participants, with 70.7% having a history of coronary heart disease, the results of which will provide evidence on the cardiovascular effects of oral semaglutide in patients with T2DM and established ASCVD [93].

8. Conclusion and Future Perspective

A multitude of studies of GLP-1-based interventions have shown beneficial effects on cardiovascular parameters that go well beyond its classical identification as a hypoglycemic drug. Several GLP-1R agonists, such as liraglutide, albiglutide, semaglutide, and dulaglutide, have been recommended to reduce the risk of major adverse cardiovascular events in patients with T2DM and established ASCVD or multiple risk factors for ASCVD [94]. Therefore, larger and longer-term randomized trials with clinical endpoints, such as cardiovascular morbidity and mortality, should be performed to verify the cardioprotective benefit of adjunctive GLP-1-based therapies during all periods of heart attack. However, the translation of experimentally promising results into clinical therapy has proven to be challenging. A new paradigm is required to overcome the barrier of translating pre-clinical findings on GLP-1-based therapies into practical applications.

Three elements are more crucial. First, patients, especially with STEMI, must be able to survive malignant arrhythmias. GLP-1 may provide an anti-arrhythmic effect, including in ischemia/reperfusion-induced ventricular arrhythmias [95]. Secondly, infarct size must be restricted. The best way to treat STEMI is with primary percutaneous coronary intervention to get the blood flow back to the coronary artery as soon as possible. However, this is paradoxically associated with reperfusion injury due to the generation of reactive oxygen metabolites and proinflammatory neutrophil infiltrates, which can exacerbate apoptosis and cell death. There is no standard therapy for myocardial protection following reperfusion in acute myocardial infarction. Even though different ways that GLP-1 protects the heart are biologically plausible and have shown promising results in experiments, they still need to be confirmed in large-scale clinical trials. Thirdly, the prognosis and quality of life of patients with CAD must be improved. A general goal in management includes improved symptoms with hemodynamic stabilization and increased use of evidence-based therapies to reduce recurrent hospitalization and mortality. Due to their inotropic and chronotropic properties [96], GLP-1R agonists might be selected as an add-on therapy to improve myocardial function and remodeling in CAD patients with or without diabetes.

In conclusion, currently, GLP-1 is a promising option with benign cardiovascular safety for the treatment of T2DM and coexistent CAD. Nevertheless, larger and longer-term well-designed randomized control trials of both cardiovascular morbidity and mortality are needed to evaluate GLP-1R agonist as a cardioprotective agent and the population in whom it will work best.

Author Contributions

QX and JW prepared the manuscript and tables. KH helped with references collection. LZ and WL designed manuscript outline and revised manuscript. All authors have read and agreed to the published version of the manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work. All authors contributed to editorial changes in the manuscript.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This study was supported by the Science and Technology Plan Project of Jiangxi Provincial Health Commission (NO. 202312257).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Libby P, Theroux P. Pathophysiology of coronary artery disease. Circulation. 2005; 111: 3481–3488.
[2]
Nesto RW, Rutter MK. Impact of the atherosclerotic process in patients with diabetes. Acta Diabetologica. 2002; 39: S22–S28.
[3]
Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Research and Clinical Practice. 2022; 183: 109119.
[4]
Diabetes Control and Complications Trial Research Group, Nathan DM, Genuth S, Lachin J, Cleary P, Crofford O, et al. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The New England Journal of Medicine. 1993; 329: 977–986.
[5]
ADVANCE Collaborative Group, Patel A, MacMahon S, Chalmers J, Neal B, Billot L, et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. The New England Journal of Medicine. 2008; 358: 2560–2572.
[6]
nullIntensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet. 1998; 352: 837–853.
[7]
Action to Control Cardiovascular Risk in Diabetes Study Group, Gerstein HC, Miller ME, Byington RP, Goff DC, Jr, Bigger JT, et al. Effects of intensive glucose lowering in type 2 diabetes. The New England Journal of Medicine. 2008; 358: 2545–2559.
