Next Article in Journal
Pleomorphic Adenoma of the Parotid Gland and the Parapharyngeal Space: Two Diametrically Opposing Surgical Philosophies for the Same Histopathologic Entity?
Next Article in Special Issue
Vascular Alterations Preceding Arterial Wall Thickening in Overweight and Obese Children
Previous Article in Journal
Effectiveness of S-O Clip-Assisted Colorectal Endoscopic Submucosal Dissection
Previous Article in Special Issue
Recent Advances on the Role and Therapeutic Potential of Regulatory T Cells in Atherosclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of SGLT2 Inhibitors on Atherosclerosis: Lessons from Cardiovascular Clinical Outcomes in Type 2 Diabetic Patients and Basic Researches

1
Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
2
Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
3
Omi Medical Center, Omi General Hospital, Kusatsu, Shiga 525-8585, Japan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2022, 11(1), 137; https://doi.org/10.3390/jcm11010137
Submission received: 1 December 2021 / Revised: 21 December 2021 / Accepted: 24 December 2021 / Published: 27 December 2021
(This article belongs to the Special Issue Current Challenges and Advances in Atherosclerosis)

Abstract

:
Atherosclerosis-caused cardiovascular diseases (CVD) are the leading cause of mortality in type 2 diabetes mellitus (T2DM). Sodium-glucose cotransporter 2 (SGLT2) inhibitors are effective oral drugs for the treatment of T2DM patients. Multiple pre-clinical and clinical studies have indicated that SGLT2 inhibitors not only reduce blood glucose but also confer benefits with regard to body weight, insulin resistance, lipid profiles and blood pressure. Recently, some cardiovascular outcome trials have demonstrated the safety and cardiovascular benefits of SGLT2 inhibitors beyond glycemic control. The SGLT2 inhibitors empagliflozin, canagliflozin, dapagliflozin and ertugliflozin reduce the rates of major adverse cardiovascular events and of hospitalization for heart failure in T2DM patients regardless of CVD. The potential mechanisms of SGLT2 inhibitors on cardioprotection may be involved in improving the function of vascular endothelial cells, suppressing oxidative stress, inhibiting inflammation and regulating autophagy, which further protect from the progression of atherosclerosis. Here, we summarized the pre-clinical and clinical evidence of SGLT2 inhibitors on cardioprotection and discussed the potential molecular mechanisms of SGLT2 inhibitors in preventing the pathogenesis of atherosclerosis and CVD.

1. Introduction

Atherosclerosis is a chronic progressive disease characterized by the accumulation and deposition of lipids and fibrous elements in large arteries [1]. Generally, the formation of atherosclerotic plaques is divided into four stages: fatty streaks, atheromatous plaques, complicated atheromatous plaques and clinical complications [2]. Plaque rupture and thromboembolism may lead to severe cardiovascular diseases (CVD), including acute coronary syndrome (ACS), myocardial infarction (MI) or stroke [1,3]. CVD caused by atherosclerosis is the main cause of mortality in metabolic-related diseases, especially in type 2 diabetes mellitus (T2DM) [4,5,6]. Correspondingly, compared with non-diabetic individuals, T2DM patients have a higher risk of atherosclerosis and CVD [7,8]. Over the last decade, CVD globally affected approximately 32.2% of T2DM patients, accounting for 9.9% of deaths among them [9]. Therefore, in recent years, clinical trials of anti-diabetic agents have not only focused on hypoglycemic benefits but on cardiovascular safety and cardioprotective effects as well.
Sodium-glucose cotransporter 2 (SGLT2) is mainly localized in the proximal tubule, which is responsible for reabsorbing 80–90% of filtered glucose under physiological conditions [10,11]. The direct effect of SGLT2 inhibitors is to block glucose transportation in the kidney, which may depend on two distinct mechanisms: (1) by augmenting renal glucose excretion and (2) by ameliorating glucotoxicity [10]. Multiple clinical trials have demonstrated that SGLT2 inhibitors reduce blood glucose by inhibiting the reabsorption of filtered glucose in the proximal tubules independent of insulin. SGLT2 inhibitors slow the progression of macroalbuminuria, reduce the doubling of the serum creatinine level, sustain 40% reduction in the estimated glomerular filtration rate (eGFR) and further protect from inevitable renal replacement therapy or death from renal disease [12,13,14,15,16]. These renoprotective benefits of SGLT2 inhibitors may be attributed to glycemic control, reducing body weight, lipid profiles, blood pressure and uric acid and improving insulin resistance. Moreover, due to the improvement of these cardiovascular risk factors, SGLT2 inhibitors also present cardioprotective effects [10,11,12,13,14,15,16,17,18,19].
In this review, we summarized the clinical evidence of SGLT2 inhibitors on cardioprotection and discussed the potential molecular mechanisms of SGLT2 inhibitors in preventing the pathogenesis of atherosclerosis and CVD.

2. Pathophysiological and Pharmacologic Roles of SGLT2 Inhibition

The kidney is a crucial organ for maintaining glucose metabolism. In healthy individuals, the human kidney filters more than 180 g of glucose per day. SGLT2 is a 75 kilodalton (kDa) protein coded by the solute carrier family 5 (SLC5A2) gene in humans and is mainly localized in the kidney proximal tubule [20]. A total of 80–90% of filtered glucose is reabsorbed by SGLT2 in the early proximal tubule, while the remaining 10–20% is absorbed by SGLT1. The energy for the transport capacity of SGLT2 and SGLT1 is derived from the Na+/K+ ATPase pump located in the basolateral membrane of the proximal tubule. Therefore, the reabsorption of filtered glucose is also accompanied by Na+ reabsorption [10,18,21]. SGLT2 knockout mice had glucosuria and polyuria compared with wildtype mice [22]. In patients with T2DM, the reabsorption of filtered glucose in the proximal tubules is significantly increased, contributing to elevated SGLT2 expression [23,24]. Meanwhile, the elevated reabsorption of Na+ activates the local renin-angiotensin system, stimulating the constriction of the adjacent efferent arteriole and dilation of the afferent arteriole, which leads to increases in intraglomerular pressure and glomerular filtration rate (GFR), which ultimately results in glomerular damage [10]. This evidence indicates that the inhibition of SGLT2 is a potential target for glycemic control and conferring renoprotection in treating T2DM.
The first natural SGLT inhibitor, called phlorizin, is derived from fruit trees. Phlorizin functions as a competitive inhibitor of SGLT1/2 to block glucose absorption in the proximal renal tubule and mucosa of the small intestine [25,26]. However, due to poor solubility in water, poor oral bioavailability, the non-selective inhibition of both SGLT1 and SGLT2 and a short half-life, effective studies on the mechanisms of specific SGLT2 inhibition by phlorizin are limited [25,27]. Subsequently, more novel selective SGLT2 inhibitors have been identified. Compared with phlorizin, these SGLT2 inhibitors are structurally improved. Dapagliflozin contains C-glucoside, while canagliflozin and empagliflozin contain C-glycosylated diarylmethane pharmacophore. These structures are resistant to hydrolysis by β-glucosidases to increase their half-life and specificity to SGLT2 [27,28]. To date, empagliflozin, canagliflozin, dapagliflozin and ertugliflozin have been approved by the United States Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for the treatment of T2DM [26]. Some other novel SGLT2 inhibitors, such as ipragliflozin and luseogliflozin, have been approved in Japan and other countries [29,30].

3. Mechanisms of SGLT2 Inhibitors against Atherosclerosis

During the formation of atherosclerotic plaques, three types of cells, endothelial cells (ECs), vascular smooth muscle cells (VSMCs) and monocytes/macrophages, play crucial roles. At the first stage, due to endothelial dysfunction, excessive lipids and lipoproteins accumulate in the subendothelial matrix. This process is triggered by the accumulation of oxidized low-density lipoprotein (Ox-LDL), which stimulates the release of pro-inflammatory cytokines, including interleukin 8 (IL-8) and adhesion molecules such as intercellular cell adhesion molecule-1 (ICAM-1), Vascular cell adhesion protein 1 (VCAM-1), monocyte chemoattractant protein 1 (MCP-1) and macrophage colony-stimulating factor (M-CSF). Then, circulating monocytes migrate to the intima, where they proliferate, differentiate into macrophages and combine with lipoproteins to form foam cells. Monocytes/macrophages express pro-inflammatory cytokines, including IL-1 and tumor necrosis factor α (TNFα). VSMCs migrating from the medial layer can also secret pro-fibrosis molecules. With the continuous migration of monocytes into the intima and the accumulation of lipids and lipoproteins, foam cells and SMCs die to form necrotic cores. Platelets will recruit to form thrombi and eventually develop atherosclerotic plaques. Vascular occlusion or plaque rupture may result in acute cardiovascular and cerebrovascular events [1,3,31].
Some metabolic characteristics, including obesity, dyslipidemia, hyperglycemia and hypertension, are independent risk factors for atherosclerosis [32]. These metabolic disorders, which may lead to endothelial dysfunction, oxidative stress, inflammation and the impairment of autophagy, participate in the formation of plaques and the pathogenesis of atherosclerosis [33]. A series of basic studies have confirmed that SGLT2 inhibitors have cardioprotective effects beyond glucose control in animal models of atherosclerosis [34,35,36,37,38]. The underlying mechanisms of SGLT2 inhibitors may be related to protecting endothelial function, anti-oxidative stress, anti-inflammation and maintaining basal and adaptive autophagy (Figure 1).

3.1. Maintaining Endothelial Function

Endogenous nitric oxide (NO) is a vasodilatory molecule produced by NO synthase (NOS), which presents antiatherosclerotic effects [39]. SGLT2 inhibitors inhibit cardiac Na+/K+ exchange to induce vasodilation [40]. Previous studies have shown that SGLT2 inhibitors can participate in the regulation of vasodilation by increasing the anabolism and bioavailability of NO. Both empagliflozin and canagliflozin ameliorated aortic stiffness and improved vasodilation via endothelial NOS (eNOS) phosphorylation in db/db mice and non-diabetic rats [41,42]. Empagliflozin and dapagliflozin restored NO bioavailability by suppressing reactive oxygen species (ROS) generation in TNFα-induced ECs [43].

3.2. Anti-Oxidative Stress

An imbalance between the production and scavenging of ROS results in oxidative stress, which plays crucial roles in the progression of atherosclerosis [44]. Multiple risk factors of atherosclerosis, such as high glucose, elevated free fatty acids (FFA) and triglyceride (TG), can increase ROS production via activating nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX1-5, p22phox, p47phox), inhibiting NOS and suppressing glyceraldehyde 3-phosphate dehydrogenase (GAPDH) activity, and then by further amplifying oxidative stress in macrophages, ECs and adipocytes and accelerating the formation of atherosclerotic plaques [45,46,47,48].
Both in vitro and in vivo experiments have confirmed the anti-oxidative stress effects of SGLT2 inhibitors. Angiotensin II can induce SGLT2 expression in ECs, which leads to the activation of oxidative stress due to NADPH oxidase. Empagliflozin presented anti-oxidant effects to protect against endothelial senescence and dysfunction via suppressing the NADPH oxidase/SGLT2 pathway [49]. Canagliflozin reduced the expression of NADPH oxidase, including NOX2, NOX4, p22phox and p47phox, and reduced the urinary excretion of 8-hydroxy-2′ -deoxyguanosine (8-OHdG) in diabetic Apolipoprotein E-deficient (ApoE−/−) mice [50]. Both empagliflozin and dapagliflozin inhibited the production of ROS, ameliorated TNFα-induced oxidative stress and restored impaired NO bioavailability in human umbilical vein endothelial cells (HUVECs) and human coronary arterial endothelial cells (HCAECs) [43]. SGLT2 inhibitors, including canagliflozin, dapagliflozin and empagliflozin, protected from high-glucose-induced vasodilation disorders via suppressing NAPDH oxidase/ROS signaling in cultured ECs [51].

