Next Article in Journal
Preoperative Predictors of Late Aortic Expansion in Acute Type B Aortic Dissection Treated with TEVAR
Next Article in Special Issue
Canagliflozin Attenuates PromarkerD Diabetic Kidney Disease Risk Prediction Scores
Previous Article in Journal
Vitamin D, Albumin, and D-Dimer as Significant Prognostic Markers in Early Hospitalization in Patients with COVID-19
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pleiotropic Effects of Sodium-Glucose Cotransporter-2 Inhibitors in Cardiovascular Disease and Chronic Kidney Disease

by
Anjay Rastogi
1,* and
James L. Januzzi, Jr.
2,3,4
1
David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
2
Massachusetts General Hospital, Boston, MA 02114, USA
3
Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
4
Baim Institute for Clinical Research, Boston, MA 02215, USA
*
Author to whom correspondence should be addressed.
Submission received: 26 January 2023 / Revised: 20 March 2023 / Accepted: 3 April 2023 / Published: 12 April 2023

Abstract

:
Sodium-glucose cotransporter-2 inhibitors (SGLT2is) have been shown to improve cardiovascular and renal outcomes in patients with established cardiovascular disease, chronic kidney disease (CKD), and heart failure (HF) with reduced or preserved ejection fraction. Clinical benefit has been substantiated in patients with and without type 2 diabetes (T2D). Consequently, SGLT2is have an increasingly important role in HF and CKD management that extends beyond T2D treatment. Their pleiotropic pharmacological effects underlying their cardiovascular and renal benefits are not completely understood but include significant effects beyond blood glucose reduction. SGLT2is inhibit the reabsorption of glucose and sodium in the proximal tubule which, in addition to lowering blood glucose, activates tubuloglomerular feedback, leading to reduced glomerular hydrostatic pressure and the mitigation of glomerular filtration rate loss. SGLT2is have diuretic and natriuretic effects, leading to decreased blood pressure, preload, and left ventricular (LV) filling pressure, and improvements in other surrogates of afterload. In HF, SGLT2is mitigate the risks of hyperkalemia and ventricular arrhythmia and improve LV dysfunction. SGLT2is also reduce sympathetic tone and uric acid levels, increase hemoglobin levels, and are postulated to have anti-inflammatory properties. This narrative review discusses the multifactorial and interrelated pharmacological mechanisms underlying the cardiovascular and renal benefits of SGLT2is.

1. Introduction

Cardiovascular disease (CVD), chronic kidney disease (CKD), and type 2 diabetes (T2D) are chronic progressive diseases with considerable overlap. Almost half (49%) of US adults aged ≥20 years have CVD (defined as coronary heart disease, heart failure [HF], stroke, and hypertension); the prevalence of CKD and T2D among US adults is approximately 15% and 10%, respectively [1].
Pathologies of the cardiovascular and renal systems are linked, with disorders of one system frequently involving the other in a cardiorenal continuum [2]. Patients with T2D have increased risk of developing macrovascular (e.g., myocardial infarction, stroke, and peripheral artery disease) and microvascular (i.e., nephropathy, retinopathy, and neuropathy) complications that cause significant morbidity and mortality [3,4,5]. T2D is an independent risk factor for HF, and the incidence of HF is approximately fourfold higher in patients with T2D compared with that in the general population; furthermore, outcomes are worse when these conditions occur in the same patient [6]. T2D is the most common cause of CKD [7], and CKD is associated with an increased risk of cardiovascular events [8,9].
In the last decade, sodium-glucose cotransporter-2 inhibitors (SGLT2is) have been shown to provide clinical benefits across the cardiorenal spectrum. SGLT2is were originally developed and approved as glucose-lowering drugs for patients with T2D [10,11,12,13]. Subsequently, cardiovascular safety trials mandated by the US Food and Drug Administration found that SGLT2i treatment was associated with improved cardiovascular outcomes in patients with T2D [14,15,16]. These cardiovascular outcomes trials (CVOTs) and additional renal outcomes studies found that SGLT2is reduced the risk of major adverse cardiovascular events (MACEs) and renal events in patients with T2D and established atherosclerotic CVD, multiple cardiovascular risk factors, or CKD (Table 1) [14,15,17,18,19,20,21]. More recently, SGLT2is have been shown to confer benefit in patients with HF across the full spectrum of ejection fractions (EFs) [22,23,24,25,26] and in patients with CKD, regardless of their glycemic status (Table 1) [27,28,29]. On the basis of the findings from these outcome studies, drugs in this class have been approved for use in patients with established CVD or multiple risk factors [10,11], CKD [10,12], HF with reduced EF (HFrEF) [10,11], and HF with preserved EF (HFpEF) [11], including in patients without T2D [10,12], for the purpose of reducing the risk of cardiovascular death or hospitalization and preventing disease progression.
US and European treatment guidelines recommend SGLT2is to reduce the risk of HF, MACE, cardiovascular death, and progression of kidney disease in patients with T2D with established CVD or multiple risk factors for CVD [31,32,33,34,35,36] and for the treatment of HF and CKD in patients with or without T2D [34,35,36]. Although the first SGLT2i approvals for HF were for HFrEF, the updated 2022 American Heart Association (AHA)/American College of Cardiology (ACC)/Heart Failure Society of America (HFSA) guideline for the management of HF recommends the use of an SGLT2i across the full spectrum of HF, including HFrEF (defined as left ventricular EF [LVEF] ≤ 40%), HF with mildly reduced EF (LVEF 41–49%), and HFpEF (LVEF ≥50%), irrespective of the presence of T2D [36].
SGLT2is have pleiotropic pharmacological effects that include those related to their glucose-lowering effects and those that are independent of glucose lowering effects. Some pharmacological mechanisms, such as the effects on sodium-glucose cotransporter-2 (SGLT2), are well understood and linked to evidence in clinical outcome studies. Other mechanisms are putative, based on small clinical studies or preclinical data, and their relationship to clinical outcomes is less certain. In particular, the mechanistic links between SGLT2is and the clinical benefits observed in large CVOTs have not been fully elucidated. The aim of this narrative review is to provide an overview of the glucose-independent mechanisms that may contribute to the cardiovascular and renal protective effects of SGLT2is irrespective of a patient’s glycemic status.

2. Materials and Methods

A targeted literature search of PubMed was conducted to identify relevant English-language publications using the following search terms: (“SGLT2 inhibitors”, “SGLT-2i”, “SGLT2i”, “sodium-glucose cotransporter 2 inhibitors”, “sodium-glucose cotransporter-2 inhibitors”, “canagliflozin”, “dapagliflozin”, “empagliflozin”, “ertugliflozin”, “empagliflozin”, “sotagliflozin”) AND (“heart failure”, “chronic kidney disease”, “type 2 diabetes mellitus”, “non-insulin dependent diabetes”) AND (“mechanism of action”, “glomerular pressure”, “sympathetic nervous system”, “hematocrit”, “uric acid”, “electrolytes”, “preload/afterload”). English-language papers published between July 2015 and December 2022 reporting clinical studies and meta-analyses of SGLT2is were identified. Additional relevant studies were identified from the bibliographies of these publications.

3. SGLT2i Mechanism of Action

3.1. Inhibition of SGLT2 and Glucose-Lowering Effects

Under normal physiological conditions, the kidneys contribute to glucose homeostasis by reabsorbing almost all of the glucose in the glomerular filtrate with a negligible amount excreted in urine [37]. This is achieved primarily by SGLT2 (encoded by SLC5A2), which is expressed almost exclusively in the kidney, whereas sodium-glucose cotransporter-1 (SLC5A1) is expressed mainly in the small intestine and to a small extent in the kidney [38]. SGLT2 is a membrane transport protein located exclusively in the apical membrane of epithelial cells in the proximal tubule that facilitates reabsorption of approximately 90% of filtered glucose coupled with sodium transport in a 1:1 ratio. This passive process involves an electrochemical gradient—sodium ions flow from a region of high concentration in the glomerular filtrate to a region of low concentration in tubular epithelial cells—maintained by the basolateral Na+/K+ ATPase pump [39]. After being reabsorbed from the glomerular filtrate, glucose is transported from the tubular epithelial cells to the interstitial fluid via glucose transporter 2, the main glucose transporter in the kidney, which is located in the basolateral membrane [40,41].
Inhibition of SGLT2 blocks reabsorption and increases renal excretion of glucose [42]. This results in reductions in plasma glucose levels in patients with T2D that are reflected by reductions in glycated hemoglobin (A1C) in the order of ~1% [43,44,45,46]. This effect is markedly reduced as the glomerular filtration rate (GFR) decreases to 45 mL/min/1.73 m2 and is negligible in patients with a GFR of 30 mL/min/1.73 m2 or lower [47,48,49]. Inhibition of renal glucose reabsorption and reductions in plasma glucose levels achieved with SGLT2is occur independent of insulin levels or peripheral insulin resistance; thus, the risk of hypoglycemia is low with SGLT2i therapy [5,50]. By promoting glucosuria, treatment with an SGLT2i is associated with a daily caloric loss of approximately 200 kcal/day and weight loss of approximately 2–3 kg over 6 months [51,52].

3.2. Reduction in Intraglomerular Pressure and Preservation of Renal Function

The cardiorenal effects of SGLT2is are not the result of glucose-lowering activity alone, because they are observed sooner than would be expected for a glucose-mediated mechanism of action, are sustained with continued use, and are independent of T2D status. Moreover, cardiorenal benefits are not observed with most classes of anti-hyperglycemic drugs [53]. This review focuses on the mechanisms for which clinical data are available (Figure 1).
Glomerular filtration is regulated, in part, by tubuloglomerular feedback. Increasing sodium chloride concentrations in the distal tubule cause a release of adenosine from cells in the macula densa, which results in constriction of afferent (precapillary) glomerular arterioles and a reduction in the GFR. The expression and activity of SGLT2 are increased in patients with T2D [54], resulting in increased renal reabsorption of glucose and sodium in these individuals. Increased reabsorption of sodium in the proximal tubule reduces the concentration of sodium chloride in the macula densa and leads to the deactivation of tubuloglomerular feedback and dilatation of afferent arterioles [55,56]. Concurrent activation of the renin–angiotensin–aldosterone system (RAAS) induces constriction of efferent (postcapillary) glomerular arterioles, and together these changes cause persistent increases in single-nephron GFR, glomerular hyperfiltration, and glomerular hypertension [56]. Glomerular hyperfiltration is a common cause of kidney injury and leads to progressive reduction of kidney function [57].
SGLT2i therapy mitigates these effects by blocking the reabsorption of glucose and sodium in the proximal tubule. Solute delivery to the macula densa is restored and tubuloglomerular feedback is activated, which results in constriction of afferent arterioles, reduced intraglomerular pressure, and decreased hyperfiltration (Figure 2) [37,57,58,59]. Reduced glomerular hydrostatic pressure preserves renal function in the long term [59].
Clinically, the restoration of tubuloglomerular feedback manifests as an acute decrease in the estimated glomerular filtration rate (eGFR) of ~5 mL/min/1.73 m2 during the first 2–6 weeks of SGLT2i treatment [30,60,61,62,63,64,65,66,67]. After this initial decline, the eGFR recovers and returns to baseline, indicating that the initial drop is the result of altered hemodynamics rather than glomerular damage [53,62,63,64]. This initial eGFR decrease was not found to be associated with greater long-term decreases in eGFR [66,67]. SGLT2is confer renoprotective effects irrespective of baseline eGFR and CKD stage [30,68], and long-term SGLT2i therapy preserves kidney function in patients with and without T2D [60,69]. In DECLARE-TIMI 58, treatment with dapagliflozin significantly reduced the progression of kidney disease relative to a placebo in patients with T2D. Over a median of 4.2 years of follow-up, significantly fewer dapagliflozin-treated versus placebo-treated patients experienced a sustained decrease in eGFR, defined as a 40% decrease from baseline to <60 mL/min/1.73 m2 or progression to end-stage kidney disease [69]. Therefore, a transient decrease in eGFR after treatment initiation should not prompt interruption or discontinuation of SGLT2i therapy [34]. Rather, the data suggest that treatment with an SGLT2i prevents CKD in patients with T2D.
Data from EMPA-REG OUTCOME [14,63], DECLARE-TIMI 58 [18,69], DELIVER [25,30], VERTIS-CV [19,62], and CREDENCE [28,64] suggest that the early eGFR decrease should not be cause for concern unless there are signs and symptoms of volume depletion (orthostatic hypotension, blood pressure [BP] <120/70 mmHg), specifically in older patients (>65–70 years) or in those receiving high-dose diuretics [70].
SGLT2i treatment significantly reduces albumin excretion in patients with T2D and microalbuminuria (urinary albumin-to-creatinine ratio [UACR] of 30–300 mg/g) or macroalbuminuria (UACR >300 mg/g), including patients receiving drugs that block the RAAS [71,72,73]. A prespecified analysis of the DAPA-CKD study showed that significant reductions in UACR occurred in patients with and without T2D (with larger reductions in patients with T2D) [74]. Similarly, an exploratory analysis of DECLARE-TIMI 58 showed that dapagliflozin therapy slowed the increase in UACR over 4 years, regardless of baseline eGFR and UACR, including in non-albuminuric patients [75]. The positive effect on albumin excretion is reversed if SGLT2i therapy is discontinued, which indicates that it is the result of altered renal hemodynamics [71,72,73]. SGLT2is have also been shown to slow the long-term decline in eGFR in patients with T2D and microalbuminuria or macroalbuminuria [76].
Acute kidney injury (AKI) has been reported in the post-marketing surveillance of patients taking SGLT2is, although a causal relationship has not been established [10,11,12,13]. It is possible that SGLT2i-associated AKI is related to volume depletion, an uncommon adverse event in patients taking SGLT2is. Consistent with this hypothesis, the risk of AKI was low in patients receiving an SGLT2i in CVOTs [15,17,18,27,28]. Moreover, a meta-analysis of data from CVOTs showed a 34% reduction in the risk of AKI during SGLT2i therapy, relative to placebo [77]. Similarly, prespecified analyses of DAPA-CKD [78] and DAPA-HF [79] showed that dapagliflozin reduced the risk of abrupt declines in kidney function, defined as a doubling of serum creatinine by 32% [78] and 44% [79], relative to placebo.