[8]
Ferro EG, Michos ED, Bhatt DL, Lincoff AM, Elshazly MB. New Decade, New FDA Guidance for Diabetes Drug Development: Lessons Learned and Future Directions. Journal of the American College of Cardiology. 2020; 76: 2522–2526.
[9]
Drucker DJ. Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. Cell Metabolism. 2018; 27: 740–756.
[10]
Cowie MR, Fisher M. SGLT2 inhibitors: mechanisms of cardiovascular benefit beyond glycaemic control. Nature Reviews. Cardiology. 2020; 17: 761–772.
[11]
Deacon CF, Johnsen AH, Holst JJ. Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo. The Journal of Clinical Endocrinology and Metabolism. 1995; 80: 952–957.
[12]
Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007; 132: 2131–2157.
[13]
Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006; 368: 1696–1705.
[14]
American Diabetes Association. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes-2019. Diabetes Care. 2019; 42: S90–S102.
[15]
Wei Y, Mojsov S. Tissue-specific expression of the human receptor for glucagon-like peptide-I: brain, heart and pancreatic forms have the same deduced amino acid sequences. FEBS Letters. 1995; 358: 219–224.
[16]
Drucker DJ. The biology of incretin hormones. Cell Metabolism. 2006; 3: 153–165.
[17]
Pussinen PJ, Tuomisto K, Jousilahti P, Havulinna AS, Sundvall J, Salomaa V. Endotoxemia, immune response to periodontal pathogens, and systemic inflammation associate with incident cardiovascular disease events. Arteriosclerosis, Thrombosis, and Vascular Biology. 2007; 27: 1433–1439.
[18]
Noyan-Ashraf MH, Shikatani EA, Schuiki I, Mukovozov I, Wu J, Li RK, et al. A glucagon-like peptide-1 analog reverses the molecular pathology and cardiac dysfunction of a mouse model of obesity. Circulation. 2013; 127: 74–85.
[19]
Dozier KC, Cureton EL, Kwan RO, Curran B, Sadjadi J, Victorino GP. Glucagon-like peptide-1 protects mesenteric endothelium from injury during inflammation. Peptides. 2009; 30: 1735–1741.
[20]
Shiraki A, Oyama JI, Komoda H, Asaka M, Komatsu A, Sakuma M, et al. The glucagon-like peptide 1 analog liraglutide reduces TNF-α-induced oxidative stress and inflammation in endothelial cells. Atherosclerosis. 2012; 221: 375–382.
[21]
Lappas M. Anti-inflammatory properties of sirtuin 6 in human umbilical vein endothelial cells. Mediators of Inflammation. 2012; 2012: 597514.
[22]
Krasner NM, Ido Y, Ruderman NB, Cacicedo JM. Glucagon-like peptide-1 (GLP-1) analog liraglutide inhibits endothelial cell inflammation through a calcium and AMPK dependent mechanism. PLoS ONE. 2014; 9: e97554.
[23]
Fan SH, Xiong QF, Wang L, Zhang LH, Shi YW. Glucagon-like peptide 1 treatment reverses vascular remodelling by downregulating matrix metalloproteinase 1 expression through inhibition of the ERK1/2/NF-κB signalling pathway. Molecular and Cellular Endocrinology. 2020; 518: 111005.
[24]
Chaudhuri A, Ghanim H, Vora M, Sia CL, Korzeniewski K, Dhindsa S, et al. Exenatide exerts a potent antiinflammatory effect. The Journal of Clinical Endocrinology and Metabolism. 2012; 97: 198–207.
[25]
Burgmaier M, Liberman A, Möllmann J, Kahles F, Reith S, Lebherz C, et al. Glucagon-like peptide-1 (GLP-1) and its split products 9-37) and GLP-1(28-37) stabilize atherosclerotic lesions in apoe⁻/⁻ mice. Atherosclerosis. 2013; 231: 427–435.
[26]
Song X, Jia H, Jiang Y, Wang L, Zhang Y, Mu Y, et al. Anti-atherosclerotic effects of the glucagon-like peptide-1 (GLP-1) based therapies in patients with type 2 Diabetes Mellitus: A meta-analysis. Scientific Reports. 2015; 5: 10202.