3.3. Anti-Inflammation

Inflammatory cytokines and inflammatory cascade reactions throughout participate in the formation, progression and rupture of atherosclerotic plaque [1,31]. Previous studies have revealed that SGLT2 inhibitors, including dapagliflozin, empagliflozin, canagliflozin and luseogliflozin, reduced a series of pro-inflammatory cytokines, including IL-1β, IL-18 [34], IL-6 and TNFα [35], and adhesion molecules such as MCP-1 [35,38,52,53], ICAM-1 [36,50] and VCAM-1 [50,52,53] in atherosclerosis animal models with or without diabetes. In vitro studies have explored the potential mechanisms of the anti-inflammatory effects of SGLT2 inhibitors. Dapagliflozin may inhibit high glucose-induced TNFα, MCP-1 and VCAM-1 via suppressing the nuclear factor kappa-B (NF-κB) pathway in human vascular endothelial cells [54]. Canagliflozin suppressed IL-1β-activated cytokine and chemokine, such as IL-6 and MCP-1, partly via AMP-activated protein kinase (AMPK) activation [55].
In addition to inflammatory pathways, inflammasome-mediated inflammatory pathways are also involved in the pathogenesis of atherosclerosis [56,57,58]. Ox-LDL and high glucose are responsible for the activation of the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome [56,59]. NLRP3 is essential for activating the precursors of IL-1β and IL-18 into their mature forms, which recruit in vascular endothelial cells, leading to atherothrombosis [58]. NF-κB promotes the transcription of NLRP3 and participates in cardiac inflammation [60], which links inflammasome and the inflammatory pathway. Moreover, TG and very low-density lipoprotein (VLDL)-related arterial inflammation are closely related to the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 1 (NLRP1) inflammasome activation in ECs [61]. Dapagliflozin inhibited IL-1β expression via NLRP3/caspase 1 signaling in streptozotocin (STZ)-induced diabetic ApoE−/− mice and T2DM rodent models [34,62,63]. Empagliflozin suppressed IL-17A-induced IL-1β and IL-18 secretions via NLRP3/caspase1 signaling and further inhibited the cell proliferation and migration of human aortic SMCs [64] and decreased the expression of IL-1β via suppressing NF-κB phosphorylation/NLRP3 signaling in human macrophages [62].

3.4. Regulation of Autophagy

Autophagy plays a complex role in the pathogenesis of atherosclerosis. Basal and mild adaptive autophagy to stress can maintain the endothelial functions of ECs, VSMCs and macrophages and protect against the formation of atherosclerotic plaques, while both deficient and excessive autophagy are related to inflammation, oxidative stress and apoptosis, which may contribute to autophagy-dependent cell death, aggravate vascular injury and lead to plaque instability or rupture [33,65,66]. Therefore, the precise regulation of autophagy is essential to prevent the development of atherosclerosis.
The SGLT2 inhibitors empagliflozin and dapagliflozin have been identified to restore autophagy deficiency in diabetic or obese rodent models, which mainly depends on the activation of nutrient-sensing pathways, such as the AMPK/mTOR (mechanistic target of rapamycin) signaling pathway [67,68]. In vitro studies have also identified several potential mechanisms. Empagliflozin restored autophagic flux impairment via activating AMPK and suppressing mTOR in H9c2 cells (rat cardiac myoblast) [69], RAW264.7 and THP-1 cells (macrophage cell lines) [70]. Our previous study also indicated that dapagliflozin had similar effects on autophagy restoration via AMPK/mTOR signaling in high-glucose-treated proximal tubular cells [71].

4. Improvements of Indicators Related to CVD by SGLT2 Inhibitor

Disorders of metabolic-related characteristics, including glucose, lipid profiles, blood pressure, uric acid, etc., contribute to increased risks of developing atherosclerosis and CVD [33]. Beyond the benefits from glycemic control, multiple clinical and rodent research studies have demonstrated that the cardiovascular benefits of SGLT2 inhibitors are closely related to the improvement of these metabolic-related characteristics.

4.1. Pre-Clinical Evidence

ApoE−/− mice and low density lipoprotein receptor knockout (Ldlr−/−) mice are well-established and extensively used rodent models of atherosclerosis [72]. Multiple studies have demonstrated that SGLT2 inhibitors, including dapagliflozin, empagliflozin, canagliflozin, luseogliflzin and ipragliflozin, reduce metabolic indicators to protect from the progression of atherosclerosis (Table 1). Dapagliflozin reduced fasting blood glucose (FBG), total cholesterol (TC) and TG [34], body weight, and glycosylated hemoglobin (HbA1c) [73] in STZ-induced diabetic ApoE−/− mice and STZ-induced diabetic Ldlr−/− mice [37]. Empagliflozin ameliorated FBG, TC, heart rate, blood pressure (BP) [53], TG, LDL [74], urinary microalbumin, body weight and fat mass [35,74] in high fat diet (HFD)-fed ApoE−/− mice and reduced body weight and TG in STZ-induced diabetic ApoE−/− mice [38]. Canagliflozin decreased glucose, TC, TG, LDL and heart rate in HFD-fed diabetic ApoE−/− mice [52] and reduced TC and glucose in STZ-induced diabetic ApoE−/− mice [50]. Luseogliflozin reduced body weight, TC, TG, BP and glucose in HFD-fed mice [75] and nicotinamide in STZ-induced diabetic ApoE−/− mice [36]. Ipragliflozin decreased body weight and glucose to inhibit vascular remodeling in HFD-fed mice [76]. All these metabolic changes ameliorate vascular remodeling, reduce plaque size and increase plaque stability to protect from the progression of atherosclerosis.
In addition to these risk factors of CVD, T2DM patients treated with SGLT2 inhibitors showed elevated plasma ketone levels [77]. SGLT2 inhibitors reduce plasma glucose concentration by increasing renal glucose excretion. To meet the energy requirements of cellules, lipid oxidation is activated, leading to the increasing of acetyl coenzyme A (CoA). Acetyl CoA can enter the Krebs cycle to be converted to ketones (acetoacetate and β-hydroxybutyrate). Moreover, enhanced lipolysis in the adipocyte leads to the increase of plasma FFA, which can also be converted to acetyl CoA via β oxidation and subsequently to ketones in the liver [10]. The elevated ketones may improve the energy metabolism of the heart. Previous studies indicated that β-hydroxybutyrateas is a strong anti-inflammatory factor. Empagliflozin can significantly increase serum β-hydroxybutyrateas to inhibit NLRP3 inflammasome and reduce the expression of IL-1β levels, which benefit from CVD [62] (Figure 2).

4.2. Clinical Evidence

SGLT2 inhibitors not only reduce high glucose independent of insulin but also improve systolic blood pressure (SBP), body weight, lipid profile and uric acid, which are considered high risk factors for CVD [10] (Figure 2). The improvement of these metabolic indicators may play a crucial role in protecting against the pathogenesis of atherosclerosis and CVD. Numerous clinical trials have confirmed that SGLT2 inhibitors can effectively reduce body weight, which may be related to increases in fat utilization, reductions in adipose tissue mass and browning in white adipose tissue, further attenuating obesity-induced insulin resistance [78,79,80,81,82]. Treatment with empagliflozin [83,84,85], canagliflozin [82,86,87,88] and ertugliflozin [89] reduced SBP in patients with T2DM and hypertension or patients with T2DM and chronic kidney disease (CKD). This benefit may be related to minimal natriuresis and urinary glucose excretion, which lead to weight loss, osmotic diuresis, reduced plasma volume and arterial stiffness [90]. SGLT2 inhibitors also lead to a small decrease in plasma TG, increases in high-density lipoprotein cholesterol (HDL-C) and a small increase in LDL [82,91,92,93]. A potential mechanism of increased LDL and decreased TG with SGLT2 inhibition may be related to the reduction of LDL clearance, the greater lipolysis of triglyceride-rich lipoproteins [94] and the effect of switching energy metabolism from carbohydrate to lipid utilization [95]. Elevated circulating uric acid is associated with the risk of hypertension and CVD. Multiple studies have demonstrated that SGLT2 inhibitors can reduce circulating uric acid via enhancing urinary uric acid excretion in association with increased urinary glucose [96,97,98], which may benefit patients with CVD.

5. Clinical Evidence of SGLT2 Inhibitors against CVD

Since 2008, the FDA has required that cardiovascular outcome trials (CVOTs) be carried out for new anti-diabetic drugs due to the significance of cardioprotection on T2DM patients. All potential drugs should exclude the major adverse cardiovascular events (MACE) defined in the FDA guidance, including non-fatal myocardial infarction, non-fatal stroke and cardiovascular death [99,100]. Furthermore, the effect of SGLT2 inhibitors on cardiovascular outcomes, including hospitalization for heart failure, death and CVD, compared to other glucose-lowering drugs, was also verified in a real clinical practice [101,102].