3.3. Reduction in Preload/Afterload and Effects on BP

SGLT2i therapy has diuretic and natriuretic effects in patients with T2D. This results in an approximate 1–2 kg decrease in total body water, a 6% loss in sodium, and a decrease in plasma volume during the first few weeks of treatment [80,81,82]. However, these effects are transient and stabilize subsequently. The contraction in plasma volume decreases cardiac preload [59] and left ventricular (LV) filling pressure, leading to reductions in myocardial stretch and interstitial fibrosis [45].
Plasma volume contraction is associated with sustained reductions in systolic/diastolic BP (SBP/DBP) of approximately 5/2 mmHg without a compensatory increase in heart rate and no evidence of orthostatic hypotension [80,83,84,85]. These improvements in BP are amplified by the SGLT2i-associated reduction in body weight [80,83,84].
The diuretic effects of SGLT2is differ from those of thiazide or loop diuretics. SGLT2is do not affect plasma osmolality or reduce intravascular volume. Despite having less potent natriuretic and diuretic effects than a loop diuretic, dapagliflozin produced greater reductions in interstitial fluid volume than in blood volume [86]. This may explain why an SGLT2i may provide better control of congestion without reducing arterial filling and perfusion in patients with HF. From a clinical perspective, better control of congestion by SGLT2is may result in the lower hospitalization rates for HF observed in numerous CVOTs [14,15,18,19,21,28]. Importantly, unlike loop diuretics, the natriuretic effects of SGLT2i therapy do not result in activation of the RAAS [59]. Furthermore, reductions in BP did not affect natriuresis in studies with dapagliflozin and empagliflozin in patients with T2D and preserved kidney function [87,88]. The available data suggest the cardiovascular and renal protective effects of SGLT2is are mediated by factors other than natriuresis and volume changes and are independent of changes in mean SBP [45,53,87,88].
In patients with HFrEF, the natriuretic effects of SGLT2is are synergistic with loop diuretics and allow for reductions in the dose of loop diuretics [89,90]. For example, a significantly higher proportion of patients had their diuretic dose reduced after 6 months and 12 months of treatment with dapagliflozin in DAPA-HF [90].
SGLT2i treatment decreases sympathetic nervous system (SNS) activity (discussed later in this review), which is accompanied by reductions in central SBP, pulse pressure, mean arterial pressure (MAP), the double product (SBP multiplied by heart rate), and aortic pulse wave velocity, which are indications of reduced arterial stiffness and surrogates of afterload [91,92,93]. In addition, SGLT2i–mediated vasodilation via activation of voltage-gated potassium channels and protein kinase G may result in improved subendocardial blood flow [41,94]. A post hoc analysis of a large randomized controlled trial demonstrated favorable effects of empagliflozin on pulse pressure, MAP, and the double product after a median of 3.1 years of treatment [95].

3.4. Effects on Potassium Levels

Hyperkalemia is common in patients with HF, is associated with specific risk factors including comorbidities such as CKD and T2D and drugs such as mineralocorticoid antagonists and RAAS inhibitors [96], and increases the risk of cardiovascular-related morbidity and mortality [97]. Treatment with an SGLT2i reduced the risks of moderate hyperkalemia in patients with T2D or CKD [98] and in patients with HFrEF and HFpEF, including patients receiving mineralocorticoid antagonists, in major CVOTs [99,100,101,102]. Importantly, SGLT2i use did not increase the incidence of hypokalemia in patients with T2D or CKD receiving loop or thiazide diuretics [90,102]. It is not clear whether dose modifications of mineralocorticoid antagonists, RAAS inhibitors, or diuretics contributed to the observed differences in hyperkalemia incidence. In a meta-analysis of data from six cardiovascular and renal outcome trials, the beneficial effects of SGLT2i therapy on the incidence of serious hyperkalemia were significantly different in patients who were and were not receiving RAAS inhibitors at baseline; however, treatment with an SGLT2i as compared with a placebo was associated with significantly lower risk of serious hyperkalemia in those who were and were not receiving RAAS inhibitors at baseline [98].

3.5. Effects on LV Size and Function

In patients with T2D and a history of CVD, SGLT2is reduce LV mass index and end-diastolic volume [45,103]. In patients with HFrEF with and without T2D, empagliflozin significantly reduced LV and atrial volumes [104,105,106]. In patients with HFpEF, dapagliflozin significantly improved LV diastolic functional parameters and global longitudinal strain [107]. Canagliflozin has also been shown to improve LV parameters in patients with HFpEF [108].

3.6. Risk of Ventricular Arrhythmias

Ventricular arrhythmias are common among patients with HFrEF, resulting from chamber dilatation and cardiomyocyte stretch, and may lead to sudden death. A post hoc analysis of the DAPA-HF study, which included patients with HFrEF both with and without T2D, showed that the addition of dapagliflozin to standard therapy for HF was associated with a 21% reduction in the risk of any ventricular arrhythmia, resuscitated cardiac arrest, or sudden death relative to the placebo plus standard therapy [109]. The reductions in cardiovascular outcomes were associated with reduced cardiac chamber size and decreased N-terminal prohormone of brain natriuretic peptide levels, which suggests that SGLT2i therapy decreases wall stress [109].

3.7. Reduction in SNS Activation

Chronic activation of the SNS is prevalent in patients with T2D, ischemic heart disease, HF, hypertension, and CKD and results in fluid retention, endothelial dysfunction, and increased arterial stiffness [110]; chronic SNS activation is associated with poor prognosis [111,112,113,114,115]. The SNS is regulated, in part, by the kidneys, as renal stress results in increased signaling to the brain, stimulating the sympathetic center and enhancing sympathetic outflow [110]. It has been proposed that transient osmotic diuresis induced by SGLT2is leads to a decrease in sympathetic outflow to the kidneys and results in a correction of systemic fluid retention in patients with T2D [110].
There is evidence that SGLT2is inhibit SNS activation by suppressing renal afferent signaling to the brain and central reflex mechanisms [110]. Dapagliflozin inhibited SNS activity in a neurogenic hypertensive mouse model as indicated by reduced tyrosine hydroxylase staining in renal tissue and norepinephrine levels in the kidney [116], and empagliflozin reduced renal sympathetic nerve overactivity in a rabbit model of diabetes [117]. Luseogliflozin reduced resting heart rate in patients with T2D and a baseline heart rate >70 beats per minute [118]. Empagliflozin significantly reduced the low:high frequency ratio in heart rate variability in patients with T2D and acute myocardial infarction [119]. Dapagliflozin significantly decreased muscle sympathetic nerve activity (MSNA) correlated with a decrease in brain natriuretic peptide in a small study in patients with T2D with and without HF [120]. The reduction in MSNA was more marked in the patients with HF, which suggests that the cardioprotective effects of SGLT2is are due in part to improved sympathetic nerve activity [120].

3.8. Effects on Hematologic Parameters

Treatment with an SGLT2i is associated with improvements in hemoglobin, hematocrit, and erythropoietin levels. For example, in a short-term study in patients with T2D, serum erythropoietin levels and reticulocyte counts increased over the first 4 weeks of treatment with dapagliflozin, whereas increases in hematocrit, hemoglobin, and red cell mass were sustained over 12 weeks and were all significant when compared with those with a placebo or hydrochlorothiazide [80].
Significantly higher hematocrit and hemoglobin levels were observed over a median of 2.6 years of follow-up in patients with T2D and CKD in CREDENCE [121], over 206 weeks of follow-up in patients with T2D and CVD in EMPA-REG OUTCOME [122], over 260 weeks of follow-up in patients with T2D and CVD in VERTIS CV [123,124], and over 12 to 48 months of follow-up in patients with T2D and CVD in DECLARE-TIMI-58 [125]. Moreover, the risk of anemia, initiation of treatment with an iron preparation, or a requirement for an erythropoiesis-stimulating agent were all significantly lower in recipients of canagliflozin than in recipients of placebo in CREDENCE [121].
In patients with HFrEF enrolled in DAPA-HF, dapagliflozin treatment was associated with increases in hematocrit that plateaued after 4 months and were sustained for up to 2 years, regardless of T2D status [126], diuretic use, or baseline diuretic dose [90]. The increase in hematocrit persisted at 6 and 12 months regardless of whether the diuretic dose was increased, decreased, or remained unchanged [90].
The mechanism underlying these improvements has not been fully elucidated; however, given the weak diuretic effect of SGLT2is and the sustained nature of these hematologic changes, it seems likely that factors in addition to hemoconcentration are involved [80,90,122,125,127].
There is evidence that SGLT2is increase erythropoiesis by modulating iron regulatory proteins [128,129]. For example, in patients with T2D, dapagliflozin significantly reduced levels of hepcidin, which is a known suppressor of erythropoiesis, and increased levels of erythroferrone, which is a hepcidin inhibitor [128]. In addition, SGLT2is may improve the hypoxic microenvironment of the proximal tubule by alleviating metabolic stress, normalizing renal cortical oxygenation, and improving the tubulointerstitial function [127].
Improving hematologic status enhances the delivery of oxygen to tissues and may in part explain reductions in cardiovascular death in patients treated with SGLT2is [122,130,131].

3.9. Reduction in Uric Acid Levels

Hyperuricemia increases oxidative stress, impairs endothelial function, activates the RAAS, and is associated with an increased risk of hypertension, CVD, and CKD. Treatment with an SGLT2i is associated with sustained reductions in serum uric acid levels in patients with T2D [132,133,134] or HFrEF [135]. In patients with T2D, the magnitude of the reduction in serum uric acid levels decreased with increasing duration of disease, higher A1C levels, or decreasing eGFR [133]. Moreover, the uric acid lowering effect of SGLT2i therapy was absent in patients with an eGFR <60 mL/min/1.73 m2 [133].
Uric acid is not transported by SGLT2 [136], and SGLT2is do not interact with transporters involved in renal uric acid reabsorption (i.e., uric acid transporter 1, organic anion transporters 4 and 10, glucose transporter 9, and sodium-coupled monocarboxylate transporter) [136]. Thus, the uric acid-lowering effects of SGLT2is are thought to be mediated by the urinary excretion of uric acid secondary to glucosuria [136].
In patients with HFrEF enrolled in EMPEROR-HF, treatment with an SGLT2i was associated with more rapid reductions in serum uric acid levels than those with placebo treatment, which were reflected in a lower incidence of hyperuricemic events (acute gout, gouty arthritis, and initiation of anti-gout therapy) [137]. Moreover, the significant beneficial effects of empagliflozin therapy on the primary composite endpoint were not influenced by baseline serum uric acid levels [137].

3.10. Effects on Intracellular Sodium

Modulation of intracellular sodium through inhibition of the sodium–hydrogen exchanger (NHE) in both the kidneys and the myocardium may contribute to the cardiovascular and renal benefits of SGLT2i therapy [138,139]. Patients with T2D and HF have increased NHE activity that may contribute to resistance to diuretics and endogenous natriuretic peptides [139]. Reductions in cardiac injury, hypertrophy, fibrosis, remodeling, and systolic dysfunction observed with empagliflozin have been suggested to result from inhibition of the NHE [138]. In support of this hypothesis, empagliflozin has been shown to reduce intracellular sodium and calcium concentrations in isolated rabbit cardiomyocytes; empagliflozin, dapagliflozin, and canagliflozin reduced intracellular sodium in mouse cardiomyocytes, presumably through direct action on the NHE [140,141]; and empagliflozin reduced intracellular sodium in human umbilical vein endothelial cells and human coronary artery endothelial cells [142]. In contrast, empagliflozin and other SGLT2i agents did not inhibit the NHE and had no effect on intracellular sodium concentrations in isolated rat ventricular cardiomyocytes [143].