[27]
Heusch G. Myocardial Ischemia: Lack of Coronary Blood Flow or Myocardial Oxygen Supply/Demand Imbalance? Circulation Research. 2016; 119: 194–196.
[28]
Ban K, Noyan-Ashraf MH, Hoefer J, Bolz SS, Drucker DJ, 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.
[29]
Chai W, Dong Z, Wang N, Wang W, Tao L, Cao W, et al. Glucagon-like peptide 1 recruits microvasculature and increases glucose use in muscle via a nitric oxide-dependent mechanism. Diabetes. 2012; 61: 888–896.
[30]
Nikolaidis LA, Elahi D, Hentosz T, Doverspike A, Huerbin R, Zourelias L, et al. Recombinant glucagon-like peptide-1 increases myocardial glucose uptake and improves left ventricular performance in conscious dogs with pacing-induced dilated cardiomyopathy. Circulation. 2004; 110: 955–961.
[31]
Dokken BB, Hilwig WR, Teachey MK, Panchal RA, Hubner K, Allen D, et al. Glucagon-like peptide-1 (GLP-1) attenuates post-resuscitation myocardial microcirculatory dysfunction. Resuscitation. 2010; 81: 755–760.
[32]
Dokken BB, Piermarini CV, Teachey MK, Gura MT, Dameff CJ, Heller BD, et al. Glucagon-like peptide-1 preserves coronary microvascular endothelial function after cardiac arrest and resuscitation: potential antioxidant effects. American Journal of Physiology. Heart and Circulatory Physiology. 2013; 304: H538–H546.
[33]
Xiong QF, Fan SH, Li XW, Niu Y, Wang J, Zhang X, et al. GLP-1 Relaxes Rat Coronary Arteries by Enhancing ATP-Sensitive Potassium Channel Currents. Cardiology Research and Practice. 2019; 2019: 1968785.
[34]
Sukumaran V, Tsuchimochi H, Sonobe T, Waddingham MT, Shirai M, Pearson JT. Liraglutide treatment improves the coronary microcirculation in insulin resistant Zucker obese rats on a high salt diet. Cardiovascular Diabetology. 2020; 19: 24.
[35]
Gejl M, Søndergaard HM, Stecher C, Bibby BM, Møller N, Bøtker HE, et al. Exenatide alters myocardial glucose transport and uptake depending on insulin resistance and increases myocardial blood flow in patients with type 2 diabetes. The Journal of Clinical Endocrinology and Metabolism. 2012; 97: E1165–E1169.
[36]
Subaran SC, Sauder MA, Chai W, Jahn LA, Fowler DE, Aylor KW, et al. GLP-1 at physiological concentrations recruits skeletal and cardiac muscle microvasculature in healthy humans. Clinical Science. 2014; 127: 163–170.
[37]
Nielsen R, Jorsal A, Iversen P, Tolbod LP, Bouchelouche K, Sørensen J, et al. Effect of liraglutide on myocardial glucose uptake and blood flow in stable chronic heart failure patients: A double-blind, randomized, placebo-controlled LIVE sub-study. Journal of Nuclear Cardiology. 2019; 26: 585–597.
[38]
Breisch EA, Houser SR, Carey RA, Spann JF, Bove AA. Myocardial blood flow and capillary density in chronic pressure overload of the feline left ventricle. Cardiovascular Research. 1980; 14: 469–475.
[39]
Wang J, Fan S, Xiong Q, Niu Y, Zhang X, Qin J, et al. Glucagon-like peptide-1 attenuates cardiac hypertrophy via the AngII/AT1R/ACE2 and AMPK/mTOR/p70S6K pathways. Acta Biochimica et Biophysica Sinica. 2021; 53: 1189–1197.
[40]
Chalikias G, Tziakas D. Slow Coronary Flow: Pathophysiology, Clinical Implications, and Therapeutic Management. Angiology. 2021; 72: 808–818.