5.1. Randomized Controlled Trials (RCTs)

A meta-analysis that summarized 6 regulatory submissions and 57 published trials revealed that seven different SGLT2 inhibitors reduced the risk of Major Adverse Cardiovascular Events (MACEs) and death from any cause [103]. In this section, we summarized some landmark CVOTs of SGLT2 inhibitors (Table 2).
The Empagliflozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients–Removing Excess Glucose trial (EMPA-REG OUTCOME) was the first CVOT to determine the cardiovascular benefits of SGLT2 inhibitors. This trial enrolled T2DM patients, who were treated with either empagliflozin or placebo. The primary composite outcome was MACEs. Compared with the placebo group, the empagliflozin group showed a significant reduction in death from MACEs, hospitalization for heart failure and death from any cause. However, there were no significant benefits for MI and stroke [104]. Subsequently, two trials demonstrated cardiovascular benefits in patients with heart failure and a reduced ejection fraction. The Empagliflozin Outcome Trial in Patients with Chronic Heart Failure with Reduced Ejection Fraction (EMPEROR-Reduced) enrolled 3730 patients (50% diagnosed as diabetes) with New York Heart Association (NYHA) class II to IV heart failure or with an ejection fraction (EF) ≤40%. Compared with the placebo group, the empagliflozin group had a lower risk of any inpatient or outpatient worsening heart failure event regardless of T2DM [105,106]. The Empagliflozin Outcome Trial in Patients with Chronic Heart Failure with Preserved Ejection Fraction (EMPEROR-Preserved) trial also identified the benefits of empagliflozin on heart failure in patients with NYHA class II to IV and an EF ≥40% [107].
The cardiovascular benefits of canagliflozin were demonstrated by the Canagliflozin Cardiovascular Assessment Study (CANVAS) and Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation (CREDENCE). CANVAS involved patients with T2DM, who were treated with canagliflozin or placebo. The primary outcome was also MACEs. Similar to empagliflozin, canagliflozin showed a significant reduction in MACEs, hospitalization for heart failure and death from any cause [19]. CREDENCE confirmed the cardio and renal benefits of canagliflozin in patients with T2DM and chronic kidney disease (CKD). The primary outcome was a composite of end-stage kidney disease or death from renal or cardiovascular disease. Patients in the canagliflozin group had a lower risk of cardiovascular death, MI, stroke and hospitalization for heart failure [14].
The Dapagliflozin Effect on Cardiovascular Events–Thrombolysis in Myocardial Infarction 58 (DECLARE–TIMI 58) evaluated the cardiovascular benefits of dapagliflozin on T2DM patients. The primary outcome was MACEs. Although dapagliflozin was not found to reduce the risk of MACEs, it reduced the risk of hospitalization for heart failure [108]. Another trial, named the Dapagliflozin and Prevention of Adverse Outcomes in Heart Failure (DAPA-HF) trial, confirmed this benefit. DAPA-HF enrolled patients with heart failure and reduced ejection fraction. The primary outcome was a composite of worsening heart failure or death from cardiovascular causes. This trial also demonstrated that dapagliflozin reduced the risk of heart failure hospitalization and cardiovascular death regardless of diabetes [109].
Ertugliflozin has also completed the assessment of cardiovascular events recently. The Evaluation of Ertugliflozin Efficacy and Safety Cardiovascular Outcomes Trial (VERTIS CV) enrolled patients with T2DM and established CVD. The primary outcome was MACEs. Similar to dapagliflozin, ertugliflozin was not found to reduce the risk of MACEs, but it did reduce the risk of hospitalization for heart failure [110].
In addition to the above SGLT2 inhibitors approved by the FDA and EMA for cardiovascular indications, another SGLT2 inhibitor, sotaliflozin, also present benefits for cardiovascular death and heart failure. The Effect of Sotagliflozin on Cardiovascular Events in Patients with Type 2 Diabetes Post Worsening Heart Failure (SOLOIST-WHF) and the Effect of Sotagliflozin on Cardiovascular and Renal Events in Patients with Type 2 Diabetes and Moderate Renal Impairment Who Are at Cardiovascular Risk (SCORED) trial demonstrated that, compared with placebo, sotaliflozin treatment significantly reduced the total number of deaths from cardiovascular causes and hospitalizations and urgent visits for heart failure in T2DM patients with recent worsening heart failure [111] and T2DM patients with chronic kidney disease regardless of albuminuria [112].
According to these hallmark CVOTs, the effects of SGLT2 inhibitors on atherosclerotic cardiovascular events, such as MI and strokes, are less impressive than the effects on heart failure (Table 2). A possible reason is the heterogeneity in the CVD risk of the study populations. Besides, SGLT2 inhibitors may primarily target on ameliorating cardiac structure and function (“the pump”) and not on the “pipes” (coronary arteries) [113,114]. Patients with T2DM and cardiac hypertrophy, diastolic or systolic dysfunction or with a hypervolemic state (regardless of cardiac or renal origin) are the best candidates for treatment with SGLT2 inhibitors [114].

5.2. Multinational Observational Cohort Study

The effects of drugs may be different between the RCTs and real-world practice [115,116]. Because the characteristics of patients in the RCTs do not necessarily match the standard population with T2DM, these results are difficult to be generalized.
The Comparative Effectiveness of Cardiovascular Outcomes in New Users of SGLT2 inhibitors (CVD-REAL) Study is a multinational (USA, Sweden, Norway and Denmark) observational study. In this study, 309,056 patients newly initiated on either SGLT2 inhibitors, including empagliflozin, canagliflozin and dapagliflozin, or other glucose lowering drugs were analyzed on the risk for hospitalization for heart failure and death in patients with T2DM, using clinical data in real-world practice after propensity score matching [101]. There were 961 hospitalizations for heart failure cases during 190,164 person-years follow-up in 6 countries, and, of 215,622 patients in the United States, Norway, Denmark, Sweden and the United Kingdom, death occurred in 1334. The use of SGLT2 inhibitors was significantly associated with lower rates of hospitalization for heart failure and death, with no significant heterogeneity by country, compared to other glucose-lowering drugs (Table 3). In addition, the sub-analysis of the CVD-REAL study exhibited an association between the initiation of SGLT2 inhibitors versus other glucose-lowering drugs and the rates of MI and stroke. Overall, 205,160 patients were included, and, in the intent-to-treat analysis, over 188,551 and 188,678 person-years of follow-up (MI and stroke, respectively), there were 1077 MI and 968 stroke events. The initiation of SGLT2 inhibitors was associated with a modestly lower risk of MI and stroke [102] (Table 3).
The CVD-REAL2 study is a similar study design to the CVD-REAL and was conducted in six countries: South Korea, Japan, Singapore, Israel, Australia and Canada [117]. After propensity score matching, the risks for death, hospitalization for heart failure, MI and stroke were analyzed in 235,064 patients of each group, newly initiated on either SGLT2 inhibitors (dapagliflozin, empagliflozin, ipragliflozin, canagliflozin, tofogliflozin and luseogliflozin) or other glucose-lowering drugs. In total, 74% of patients had no history of CVD, and patient characteristics were well-balanced in both groups. The initiation of SGLT2 inhibitors significantly reduced the risk for death, hospitalization for heart failure, MI and stroke [117] (Table 3).
In addition, a sub-analysis of the CVD-REAL-2 study showed that the initiation of SGLT2 inhibitors was associated with substantially lower risks of hospitalization for heart failure (HR 0.69, 95% CI 0.61–0.77; p < 0.0001), all-cause death (0.59, 0.52–0.67; p < 0.0001) and the composite of hospitalization for heart failure or all-cause death (0.64, 0.57–0.72; p < 0.0001) compared to dipeptidyl peptide-4 (DPP-4)-4 inhibitors [118]. Furthermore, in another retrospective cohort study, each of the 11,431 T2DM patients with peripheral artery disease (PAD) taking the SGLT2 inhibitor or DPP-4 inhibitor were analyzed for the risk for ischemic stroke and acute MI after propensity score matching. The use of the SGLT2 inhibitor had comparable risks of ischemic stroke and acute MI. However, the SGLT2 inhibitor group showed lower risks of congestive heart failure (HR: 0.66; 95% CI 0.49–0.89; p = 0.0062), lower limb ischemia requiring revascularization (HR: 0.73; 95% CI 0.54–0.98; p = 0.0367) or amputation (HR: 0.43; 95% CI 0.30–0.62; p < 0.0001) and cardiovascular death (HR: 0.67; 95% CI 0.49–0.90; p = 0.0089) compared to those in the DPP-4 inhibitor group. [119].

6. Perspectives and Conclusions

In recent years, as effective anti-diabetic agents, SGLT2 inhibitors have not only achieved significant results in glycemic control via increasing urinary glucose excretion independent of insulin but have also presented cardiovascular protective effects, especially in reducing the risk of MACEs and hospitalization from heart failure. Although data from the CVOTs indicated less impressive results on atherosclerotic cardiovascular events such as MI and stroke than on heart failure, some real-world practice indicated the benefits on them (Table 2). This may be related to the significant heterogeneity in the CVD risk of the study populations [113]. Clinical studies have confirmed the cardiovascular safety of SGLT2 inhibitors [19,104,108,110]. Although some common adverse events, including polyuria, genital mycotic infections, urinary tract infections and ketoacidosis, need to be carefully monitored for [19,104,108], they will not cause severe or fatal consequences. Besides, although patients need to be alerted to some other risks, such as amputation and fractures related to canagliflozin [19], there have not been significantly higher incidences than of other anti-diabetic drugs, such as glucagon-like peptide 1 receptor agonists (GLP-1RA) and DPP-4 inhibitors [120]. Based on the safety, efficiency and cardiovascular benefits, pharmacologic recommendations from the guidelines of American Diabetes Association (ADA) recommend listing SGLT2 inhibitors for the treatment of T2DM patients with established high risk factors of atherosclerotic cardiovascular disease (ASCVD), ASCVD and heart failure [121]. The underlying mechanisms of SGLT2 inhibitors on cardioprotection may be related to improving the function of vascular endothelial cells, suppressing oxidative stress, inhibiting inflammation and regulating autophagy, which further protects from the progression of atherosclerosis. More studies are needed, however, to elucidate the underlying mechanisms.

Author Contributions

J.X., T.H., M.K. and D.K. contributed to drafting and writing the article. J.X., T.H., M.K. and D.K. contributed to the discussion of the review. M.K. and D.K. are responsible for the integrity of the content. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

Boehringer Ingelheim, Mitsubishi Tanabe Pharma, Taisho Pharmaceutical Co. and Ono Pharmaceutical Co. contributed to establishing the Division of Anticipatory Molecular Food Science and Technology. The authors declare that there are no conflicts of interest associated with this manuscript. The funders had no role in the interpretation or writing of the manuscript.