3.11. Anti-Inflammatory, Antiplatelet, and Antioxidant Effects

Cardiovascular and renal complications in patients with T2D are significantly driven by chronic low-grade inflammation mediated by the NOD-like receptor pyrin domain-containing protein 3 inflammasome and oxidative stress [144]. SGLT2is may have anti-inflammatory effects that reverse fibrosis, slow CKD progression, and provide cardioprotective effects [145,146]. Serum inflammatory markers influenced by SGLT2is are listed in Table 2 [145,146].
SGLT2is reduce inflammation and oxidative stress in vitro and in animal models [147,148,149]. The anti-inflammatory effects may result, in part, from inhibition of the NHE and attenuation of the sodium levels in human endothelial cells [142]. In patients with concomitant T2D and coronary artery disease, treatment with 10 mg/day of empagliflozin for 6 months mitigated inflammation, platelet reactivity, and oxidative stress and was associated with glycemic improvement in the randomized, double-blind, placebo-controlled EMPA-CARD study [150]. Improvements included significant reductions in markers of inflammation (interleukin-6, interleukin-1β, and high-sensitivity C-reactive protein), platelet reactivity (expression of P-selectin), and oxidative burden (reactive oxygen species) and increases in antioxidant levels (superoxide dismutase, glutathione, and total antioxidant capacity).
Short-term treatment with an SGLT2i significantly decreased platelet reactivity in patients with stable coronary artery disease and T2D who were receiving dual antiplatelet therapy (aspirin plus clopidogrel) in the open-label EFFECT (empagliflozin) [151] and EDGE (dapagliflozin) [152] pilot studies.

3.12. Effects on Lipolysis and Ketone bodies

It is hypothesized that alterations in lipolysis and ketone metabolism and the renal handling of ketone bodies may contribute to the cardio- and reno-protective effects of SGLT2is [153]. SGLT2i-induced glycosuria results in a net energy loss, thereby producing a fasting-like metabolic environment. The utilization of glucose decreases, whereas the utilization of free fatty acids (FFA), ketone bodies, and branched chain amino acids (BCAAs) increases. Elevated ketone body levels represent an additional energy source that contributes to improved performance of the heart and kidney. Empagliflozin has been shown to increase circulatory FFA, blood ketones, and BCAAs in patients with T2D [154,155,156]. In addition, experiments in animal models suggest that the SGLT2i-induced shift to a fasting-like metabolic response attenuates the mechanistic target of the rapamycin (mTOR) pathway and improves mitochondrial function [153].
The fasting-like metabolic environment and production of ketone bodies may also contribute to rare cases of euglycemic diabetic ketoacidosis during treatment with SGLT2is. The incidence of this complication was similar or lower in patients treated with an SGLT2i than that in patients receiving a placebo in several cardiovascular and renal outcome trials, including EMPEROR-Reduced [24], EMPEROR-Preserved [22], DAPA-HF [23], and DAPA CKD [27], and higher in patients treated with an SGLT2i than that in those treated with a placebo in other trials, including EMPA-KIDNEY [29], CREDENCE [28], and SCORED [20]. Given the potential seriousness of this condition, clinicians should be aware of the possibility that patients may present with diabetic ketoacidosis during treatment with an SGLT2i [157].

4. Conclusions

In patients with and without T2D, SGLT2is confer cardiovascular and renal benefits that result from complex pharmacological mechanisms. These include effects that are independent of their blood glucose-lowering activity. Enhanced tubuloglomerular feedback plays a central role in protecting the kidney and attenuating the decline in GFR and progression of albuminuria. The link between inhibition of SGLT2 in the kidney and reductions in clinical outcomes (hospitalizations, progression of renal disease, mortality) is multifactorial and involves diverse elements, including the inhibition of central SNS outflow and improved hemodynamics. Although initially developed as antihyperglycemic agents, SGLT2is play an increasingly important role in the management of HF and CKD that extends beyond the treatment of T2D.

Author Contributions

A.R. and J.L.J.J. have made a substantial contribution to the conception and design of this review article and interpretation of the relevant literature. Both authors have provided critical reviews of the manuscript during its development and approved the final version of the manuscript for submission. All authors have read and agreed to the published version of the manuscript.

Funding

The development of the manuscript was supported by AstraZeneca.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Mitali Choudhury, of inScience Communications, Springer Healthcare, and Blair Jarvis, on behalf of inScience Communications, Springer Healthcare, provided medical writing support funded by AstraZeneca.

Conflicts of Interest

Anjay Rastogi has received research grants and consulting fees from Amgen, AstraZeneca, Bayer, CSL Vifor, Gilead, GlaxoSmithKline, Janssen, Otsuka, Pfizer, and Sanofi. James L. Januzzi, Jr. is a Trustee of the American College of Cardiology and a board member of Imbria Pharmaceuticals; has received research support from Abbott, Applied Therapeutics, Innolife, Novartis Pharmaceuticals, and Roche Diagnostics; has received consulting income from Abbott, AstraZeneca, Beckman, Boehringer-Ingelheim, Bristol Myers Squibb, Janssen, Novartis Pharmaceuticals, Pfizer, Merck, Roche Diagnostics, and Siemens; and participates in clinical endpoint committee/data safety monitoring boards for Abbott, AbbVie, Bayer, CVRx, Intercept, Janssen, and Takeda.