[41]
Doyle ME, Egan JM. Mechanisms of action of glucagon-like peptide 1 in the pancreas. Pharmacology & Therapeutics. 2007; 113: 546–593.
[42]
Bose AK, Mocanu MM, Carr RD, Brand CL, Yellon DM. Glucagon-like peptide 1 can directly protect the heart against ischemia/reperfusion injury. Diabetes. 2005; 54: 146–151.
[43]
Bose AK, Mocanu MM, Carr RD, Yellon DM. Glucagon like peptide-1 is protective against myocardial ischemia/reperfusion injury when given either as a preconditioning mimetic or at reperfusion in an isolated rat heart model. Cardiovascular Drugs and Therapy. 2005; 19: 9–11.
[44]
Bose AK, Mocanu MM, Carr RD, Yellon DM. Myocardial ischaemia-reperfusion injury is attenuated by intact glucagon like peptide-1 (GLP-1) in the in vitro rat heart and may involve the p70s6K pathway. Cardiovascular Drugs and Therapy. 2007; 21: 253–256.
[45]
Huisamen B, Genade S, Lochner A. Signalling pathways activated by glucagon-like peptide-1 (7-36) amide in the rat heart and their role in protection against ischaemia. Cardiovascular Journal of Africa. 2008; 19: 77–83.
[46]
Matsubara M, Kanemoto S, Leshnower BG, Albone EF, Hinmon R, Plappert T, et al. Single dose GLP-1-Tf ameliorates myocardial ischemia/reperfusion injury. The Journal of Surgical Research. 2011; 165: 38–45.
[47]
Noyan-Ashraf MH, Momen MA, Ban K, Sadi AM, Zhou YQ, Riazi AM, et al. GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice. Diabetes. 2009; 58: 975–983.
[48]
Bao W, Aravindhan K, Alsaid H, Chendrimada T, Szapacs M, Citerone DR, et al. Albiglutide, a long lasting glucagon-like peptide-1 analog, protects the rat heart against ischemia/reperfusion injury: evidence for improving cardiac metabolic efficiency. PLoS ONE. 2011; 6: e23570.
[49]
Chang G, Zhang D, Yu H, Zhang P, Wang Y, Zheng A, et al. Cardioprotective effects of exenatide against oxidative stress-induced injury. International Journal of Molecular Medicine. 2013; 32: 1011–1020.
[50]
Wohlfart P, Linz W, Hübschle T, Linz D, Huber J, Hess S, et al. Cardioprotective effects of lixisenatide in rat myocardial ischemia-reperfusion injury studies. Journal of Translational Medicine. 2013; 11: 84.
[51]
Eid RA, Bin-Meferij MM, El-Kott AF, Eleawa SM, Zaki MSA, Al-Shraim M, et al. Exendin-4 Protects Against Myocardial Ischemia-Reperfusion Injury by Upregulation of SIRT1 and SIRT3 and Activation of AMPK. Journal of Cardiovascular Translational Research. 2021; 14: 619–635.
[52]
Sonne DP, Engstrøm T, Treiman M. Protective effects of GLP-1 analogues exendin-4 and GLP-1(9-36) amide against ischemia-reperfusion injury in rat heart. Regulatory Peptides. 2008; 146: 243–249.
[53]
Kavianipour M, Ehlers MR, Malmberg K, Ronquist G, Ryden L, Wikström G, et al. Glucagon-like peptide-1 (7-36) amide prevents the accumulation of pyruvate and lactate in the ischemic and non-ischemic porcine myocardium. Peptides. 2003; 24: 569–578.
[54]
Kristensen J, Mortensen UM, Schmidt M, Nielsen PH, Nielsen TT, Maeng M. Lack of cardioprotection from subcutaneously and preischemic administered liraglutide in a closed chest porcine ischemia reperfusion model. BMC Cardiovascular Disorders. 2009; 9: 31.
[55]
Ekström K, Dalsgaard M, Iversen K, Pedersen-Bjergaard U, Vejlstrup N, Diemar SS, et al. Effects of liraglutide and ischemic postconditioning on myocardial salvage after I/R injury in pigs. Scandinavian Cardiovascular Journal. 2017; 51: 8–14.