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. Lusis, A.J. Atherosclerosis. Nature 2000, 407, 233–241. [Google Scholar] [CrossRef] [PubMed]
  2. Hassanpour, M.; Rahbarghazi, R.; Nouri, M.; Aghamohammadzadeh, N.; Safaei, N.; Ahmadi, M. Role of autophagy in atherosclerosis: Foe or friend? J. Inflam. 2019, 16, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Weber, C.; Noels, H. Atherosclerosis: Current pathogenesis and therapeutic options. Nat. Med. 2011, 17, 1410–1422. [Google Scholar] [CrossRef]
  4. Schneider, A.L.; Kalyani, R.R.; Golden, S.; Stearns, S.C.; Wruck, L.; Yeh, H.C.; Coresh, J.; Selvin, E. Diabetes and Prediabetes and Risk of Hospitalization: The Atherosclerosis Risk in Communities (ARIC) Study. Diabetes Care 2016, 39, 772–779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lovren, F.; Teoh, H.; Verma, S. Obesity and atherosclerosis: Mechanistic insights. Can. J. Cardiol. 2015, 31, 177–183. [Google Scholar] [CrossRef]
  6. Frohlich, E.D.; Susic, D. Blood pressure, large arteries and atherosclerosis. Adv. Cardiol. 2007, 44, 117–124. [Google Scholar] [CrossRef]
  7. Low Wang, C.C.; Hess, C.N.; Hiatt, W.R.; Goldfine, A.B. Clinical Update: Cardiovascular Disease in Diabetes Mellitus: Atherosclerotic Cardiovascular Disease and Heart Failure in Type 2 Diabetes Mellitus—Mechanisms, Management, and Clinical Considerations. Circulation 2016, 133, 2459–2502. [Google Scholar] [CrossRef] [PubMed]
  8. Beckman, J.A.; Creager, M.A. Vascular Complications of Diabetes. Circul. Res. 2016, 118, 1771–1785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Einarson, T.R.; Acs, A.; Ludwig, C.; Panton, U.H. Prevalence of cardiovascular disease in type 2 diabetes: A systematic literature review of scientific evidence from across the world in 2007–2017. Cardiovasc. Diabetol. 2018, 17, 83. [Google Scholar] [CrossRef] [Green Version]
  10. DeFronzo, R.A.; Norton, L.; Abdul-Ghani, M. Renal, metabolic and cardiovascular considerations of SGLT2 inhibition. Nat. Rev. Nephrol. 2017, 13, 11–26. [Google Scholar] [CrossRef]
  11. Fioretto, P.; Zambon, A.; Rossato, M.; Busetto, L.; Vettor, R. SGLT2 Inhibitors and the Diabetic Kidney. Diabetes Care 2016, 39 (Suppl. 2), S165–S171. [Google Scholar] [CrossRef] [Green Version]
  12. Wanner, C.; Inzucchi, S.E.; Lachin, J.M.; Fitchett, D.; von Eynatten, M.; Mattheus, M.; Johansen, O.E.; Woerle, H.J.; Broedl, U.C.; Zinman, B. Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes. N. Engl. J. Med. 2016, 375, 323–334. [Google Scholar] [CrossRef] [PubMed]
  13. Mosenzon, O.; Wiviott, S.D.; Cahn, A.; Rozenberg, A.; Yanuv, I.; Goodrich, E.L.; Murphy, S.A.; Heerspink, H.J.L.; Zelniker, T.A.; Dwyer, J.P.; et al. Effects of dapagliflozin on development and progression of kidney disease in patients with type 2 diabetes: An analysis from the DECLARE-TIMI 58 randomised trial. Lancet. Diabetes Endocrinol. 2019, 7, 606–617. [Google Scholar] [CrossRef]
  14. Perkovic, V.; Jardine, M.J.; Neal, B.; Bompoint, S.; Heerspink, H.J.L.; Charytan, D.M.; Edwards, R.; Agarwal, R.; Bakris, G.; Bull, S.; et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N. Engl. J. Med. 2019, 380, 2295–2306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Cherney, D.Z.I.; Cosentino, F.; Dagogo-Jack, S.; McGuire, D.K.; Pratley, R.; Frederich, R.; Maldonado, M.; Liu, C.C.; Liu, J.; Pong, A.; et al. Ertugliflozin and Slope of Chronic eGFR: Prespecified Analyses from the Randomized VERTIS CV Trial. Clin. J. Am. Soc. Nephrol. CJASN 2021, 16, 1345–1354. [Google Scholar] [CrossRef]
  16. Perkovic, V.; de Zeeuw, D.; Mahaffey, K.W.; Fulcher, G.; Erondu, N.; Shaw, W.; Barrett, T.D.; Weidner-Wells, M.; Deng, H.; Matthews, D.R.; et al. Canagliflozin and renal outcomes in type 2 diabetes: Results from the CANVAS Program randomised clinical trials. Lancet Diabetes Endocrinol. 2018, 6, 691–704. [Google Scholar] [CrossRef]
  17. Vallon, V.; Thomson, S.C. Targeting renal glucose reabsorption to treat hyperglycaemia: The pleiotropic effects of SGLT2 inhibition. Diabetologia 2017, 60, 215–225. [Google Scholar] [CrossRef] [PubMed]
  18. Sen, T.; Heerspink, H.J.L. A kidney perspective on the mechanism of action of sodium glucose co-transporter 2 inhibitors. Cell Metab. 2021. [Google Scholar] [CrossRef]
  19. 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] [PubMed]
  20. Wright, E.M. Renal Na+-glucose cotransporters. Am. J. Physiol. Renal. Physiol. 2001, 280, F10–F18. [Google Scholar] [CrossRef]
  21. DeFronzo, R.A.; Reeves, W.B.; Awad, A.S. Pathophysiology of diabetic kidney disease: Impact of SGLT2 inhibitors. Nat. Rev. Nephrol. 2021, 17, 319–334. [Google Scholar] [CrossRef]
  22. Vallon, V.; Platt, K.A.; Cunard, R.; Schroth, J.; Whaley, J.; Thomson, S.C.; Koepsell, H.; Rieg, T. SGLT2 mediates glucose reabsorption in the early proximal tubule. J. Am. Soc. Nephrol. 2011, 22, 104–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wang, X.X.; Levi, J.; Luo, Y.; Myakala, K.; Herman-Edelstein, M.; Qiu, L.; Wang, D.; Peng, Y.; Grenz, A.; Lucia, S.; et al. SGLT2 Protein Expression Is Increased in Human Diabetic Nephropathy: Sglt2 Protein Inhibition Decreases Renal Lipid Accumulation, Inflammation, And The Development Of Nephropathy In Diabetic Mice. J. Biol. Chem. 2017, 292, 5335–5348. [Google Scholar] [CrossRef] [Green Version]
  24. Rahmoune, H.; Thompson, P.W.; Ward, J.M.; Smith, C.D.; Hong, G.; Brown, J. Glucose transporters in human renal proximal tubular cells isolated from the urine of patients with non-insulin-dependent diabetes. Diabetes 2005, 54, 3427–3434. [Google Scholar] [CrossRef] [Green Version]
  25. Ehrenkranz, J.R.; Lewis, N.G.; Kahn, C.R.; Roth, J. Phlorizin: A review. Diabetes Metabol. Res. Rev. 2005, 21, 31–38. [Google Scholar] [CrossRef]
  26. Choi, C.I. Sodium-Glucose Cotransporter 2 (SGLT2) Inhibitors from Natural Products: Discovery of Next-Generation Antihyperglycemic Agents. Molecules 2016, 21, 1136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Rieg, T.; Vallon, V. Development of SGLT1 and SGLT2 inhibitors. Diabetologia 2018, 61, 2079–2086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Grempler, R.; Thomas, L.; Eckhardt, M.; Himmelsbach, F.; Sauer, A.; Sharp, D.E.; Bakker, R.A.; Mark, M.; Klein, T.; Eickelmann, P. Empagliflozin, a novel selective sodium glucose cotransporter-2 (SGLT-2) inhibitor: Characterisation and comparison with other SGLT-2 inhibitors. Diabetes, Obes. Metabol. 2012, 14, 83–90. [Google Scholar] [CrossRef]
  29. Poole, R.M.; Dungo, R.T. Ipragliflozin: First global approval. Drugs 2014, 74, 611–617. [Google Scholar] [CrossRef]
  30. Markham, A.; Elkinson, S. Luseogliflozin: First global approval. Drugs 2014, 74, 945–950. [Google Scholar] [CrossRef]
  31. Mizuno, Y.; Jacob, R.F.; Mason, R.P. Inflammation and the development of atherosclerosis. J. Atherosc. Thromb. 2011, 18, 351–358. [Google Scholar] [CrossRef] [Green Version]
  32. Aboonabi, A.; Meyer, R.R.; Singh, I. The association between metabolic syndrome components and the development of atherosclerosis. J. Hum. Hypertens. 2019, 33, 844–855. [Google Scholar] [CrossRef]
  33. Xu, J.; Kitada, M.; Ogura, Y.; Koya, D. Relationship Between Autophagy and Metabolic Syndrome Characteristics in the Pathogenesis of Atherosclerosis. Front. Cell Dev. Biol. 2021, 9, 641852. [Google Scholar] [CrossRef]
  34. Leng, W.; Ouyang, X.; Lei, X.; Wu, M.; Chen, L.; Wu, Q.; Deng, W.; Liang, Z. The SGLT-2 Inhibitor Dapagliflozin Has a Therapeutic Effect on Atherosclerosis in Diabetic ApoE−/− Mice. Med. Inflamm. 2016, 2016, 6305735. [Google Scholar] [CrossRef] [Green Version]
  35. Han, J.H.; Oh, T.J.; Lee, G.; Maeng, H.J.; Lee, D.H.; Kim, K.M.; Choi, S.H.; Jang, H.C.; Lee, H.S.; Park, K.S.; et al. The beneficial effects of empagliflozin, an SGLT2 inhibitor, on atherosclerosis in ApoE−/− mice fed a western diet. Diabetologia 2017, 60, 364–376. [Google Scholar] [CrossRef] [Green Version]
  36. Nakatsu, Y.; Kokubo, H.; Bumdelger, B.; Yoshizumi, M.; Yamamotoya, T.; Matsunaga, Y.; Ueda, K.; Inoue, Y.; Inoue, M.K.; Fujishiro, M.; et al. The SGLT2 Inhibitor Luseogliflozin Rapidly Normalizes Aortic mRNA Levels of Inflammation-Related but Not Lipid-Metabolism-Related Genes and Suppresses Atherosclerosis in Diabetic ApoE KO Mice. Int. J. Mol. Sci. 2017, 18, 1704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Al-Sharea, A.; Murphy, A.J.; Huggins, L.A.; Hu, Y.; Goldberg, I.J.; Nagareddy, P.R. SGLT2 inhibition reduces atherosclerosis by enhancing lipoprotein clearance in Ldlr−/− type 1 diabetic mice. Atherosclerosis 2018, 271, 166–176. [Google Scholar] [CrossRef]
  38. Ganbaatar, B.; Fukuda, D.; Shinohara, M.; Yagi, S.; Kusunose, K.; Yamada, H.; Soeki, T.; Hirata, K.I.; Sata, M. Empagliflozin ameliorates endothelial dysfunction and suppresses atherogenesis in diabetic apolipoprotein E-deficient mice. Eur. J. Pharmacol. 2020, 875, 173040. [Google Scholar] [CrossRef]