References

  1. Tsao, C.W.; Aday, A.W.; Almarzooq, Z.I.; Alonso, A.; Beaton, A.Z.; Bittencourt, M.S.; Boehme, A.K.; Buxton, A.E.; Carson, A.P.; Commodore-Mensah, Y.; et al. Heart Disease and Stroke Statistics-2022 Update: A Report from the American Heart Association. Circulation 2022, 145, e153–e639. [Google Scholar] [CrossRef]
  2. Fontes-Carvalho, R.; Santos-Ferreira, D.; Raz, I.; Marx, N.; Ruschitzka, F.; Cosentino, F. Protective Effects of SGLT-2 Inhibitors Across the Cardiorenal Continuum: Two Faces of the Same Coin. Eur. J. Prev. Cardiol. 2022, 29, 1352–1360. [Google Scholar] [CrossRef] [PubMed]
  3. The Emerging Risk Factors Collaboration. Diabetes Mellitus, Fasting Blood Glucose Concentration, and Risk of Vascular Disease: A Collaborative Meta-Analysis of 102 Prospective Studies. Lancet 2010, 375, 2215–2222. [Google Scholar] [CrossRef] [Green Version]
  4. Rydén, L.; Grant, P.J.; Anker, S.D.; Berne, C.; Cosentino, F.; Danchin, N.; Deaton, C.; Escaned, J.; Hammes, H.-P.; Huikuri, H.; et al. ESC Guidelines on Diabetes, pre-Diabetes, and Cardiovascular Diseases Developed in Collaboration with the EASD: The Task Force on Diabetes, pre-Diabetes, and Cardiovascular Diseases of the European Society of Cardiology (ESC) and Developed in Collaboration with the European Association for the Study of Diabetes (EASD). Eur. Heart J. 2013, 34, 3035–3087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Seufert, J. SGLT2 Inhibitors—An Insulin-Independent Therapeutic Approach for Treatment of Type 2 Diabetes: Focus on Canagliflozin. Diabetes Metab. Syndr. Obes. 2015, 8, 543–554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Dunlay, S.M.; Givertz, M.M.; Aguilar, D.; Allen, L.A.; Chan, M.; Desai, A.S.; Deswal, A.; Dickson, V.V.; Kosiborod, M.N.; Lekavich, C.L.; et al. Type 2 Diabetes Mellitus and Heart Failure: A Scientific Statement from the American Heart Association and the Heart Failure Society of America: This statement does not represent an update of the 2017 ACC/AHA/HFSA heart failure guideline update. Circulation 2019, 140, e294–e324. [Google Scholar] [CrossRef]
  7. Jha, V.; Garcia-Garcia, G.; Iseki, K.; Li, Z.; Naicker, S.; Plattner, B.; Saran, R.; Wang, A.Y.-M.; Yang, C.W. Chronic Kidney Disease: Global Dimension and Perspectives. Lancet 2013, 382, 260–272. [Google Scholar] [CrossRef]
  8. Matsushita, K.; van der Velde, M.; Astor, B.C.; Woodward, M.; Levey, A.S.; de Jong, P.E.; Coresh, J.; Gansevoort, R.T. Association of Estimated Glomerular Filtration Rate and Albuminuria with All-Cause and Cardiovascular Mortality in General Population Cohorts: A Collaborative Meta-Analysis. Lancet 2010, 375, 2073–2081. [Google Scholar] [CrossRef] [Green Version]
  9. Scirica, B.M.; Mosenzon, O.; Bhatt, D.L.; Udell, J.A.; Steg, P.G.; McGuire, D.K.; Im, K.; Kanevsky, E.; Stahre, C.; Sjöstrand, M.; et al. Cardiovascular Outcomes According to Urinary Albumin and Kidney Disease in Patients with Type 2 Diabetes at High Cardiovascular Risk: Observations from the SAVOR-TIMI 53 Trial. JAMA Cardiol. 2018, 3, 155–163. [Google Scholar] [CrossRef] [Green Version]
  10. AstraZeneca. Farxiga® (Dapagliflozin) Tablets, for Oral Use [Prescribing Information]. Available online: https://den8dhaj6zs0e.cloudfront.net/50fd68b9-106b-4550-b5d0-12b045f8b184/0be9cb1b-3b33-41c7-bfc2-04c9f718e442/0be9cb1b-3b33-41c7-bfc2-04c9f718e442_viewable_rendition__v.pdf (accessed on 28 February 2023).
  11. Boehringer Ingelheim. Jardiance® (Empagliflozin) Tablets, for Oral Use [Prescribing Information]. Available online: https://docs.boehringer-ingelheim.com/Prescribing%20Information/PIs/Jardiance/jardiance.pdf (accessed on 28 March 2022).
  12. Janssen Pharmaceuticals. Invokana (Canagliflozin) Tablets, for Oral Use [Prescribing Information]. Available online: https://www.janssenlabels.com/package-insert/product-monograph/prescribing-information/INVOKANA-pi.pdf (accessed on 29 March 2022).
  13. Merck & Co. Steglatro™ (Ertugliflozin) Tablets, for Oral Use [Prescribing Information]. Available online: https://www.merck.com/product/usa/pi_circulars/s/steglatro/steglatro_pi.pdf (accessed on 28 March 2022).
  14. 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] [Green Version]
  15. 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]
  16. Cefalu, W.T.; Kaul, S.; Gerstein, H.C.; Holman, R.R.; Zinman, B.; Skyler, J.S.; Green, J.B.; Buse, J.B.; Inzucchi, S.E.; Leiter, L.A.; et al. Cardiovascular Outcomes Trials in Type 2 Diabetes: Where Do We Go from Here? Reflections from a Diabetes Care Editors’ Expert Forum. Diabetes Care 2018, 41, 14–31. [Google Scholar] [CrossRef] [Green Version]
  17. 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]
  18. 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]
  19. 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] [PubMed]
  20. 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]
  21. 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]
  22. 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]
  23. 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] [Green Version]
  24. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Pocock, S.J.; Carson, P.; Januzzi, J.; Verma, S.; Tsutsui, H.; Brueckmann, M.; et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med. 2020, 383, 1413–1424. [Google Scholar] [CrossRef]
  25. Solomon, S.D.; McMurray, J.J.V.; Claggett, B.; de Boer, R.A.; DeMets, D.; Hernandez, A.F.; Inzucchi, S.E.; Kosiborod, M.N.; Lam, C.S.P.; Martinez, F.; et al. Dapagliflozin in Heart Failure with Mildly Reduced or Preserved Ejection Fraction. N. Engl. J. Med. 2022, 387, 1089–1098. [Google Scholar] [CrossRef]
  26. Jhund, P.S.; Kondo, T.; Butt, J.H.; Docherty, K.F.; Claggett, B.L.; Desai, A.S.; Vaduganathan, M.; Gasparyan, S.B.; Bengtsson, O.; Lindholm, D.; et al. Dapagliflozin Across the Range of Ejection Fraction in Patients with Heart Failure: A Patient-Level, Pooled Meta-Analysis of DAPA-HF and DELIVER. Nat. Med. 2022, 28, 1956–1964. [Google Scholar] [CrossRef] [PubMed]
  27. Heerspink, H.J.L.; Stefánsson, B.V.; Correa-Rotter, R.; Chertow, G.M.; Greene, T.; Hou, F.F.; Mann, J.F.E.; McMurray, J.J.V.; Lindberg, M.; Rossing, P.; et al. Dapagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2020, 383, 1436–1446. [Google Scholar] [CrossRef] [PubMed]
  28. 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] [Green Version]
  29. The EMPA-KIDNEY Collaborative Group; Herrington, W.G.; Staplin, N.; Wanner, C.; Green, J.B.; Hauske, S.J.; Emberson, J.R.; Preiss, D.; Judge, P.; Mayne, K.J.; et al. Empagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2023, 388, 117–127. [Google Scholar] [CrossRef]
  30. Mc Causland, F.R.; Claggett, B.L.; Vaduganathan, M.; Desai, A.S.; Jhund, P.; de Boer, R.A.; Docherty, K.; Fang, J.; Hernandez, A.F.; Inzucchi, S.E.; et al. Dapagliflozin and Kidney Outcomes in Patients with Heart Failure with Mildly Reduced or Preserved Ejection Fraction: A Prespecified Analysis of the DELIVER Randomized Clinical Trial. JAMA Cardiol. 2023, 8, 56–65. [Google Scholar] [CrossRef]
  31. Arnett, D.K.; Blumenthal, R.S.; Albert, M.A.; Buroker, A.B.; Goldberger, Z.D.; Hahn, E.J.; Himmelfarb, C.D.; Khera, A.; Lloyd-Jones, D.; McEvoy, J.W.; et al. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J. Am. Coll. Cardiol. 2019, 74, 1376–1414. [Google Scholar] [CrossRef]
  32. Buse, J.B.; Wexler, D.J.; Tsapas, A.; Rossing, P.; Mingrone, G.; Mathieu, C.; D’Alessio, D.A.; Davies, M.J. 2019 Update to: Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 2020, 43, 487–493. [Google Scholar] [CrossRef] [Green Version]
  33. Cosentino, F.; Grant, P.J.; Aboyans, V.; Bailey, C.J.; Ceriello, A.; Delgado, V.; Federici, M.; Filippatos, G.; Grobbee, D.E.; Hansen, T.B.; et al. 2019 ESC Guidelines on Diabetes, pre-Diabetes, and Cardiovascular Diseases Developed in Collaboration with the EASD. Eur. Heart J. 2020, 41, 255–323. [Google Scholar] [CrossRef] [Green Version]
  34. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Böhm, M.; Burri, H.; Butler, J.; Čelutkienė, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the Diagnosis and Treatment of Acute and Chronic Heart Failure. Eur. Heart J. 2021, 42, 3599–3726. [Google Scholar] [CrossRef] [PubMed]
  35. UK Kidney Association. UK Kidney Association Clinical Practice Guideline: Sodium-Glucose Co-Transporter-2 (SGLT-2) Inhibition in Adults with Kidney Disease. Available online: https://ukkidney.org/sites/renal.org/files/UKKA%20guideline_SGLT2i%20in%20adults%20with%20kidney%20disease%20v1%2020.10.21.pdf (accessed on 26 March 2022).
  36. Heidenreich, P.A.; Bozkurt, B.; Aguilar, D.; Allen, L.A.; Byun, J.J.; Colvin, M.M.; Deswal, A.; Drazner, M.H.; Dunlay, S.M.; Evers, L.R.; et al. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation 2022, 145, e895–e1032. [Google Scholar] [CrossRef]
  37. Wanner, C. EMPA-REG OUTCOME: The Nephrologist’s Point of View. Am. J. Cardiol. 2017, 120, S59–S67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Kshirsagar, R.P.; Kulkarni, A.A.; Chouthe, R.S.; Pathan, S.K.; Une, H.D.; Reddy, G.B.; Diwan, P.V.; Ansari, S.A.; Sangshetti, J.N. SGLT Inhibitors as Antidiabetic Agents: A Comprehensive Review. RSC Adv. 2020, 10, 1733–1756. [Google Scholar] [CrossRef] [Green Version]
  39. Zelniker, T.A.; Braunwald, E. Mechanisms of Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2020, 75, 422–434. [Google Scholar] [CrossRef] [PubMed]
  40. Shepard, B.D.; Pluznick, J.L. Saving the Sweetness: Renal Glucose Handling in Health and Disease. Am. J. Physiol. Renal. Physiol. 2017, 313, F55–F61. [Google Scholar] [CrossRef] [Green Version]
  41. Lytvyn, Y.; Bjornstad, P.; Udell, J.A.; Lovshin, J.A.; Cherney, D.Z.I. Sodium Glucose Cotransporter-2 Inhibition in Heart Failure: Potential Mechanisms, Clinical Applications, and Summary of Clinical Trials. Circulation 2017, 136, 1643–1658. [Google Scholar] [CrossRef]
  42. Wilcox, T.; De Block, C.; Schwartzbard, A.Z.; Newman, J.D. Diabetic Agents, from Metformin to SGLT2 Inhibitors and GLP1 Receptor Agonists: JACC Focus Seminar. J. Am. Coll. Cardiol. 2020, 75, 1956–1974. [Google Scholar] [CrossRef] [PubMed]
  43. Sun, Y.-N.; Zhou, Y.; Chen, X.; Che, W.-S.; Leung, S.-W. The Efficacy of Dapagliflozin Combined with Hypoglycaemic Drugs in Treating Type 2 Diabetes Mellitus: Meta-Analysis of Randomised Controlled Trials. BMJ Open 2014, 4, e004619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Wang, Z.; Sun, J.; Han, R.; Fan, D.; Dong, X.; Luan, Z.; Xiang, R.; Zhao, M.; Yang, J. Efficacy and Safety of Sodium-Glucose Cotransporter-2 Inhibitors Versus Dipeptidyl Peptidase-4 Inhibitors as Monotherapy or Add-on to Metformin in Patients with Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Diabetes Obes. Metab. 2018, 20, 113–120. [Google Scholar] [CrossRef]