[56]
Timmers L, Henriques JPS, de Kleijn DPV, Devries JH, Kemperman H, Steendijk P, et al. Exenatide reduces infarct size and improves cardiac function in a porcine model of ischemia and reperfusion injury. Journal of the American College of Cardiology. 2009; 53: 501–510.
[57]
Siraj MA, Mundil D, Beca S, Momen A, Shikatani EA, Afroze T, et al. Cardioprotective GLP-1 metabolite prevents ischemic cardiac injury by inhibiting mitochondrial trifunctional protein-α. The Journal of Clinical Investigation. 2020; 130: 1392–1404.
[58]
Ban K, Kim KH, Cho CK, Sauvé M, Diamandis EP, Backx PH, et al. Glucagon-like peptide (GLP)-1(9-36)amide-mediated cytoprotection is blocked by exendin(9-39) yet does not require the known GLP-1 receptor. Endocrinology. 2010; 151: 1520–1531.
[59]
Besch G, Perrotti A, Salomon du Mont L, Puyraveau M, Ben-Said X, Baltres M, et al. Impact of intravenous exenatide infusion for perioperative blood glucose control on myocardial ischemia-reperfusion injuries after coronary artery bypass graft surgery: sub study of the phase II/III ExSTRESS randomized trial. Cardiovascular Diabetology. 2018; 17: 140.
[60]
Carvalho G, Pelletier P, Albacker T, Lachapelle K, Joanisse DR, Hatzakorzian R, et al. Cardioprotective effects of glucose and insulin administration while maintaining normoglycemia (GIN therapy) in patients undergoing coronary artery bypass grafting. The Journal of Clinical Endocrinology and Metabolism. 2011; 96: 1469–1477.
[61]
Nakadate Y, Sato H, Oguchi T, Sato T, Kawakami A, Ishiyama T, et al. Glycemia and the cardioprotective effects of insulin pre-conditioning in the isolated rat heart. Cardiovascular Diabetology. 2017; 16: 43.
[62]
Read PA, Hoole SP, White PA, Khan FZ, O’Sullivan M, West NEJ, et al. A pilot study to assess whether glucagon-like peptide-1 protects the heart from ischemic dysfunction and attenuates stunning after coronary balloon occlusion in humans. Circulation. Cardiovascular Interventions. 2011; 4: 266–272.
[63]
McCormick LM, Hoole SP, White PA, Read PA, Axell RG, Clarke SJ, et al. Pre-treatment with glucagon-like Peptide-1 protects against ischemic left ventricular dysfunction and stunning without a detected difference in myocardial substrate utilization. JACC. Cardiovascular Interventions. 2015; 8: 292–301.
[64]
Read PA, Khan FZ, Dutka DP. Cardioprotection against ischaemia induced by dobutamine stress using glucagon-like peptide-1 in patients with coronary artery disease. Heart. 2012; 98: 408–413.
[65]
McCormick LM, Heck PM, Ring LS, Kydd AC, Clarke SJ, Hoole SP, et al. Glucagon-like peptide-1 protects against ischemic left ventricular dysfunction during hyperglycemia in patients with coronary artery disease and type 2 diabetes mellitus. Cardiovascular Diabetology. 2015; 14: 102.
[66]
Kumarathurai P, Anholm C, Nielsen OW, Kristiansen OP, Mølvig J, Madsbad S, et al. Effects of the glucagon-like peptide-1 receptor agonist liraglutide on systolic function in patients with coronary artery disease and type 2 diabetes: a randomized double-blind placebo-controlled crossover study. Cardiovascular Diabetology. 2016; 15: 105.
[67]
Kumarathurai P, Sajadieh A, Anholm C, Kristiansen OP, Haugaard SB, Nielsen OW. Effects of liraglutide on diastolic function parameters in patients with type 2 diabetes and coronary artery disease: a randomized crossover study. Cardiovascular Diabetology. 2021; 20: 12.