  39. Förstermann, U.; Sessa, W.C. Nitric oxide synthases: Regulation and function. Eur. Heart J. 2012, 33, 829–837, 837a–837d. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Uthman, L.; Baartscheer, A.; Bleijlevens, B.; Schumacher, C.A.; Fiolet, J.W.T.; Koeman, A.; Jancev, M.; Hollmann, M.W.; Weber, N.C.; Coronel, R.; et al. Class effects of SGLT2 inhibitors in mouse cardiomyocytes and hearts: Inhibition of Na+/H+ exchanger, lowering of cytosolic Na+ and vasodilation. Diabetologia 2018, 61, 722–726. [Google Scholar] [CrossRef] [Green Version]
  41. Aroor, A.R.; Das, N.A.; Carpenter, A.J.; Habibi, J.; Jia, G.; Ramirez-Perez, F.I.; Martinez-Lemus, L.; Manrique-Acevedo, C.M.; Hayden, M.R.; Duta, C.; et al. Glycemic control by the SGLT2 inhibitor empagliflozin decreases aortic stiffness, renal resistivity index and kidney injury. Cardiovasc. Diabetol. 2018, 17, 108. [Google Scholar] [CrossRef]
  42. Sayour, A.A.; Korkmaz-Icöz, S.; Loganathan, S.; Ruppert, M.; Sayour, V.N.; Oláh, A.; Benke, K.; Brune, M.; Benkő, R.; Horváth, E.M.; et al. Acute canagliflozin treatment protects against in vivo myocardial ischemia-reperfusion injury in non-diabetic male rats and enhances endothelium-dependent vasorelaxation. J. Trans. Med. 2019, 17, 127. [Google Scholar] [CrossRef] [PubMed]
  43. Uthman, L.; Homayr, A.; Juni, R.P.; Spin, E.L.; Kerindongo, R.; Boomsma, M.; Hollmann, M.W.; Preckel, B.; Koolwijk, P.; van Hinsbergh, V.W.M.; et al. Empagliflozin and Dapagliflozin Reduce ROS Generation and Restore NO Bioavailability in Tumor Necrosis Factor α-Stimulated Human Coronary Arterial Endothelial Cells. Cell Physiol. Biochem. 2019, 53, 865–886. [Google Scholar] [CrossRef]
  44. Yang, X.; Li, Y.; Li, Y.; Ren, X.; Zhang, X.; Hu, D.; Gao, Y.; Xing, Y.; Shang, H. Oxidative Stress-Mediated Atherosclerosis: Mechanisms and Therapies. Front. Physiol. 2017, 8, 600. [Google Scholar] [CrossRef] [Green Version]
  45. Rosenblat, M.; Volkova, N.; Paland, N.; Aviram, M. Triglyceride accumulation in macrophages upregulates paraoxonase 2 (PON2) expression via ROS-mediated JNK/c-Jun signaling pathway activation. BioFactors 2012, 38, 458–469. [Google Scholar] [CrossRef] [PubMed]
  46. Schaffer, S.W.; Jong, C.J.; Mozaffari, M. Role of oxidative stress in diabetes-mediated vascular dysfunction: Unifying hypothesis of diabetes revisited. Vasc. Pharmacol. 2012, 57, 139–149. [Google Scholar] [CrossRef]
  47. Furukawa, S.; Fujita, T.; Shimabukuro, M.; Iwaki, M.; Yamada, Y.; Nakajima, Y.; Nakayama, O.; Makishima, M.; Matsuda, M.; Shimomura, I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J. Clin. Investig. 2004, 114, 1752–1761. [Google Scholar] [CrossRef] [PubMed]
  48. Di Marco, E.; Gray, S.P.; Chew, P.; Koulis, C.; Ziegler, A.; Szyndralewiez, C.; Touyz, R.M.; Schmidt, H.H.; Cooper, M.E.; Slattery, R.; et al. Pharmacological inhibition of NOX reduces atherosclerotic lesions, vascular ROS and immune-inflammatory responses in diabetic Apoe−/− mice. Diabetologia 2014, 57, 633–642. [Google Scholar] [CrossRef]
  49. Park, S.H.; Belcastro, E.; Hasan, H.; Matsushita, K.; Marchandot, B.; Abbas, M.; Toti, F.; Auger, C.; Jesel, L.; Ohlmann, P.; et al. Angiotensin II-induced upregulation of SGLT1 and 2 contributes to human microparticle-stimulated endothelial senescence and dysfunction: Protective effect of gliflozins. Cardiovasc. Diabetol. 2021, 20, 65. [Google Scholar] [CrossRef] [PubMed]
  50. Rahadian, A.; Fukuda, D.; Salim, H.M.; Yagi, S.; Kusunose, K.; Yamada, H.; Soeki, T.; Sata, M. Canagliflozin Prevents Diabetes-Induced Vascular Dysfunction in ApoE-Deficient Mice. J. Atherosc. Thrombosis 2020, 27, 1141–1151. [Google Scholar] [CrossRef] [Green Version]
  51. El-Daly, M.; Pulakazhi Venu, V.K.; Saifeddine, M.; Mihara, K.; Kang, S.; Fedak, P.W.M.; Alston, L.A.; Hirota, S.A.; Ding, H.; Triggle, C.R.; et al. Hyperglycaemic impairment of PAR2-mediated vasodilation: Prevention by inhibition of aortic endothelial sodium-glucose-co-Transporter-2 and minimizing oxidative stress. Vasc. Pharmacol. 2018, 109, 56–71. [Google Scholar] [CrossRef]
  52. Nasiri-Ansari, Ν.; Dimitriadis, G.K.; Agrogiannis, G.; Perrea, D.; Kostakis, I.D.; Kaltsas, G.; Papavassiliou, A.G.; Randeva, H.S.; Kassi, E. Canagliflozin attenuates the progression of atherosclerosis and inflammation process in APOE knockout mice. Cardiovasc. Diabetol. 2018, 17, 106. [Google Scholar] [CrossRef] [Green Version]
  53. Dimitriadis, G.K.; Nasiri-Ansari, N.; Agrogiannis, G.; Kostakis, I.D.; Randeva, M.S.; Nikiteas, N.; Patel, V.H.; Kaltsas, G.; Papavassiliou, A.G.; Randeva, H.S.; et al. Empagliflozin improves primary haemodynamic parameters and attenuates the development of atherosclerosis in high fat diet fed APOE knockout mice. Mol. Cellul. Endoc. 2019, 494, 110487. [Google Scholar] [CrossRef]
  54. Gaspari, T.; Spizzo, I.; Liu, H.; Hu, Y.; Simpson, R.W.; Widdop, R.E.; Dear, A.E. Dapagliflozin attenuates human vascular endothelial cell activation and induces vasorelaxation: A potential mechanism for inhibition of atherogenesis. Diab. Vasc. Dis. Res. 2018, 15, 64–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Mancini, S.J.; Boyd, D.; Katwan, O.J.; Strembitska, A.; Almabrouk, T.A.; Kennedy, S.; Palmer, T.M.; Salt, I.P. Canagliflozin inhibits interleukin-1β-stimulated cytokine and chemokine secretion in vascular endothelial cells by AMP-activated protein kinase-dependent and -independent mechanisms. Sci. Rep. 2018, 8, 5276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Duewell, P.; Kono, H.; Rayner, K.J.; Sirois, C.M.; Vladimer, G.; Bauernfeind, F.G.; Abela, G.S.; Franchi, L.; Nuñez, G.; Schnurr, M.; et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010, 464, 1357–1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Razani, B.; Feng, C.; Coleman, T.; Emanuel, R.; Wen, H.; Hwang, S.; Ting, J.P.; Virgin, H.W.; Kastan, M.B.; Semenkovich, C.F. Autophagy links inflammasomes to atherosclerotic progression. Cell Metab. 2012, 15, 534–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Satish, M.; Agrawal, D.K. Atherothrombosis and the NLRP3 inflammasome-endogenous mechanisms of inhibition. Trans. Res. J. Lab. Clin. Med. 2020, 215, 75–85. [Google Scholar] [CrossRef] [PubMed]
  59. Qiu, Y.Y.; Tang, L.Q. Roles of the NLRP3 inflammasome in the pathogenesis of diabetic nephropathy. Pharmacol. Res. 2016, 114, 251–264. [Google Scholar] [CrossRef] [PubMed]
  60. Luo, B.; Li, B.; Wang, W.; Liu, X.; Xia, Y.; Zhang, C.; Zhang, M.; Zhang, Y.; An, F. NLRP3 gene silencing ameliorates diabetic cardiomyopathy in a type 2 diabetes rat model. PLoS ONE 2014, 9, e104771. [Google Scholar] [CrossRef]
  61. Bleda, S.; de Haro, J.; Varela, C.; Ferruelo, A.; Acin, F. Elevated levels of triglycerides and vldl-cholesterol provoke activation of nlrp1 inflammasome in endothelial cells. Int. J. Cardiol. 2016, 220, 52–55. [Google Scholar] [CrossRef]
  62. Kim, S.R.; Lee, S.G.; Kim, S.H.; Kim, J.H.; Choi, E.; Cho, W.; Rim, J.H.; Hwang, I.; Lee, C.J.; Lee, M.; et al. SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nat. Commun. 2020, 11, 2127. [Google Scholar] [CrossRef]
  63. Ye, Y.; Bajaj, M.; Yang, H.C.; Perez-Polo, J.R.; Birnbaum, Y. SGLT-2 Inhibition with Dapagliflozin Reduces the Activation of the Nlrp3/ASC Inflammasome and Attenuates the Development of Diabetic Cardiomyopathy in Mice with Type 2 Diabetes. Further Augmentation of the Effects with Saxagliptin, a DPP4 Inhibitor. Cardiovasc. Drugs Therapy 2017, 31, 119–132. [Google Scholar] [CrossRef] [PubMed]
  64. Sukhanov, S.; Higashi, Y.; Yoshida, T.; Mummidi, S.; Aroor, A.R.; Jeffrey Russell, J.; Bender, S.B.; DeMarco, V.G.; Chandrasekar, B. The SGLT2 inhibitor Empagliflozin attenuates interleukin-17A-induced human aortic smooth muscle cell proliferation and migration by targeting TRAF3IP2/ROS/NLRP3/Caspase-1-dependent IL-1β and IL-18 secretion. Cell. Signal. 2021, 77, 109825. [Google Scholar] [CrossRef]
  65. Martinet, W.; De Meyer, G.R. Autophagy in atherosclerosis: A cell survival and death phenomenon with therapeutic potential. Circul. Res. 2009, 104, 304–317. [Google Scholar] [CrossRef] [Green Version]
  66. Kitada, M.; Ogura, Y.; Koya, D. The protective role of Sirt1 in vascular tissue: Its relationship to vascular aging and atherosclerosis. Aging 2016, 8, 2290–2307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Aragón-Herrera, A.; Feijóo-Bandín, S.; Otero Santiago, M.; Barral, L.; Campos-Toimil, M.; Gil-Longo, J.; Costa Pereira, T.M.; García-Caballero, T.; Rodríguez-Segade, S.; Rodríguez, J.; et al. Empagliflozin reduces the levels of CD36 and cardiotoxic lipids while improving autophagy in the hearts of Zucker diabetic fatty rats. Biochem. Pharmacol. 2019, 170, 113677. [Google Scholar] [CrossRef]
  68. Jaikumkao, K.; Promsan, S.; Thongnak, L.; Swe, M.T.; Tapanya, M.; Htun, K.T.; Kothan, S.; Intachai, N.; Lungkaphin, A. Dapagliflozin ameliorates pancreatic injury and activates kidney autophagy by modulating the AMPK/mTOR signaling pathway in obese rats. J. Cellul. Physiol. 2021, 236, 6424–6440. [Google Scholar] [CrossRef]