  45. Tadic, M.; Sala, C.; Saeed, S.; Grassi, G.; Mancia, G.; Rottbauer, W.; Cuspidi, C. New Antidiabetic Therapy and HFpEF: Light at the End of Tunnel? Heart Fail. Rev. 2022, 27, 1137–1146. [Google Scholar] [CrossRef]
  46. Caruso, I.; Giorgino, F. SGLT-2 Inhibitors as Cardio-Renal Protective Agents. Metabolism 2022, 127, 154937. [Google Scholar] [CrossRef] [PubMed]
  47. Ferrannini, E.; Veltkamp, S.A.; Smulders, R.A.; Kadokura, T. Renal Glucose Handling: Impact of Chronic Kidney Disease and Sodium-Glucose Cotransporter 2 Inhibition in Patients with Type 2 Diabetes. Diabetes Care 2013, 36, 1260–1265. [Google Scholar] [CrossRef] [Green Version]
  48. Fioretto, P.; Zambon, A.; Rossato, M.; Busetto, L.; Vettor, R. SGLT2 Inhibitors and the Diabetic Kidney. Diabetes Care 2016, 39, S165–S171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Cherney, D.Z.I.; Cooper, M.E.; Tikkanen, I.; Pfarr, E.; Johansen, O.E.; Woerle, H.J.; Broedl, U.C.; Lund, S.S. Pooled analysis of Phase III trials indicate contrasting influences of renal function on blood pressure, body weight, and HbA1c reductions with empagliflozin. Kidney Int. 2018, 93, 231–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Baker, M.L.; Perazella, M.A. SGLT2 Inhibitor Therapy in Patients with Type-2 Diabetes Mellitus: Is Acute Kidney Injury a Concern? J. Nephrol. 2020, 33, 985–994. [Google Scholar] [CrossRef]
  51. List, J.F.; Woo, V.; Morales, E.; Tang, W.; Fiedorek, F.T. Sodium-Glucose Cotransport Inhibition with Dapagliflozin in Type 2 Diabetes. Diabetes Care 2009, 32, 650–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Clar, C.; Gill, J.A.; Court, R.; Waugh, N. Systematic Review of SGLT2 Receptor Inhibitors in Dual or Triple Therapy in Type 2 Diabetes. BMJ Open 2012, 2, e001007. [Google Scholar] [CrossRef] [PubMed]
  53. Tamargo, J. Sodium-glucose Cotransporter 2 Inhibitors in Heart Failure: Potential Mechanisms of Action, Adverse Effects and Future Developments. Eur. Cardiol. 2019, 14, 23–32. [Google Scholar] [CrossRef] [Green Version]
  54. 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]
  55. Anders, H.-J.; Davis, J.M.; Thurau, K. Nephron Protection in Diabetic Kidney Disease. N. Engl. J. Med. 2016, 375, 2096–2098. [Google Scholar] [CrossRef]
  56. Anders, H.-J.; Huber, T.B.; Isermann, B.; Schiffer, M. CKD in Diabetes: Diabetic Kidney Disease Versus Nondiabetic Kidney Disease. Nat. Rev. Nephrol. 2018, 14, 361–377. [Google Scholar] [CrossRef]
  57. Sen, T.; Heerspink, H.J.L. A Kidney Perspective on the Mechanism of Action of Sodium Glucose Co-Transporter 2 Inhibitors. Cell Metab. 2021, 33, 732–739. [Google Scholar] [CrossRef] [PubMed]
  58. 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] [PubMed]
  59. Zelniker, T.A.; Braunwald, E. Cardiac and Renal Effects of Sodium-Glucose Co-Transporter 2 Inhibitors in Diabetes: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2018, 72, 1845–1855. [Google Scholar] [CrossRef] [PubMed]
  60. Wanner, C.; Heerspink, H.J.L.; Zinman, B.; Inzucchi, S.E.; Koitka-Weber, A.; Mattheus, M.; Hantel, S.; Woerle, H.-J.; Broedl, U.C.; von Eynatten, M.; et al. Empagliflozin and Kidney Function Decline in Patients with Type 2 Diabetes: A Slope Analysis from the EMPA-REG OUTCOME Trial. J. Am. Soc. Nephrol. 2018, 29, 2755–2769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. van Bommel, E.J.M.; Muskiet, M.H.A.; van Baar, M.J.B.; Tonneijck, L.; Smits, M.M.; Emanuel, A.L.; Bozovic, A.; Danser, A.H.J.; Geurts, F.; Hoorn, E.J.; et al. The Renal Hemodynamic Effects of the SGLT2 Inhibitor Dapagliflozin Are Caused by Post-Glomerular Vasodilatation Rather than pre-Glomerular Vasoconstriction in Metformin-Treated Patients with Type 2 Diabetes in the Randomized, Double-Blind RED Trial. Kidney Int. 2020, 97, 202–212. [Google Scholar] [CrossRef]
  62. 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. Clin. J. Am. Soc. Nephrol. 2021, 16, 1345–1354. [Google Scholar] [CrossRef]
  63. Kraus, B.J.; Weir, M.R.; Bakris, G.L.; Mattheus, M.; Cherney, D.Z.I.; Sattar, N.; Heerspink, H.J.L.; Ritter, I.; von Eynatten, M.; Zinman, B.; et al. Characterization and Implications of the Initial Estimated Glomerular Filtration Rate ‘Dip’ upon Sodium-Glucose Cotransporter-2 Inhibition with Empagliflozin in the EMPA-REG OUTCOME Trial. Kidney Int. 2021, 99, 750–762. [Google Scholar] [CrossRef]
  64. Oshima, M.; Jardine, M.J.; Agarwal, R.; Bakris, G.; Cannon, C.P.; Charytan, D.M.; de Zeeuw, D.; Edwards, R.; Greene, T.; Levin, A.; et al. Insights From CREDENCE Trial Indicate an Acute Drop in Estimated Glomerular Filtration Rate during Treatment with Canagliflozin with Implications for Clinical Practice. Kidney Int. 2021, 99, 999–1009. [Google Scholar] [CrossRef]
  65. Adamson, C.; Docherty, K.F.; Heerspink, H.J.L.; de Boer, R.A.; Damman, K.; Inzucchi, S.E.; Kober, L.; Kosiborod, M.N.; Martinez, F.A.; Petrie, M.C.; et al. Initial Decline (Dip) in Estimated Glomerular Filtration Rate after Initiation of Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction: Insights from DAPA-HF. Circulation 2022, 146, 438–449. [Google Scholar] [CrossRef]
  66. Cherney, D.Z.I.; Cosentino, F.; Dagogo-Jack, S.; McGuire, D.K.; Pratley, R.E.; Frederich, R.; Maldonado, M.; Liu, C.C.; Pong, A.; Cannon, C.P.; et al. Initial eGFR Changes with Ertugliflozin and Associations with Clinical Parameters: Analyses from the VERTIS CV Trial. Am. J. Nephrol. 2022, 53, 516–525. [Google Scholar] [CrossRef] [PubMed]
  67. Jongs, N.; Chertow, G.M.; Greene, T.; McMurray, J.J.V.; Langkilde, A.M.; Correa-Rotter, R.; Kashihara, N.; Rossing, P.; Sjostrom, C.D.; Stefansson, B.V.; et al. Correlates and Consequences of an Acute Change in eGFR in Response to the SGLT2 Inhibitor Dapagliflozin in Patients with CKD. J. Am. Soc. Nephrol. 2022, 33, 2094–2107. [Google Scholar] [CrossRef] [PubMed]
  68. Chertow, G.M.; Vart, P.; Jongs, N.; Toto, R.D.; Gorriz, J.L.; Hou, F.F.; McMurray, J.J.V.; Correa-Rotter, R.; Rossing, P.; Sjöström, C.D.; et al. Effects of Dapagliflozin in Stage 4 Chronic Kidney Disease. J. Am. Soc. Nephrol. 2021, 32, 2352–2361. [Google Scholar] [CrossRef] [PubMed]
  69. 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] [PubMed]
  70. Heerspink, H.J.L.; Cherney, D.Z.I. Clinical Implications of an Acute Dip in eGFR after SGLT2 Inhibitor Initiation. Clin. J. Am. Soc. Nephrol. 2021, 16, 1278–1280. [Google Scholar] [CrossRef]
  71. Heerspink, H.J.L.; Johnsson, E.; Gause-Nilsson, I.; Cain, V.A.; Sjöström, C.D. Dapagliflozin Reduces Albuminuria in Patients with Diabetes and Hypertension Receiving Renin-Angiotensin Blockers. Diabetes Obes. Metab. 2016, 18, 590–597. [Google Scholar] [CrossRef] [Green Version]
  72. Cherney, D.Z.I.; Zinman, B.; Inzucchi, S.E.; Koitka-Weber, A.; Mattheus, M.; von Eynatten, M.; Wanner, C. Effects of Empagliflozin on the Urinary Albumin-to-Creatinine Ratio in Patients with Type 2 Diabetes and Established Cardiovascular Disease: An Exploratory Analysis from the EMPA-REG OUTCOME Randomised, Placebo-Controlled Trial. Lancet Diabetes Endocrinol. 2017, 5, 610–621. [Google Scholar] [CrossRef]
  73. Petrykiv, S.I.; Laverman, G.D.; de Zeeuw, D.; Heerspink, H.J.L. The Albuminuria-Lowering Response to Dapagliflozin Is Variable and Reproducible among Individual Patients. Diabetes Obes. Metab. 2017, 19, 1363–1370. [Google Scholar] [CrossRef]
  74. Jongs, N.; Greene, T.; Chertow, G.M.; McMurray, J.J.V.; Langkilde, A.M.; Correa-Rotter, R.; Rossing, P.; Sjöström, C.D.; Stefansson, B.V.; Toto, R.D.; et al. Effect of Dapagliflozin on Urinary Albumin Excretion in Patients with Chronic Kidney Disease with and without Type 2 Diabetes: A Prespecified Analysis from the DAPA-CKD Trial. Lancet Diabetes Endocrinol. 2021, 9, 755–766. [Google Scholar] [CrossRef]
  75. Mosenzon, O.; Wiviott, S.D.; Heerspink, H.J.L.; Dwyer, J.P.; Cahn, A.; Goodrich, E.L.; Rozenberg, A.; Schechter, M.; Yanuv, I.; Murphy, S.A.; et al. The Effect of Dapagliflozin on Albuminuria in DECLARE-TIMI 58. Diabetes Care 2021, 44, 1805–1815. [Google Scholar] [CrossRef]
  76. Neuen, B.L.; Ohkuma, T.; Neal, B.; Matthews, D.R.; de Zeeuw, D.; Mahaffey, K.W.; Fulcher, G.; Li, Q.; Jardine, M.; Oh, R.; et al. Effect of Canagliflozin on Renal and Cardiovascular Outcomes across Different Levels of Albuminuria: Data from the CANVAS Program. J. Am. Soc. Nephrol. 2019, 30, 2229–2242. [Google Scholar] [CrossRef] [PubMed]
  77. Gilbert, R.E.; Thorpe, K.E. Acute Kidney Injury with Sodium-Glucose Co-Transporter-2 Inhibitors: A Meta-Analysis of Cardiovascular Outcome Trials. Diabetes Obes. Metab. 2019, 21, 1996–2000. [Google Scholar] [CrossRef] [PubMed]
  78. Heerspink, H.J.L.; Cherney, D.; Postmus, D.; Stefánsson, B.V.; Chertow, G.M.; Dwyer, J.P.; Greene, T.; Kosiborod, M.; Langkilde, A.M.; McMurray, J.J.V.; et al. A Pre-Specified Analysis of the Dapagliflozin and Prevention of Adverse Outcomes in Chronic Kidney Disease (DAPA-CKD) Randomized Controlled Trial on the Incidence of Abrupt Declines in Kidney Function. Kidney Int. 2022, 101, 174–184. [Google Scholar] [CrossRef] [PubMed]
  79. Jhund, P.S.; Solomon, S.D.; Docherty, K.F.; Heerspink, H.J.L.; Anand, I.S.; Böhm, M.; Chopra, V.; de Boer, R.A.; Desai, A.S.; Ge, J.; et al. Efficacy of Dapagliflozin on Renal Function and Outcomes in Patients with Heart Failure with Reduced Ejection Fraction: Results of DAPA-HF. Circulation 2021, 143, 298–309. [Google Scholar] [CrossRef] [PubMed]
  80. Lambers Heerspink, H.J.; de Zeeuw, D.; Wie, L.; Leslie, B.; List, J. Dapagliflozin a Glucose-Regulating Drug with Diuretic Properties in Subjects with Type 2 Diabetes. Diabetes Obes. Metab. 2013, 15, 853–862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Sha, S.; Polidori, D.; Heise, T.; Natarajan, J.; Farrell, K.; Wang, S.-S.; Sica, D.; Rothenberg, P.; Plum-Mörschel, L. Effect of the Sodium Glucose Co-Transporter 2 Inhibitor Canagliflozin on Plasma Volume in Patients with Type 2 Diabetes Mellitus. Diabetes Obes. Metab. 2014, 16, 1087–1095. [Google Scholar] [CrossRef]
  82. Karg, M.V.; Bosch, A.; Kannenkeril, D.; Striepe, K.; Ott, C.; Schneider, M.P.; Boemke-Zelch, F.; Linz, P.; Nagel, A.M.; Titze, J.; et al. SGLT-2-Inhibition with Dapagliflozin Reduces Tissue Sodium Content: A Randomised Controlled Trial. Cardiovasc. Diabetol. 2018, 17, 5. [Google Scholar] [CrossRef] [Green Version]
  83. Baker, W.L.; Smyth, L.R.; Riche, D.M.; Bourret, E.M.; Chamberlin, K.W.; White, W.B. Effects of Sodium-Glucose Co-Transporter 2 Inhibitors on Blood Pressure: A Systematic Review and Meta-Analysis. J. Am. Soc. Hypertens. 2014, 8, 262–275. [Google Scholar] [CrossRef]
  84. Liu, X.-Y.; Zhang, N.; Chen, R.; Zhao, J.-G.; Yu, P. Efficacy and Safety of Sodium-Glucose Cotransporter 2 Inhibitors in Type 2 Diabetes: A Meta-Analysis of Randomized Controlled Trials for 1 to 2years. J. Diabetes Complicat. 2015, 29, 1295–1303. [Google Scholar] [CrossRef]