[68]
Myat A, Redwood SR, Arri S, Gersh BJ, Bhatt DL, Marber MS. Liraglutide to Improve corONary haemodynamics during Exercise streSS (LIONESS): a double-blind randomised placebo-controlled crossover trial. Diabetology & Metabolic Syndrome. 2021; 13: 17.
[69]
Suhrs HE, Raft KF, Bové K, Madsbad S, Holst JJ, Zander M, et al. Effect of liraglutide on body weight and microvascular function in non-diabetic overweight women with coronary microvascular dysfunction. International Journal of Cardiology. 2019; 283: 28–34.
[70]
Chen WR, Tian F, Chen YD, Wang J, Yang JJ, Wang ZF, et al. Effects of liraglutide on no-reflow in patients with acute ST-segment elevation myocardial infarction. International Journal of Cardiology. 2016; 208: 109–114.
[71]
Lønborg J, Kelbæk H, Vejlstrup N, Bøtker HE, Kim WY, Holmvang L, et al. Exenatide reduces final infarct size in patients with ST-segment-elevation myocardial infarction and short-duration of ischemia. Circulation. Cardiovascular Interventions. 2012; 5: 288–295.
[72]
Lønborg J, Vejlstrup N, Kelbæk H, Bøtker HE, Kim WY, Mathiasen AB, et al. Exenatide reduces reperfusion injury in patients with ST-segment elevation myocardial infarction. European Heart Journal. 2012; 33: 1491–1499.
[73]
Nikolaidis LA, Mankad S, Sokos GG, Miske G, Shah A, Elahi D, et al. Effects of glucagon-like peptide-1 in patients with acute myocardial infarction and left ventricular dysfunction after successful reperfusion. Circulation. 2004; 109: 962–965.
[74]
Chen WR, Chen YD, Tian F, Yang N, Cheng LQ, Hu SY, et al. Effects of Liraglutide on Reperfusion Injury in Patients With ST-Segment-Elevation Myocardial Infarction. Circulation. Cardiovascular Imaging. 2016; 9: e005146.
[75]
Chen WR, Hu SY, Chen YD, Zhang Y, Qian G, Wang J, et al. Effects of liraglutide on left ventricular function in patients with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention. American Heart Journal. 2015; 170: 845–854.
[76]
Chen WR, Shen XQ, Zhang Y, Chen YD, Hu SY, Qian G, et al. Effects of liraglutide on left ventricular function in patients with non-ST-segment elevation myocardial infarction. Endocrine. 2016; 52: 516–526.
[77]
Woo JS, Kim W, Ha SJ, Kim JB, Kim SJ, Kim WS, et al. Cardioprotective effects of exenatide in patients with ST-segment-elevation myocardial infarction undergoing primary percutaneous coronary intervention: results of exenatide myocardial protection in revascularization study. Arteriosclerosis, Thrombosis, and Vascular Biology. 2013; 33: 2252–2260.
[78]
Roos ST, Timmers L, Biesbroek PS, Nijveldt R, Kamp O, van Rossum AC, et al. No benefit of additional treatment with exenatide in patients with an acute myocardial infarction. International Journal of Cardiology. 2016; 220: 809–814.
[79]
Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Køber LV, et al. Lixisenatide in Patients with Type 2 Diabetes and Acute Coronary Syndrome. The New England Journal of Medicine. 2015; 373: 2247–2257.
[80]
Myat A, Arri S, Bhatt DL, Gersh BJ, Redwood SR, Marber MS. Design and rationale for the randomised, double-blinded, placebo-controlled Liraglutide to Improve corONary haemodynamics during Exercise streSS (LIONESS) crossover study. Cardiovascular Diabetology. 2015; 14: 27.
[81]
Kalogeris T, Baines CP, Krenz M, Korthuis RJ. Cell biology of ischemia/reperfusion injury. International Review of Cell and Molecular Biology. 2012; 298: 229–317.