  69. Ren, C.; Sun, K.; Zhang, Y.; Hu, Y.; Hu, B.; Zhao, J.; He, Z.; Ding, R.; Wang, W.; Liang, C. Sodium-Glucose CoTransporter-2 Inhibitor Empagliflozin Ameliorates Sunitinib-Induced Cardiac Dysfunction via Regulation of AMPK-mTOR Signaling Pathway-Mediated Autophagy. Front. Pharmacol. 2021, 12, 664181. [Google Scholar] [CrossRef]
  70. Xu, C.; Wang, W.; Zhong, J.; Lei, F.; Xu, N.; Zhang, Y.; Xie, W. Canagliflozin exerts anti-inflammatory effects by inhibiting intracellular glucose metabolism and promoting autophagy in immune cells. Biochem. Pharmacol. 2018, 152, 45–59. [Google Scholar] [CrossRef]
  71. Xu, J.; Kitada, M.; Ogura, Y.; Liu, H.; Koya, D. Dapagliflozin Restores Impaired Autophagy and Suppresses Inflammation in High Glucose-Treated HK-2 Cells. Cells 2021, 10, 1457. [Google Scholar] [CrossRef] [PubMed]
  72. Véniant, M.M.; Withycombe, S.; Young, S.G. Lipoprotein size and atherosclerosis susceptibility in Apoe−/− and Ldlr−/− mice. Arteriosc. Thromb. Vasc. Biol. 2001, 21, 1567–1570. [Google Scholar] [CrossRef] [Green Version]
  73. Terasaki, M.; Hiromura, M.; Mori, Y.; Kohashi, K.; Nagashima, M.; Kushima, H.; Watanabe, T.; Hirano, T. Amelioration of Hyperglycemia with a Sodium-Glucose Cotransporter 2 Inhibitor Prevents Macrophage-Driven Atherosclerosis through Macrophage Foam Cell Formation Suppression in Type 1 and Type 2 Diabetic Mice. PLoS ONE 2015, 10, e0143396. [Google Scholar] [CrossRef]
  74. Liu, Y.; Xu, J.; Wu, M.; Xu, B.; Kang, L. Empagliflozin protects against atherosclerosis progression by modulating lipid profiles and sympathetic activity. Lipids. Health Dis. 2021, 20, 5. [Google Scholar] [CrossRef] [PubMed]
  75. Mori, Y.; Terasaki, M.; Hiromura, M.; Saito, T.; Kushima, H.; Koshibu, M.; Osaka, N.; Ohara, M.; Fukui, T.; Ohtaki, H.; et al. Luseogliflozin attenuates neointimal hyperplasia after wire injury in high-fat diet-fed mice via inhibition of perivascular adipose tissue remodeling. Cardiovasc. Diabetol. 2019, 18, 143. [Google Scholar] [CrossRef]
  76. Mori, K.; Tsuchiya, K.; Nakamura, S.; Miyachi, Y.; Shiba, K.; Ogawa, Y.; Kitamura, K. Ipragliflozin-induced adipose expansion inhibits cuff-induced vascular remodeling in mice. Cardiovasc. Diabetol. 2019, 18, 83. [Google Scholar] [CrossRef]
  77. Prattichizzo, F.; De Nigris, V.; Micheloni, S.; La Sala, L.; Ceriello, A. Increases in circulating levels of ketone bodies and cardiovascular protection with SGLT2 inhibitors: Is low-grade inflammation the neglected component? Diabetes Obes. Metab. 2018, 20, 2515–2522. [Google Scholar] [CrossRef]
  78. Xu, L.; Ota, T. Emerging roles of SGLT2 inhibitors in obesity and insulin resistance: Focus on fat browning and macrophage polarization. Adipocyte 2018, 7, 121–128. [Google Scholar] [CrossRef] [Green Version]
  79. Rosenstock, J.; Frias, J.; Páll, D.; Charbonnel, B.; Pascu, R.; Saur, D.; Darekar, A.; Huyck, S.; Shi, H.; Lauring, B.; et al. Effect of ertugliflozin on glucose control, body weight, blood pressure and bone density in type 2 diabetes mellitus inadequately controlled on metformin monotherapy (VERTIS MET). Diabetes Obes. Metab. 2018, 20, 520–529. [Google Scholar] [CrossRef] [Green Version]
  80. Schork, A.; Saynisch, J.; Vosseler, A.; Jaghutriz, B.A.; Heyne, N.; Peter, A.; Häring, H.U.; Stefan, N.; Fritsche, A.; Artunc, F. Effect of SGLT2 inhibitors on body composition, fluid status and renin-angiotensin-aldosterone system in type 2 diabetes: A prospective study using bioimpedance spectroscopy. Cardiovasc. Diabetol. 2019, 18, 46. [Google Scholar] [CrossRef] [PubMed]
  81. Sakai, S.; Kaku, K.; Seino, Y.; Inagaki, N.; Haneda, M.; Sasaki, T.; Fukatsu, A.; Kakiuchi, H.; Samukawa, Y. Efficacy and Safety of the SGLT2 Inhibitor Luseogliflozin in Japanese Patients With Type 2 Diabetes Mellitus Stratified According to Baseline Body Mass Index: Pooled Analysis of Data From 52-Week Phase III Trials. Clin. Therap. 2016, 38, 843–862.e849. [Google Scholar] [CrossRef] [Green Version]
  82. Bode, B.; Stenlöf, K.; Harris, S.; Sullivan, D.; Fung, A.; Usiskin, K.; Meininger, G. Long-term efficacy and safety of canagliflozin over 104 weeks in patients aged 55–80 years with type 2 diabetes. Diabetes Obes. Metab. 2015, 17, 294–303. [Google Scholar] [CrossRef]
  83. Mancia, G.; Cannon, C.P.; Tikkanen, I.; Zeller, C.; Ley, L.; Woerle, H.J.; Broedl, U.C.; Johansen, O.E. Impact of Empagliflozin on Blood Pressure in Patients With Type 2 Diabetes Mellitus and Hypertension by Background Antihypertensive Medication. Hypertension 2016, 68, 1355–1364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Chilton, R.; Tikkanen, I.; Hehnke, U.; Woerle, H.J.; Johansen, O.E. Impact of empagliflozin on blood pressure in dipper and non-dipper patients with type 2 diabetes mellitus and hypertension. Diabetes Obes. Metab. 2017, 19, 1620–1624. [Google Scholar] [CrossRef] [PubMed]
  85. Kario, K.; Okada, K.; Kato, M.; Nishizawa, M.; Yoshida, T.; Asano, T.; Uchiyama, K.; Niijima, Y.; Katsuya, T.; Urata, H.; et al. Twenty-Four-Hour Blood Pressure-Lowering Effect of an SGLT-2 Inhibitor in Patients with Diabetes and Uncontrolled Nocturnal Hypertension: Results from the Randomized, Placebo-Controlled SACRA Study. Circulation 2018, 139, 2089–2097. [Google Scholar] [CrossRef] [PubMed]
  86. Cefalu, W.T.; Stenlöf, K.; Leiter, L.A.; Wilding, J.P.; Blonde, L.; Polidori, D.; Xie, J.; Sullivan, D.; Usiskin, K.; Canovatchel, W.; et al. Effects of canagliflozin on body weight and relationship to HbA1c and blood pressure changes in patients with type 2 diabetes. Diabetologia 2015, 58, 1183–1187. [Google Scholar] [CrossRef] [PubMed]
  87. Ramirez, A.J.; Sanchez, M.J.; Sanchez, R.A. Diabetic patients with essential hypertension treated with amlodipine: Blood pressure and arterial stiffness effects of canagliflozin or perindopril. J. Hypertens. 2019, 37, 636–642. [Google Scholar] [CrossRef]
  88. Ye, N.; Jardine, M.J.; Oshima, M.; Hockham, C.; Heerspink, H.J.L.; Agarwal, R.; Bakris, G.; Schutte, A.E.; Arnott, C.; Chang, T.I.; et al. Blood Pressure Effects of Canagliflozin and Clinical Outcomes in Type 2 Diabetes and Chronic Kidney Disease: Insights From the CREDENCE Trial. Circulation 2021, 143, 1735–1749. [Google Scholar] [CrossRef]
  89. Liu, J.; Pong, A.; Gallo, S.; Darekar, A.; Terra, S.G. Effect of ertugliflozin on blood pressure in patients with type 2 diabetes mellitus: A post hoc pooled analysis of randomized controlled trials. Cardiovasc. Diabetol. 2019, 18, 59. [Google Scholar] [CrossRef] [Green Version]
  90. Briasoulis, A.; Al Dhaybi, O.; Bakris, G.L. SGLT2 Inhibitors and Mechanisms of Hypertension. Curr. Cardiol. Rep. 2018, 20, 1. [Google Scholar] [CrossRef]
  91. Stenlöf, K.; Cefalu, W.T.; Kim, K.A.; Jodar, E.; Alba, M.; Edwards, R.; Tong, C.; Canovatchel, W.; Meininger, G. Long-term efficacy and safety of canagliflozin monotherapy in patients with type 2 diabetes inadequately controlled with diet and exercise: Findings from the 52-week CANTATA-M study. Curr. Med. Res. Opin. 2014, 30, 163–175. [Google Scholar] [CrossRef] [PubMed]
  92. Cha, S.A.; Park, Y.M.; Yun, J.S.; Lim, T.S.; Song, K.H.; Yoo, K.D.; Ahn, Y.B.; Ko, S.H. A comparison of effects of DPP-4 inhibitor and SGLT2 inhibitor on lipid profile in patients with type 2 diabetes. Lipids. Health Dis. 2017, 16, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Jojima, T.; Sakurai, S.; Wakamatsu, S.; Iijima, T.; Saito, M.; Tomaru, T.; Kogai, T.; Usui, I.; Aso, Y. Empagliflozin increases plasma levels of campesterol, a marker of cholesterol absorption, in patients with type 2 diabetes: Association with a slight increase in high-density lipoprotein cholesterol. Int. J. Cardiol. 2021, 331, 243–248. [Google Scholar] [CrossRef]
  94. Basu, D.; Huggins, L.A.; Scerbo, D.; Obunike, J.; Mullick, A.E.; Rothenberg, P.L.; Di Prospero, N.A.; Eckel, R.H.; Goldberg, I.J. Mechanism of Increased LDL (Low-Density Lipoprotein) and Decreased Triglycerides With SGLT2 (Sodium-Glucose Cotransporter 2) Inhibition. Arteriosc. Thromb. Vasc. Biol. 2018, 38, 2207–2216. [Google Scholar] [CrossRef] [PubMed]
  95. Briand, F.; Mayoux, E.; Brousseau, E.; Burr, N.; Urbain, I.; Costard, C.; Mark, M.; Sulpice, T. Empagliflozin, via Switching Metabolism Toward Lipid Utilization, Moderately Increases LDL Cholesterol Levels Through Reduced LDL Catabolism. Diabetes 2016, 65, 2032–2038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Bailey, C.J. Uric acid and the cardio-renal effects of SGLT2 inhibitors. Diabetes Obes. Metab. 2019, 21, 1291–1298. [Google Scholar] [CrossRef] [Green Version]
  97. Chino, Y.; Kuwabara, M.; Hisatome, I. Factors Influencing Change in Serum Uric Acid after Administration of the Sodium-Glucose Cotransporter 2 Inhibitor Luseogliflozin in Patients With Type 2 Diabetes Mellitus. J. Clin. Pharmacol. 2021. [Google Scholar] [CrossRef]