  85. Staels, B. Cardiovascular Protection by Sodium Glucose Cotransporter 2 Inhibitors: Potential Mechanisms. Am. J. Med. 2017, 130, S30–S39. [Google Scholar] [CrossRef] [Green Version]
  86. Hallow, K.M.; Helmlinger, G.; Greasley, P.J.; McMurray, J.J.V.; Boulton, D.W. Why do SGLT2 inhibitors reduce heart failure hospitalization? A differential volume regulation hypothesis. Diabetes Obes. Metab. 2018, 20, 479–487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Scholtes, R.A.; Muskiet, M.H.A.; van Baar, M.J.B.; Hesp, A.C.; Greasley, P.J.; Karlsson, C.; Hammarstedt, A.; Arya, N.; van Raalte, D.H.; Heerspink, H.J.L. Natriuretic Effect of Two Weeks of Dapagliflozin Treatment in Patients with Type 2 Diabetes and Preserved Kidney Function during Standardized Sodium Intake: Results of the DAPASALT Trial. Diabetes Care 2021, 44, 440–447. [Google Scholar] [CrossRef] [PubMed]
  88. Böhm, M.; Fitchett, D.; Ofstad, A.P.; Brueckmann, M.; Kaspers, S.; George, J.T.; Zwiener, I.; Zinman, B.; Wanner, C.; Marx, N.; et al. Heart Failure and Renal Outcomes According to Baseline and Achieved Blood Pressure in Patients with Type 2 Diabetes: Results from EMPA-REG OUTCOME. J. Hypertens. 2020, 38, 1829–1840. [Google Scholar] [CrossRef] [PubMed]
  89. Griffin, M.; Rao, V.S.; Ivey-Miranda, J.; Fleming, J.; Mahoney, D.; Maulion, C.; Suda, N.; Siwakoti, K.; Ahmad, T.; Jacoby, D.; et al. Empagliflozin in Heart Failure: Diuretic and Cardiorenal Effects. Circulation 2020, 142, 1028–1039. [Google Scholar] [CrossRef] [PubMed]
  90. Jackson, A.; Dewan, P.; Anand, I.; Belohlavek, J.; Bengtsson, O.; de Boer, R.; Böhm, M.; Boulton, D.; Chopra, V.; Demets, D.; et al. Dapagliflozin and Diuretic Use in Patients with Heart Failure and Reduced Ejection Fraction in DAPA-HF. Circulation 2020, 142, 1040–1054. [Google Scholar] [CrossRef]
  91. Chilton, R.; Tikkanen, I.; Cannon, C.P.; Crowe, S.; Woerle, H.J.; Broedl, U.C.; Johansen, O.E. Effects of Empagliflozin on Blood Pressure and Markers of Arterial Stiffness and Vascular Resistance in Patients with Type 2 Diabetes. Diabetes Obes. Metab. 2015, 17, 1180–1193. [Google Scholar] [CrossRef] [Green Version]
  92. Solini, A.; Giannini, L.; Seghieri, M.; Vitolo, E.; Taddei, S.; Ghiadoni, L.; Bruno, R.M. Dapagliflozin Acutely Improves Endothelial Dysfunction, Reduces Aortic Stiffness and Renal Resistive Index in Type 2 Diabetic Patients: A Pilot Study. Cardiovasc. Diabetol. 2017, 16, 138. [Google Scholar] [CrossRef] [Green Version]
  93. Striepe, K.; Jumar, A.; Ott, C.; Karg, M.V.; Schneider, M.P.; Kannenkeril, D.; Schmieder, R.E. Effects of the Selective Sodium-Glucose Cotransporter 2 Inhibitor Empagliflozin on Vascular Function and Central Hemodynamics in Patients with Type 2 Diabetes Mellitus. Circulation 2017, 136, 1167–1169. [Google Scholar] [CrossRef]
  94. Li, H.; Shin, S.E.; Seo, M.S.; An, J.R.; Choi, I.-W.; Jung, W.K.; Firth, A.L.; Lee, D.S.; Yim, M.J.; Choi, G.; et al. The anti-Diabetic Drug Dapagliflozin Induces Vasodilation via Activation of PKG and Kv Channels. Life Sci. 2018, 197, 46–55. [Google Scholar] [CrossRef]
  95. Chilton, R.J.; Gullestad, L.; Fitchett, D.; Inzucchi, S.E.; Mattheus, M.; Woerle, H.J.; Johansen, O.E. Empagliflozin Reduces Markers of Arterial Stiffness, Vascular Resistance and Cardiac Workload in EMPA-REG OUTCOME. Circulation 2016, 134, 13520. [Google Scholar]
  96. Michel, A.; Martín-Pérez, M.; Ruigómez, A.; García Rodríguez, L.A. Risk Factors for Hyperkalaemia in a Cohort of Patients with Newly Diagnosed Heart Failure: A Nested Case-Control Study in UK General Practice. Eur. J. Heart Fail. 2015, 17, 205–213. [Google Scholar] [CrossRef] [PubMed]
  97. Toledo, C.C.; Vellosa Schwartzmann, P.; Miguel Silva, L.; da Silva Ferreira, G.; Bianchini Cardoso, F.; Citelli Ribeiro, V.; Paim, L.R.; Antunes-Correa, L.M.; Carvalho Sposito, A.; Matos Souza, J.R.; et al. Serum Potassium Levels Provide Prognostic Information in Symptomatic Heart Failure Beyond Traditional Clinical Variables. ESC Heart Fail. 2021, 8, 2133–2143. [Google Scholar] [CrossRef] [PubMed]
  98. Neuen, B.L.; Oshima, M.; Agarwal, R.; Arnott, C.; Cherney, D.Z.; Edwards, R.; Langkilde, A.M.; Mahaffey, K.W.; McGuire, D.K.; Neal, B.; et al. Sodium-Glucose Cotransporter 2 Inhibitors and Risk of Hyperkalemia in People with Type 2 Diabetes: A Meta-Analysis of Individual Participant Data from Randomized, Controlled Trials. Circulation 2022, 145, 1460–1470. [Google Scholar] [CrossRef] [PubMed]
  99. Ferreira, J.P.; Zannad, F.; Pocock, S.J.; Anker, S.D.; Butler, J.; Filippatos, G.; Brueckmann, M.; Jamal, W.; Steubl, D.; Schueler, E.; et al. Interplay of Mineralocorticoid Receptor Antagonists and Empagliflozin in Heart Failure: EMPEROR-Reduced. J. Am. Coll. Cardiol. 2021, 77, 1397–1407. [Google Scholar] [CrossRef]
  100. Ferreira, J.P.; Butler, J.; Zannad, F.; Filippatos, G.; Schueler, E.; Steubl, D.; Zeller, C.; Januzzi, J.L.; Pocock, S.; Packer, M.; et al. Mineralocorticoid Receptor Antagonists and Empagliflozin in Patients with Heart Failure and Preserved Ejection Fraction. J. Am. Coll. Cardiol. 2022, 79, 1129–1137. [Google Scholar] [CrossRef] [PubMed]
  101. Shen, L.; Kristensen, S.L.; Bengtsson, O.; Böhm, M.; de Boer, R.A.; Docherty, K.F.; Inzucchi, S.E.; Katova, T.; Køber, L.; Kosiborod, M.N.; et al. Dapagliflozin in HFrEF Patients Treated with Mineralocorticoid Receptor Antagonists: An Analysis of DAPA-HF. JACC Heart Fail. 2021, 9, 254–264. [Google Scholar] [CrossRef]
  102. Ferreira, J.P.; Zannad, F.; Butler, J.; Filipattos, G.; Ritter, I.; Schüler, E.; Kraus, B.J.; Pocock, S.J.; Anker, S.D.; Packer, M. Empagliflozin and Serum Potassium in Heart Failure: An Analysis from EMPEROR-Pooled. Eur. Heart J. 2022, 43, 2984–2993. [Google Scholar] [CrossRef]
  103. Brown, A.J.M.; Gandy, S.; McCrimmon, R.; Houston, J.G.; Struthers, A.D.; Lang, C.C. A Randomized Controlled Trial of Dapagliflozin on Left Ventricular Hypertrophy in People with Type Two Diabetes: The DAPA-LVH Trial. Eur. Heart J. 2020, 41, 3421–3432. [Google Scholar] [CrossRef]
  104. Lee, M.M.Y.; Brooksbank, K.J.M.; Wetherall, K.; Mangion, K.; Roditi, G.; Campbell, R.T.; Berry, C.; Chong, V.; Coyle, L.; Docherty, K.F.; et al. Effect of Empagliflozin on Left Ventricular Volumes in Patients with Type 2 Diabetes, or Prediabetes, and Heart Failure with Reduced Ejection Fraction (SUGAR-DM-HF). Circulation 2021, 143, 516–525. [Google Scholar] [CrossRef]
  105. Omar, M.; Jensen, J.; Ali, M.; Frederiksen, P.H.; Kistorp, C.; Videbæk, L.; Poulsen, M.K.; Tuxen, C.D.; Möller, S.; Gustafsson, F.; et al. Associations of Empagliflozin with Left Ventricular Volumes, Mass, and Function in Patients with Heart Failure and Reduced Ejection Fraction: A Substudy of the Empire HF Randomized Clinical Trial. JAMA Cardiol. 2021, 6, 836–840. [Google Scholar] [CrossRef]
  106. Santos-Gallego, C.G.; Vargas-Delgado, A.P.; Requena-Ibanez, J.A.; Garcia-Ropero, A.; Mancini, D.; Pinney, S.; Macaluso, F.; Sartori, S.; Roque, M.; Sabatel-Perez, F.; et al. Randomized Trial of Empagliflozin in Nondiabetic Patients with Heart Failure and Reduced Ejection Fraction. J. Am. Coll. Cardiol. 2021, 77, 243–255. [Google Scholar] [CrossRef] [PubMed]
  107. Tanaka, H.; Soga, F.; Tatsumi, K.; Mochizuki, Y.; Sano, H.; Toki, H.; Matsumoto, K.; Shite, J.; Takaoka, H.; Doi, T.; et al. Positive Effect of Dapagliflozin on Left Ventricular Longitudinal Function for Type 2 Diabetic Mellitus Patients with Chronic Heart Failure. Cardiovasc. Diabetol. 2020, 19, 6. [Google Scholar]
  108. Sezai, A.; Sekino, H.; Unosawa, S.; Taoka, M.; Osaka, S.; Tanaka, M. Canagliflozin for Japanese Patients with Chronic Heart Failure and Type II Diabetes. Cardiovasc. Diabetol. 2019, 18, 76. [Google Scholar] [CrossRef] [Green Version]
  109. Curtain, J.P.; Docherty, K.F.; Jhund, P.S.; Petrie, M.C.; Inzucchi, S.E.; Køber, L.; Kosiborod, M.N.; Martinez, F.A.; Ponikowski, P.; Sabatine, M.S.; et al. Effect of Dapagliflozin on Ventricular Arrhythmias, Resuscitated Cardiac Arrest, or Sudden Death in DAPA-HF. Eur. Heart J. 2021, 42, 3727–3738. [Google Scholar] [CrossRef] [PubMed]
  110. Sano, M. A New Class of Drugs for Heart Failure: SGLT2 Inhibitors Reduce Sympathetic Overactivity. J. Cardiol. 2018, 71, 471–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Zoccali, C.; Mallamaci, F.; Parlongo, S.; Cutrupi, S.; Benedetto, F.A.; Tripepi, G.; Bonanno, G.; Rapisarda, F.; Fatuzzo, P.; Seminara, G.; et al. Plasma Norepinephrine Predicts Survival and Incident Cardiovascular Events in Patients with End-Stage Renal Disease. Circulation 2002, 105, 1354–1359. [Google Scholar] [CrossRef] [Green Version]
  112. Barretto, A.C.P.; Santos, A.C.; Munhoz, R.; Rondon, M.U.P.B.; Franco, F.G.; Trombetta, I.C.; Roveda, F.; de Matos, L.N.J.; Braga, A.M.W.; Middlekauff, H.R.; et al. Increased Muscle Sympathetic Nerve Activity Predicts Mortality in Heart Failure Patients. Int. J. Cardiol. 2009, 135, 302–307. [Google Scholar] [CrossRef]
  113. Amami, K.; Yamada, S.; Yoshihisa, A.; Kaneshiro, T.; Hijioka, N.; Nodera, M.; Nehashi, T.; Takeishi, Y. Predictive Impacts of Chronic Kidney Disease and Cardiac Sympathetic Nervous Activity on Lethal Arrhythmic Events in Chronic Heart Failure. Ann. Noninvasive Electrocardiol. 2022, 27, e12900. [Google Scholar] [CrossRef]
  114. Hillis, G.S.; Hata, J.; Woodward, M.; Perkovic, V.; Arima, H.; Chow, C.K.; Zoungas, S.; Patel, A.; Poulter, N.R.; Mancia, G.; et al. Resting Heart Rate and the Risk of Microvascular Complications in Patients with Type 2 Diabetes Mellitus. J. Am. Heart Assoc. 2012, 1, e002832. [Google Scholar] [CrossRef] [Green Version]
  115. Vazir, A.; Claggett, B.; Pitt, B.; Anand, I.; Sweitzer, N.; Fang, J.; Fleg, J.; Rouleau, J.; Shah, S.; Pfeffer, M.A.; et al. Prognostic Importance of Temporal Changes in Resting Heart Rate in Heart Failure and Preserved Ejection Fraction: From the TOPCAT Study. JACC Heart Fail. 2017, 5, 782–791. [Google Scholar] [CrossRef]
  116. Herat, L.Y.; Magno, A.L.; Rudnicka, C.; Hricova, J.; Carnagarin, R.; Ward, N.C.; Arcambal, A.; Kiuchi, M.G.; Head, G.A.; Schlaich, M.P.; et al. SGLT2 Inhibitor–Induced Sympathoinhibition: A Novel Mechanism for Cardiorenal Protection. JACC Basic Transl. Sci. 2020, 5, 169–179. [Google Scholar] [CrossRef] [PubMed]
  117. Gueguen, C.; Burke, S.L.; Barzel, B.; Eikelis, N.; Watson, A.M.D.; Jha, J.C.; Jackson, K.L.; Sata, Y.; Lim, K.; Lambert, G.W.; et al. Empagliflozin modulates renal sympathetic and heart rate baroreflexes in a rabbit model of diabetes. Diabetologia 2020, 63, 1424–1434. [Google Scholar] [CrossRef] [PubMed]