[82]
Bentley-Lewis R, Aguilar D, Riddle MC, Claggett B, Diaz R, Dickstein K, et al. Rationale, design, and baseline characteristics in Evaluation of LIXisenatide in Acute Coronary Syndrome, a long-term cardiovascular end point trial of lixisenatide versus placebo. American Heart Journal. 2015; 169: 631–638.e7.
[83]
Marfella R, Sardu C, Balestrieri ML, Siniscalchi M, Minicucci F, Signoriello G, et al. Effects of incretin treatment on cardiovascular outcomes in diabetic STEMI-patients with culprit obstructive and multivessel non obstructive-coronary-stenosis. Diabetology & Metabolic Syndrome. 2018; 10: 1.
[84]
Marfella R, Sardu C, Calabrò P, Siniscalchi M, Minicucci F, Signoriello G, et al. Non-ST-elevation myocardial infarction outcomes in patients with type 2 diabetes with non-obstructive coronary artery stenosis: Effects of incretin treatment. Diabetes, Obesity & Metabolism. 2018; 20: 723–729.
[85]
Huang M, Wei R, Wang Y, Su T, Li Q, Yang X, et al. Protective effect of glucagon-like peptide-1 agents on reperfusion injury for acute myocardial infarction: a meta-analysis of randomized controlled trials. Annals of Medicine. 2017; 49: 552–561.
[86]
Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JFE, Nauck MA, et al. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. The New England Journal of Medicine. 2016; 375: 311–322.
[87]
Christensen RM, Juhl CR, Torekov SS. Benefit-Risk Assessment of Obesity Drugs: Focus on Glucagon-like Peptide-1 Receptor Agonists. Drug Safety. 2019; 42: 957–971.
[88]
Yamada Y, Katagiri H, Hamamoto Y, Deenadayalan S, Navarria A, Nishijima K, et al. Dose-response, efficacy, and safety of oral semaglutide monotherapy in Japanese patients with type 2 diabetes (PIONEER 9): a 52-week, phase 2/3a, randomised, controlled trial. The Lancet. Diabetes & Endocrinology. 2020; 8: 377–391.
[89]
Wilding JPH, Overgaard RV, Jacobsen LV, Jensen CB, le Roux CW. Exposure-response analyses of liraglutide 3.0 mg for weight management. Diabetes, Obesity & Metabolism. 2016; 18: 491–499.
[90]
Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, et al. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. The New England Journal of Medicine. 2016; 375: 1834–1844.
[91]
Husain M, Birkenfeld AL, Donsmark M, Dungan K, Eliaschewitz FG, Franco DR, et al. Oral Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. The New England Journal of Medicine. 2019; 381: 841–851.
[92]
Singh G, Krauthamer M, Bjalme-Evans M. Wegovy (semaglutide): a new weight loss drug for chronic weight management. Journal of Investigative Medicine: the Official Publication of the American Federation for Clinical Research. 2022; 70: 5–13.
[93]
McGuire DK, Busui RP, Deanfield J, Inzucchi SE, Mann JFE, Marx N, et al. Effects of oral semaglutide on cardiovascular outcomes in individuals with type 2 diabetes and established atherosclerotic cardiovascular disease and/or chronic kidney disease: Design and baseline characteristics of SOUL, a randomized trial. Diabetes, Obesity & Metabolism. 2023; 25: 1932–1941.
[94]
American Diabetes Association. 10. Cardiovascular Disease and Risk Management: Standards of Medical Care in Diabetes-2020. Diabetes Care. 2020; 43: S111–S134.
[95]
Hu X, Yang X, Jiang H. Glucagon-like peptide-1 and related agents: novel anti-arrhythmic agents during myocardial ischemia and reperfusion. International Journal of Cardiology. 2013; 168: 3119–3120.
[96]
Wallner M, Kolesnik E, Ablasser K, Khafaga M, Wakula P, Ljubojevic S, et al. Exenatide exerts a PKA-dependent positive inotropic effect in human atrial myocardium: GLP-1R mediated effects in human myocardium. Journal of Molecular and Cellular Cardiology. 2015; 89: 365–375.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top