  98. Davies, M.J.; Trujillo, A.; Vijapurkar, U.; Damaraju, C.V.; Meininger, G. Effect of canagliflozin on serum uric acid in patients with type 2 diabetes mellitus. Diabetes Obes. Metab. 2015, 17, 426–429. [Google Scholar] [CrossRef] [Green Version]
  99. McGuire, D.K.; Marx, N.; Johansen, O.E.; Inzucchi, S.E.; Rosenstock, J.; George, J.T. FDA guidance on antihyperglyacemic therapies for type 2 diabetes: One decade later. Diabetes Obes. Metab. 2019, 21, 1073–1078. [Google Scholar] [CrossRef]
  100. Rangaswami, J.; Bhalla, V.; de Boer, I.H.; Staruschenko, A.; Sharp, J.A.; Singh, R.R.; Lo, K.B.; Tuttle, K.; Vaduganathan, M.; Ventura, H.; et al. Cardiorenal Protection With the Newer Antidiabetic Agents in Patients With Diabetes and Chronic Kidney Disease: A Scientific Statement From the American Heart Association. Circulation 2020, 142, e265–e286. [Google Scholar] [CrossRef]
  101. Kosiborod, M.; Cavender, M.A.; Fu, A.Z.; Wilding, J.P.; Khunti, K.; Holl, R.W.; Norhammar, A.; Birkeland, K.I.; Jørgensen, M.E.; Thuresson, M.; et al. Lower Risk of Heart Failure and Death in Patients Initiated on Sodium-Glucose Cotransporter-2 Inhibitors Versus Other Glucose-Lowering Drugs: The CVD-REAL Study (Comparative Effectiveness of Cardiovascular Outcomes in New Users of Sodium-Glucose Cotransporter-2 Inhibitors). Circulation 2017, 136, 249–259. [Google Scholar] [CrossRef]
  102. Kosiborod, M.; Birkeland, K.I.; Cavender, M.A.; Fu, A.Z.; Wilding, J.P.; Khunti, K.; Holl, R.W.; Norhammar, A.; Jørgensen, M.E.; Wittbrodt, E.T.; et al. Rates of myocardial infarction and stroke in patients initiating treatment with SGLT2-inhibitors versus other glucose-lowering agents in real-world clinical practice: Results from the CVD-REAL study. Diabetes Obes. Metab. 2018, 20, 1983–1987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Wu, J.H.; Foote, C.; Blomster, J.; Toyama, T.; Perkovic, V.; Sundström, J.; Neal, B. Effects of sodium-glucose cotransporter-2 inhibitors on cardiovascular events, death, and major safety outcomes in adults with type 2 diabetes: A systematic review and meta-analysis. Lancet Diabetes Endocrinol. 2016, 4, 411–419. [Google Scholar] [CrossRef]
  104. Zinman, B.; Wanner, C.; Lachin, J.M.; Fitchett, D.; Bluhmki, E.; Hantel, S.; Mattheus, M.; Devins, T.; Johansen, O.E.; Woerle, H.J.; et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N. Engl. J. Med 2015, 373, 2117–2128. [Google Scholar] [CrossRef] [PubMed]
  105. Anker, S.D.; Butler, J.; Filippatos, G.; Khan, M.S.; Marx, N.; Lam, C.S.P.; Schnaidt, S.; Ofstad, A.P.; Brueckmann, M.; Jamal, W.; et al. Effect of Empagliflozin on Cardiovascular and Renal Outcomes in Patients With Heart Failure by Baseline Diabetes Status: Results From the EMPEROR-Reduced Trial. Circulation 2021, 143, 337–349. [Google Scholar] [CrossRef] [PubMed]
  106. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Ferreira, J.P.; Pocock, S.J.; Carson, P.; Anand, I.; Doehner, W.; Haass, M.; et al. Effect of Empagliflozin on the Clinical Stability of Patients With Heart Failure and a Reduced Ejection Fraction: The EMPEROR-Reduced Trial. Circulation 2021, 143, 326–336. [Google Scholar] [CrossRef]
  107. Anker, S.D.; Butler, J.; Filippatos, G.; Ferreira, J.P.; Bocchi, E.; Böhm, M.; Brunner-La Rocca, H.P.; Choi, D.J.; Chopra, V.; Chuquiure-Valenzuela, E.; et al. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N. Engl. J. Med. 2021, 385, 1451–1461. [Google Scholar] [CrossRef]
  108. Wiviott, S.D.; Raz, I.; Bonaca, M.P.; Mosenzon, O.; Kato, E.T.; Cahn, A.; Silverman, M.G.; Zelniker, T.A.; Kuder, J.F.; Murphy, S.A.; et al. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2019, 380, 347–357. [Google Scholar] [CrossRef]
  109. McMurray, J.J.V.; Solomon, S.D.; Inzucchi, S.E.; Køber, L.; Kosiborod, M.N.; Martinez, F.A.; Ponikowski, P.; Sabatine, M.S.; Anand, I.S.; Bělohlávek, J.; et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N. Engl. J. Med. 2019, 381, 1995–2008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Cannon, C.P.; Pratley, R.; Dagogo-Jack, S.; Mancuso, J.; Huyck, S.; Masiukiewicz, U.; Charbonnel, B.; Frederich, R.; Gallo, S.; Cosentino, F.; et al. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N. Engl. J. Med. 2020, 383, 1425–1435. [Google Scholar] [CrossRef]
  111. Bhatt, D.L.; Szarek, M.; Steg, P.G.; Cannon, C.P.; Leiter, L.A.; McGuire, D.K.; Lewis, J.B.; Riddle, M.C.; Voors, A.A.; Metra, M.; et al. Sotagliflozin in Patients with Diabetes and Recent Worsening Heart Failure. N. Engl. J. Med. 2021, 384, 117–128. [Google Scholar] [CrossRef] [PubMed]
  112. Bhatt, D.L.; Szarek, M.; Pitt, B.; Cannon, C.P.; Leiter, L.A.; McGuire, D.K.; Lewis, J.B.; Riddle, M.C.; Inzucchi, S.E.; Kosiborod, M.N.; et al. Sotagliflozin in Patients with Diabetes and Chronic Kidney Disease. N. Engl. J. Med. 2021, 384, 129–139. [Google Scholar] [CrossRef]
  113. Hupfeld, C.; Mudaliar, S. Navigating the "MACE" in Cardiovascular Outcomes Trials and decoding the relevance of Atherosclerotic Cardiovascular Disease benefits versus Heart Failure benefits. Diabetes Obes. Metab. 2019, 21, 1780–1789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Natali, A.; Nesti, L.; Tricò, D.; Ferrannini, E. Effects of GLP-1 receptor agonists and SGLT-2 inhibitors on cardiac structure and function: A narrative review of clinical evidence. Cardiovasc. Diabetol. 2021, 20, 196. [Google Scholar] [CrossRef] [PubMed]
  115. Shao, S.C.; Lin, Y.H.; Chang, K.C.; Chan, Y.Y.; Hung, M.J.; Kao Yang, Y.H.; Lai, E.C. Sodium glucose co-transporter 2 inhibitors and cardiovascular event protections: How applicable are clinical trials and observational studies to real-world patients? BMJ Open Diabetes Res. Care 2019, 7, e000742. [Google Scholar] [CrossRef] [Green Version]
  116. Birkeland, K.I.; Bodegard, J.; Norhammar, A.; Kuiper, J.G.; Georgiado, E.; Beekman-Hendriks, W.L.; Thuresson, M.; Pignot, M.; Herings, R.M.C.; Kooy, A. How representative of a general type 2 diabetes population are patients included in cardiovascular outcome trials with SGLT2 inhibitors? A large European observational study. Diabetes Obes. Metab. 2019, 21, 968–974. [Google Scholar] [CrossRef] [Green Version]
  117. Kosiborod, M.; Lam, C.S.P.; Kohsaka, S.; Kim, D.J.; Karasik, A.; Shaw, J.; Tangri, N.; Goh, S.Y.; Thuresson, M.; Chen, H.; et al. Cardiovascular Events Associated With SGLT-2 Inhibitors Versus Other Glucose-Lowering Drugs: The CVD-REAL 2 Study. J. Am. College Cardiol. 2018, 71, 2628–2639. [Google Scholar] [CrossRef]
  118. Kohsaka, S.; Lam, C.S.P.; Kim, D.J.; Cavender, M.A.; Norhammar, A.; Jørgensen, M.E.; Birkeland, K.I.; Holl, R.W.; Franch-Nadal, J.; Tangri, N.; et al. Risk of cardiovascular events and death associated with initiation of SGLT2 inhibitors compared with DPP-4 inhibitors: An analysis from the CVD-REAL 2 multinational cohort study. Lancet. Diabetes Endocrinol. 2020, 8, 606–615. [Google Scholar] [CrossRef]
  119. Lee, H.F.; Chen, S.W.; Liu, J.R.; Li, P.R.; Wu, L.S.; Chang, S.H.; Yeh, Y.H.; Kuo, C.T.; Chan, Y.H.; See, L.C. Major adverse cardiovascular and limb events in patients with diabetes and concomitant peripheral artery disease treated with sodium glucose cotransporter 2 inhibitor versus dipeptidyl peptidase-4 inhibitor. Cardiovasc. Diabetol. 2020, 19, 160. [Google Scholar] [CrossRef]
  120. Paul, S.K.; Bhatt, D.L.; Montvida, O. The association of amputations and peripheral artery disease in patients with type 2 diabetes mellitus receiving sodium-glucose cotransporter type-2 inhibitors: Real-world study. Eur. Heart J. 2021, 42, 1728–1738. [Google Scholar] [CrossRef]
  121. American Diabetes Association. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes-2021. Diabetes Care 2021, 44, S111–S124. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms of SGLT2 inhibitors against atherosclerosis. Elevated metabolic characteristics including glucose, TC, TG, FFA, BP and angiotensin II accelerate the progression of atherosclerosis via the dysregulation of endothelial function, vasodilation and autophagy and activating oxidative stress and inflammation. SGLT2 inhibitors attenuate the alterations caused by these metabolic changes. TC, total cholesterol; TG, triglyceride; FFA, free fatty acid; BP, blood pressure; NADPH, nicotinamide adenine dinucleotide phosphate; NO, nitric oxide; eNOS, endothelial nitric oxide synthase; ROS, reactive oxygen species; NF-κB, nuclear factor kappa-B; NLRP3, nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3; IL, interleukin; TNFα, tumor necrosis factor α; MCP-1, monocyte chemoattractant protein 1; ICAM-1, intercellular cell adhesion molecule-1; VCAM-1, vascular cell adhesion protein 1; AMPK, AMP-activated protein kinase; mTOR, mechanistic target of rapamycin; , increase; , decrease.