  118. Sano, M.; Chen, S.; Imazeki, H.; Ochiai, H.; Seino, Y. Changes in Heart Rate in Patients with Type 2 Diabetes Mellitus after Treatment with Luseogliflozin: Subanalysis of Placebo-Controlled, Double-Blind Clinical Trials. J. Diabetes Investig. 2018, 9, 638–641. [Google Scholar] [CrossRef] [PubMed]
  119. Shimizu, W.; Kubota, Y.; Hoshika, Y.; Mozawa, K.; Tara, S.; Tokita, Y.; Yodogawa, K.; Iwasaki, Y.-K.; Yamamoto, T.; Takano, H.; et al. Effects of Empagliflozin Versus Placebo on cardiac Sympathetic Activity in Acute Myocardial Infarction Patients with Type 2 Diabetes Mellitus: The EMBODY Trial. Cardiovasc. Diabetol. 2020, 19, 148. [Google Scholar] [CrossRef] [PubMed]
  120. Hamaoka, T.; Murai, H.; Hirai, T.; Sugimoto, H.; Mukai, Y.; Inoue, O.; Takashima, S.; Kato, T.; Takata, S.; Usui, S.; et al. Different Responses of Muscle Sympathetic Nerve Activity to Dapagliflozin between Patients with Type 2 Diabetes with and without Heart Failure. J. Am. Heart Assoc. 2021, 10, e022637. [Google Scholar] [CrossRef]
  121. Oshima, M.; Neuen, B.L.; Jardine, M.J.; Bakris, G.; Edwards, R.; Levin, A.; Mahaffey, K.W.; Neal, B.; Pollock, C.; Rosenthal, N.; et al. Effects of Canagliflozin on Anaemia in Patients with Type 2 Diabetes and Chronic Kidney Disease: A Post-Hoc Analysis From the CREDENCE Trial. Lancet Diabetes Endocrinol. 2020, 8, 903–914. [Google Scholar] [CrossRef]
  122. Inzucchi, S.E.; Zinman, B.; Fitchett, D.; Wanner, C.; Ferrannini, E.; Schumacher, M.; Schmoor, C.; Ohneberg, K.; Johansen, O.E.; George, J.T.; et al. How Does Empagliflozin Reduce Cardiovascular Mortality? Insights from a Mediation Analysis of the EMPA-REG OUTCOME Trial. Diabetes Care 2018, 41, 356–363. [Google Scholar] [CrossRef] [Green Version]
  123. Cherney, D.Z.I.; Cosentino, F.; Pratley, R.E.; Dagogo-Jack, S.; Frederich, R.; Maldonado, M.; Liu, J.; Pong, A.; Liu, C.C.; Cannon, C.P.; et al. The Differential Effects of Ertugliflozin on Glucosuria and Natriuresis Biomarkers: Prespecified Analyses from VERTIS CV. Diabetes Obes. Metab. 2022, 24, 1114–1122. [Google Scholar] [CrossRef]
  124. Segar, M.W.; Kolkailah, A.A.; Frederich, R.; Pong, A.; Cannon, C.P.; Cosentino, F.; Dagogo-Jack, S.; McGuire, D.K.; Pratley, R.E.; Liu, C.C.; et al. Mediators of Ertugliflozin Effects on Heart Failure and Kidney Outcomes among Patients with Type 2 Diabetes Mellitus. Diabetes Obes. Metab. 2022, 24, 1829–1839. [Google Scholar] [CrossRef]
  125. Kolkailah, A.A.; Wiviott, S.D.; Raz, I.; Murphy, S.A.; Mosenzon, O.; Bhatt, D.L.; Leiter, L.A.; Wilding, J.P.H.; Gause-Nilsson, I.; Sabatine, M.S.; et al. Effect of Dapagliflozin on Hematocrit in Patients with Type 2 Diabetes at High Cardiovascular Risk: Observations from DECLARE-TIMI 58. Diabetes Care 2022, 45, e27–e29. [Google Scholar] [CrossRef]
  126. Petrie, M.C.; Verma, S.; Docherty, K.F.; Inzucchi, S.E.; Anand, I.; Belohlávek, J.; Böhm, M.; Chiang, C.E.; Chopra, V.K.; de Boer, R.A.; et al. Effect of Dapagliflozin on Worsening Heart Failure and Cardiovascular Death in Patients with Heart Failure with and without Diabetes. JAMA 2020, 323, 1353–1368. [Google Scholar] [CrossRef] [PubMed]
  127. Sano, M.; Goto, S. Possible Mechanism of Hematocrit Elevation by Sodium Glucose Cotransporter 2 Inhibitors and Associated Beneficial Renal and Cardiovascular Effects. Circulation 2019, 139, 1985–1987. [Google Scholar] [CrossRef] [PubMed]
  128. Ghanim, H.; Abuaysheh, S.; Hejna, J.; Green, K.; Batra, M.; Makdissi, A.; Chaudhuri, A.; Dandona, P. Dapagliflozin Suppresses Hepcidin and Increases Erythropoiesis. J. Clin. Endocrinol. Metab. 2020, 105, e1056–e1063. [Google Scholar] [CrossRef] [PubMed]
  129. Mazer, C.D.; Hare, G.M.T.; Connelly, P.W.; Gilbert, R.E.; Shehata, N.; Quan, A.; Teoh, H.; Leiter, L.A.; Zinman, B.; Jüni, P.; et al. Effect of Empagliflozin on Erythropoietin Levels, Iron Stores, and Red Blood Cell Morphology in Patients with Type 2 Diabetes Mellitus and Coronary Artery Disease. Circulation 2020, 141, 704–707. [Google Scholar] [CrossRef]
  130. Li, J.; Woodward, M.; Perkovic, V.; Figtree, G.A.; Heerspink, H.J.L.; Mahaffey, K.W.; de Zeeuw, D.; Vercruysse, F.; Shaw, W.; Matthews, D.R.; et al. Mediators of the Effects of Canagliflozin on Heart Failure in Patients with Type 2 Diabetes. JACC Heart Fail. 2020, 8, 57–66. [Google Scholar] [CrossRef]
  131. Sano, M.; Takei, M.; Shiraishi, Y.; Suzuki, Y. Increased Hematocrit during Sodium-Glucose Cotransporter 2 Inhibitor Therapy Indicates Recovery of Tubulointerstitial Function in Diabetic Kidneys. J. Clin. Med. Res. 2016, 8, 844–847. [Google Scholar] [CrossRef] [Green Version]
  132. Zhao, D.; Liu, H.; Dong, P. Empagliflozin Reduces Blood Pressure and Uric Acid in Patients with Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. J. Hum. Hypertens. 2019, 33, 327–339. [Google Scholar] [CrossRef]
  133. Zhao, Y.; Xu, L.; Tian, D.; Xia, P.; Zheng, H.; Wang, L.; Chen, L. Effects of Sodium-Glucose Co-Transporter 2 (SGLT2) Inhibitors on Serum Uric Acid Level: A Meta-Analysis of Randomized Controlled Trials. Diabetes Obes. Metab. 2018, 20, 458–462. [Google Scholar] [CrossRef]
  134. Akbari, A.; Rafiee, M.; Sathyapalan, T.; Sahebkar, A. Impacts of Sodium/Glucose Cotransporter-2 Inhibitors on Circulating Uric Acid Concentrations: A Systematic Review and Meta-Analysis. J. Diabetes Res. 2022, 2022, 7520632. [Google Scholar] [CrossRef]
  135. McDowell, K.; Welsh, P.; Docherty, K.F.; Morrow, D.A.; Jhund, P.S.; De Boer, R.A.; O’Meara, E.; Inzucchi, S.E.; Køber, L.; Kosiborod, M.N.; et al. Dapagliflozin Reduces Uric Acid Concentration, an Independent Predictor of Adverse Outcomes in DAPA-HF. Eur. J. Heart Fail. 2022, 24, 1066–1076. [Google Scholar] [CrossRef]
  136. Chino, Y.; Samukawa, Y.; Sakai, S.; Nakai, Y.; Yamaguchi, J.; Nakanishi, T.; Tamai, I. SGLT2 Inhibitor Lowers Serum Uric Acid through Alteration of Uric Acid Transport Activity in Renal Tubule by Increased Glycosuria. Biopharm. Drug Dispos. 2014, 35, 391–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Doehner, W.; Anker, S.D.; Butler, J.; Zannad, F.; Filippatos, G.; Ferreira, J.P.; Salsali, A.; Kaempfer, C.; Brueckmann, M.; Pocock, S.J.; et al. Uric Acid and Sodium-Glucose Cotransporter-2 Inhibition with Empagliflozin in Heart Failure with Reduced Ejection Fraction: The EMPEROR-Reduced Trial. Eur. Heart J. 2022, 43, 3435–3446. [Google Scholar] [CrossRef] [PubMed]
  138. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Zannad, F. Effects of Sodium-Glucose Cotransporter 2 Inhibitors for the Treatment of Patients with Heart Failure: Proposal of a Novel Mechanism of Action. JAMA Cardiol. 2017, 2, 1025–1029. [Google Scholar] [CrossRef]
  139. Packer, M. Activation and Inhibition of Sodium-Hydrogen Exchanger Is a Mechanism that Links the Pathophysiology and Treatment of Diabetes Mellitus with that of Heart Failure. Circulation 2017, 136, 1548–1559. [Google Scholar] [CrossRef] [PubMed]
  140. Baartscheer, A.; Schumacher, C.A.; Wüst, R.C.I.; Fiolet, J.W.T.; Stienen, G.J.M.; Coronel, R.; Zuurbier, C.J. Empagliflozin Decreases Myocardial Cytoplasmic Na(+) through Inhibition of the Cardiac Na(+)/H(+) Exchanger in Rats and Rabbits. Diabetologia 2017, 60, 568–573. [Google Scholar] [CrossRef] [Green Version]
  141. 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]
  142. Uthman, L.; Li, X.; Baartscheer, A.; Schumacher, C.A.; Baumgart, P.; Hermanides, J.; Preckel, B.; Hollmann, M.W.; Coronel, R.; Zuurbier, C.J.; et al. Empagliflozin Reduces Oxidative Stress through Inhibition of the Novel Inflammation/NHE/[Na(+)](c)/ROS-Pathway in Human Endothelial Cells. Biomed. Pharmacother. 2022, 146, 112515. [Google Scholar] [CrossRef]
  143. Chung, Y.J.; Park, K.C.; Tokar, S.; Eykyn, T.R.; Fuller, W.; Pavlovic, D.; Swietach, P.; Shattock, M.J. Off-Target Effects of Sodium-Glucose Co-Transporter 2 Blockers: Empagliflozin Does Not Inhibit Na+/H+ Exchanger-1 or Lower [Na+]i in the Heart. Cardiovasc. Res. 2021, 117, 2794–2806. [Google Scholar] [CrossRef]
  144. Sharma, A.; Tate, M.; Mathew, G.; Vince, J.E.; Ritchie, R.H.; de Haan, J.B. Oxidative Stress and NLRP3-Inflammasome Activity as Significant Drivers of Diabetic Cardiovascular Complications: Therapeutic Implications. Front. Physiol. 2018, 9, 114. [Google Scholar] [CrossRef] [Green Version]
  145. Bonnet, F.; Scheen, A.J. Effects of SGLT2 Inhibitors on Systemic and Tissue Low-Grade Inflammation: The Potential Contribution to Diabetes Complications and Cardiovascular Disease. Diabetes Metab. 2018, 44, 457–464. [Google Scholar] [CrossRef]
  146. Heerspink, H.J.L.; Perco, P.; Mulder, S.; Leierer, J.; Hansen, M.K.; Heinzel, A.; Mayer, G. Canagliflozin Reduces Inflammation and Fibrosis Biomarkers: A Potential Mechanism of Action for Beneficial Effects of SGLT2 Inhibitors in Diabetic Kidney Disease. Diabetologia 2019, 62, 1154–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Aragón-Herrera, A.; Feijóo-Bandín, S.; Otero Santiago, M.; Barral, L.; Campos-Toimil, M.; Gil-Longo, J.; Costa Pereira, T.M.; García-Caballero, T.; Rodriguez-Segade, S.; Rodriguez, 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]
  148. Lee, N.; Heo, Y.J.; Choi, S.E.; Jeon, J.Y.; Han, S.J.; Kim, D.J.; Kang, Y.; Lee, K.W.; Kim, H.J. Anti-inflammatory Effects of Empagliflozin and Gemigliptin on LPS-Stimulated Macrophage via the IKK/NF-kappaB, MKK7/JNK, and JAK2/STAT1 Signalling Pathways. J. Immunol. Res. 2021, 2021, 9944880. [Google Scholar] [CrossRef] [PubMed]
  149. Steven, S.; Oelze, M.; Hanf, A.; Kröller-Schön, S.; Kashani, F.; Roohani, S.; Welschof, P.; Kopp, M.; Godtel-Armbrust, U.; Xia, N.; et al. The SGLT2 Inhibitor Empagliflozin Improves the Primary Diabetic Complications in ZDF Rats. Redox Biol. 2017, 13, 370–385. [Google Scholar] [CrossRef] [PubMed]
  150. Gohari, S.; Reshadmanesh, T.; Khodabandehloo, H.; Karbalaee-Hasani, A.; Ahangar, H.; Arsang-Jang, S.; Ismail-Beigi, F.; Dadashi, M.; Ghanbari, S.; Taheri, H.; et al. The Effect of EMPAgliflozin on Markers of Inflammation in Patients with Concomitant Type 2 Diabetes Mellitus and Coronary ARtery Disease: The EMPA-CARD Randomized Controlled Trial. Diabetol. Metab. Syndr. 2022, 14, 170. [Google Scholar] [CrossRef]
  151. Seecheran, N.; Ramdeen, A.; Debideen, N.; Ali, K.; Grimaldos, K.; Grimaldos, G.; Karan, A.; Seecheran, R.; Seecheran, V.; Persad, S.; et al. The Effect of Empagliflozin on Platelet Function Profiles in Patients with Stable Coronary Artery Disease in Trinidad: The EFFECT Pilot Study. Cardiol. Ther. 2021, 10, 189–199. [Google Scholar] [CrossRef]
  152. Seecheran, N.; Grimaldos, K.; Ali, K.; Grimaldos, G.; Richard, S.; Ishmael, A.; Gomes, C.; Karan, A.; Seecheran, R.; Seecheran, V.; et al. The Effect of Dapagliflozin on Platelet Function Testing Profiles in Diabetic Patients: The EDGE Pilot Study. Cardiol. Ther. 2021, 10, 561–568. [Google Scholar] [CrossRef]