Figure 1. Mechanisms of SGLT2 inhibitors against atherosclerosis. Elevated metabolic characteristics including glucose, TC, TG, FFA, BP and angiotensin II accelerate the progression of atherosclerosis via the dysregulation of endothelial function, vasodilation and autophagy and activating oxidative stress and inflammation. SGLT2 inhibitors attenuate the alterations caused by these metabolic changes. TC, total cholesterol; TG, triglyceride; FFA, free fatty acid; BP, blood pressure; NADPH, nicotinamide adenine dinucleotide phosphate; NO, nitric oxide; eNOS, endothelial nitric oxide synthase; ROS, reactive oxygen species; NF-κB, nuclear factor kappa-B; NLRP3, nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3; IL, interleukin; TNFα, tumor necrosis factor α; MCP-1, monocyte chemoattractant protein 1; ICAM-1, intercellular cell adhesion molecule-1; VCAM-1, vascular cell adhesion protein 1; AMPK, AMP-activated protein kinase; mTOR, mechanistic target of rapamycin; , increase; , decrease.
Jcm 11 00137 g001
Figure 2. Improvements of some CVD-related clinical indicators by SGLT2 inhibitors. SGLT2 inhibitors not only reduce high glucose independent of insulin but also improve blood pressure, body weight, lipid profile and ketones and uric acid to present cardioprotective effects; , increase; , decrease.
Figure 2. Improvements of some CVD-related clinical indicators by SGLT2 inhibitors. SGLT2 inhibitors not only reduce high glucose independent of insulin but also improve blood pressure, body weight, lipid profile and ketones and uric acid to present cardioprotective effects; , increase; , decrease.
Jcm 11 00137 g002
Table 1. Benefits of SGLT2 inhibitors in atherosclerotic rodents.
Table 1. Benefits of SGLT2 inhibitors in atherosclerotic rodents.
SGLT2 InhibitorsAtherosclerotic RodentsChanges of Metabolic CharacteristicsReferences
DapagliflozinSTZ-induced diabetic ApoE−/− miceFBG↓,
TC↓,
TG↓ body weight↓,
HbA1c↓
[34,73]
DapagliflozinSTZ-induced diabetic Ldlr−/− miceFBG↓,
TC↓,
TG↓ body weight↓
[37]
EmpagliflozinHFD-fed ApoE−/− miceFBG↓,
TC↓, heart rate↓,
BP↓
TG↓,
LDL↓, urinary microalbumin↓, body weight↓
[35,53,74]
EmpagliflozinSTZ-induced diabetic ApoE−/− micebody weight↓,
TG↓
[38]
CanagliflozinHFD-fed diabetic ApoE−/− miceglucose↓,
TC↓,
TG↓,
LDL↓, heart rate↓
[52]
CanagliflozinSTZ-induced diabetic ApoE−/− miceTC↓, glucose↓[50]
LuseogliflozinHFD-fed micebody weight↓,
TC↓,
TG↓,
BP↓, glucose↓
[75]
LuseogliflozinSTZ-induced diabetic ApoE−/− micebody weight↓,
TC↓,
TG↓, BP↓, glucose↓
[36]
IpragliflozinHFD-fed micebody weight↓, glucose↓[76]
↓, decrease; STZ, streptozotocin; Apo E, Apolipoprotein E; Ldlr, low density lipoprotein receptor; HFD, high fat diet; FBG, fasting blood glucose; TC, total cholesterol; TG, triglycerides; HbA1c, glycosylated hemoglobin; LDL, low-density lipoprotein; BP, blood pressure.
Table 2. Reported cardiovascular outcome trials of SGLT2 inhibitors.
Table 2. Reported cardiovascular outcome trials of SGLT2 inhibitors.
EMPA-REG OUTCOME [104]EMPEROR- Reduced [105,106]EMPEROR-Preserved [107]CANVAS [19]CREDENCE [14]DECLARE- TIMI 58 [108]DAPA-HF [109]VERTIS CV [110]SOLOIST-WHF [111]SCORED [112]
Drugempagliflozinempagliflozinempagliflozincanagliflozincanagliflozindapagliflozindapagliflozinertugliflozinsotaglflozinsotaglflozin
Patients7020
(T2DM and CVD)
3730
(NYHA class II, III, or IV heart failure, EF ≤ 40%, 50% were DM)
5988 (NYHA class II-IV and EF ≥ 40%)10,142 (T2DM)4401
(T2DM and CKD)
17,160
(T2DM)
4744
(NYHA class II, III, or IV heart failure, EF ≤ 40%, 45% were T2DM)
8238
(T2DM and CVD)
1222 (T2DM and
heart failure
10,584 (T2DM and CKD)
Duration of diabetes≥10 years--13.5 ± 7.815.8 ± 8.611 (6–16)-12.9 ± 8.3--
Median follow-up3.1 years16 months26.2 months126.1 weeks2.62 years4.2 years3.5 years3.5 years9.0 months16 months
Primary outcome *0.86
(0.74–0.99)
0.76
(0.67–0.87)
0.79 (0.69–0.90)0.86
(0.75–0.97)
0.70
(0.59–0.82)
0.93
(0.84–1.03)
0.74
(0.65–0.85)
0.97
(0.85–1.11)
0.67 (0.52–0.85)0.74 (0.63–0.88)
Cardiovascular death0.62
(0.49–0.77)
-0.91 (0.76–1.09)0.87
(0.72–1.06)
0.78
(0.61–1.00)
0.98 (0.82–1.17)0.82
(0.69–0.98)
0.92 (0.77–1.11)0.84 (0.58–1.22)0.90 (0.73–1.12)
Nonfatal
MI
0.87
(0.70–1.09)
--0.85
(0.69–1.05)
-0.89 (0.77–1.01)-1.04
(0.86–1. 27)
--
Nonfatal stroke1.24
(0.92–1.67)
--0.90
(0.71–1.15)
-1.01 (0.84–1.21)-1.00
(0.76–1.32)
--
Death
from any cause
0.68
(0.57–0.82)
-1.00 (0.87–1.15)0.87
(0.74–1.01)
0.83
(0.68–1.02)
0.93 (0.82–1.04)0.83
(0.71–0.97)
0.93
(0.80–1.08)
0.82 (0.59–1.14)0.99 (0.83–1.18)
Hospitalization
for heart failure
0.65
(0.50–0.85)
0.70
(0.58–0.85)
0.71 (0.60–0.83)0.67
(0.52–0.87)
0.61
(0.47–0.80)
0.73 (0.61–0.88)0.70
(0.59–0.83)
0.70
(0.54–0.90)
0.64 (0.49–0.83)0.67 (0.55–0.82)
EMPA-REG OUTCOME, Empagliflozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients–Removing Excess Glucose; EMPEROR-Reduced, Empagliflozin Outcome Trial in Patients with Chronic Heart Failure with Reduced Ejection Fraction; EMPEROR-Preserved, Empagliflozin Outcome Trial in Patients with Chronic Heart Failure with Preserved Ejection Fraction; CANVAS, Canagliflozin Cardiovascular Assessment Study; CREDENCE, Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation; DECLARE–TIMI 58, Dapagliflozin Effect on Cardiovascular Events–Thrombolysis in Myocardial Infarction 58; DAPA-HF, Dapagliflozin and Prevention of Adverse Outcomes in Heart Failure; VERTIS CV, Ertugliflozin Efficacy and Safety Cardiovascular Outcomes Trial; SOLOIST-WHF, the Effect of Sotagliflozin on Cardiovascular Events in Patients with Type 2 Diabetes Post Worsening Heart Failure; SCORED, the Effect of Sotagliflozin on Cardiovascular and Renal Events in Patients with Type 2 Diabetes and Moderate Renal Impairment Who Are at Cardiovascular Risk; T2DM, type 2 diabetes mellitus; CKD, chronic kidney diseases; NYHA, New York Heart Association; CVD, cardiovascular diseases; EF, ejection fraction; MI, myocardial infarction. * Primary outcomes of RCTs: EMPA-REG OUTCOME, CANVAS, CREDENCE, DECLARE–TIMI 58, VERTIS CV: MACE (the major adverse cardiovascular events including a composite of death from cardiovascular causes, nonfatal myocardial infarction or nonfatal stroke); EMPEROR-Reduced, EMPEROR-Preserved: the composite of cardiovascular death or hospitalization for heart failure; DAPA-HF: a composite of worsening heart failure (hospitalization or an urgent visit resulting in intravenous therapy for heart failure) or cardiovascular death; SOLOIST-WHF, SCORED: the total number of deaths from cardiovascular causes and hospitalizations and urgent visits for heart failure.
Table 3. Reported real world evidence of SGLT2 inhibitors for CVD.
Table 3. Reported real world evidence of SGLT2 inhibitors for CVD.
CVD-REAL [101]Sub-Analysis of CVD-REAL [102]CVD-REAL 2 [117]Sub-Analysis of CVD-REAL 2 Study [118]Retrospective Cohort Study on PAD Patients [119]
Drugempagliflozin, canagliflozin and dapagliflozinempagliflozin, canagliflozin and dapagliflozincanagliflozin, dapagliflozin, empagliflozin, in all countries; ipragliflozin in South Korea and Japan; tofogliflozin, luseogliflozin in Japancanagliflozin,
dapagliflozin and empagliflozin in all countries apart from
South Korea; ipragliflozin in South Korea and Japan;
Tofogliflozin and luseogliflozin in Japan only
empagliflozin and dapa gliflozin
Patients309,056205,160470,128386,24822,862
RegionsEuropean and North American regionsEuropean and North American regionsthe Asia-Pacific, Middle East and North American regionsthe Asia-Pacific, Middle East, European and North American regionsthe Asia-Pacific region
Duration of diabetes≥1 years≥1 years≥1 years≥1 years-
Mean follow-up *239 days/
211 days
254 days/
232 days
374 days/
392 days
Varied by country and regions0.96 years/
0.66 years
MI-0.85 (0.72–1.00)0.81 (0.74–0.88)0.88 (0.80–0.98)0.84 (0.58–1.23)
Stroke-0.83 (0.71–0.97)0.68 (0.55–0.84)0.85 (0.77–0.93)0.81 (0.62–1.06)
Death
from any cause
0.49 (0.41–0.57)-0.51(0.37–0.70)0.59 (0.52–0.67)0.58 (0.49–0.67)
Hospitalizationfor heart failure0.61 (0.51–0.73)-0.64 (0.50–0.82)0.69 (0.61–0.77)0.66 (0.49–0.89)
CVD-REAL, The Comparative Effectiveness of Cardiovascular Outcomes in New Users of SGLT2 inhibitors; PAD, peripheral artery disease; MI, myocardial infarction.* Mean follow-up: the mean follow-up time of SGLT2 inhibitors/ other glucose lowering drugs.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Xu, J.; Hirai, T.; Koya, D.; Kitada, M. Effects of SGLT2 Inhibitors on Atherosclerosis: Lessons from Cardiovascular Clinical Outcomes in Type 2 Diabetic Patients and Basic Researches. J. Clin. Med. 2022, 11, 137. https://doi.org/10.3390/jcm11010137

AMA Style

Xu J, Hirai T, Koya D, Kitada M. Effects of SGLT2 Inhibitors on Atherosclerosis: Lessons from Cardiovascular Clinical Outcomes in Type 2 Diabetic Patients and Basic Researches. Journal of Clinical Medicine. 2022; 11(1):137. https://doi.org/10.3390/jcm11010137

Chicago/Turabian Style

Xu, Jing, Taro Hirai, Daisuke Koya, and Munehiro Kitada. 2022. "Effects of SGLT2 Inhibitors on Atherosclerosis: Lessons from Cardiovascular Clinical Outcomes in Type 2 Diabetic Patients and Basic Researches" Journal of Clinical Medicine 11, no. 1: 137. https://doi.org/10.3390/jcm11010137

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