  153. Gao, Y.M.; Feng, S.T.; Wen, Y.; Tang, T.T.; Wang, B.; Liu, B.C. Cardiorenal Protection of SGLT2 inhibitors-Perspectives from Metabolic Reprogramming. EBioMedicine 2022, 83, 104215. [Google Scholar] [CrossRef]
  154. Hiruma, S.; Shigiyama, F.; Hisatake, S.; Mizumura, S.; Shiraga, N.; Hori, M.; Ikeda, T.; Hirose, T.; Kumashiro, N. A Prospective Randomized Study Comparing Effects of Empagliflozin to Sitagliptin on Cardiac Fat Accumulation, Cardiac Function, and Cardiac Metabolism in Patients with Early-Stage Type 2 Diabetes: The ASSET Study. Cardiovasc. Diabetol. 2021, 20, 32. [Google Scholar] [CrossRef]
  155. Kappel, B.A.; Lehrke, M.; Schütt, K.; Artati, A.; Adamski, J.; Lebherz, C.; Marx, N. Effect of Empagliflozin on the Metabolic Signature of Patients with Type 2 Diabetes Mellitus and Cardiovascular Disease. Circulation 2017, 136, 969–972. [Google Scholar] [CrossRef]
  156. Nishimura, R.; Tanaka, Y.; Koiwai, K.; Ishida, K.; Salsali, A.; Kaspers, S.; Kohler, S.; Lund, S.S. Effect of Empagliflozin on Free Fatty Acids and Ketone Bodies in Japanese Patients with Type 2 Diabetes Mellitus: A Randomized Controlled Trial. Adv. Ther. 2019, 36, 2769–2782. [Google Scholar] [CrossRef] [PubMed]
  157. Bitar, Z.I.; Maadarani, O.S.; Alabdali, F.; Teama, A.; Elsawah, W.; Mohsen, M.J.; Elzoueiry, M.M. Sodium-Glucose Cotransporter-2 Inhibitors Induced Euglycemic Diabetic Ketoacidosis: Two Case Reports and a Review of the Literature. Clin. Case Rep. 2022, 10, e05440. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanisms underlying the cardio- and renoprotective effects of SGLT2is. BP, blood pressure; CV, cardiovascular; eGFR, estimated glomerular filtration rate; ESKD, end-stage kidney disease; H+, hydrogen; HHF, hospitalization for heart failure; LV, left ventricular; Na+, sodium; NT-proBNP, N-terminal prohormone of brain natriuretic peptide; SGLT2, sodium-glucose cotransporter-2; SGLT2i, sodium-glucose cotransporter-2 inhibitor; SNS, sympathetic nervous system; UACR, urinary albumin-to-creatinine ratio.
Figure 1. Mechanisms underlying the cardio- and renoprotective effects of SGLT2is. BP, blood pressure; CV, cardiovascular; eGFR, estimated glomerular filtration rate; ESKD, end-stage kidney disease; H+, hydrogen; HHF, hospitalization for heart failure; LV, left ventricular; Na+, sodium; NT-proBNP, N-terminal prohormone of brain natriuretic peptide; SGLT2, sodium-glucose cotransporter-2; SGLT2i, sodium-glucose cotransporter-2 inhibitor; SNS, sympathetic nervous system; UACR, urinary albumin-to-creatinine ratio.
Jcm 12 02824 g001
Figure 2. Glomerular effects of SGLT2is. In states of normal TGF, no glucose is delivered to the JGA. However, with glucosuria, TGF is abnormal, resulting in increased efferent arteriolar constriction and hyperfiltration. With SGLT2 inhibition, restoration of TGF occurs, lowering intraglomerular pressure and normalization of GFR. Reprintede with permission from DeFronzo RA, et al. Nat Rev Nephrol. 2017 [58]. Copyright © 2016, Nature Publishing Group, a division of Macmillan Publishers Ltd. All rights reserved. GFR, glomerular filtration rate; JGA, juxtaglomerular apparatus; Na+, sodium; SGLT1, sodium-glucose cotransporter 1; SGLT2, sodium-glucose cotransporter 2; SGLT2i, sodium-glucose cotransporter 2 inhibitor; TGF, tubular glomerular feedback.
Figure 2. Glomerular effects of SGLT2is. In states of normal TGF, no glucose is delivered to the JGA. However, with glucosuria, TGF is abnormal, resulting in increased efferent arteriolar constriction and hyperfiltration. With SGLT2 inhibition, restoration of TGF occurs, lowering intraglomerular pressure and normalization of GFR. Reprintede with permission from DeFronzo RA, et al. Nat Rev Nephrol. 2017 [58]. Copyright © 2016, Nature Publishing Group, a division of Macmillan Publishers Ltd. All rights reserved. GFR, glomerular filtration rate; JGA, juxtaglomerular apparatus; Na+, sodium; SGLT1, sodium-glucose cotransporter 1; SGLT2, sodium-glucose cotransporter 2; SGLT2i, sodium-glucose cotransporter 2 inhibitor; TGF, tubular glomerular feedback.
Jcm 12 02824 g002
Table 1. The effect of SGLT2i therapy on cardiovascular and renal outcomes in clinical trials.
Table 1. The effect of SGLT2i therapy on cardiovascular and renal outcomes in clinical trials.
Study NameDrugPopulationCV Outcomes,
HR (95% CI)
Renal Outcomes,
HR (95% CI)
CVOTs in patients with T2D
CANVAS [15]CanagliflozinAge ≥30 y with T2D and established CVD
OR
Age ≥50 y with T2D and ≥2 CVD risk factors
(N = 10,142)
MACE a,b: 0.86 (0.75–0.97)
p < 0.001 for noninferiority, p = 0.02 for superiority;
CV death or HHF:
0.78 (0.67–0.91);
HHF: 0.67 (0.52–0.87);
CV death: 0.87 (0.72–1.06)
Progression of albuminuria:
0.73 (0.67–0.79);
40% reduction in eGFR, RRT initiation, or death from renal causes:
0.60 (0.47–0.77)
DECLARE–TIMI 58 [18]Dapagliflozin Age ≥40 y with T2D and established CVD
OR
Age ≥55 y (men) or ≥60 y (women) with T2D and ≥1 CVD risk factor
(N = 17,160)
MACE a,b: 0.93 (0.84–1.03)
p < 0.001 for noninferiority,
p = 0.17 for superiority;
CV death or HHF a:
0.83 (0.73–0.95)
p = 0.005 for superiority;
HHF: 0.73 (0.61–0.88);
CV death: 0.98 (0.82–1.17)
≥40% reduction in eGFR to <60 mL/min/1.73 m2, ESKD, or death from CV or renal causes:
0.76 (0.67–0.87)
≥40% reduction in eGFR to <60 mL/min/1.73 m2, ESKD, or death from renal causes:
0.53 (0.43–0.66)
EMPA-REG OUTCOME [14,17]Empagliflozin Age ≥18 y with T2D and established CVD
(N = 7020)
MACE a,b: 0.86 (0.74–0.99)
p < 0.001 for noninferiority,
p = 0.04 for superiority;
MACE b or hospitalization for UA: 0.89 (0.78–1.01)
p < 0.001 for noninferiority,
p = 0.08 for superiority;
CV death or HHF:
0.66 (0.55–0.79)
p < 0.001
HHF: 0.65 (0.50–0.85)
p = 0.002;
CV death: 0.62 (0.49–0.77)
p < 0.001
Incident or worsening nephropathyc or CV death:
0.61 (0.55–0.69)
p < 0.001;
Incident or worsening nephropathyc:
0.61 (0.53–0.70)
p < 0.001;
Doubling of sCr with eGFR ≤45 mL/min/1.73 m2, RRT initiation, or death from renal causes:
0.54 (0.40–0.75)
p < 0.001
VERTIS CV [19]Ertugliflozin Age ≥40 y with T2D and established CVD
(N = 8246)
MACE a,b: 0.97 (0.85–1.11)
p < 0.001 for noninferiority,
CV death or HHF:
0.88 (0.75–1.03)
p = 0.11;
HHF: 0.70 (0.54–0.90);
CV death: 0.92 (0.77–1.11)
Doubling of sCr, RRT initiation, or death from renal causes:
0.81 (0.63–1.04)
Renal outcomes trials
CREDENCE [28]Canagliflozin Age ≥30 y with T2D and CKDd
(N = 4401)
MACE b: 0.80 (0.67–0.95)
p = 0.01;
CV death or HHF:
0.69 (0.57–0.83)
p < 0.001;
HHF: 0.61 (0.47–0.80)
p < 0.001;
CV death: 0.78 (0.61–1.00)
p = 0.05
ESKD, doubling of sCr, or death from renal or CV causes a:
0.66 (0.53–0.81)
p < 0.001;
Doubling of sCr:
0.60 (0.48–0.76)
p < 0.001;
ESKD: 0.68 (0.54–0.86)
p = 0.002
DAPA-CKD [27]Dapagliflozin Age ≥18 y with CKDe with or without T2D
(N = 4094)
Patients without T2D: 32.5% of total population
CV death or HHF:
0.71 (0.55–0.92)
p = 0.009;
CV death: 0.81 (0.58–1.12);
All-cause mortality:
0.69 (0.53–0.88)
p = 0.004
Sustained ≥50% reduction in eGFR, ESKD, or death from renal or CV causes a:
0.61 (0.51–0.72)
p < 0.001;
Sustained ≥50% reduction in eGFR, ESKD, or death from renal causes:
0.56 (0.45–0.68)
p < 0.001;
≥50% reduction in eGFR: 0.53 (0.42–0.67);
ESKD: 0.64 (0.50–0.82)
EMPA-KIDNEY [29]EmpagliflozinAdults with CKDf with or without T2D
(N = 6609)
Patients without T2D: 54% of total population
HHF or death from CV causes: 0.84 (0.67–1.07)
p = 0.15;
All-cause mortality:
0.87 (0.70–1.08) p = 0.21;
All-cause hospitalization 0.86 (0.78–0.95) p = 0.003;
Death from CV causes: 0.84 (0.60–1.19)
Kidney disease progression or death from CV causes: 0.72 (0.64–0.82)
p < 0.001;
Kidney disease progression: 0.71 (0.62–0.81);
ESKD or death from CV causes: 0.73 (0.59–0.89)
SCORED [20]Sotagliflozin Age ≥18 y with T2D, CKDg, and additional CV risk factorsh
(N = 10,584)
CV death, HHF, or urgent HF visit a: 0.74 (0.63–0.88)
p < 0.001
HHF or urgent HF visit:
0.67 (0.55–0.82)
p < 0.001;
CV death: 0.90 (0.73–1.12)
p = 0.35;
MACE: 0.77 (0.65–0.91)
Sustained ≥50% reduction in eGFR for ≥30 d, long-term dialysis, renal transplantation, or sustained eGFR of <15 mL/min/1.73 m2 for ≥30 d:
0.71 (0.46–1.08)
HF outcome trials
DAPA-HF [23]Dapagliflozin Age ≥18 y with NYHA class II–IV HFrEF (EF ≤40%)
(N = 4744)
Patients without T2D (including previously undiagnosed): n = 2605 (54.9% of total population)
Worsening HFh or CV deatha:
0.74 (0.65–0.85)
p < 0.001;
CV death or HHF:
0.75 (0.65–0.85)
p < 0.001;
Worsening HFi:
0.70 (0.59–0.83);
HHF: 0.70 (0.59–0.83);
Urgent HF visit:
0.43 (0.20–0.90);
CV death: 0.82 (0.69–0.98);
ΔKCCQ: 1.18 (1.11–1.26)
p < 0.001;
All-cause mortality:
0.83 (0.71–0.97)
Worsening renal function j:
0.71 (0.44–1.16)
DELIVER [25,30]DapagliflozinAge ≥40 y with NYHA class II–IV HFpEF (EF > 40%) with or without T2D
(N = 6263)
Patients without T2D: n = 3457 (55.2% of total population)
CV death, HHF or urgent HF visita: 0.82 (0.73–0.92)
p < 0.001;
HF events (HHF/urgent HF visit): 0.79 (0.69–0.91);
CV death: 0.88 (0.74–1.05);
ΔKCCQ: 1.11 (1.03–1.21)
p = 0.009
Mean difference (95% CI) in eGFR slope change per year vs. placebo (from baseline to end of trial):
0.5 mL/min/1.73 m2
(0.1–0.9)
p = 0.01;
Mean difference (95% CI) in eGFR slope change per year vs. placebo (from month 1 to end of trial):
1.4 mL/min/1.73 m2
(1.0–1.8)
p = 0.001;
Composite renal outcome k:
1.08 (0.79–1.49)
EMPEROR-Reduced [24]Empagliflozin Age ≥18 y with NYHA class II–IV HFrEF (EF ≤ 40%)
(N = 3730)
Patients without T2D: n = 1874 (50.2% of total population)
CV death or HHF a:
0.75 (0.65–0.86)
p < 0.001;
HHF: 0.69 (0.59–0.81);
CV death: 0.92 (0.75–1.12)
Composite renal outcomel:
0.50 (0.32–0.77)
EMPEROR-Preserved [22]Empagliflozin Age ≥18 y with NYHA class II–IV HFpEF (EF > 40%)
(N = 5988)
Patients without T2D: n = 3050 (50.9% of total population)
CV death or HHF a:
0.79 (0.69–0.90)
p < 0.001;
HHF: 0.71 (0.60–0.83);
CV death: 0.91 (0.76–1.09)
Mean difference (95% CI) in eGFR slope change per year vs. placebo:
1.36 mL/min/1.73 m2
(1.06–1.66)
p < 0.001;
Composite renal outcomej:
0.95 (0.73–1.24)
SOLOIST-WHF [21]Sotagliflozin Age 18–85 y with T2D and hospitalized for signs and symptoms of HF, treated with intravenous diuretics
(N = 1222)
CV death, HHF, or urgent HF visita: 0.67 (0.52–0.85)
p < 0.001;
HHF or urgent HF visit: 0.64 (0.49–0.83)
p < 0.001;
CV death: 0.84 (0.58–1.22)
p = 0.36
Difference (95% CI) in LS mean change in eGFR:
–0.16 (–1.30, +0.98) mL/min/1.73 m2
a Primary composite outcome (other outcomes are secondary). b Defined as the composite outcome of CV death, non-fatal myocardial infarction, or non-fatal stroke. c Defined as the composite outcome of progression to macroalbuminuria (UACR > 300 mg/g), doubling of sCr accompanied by an eGFR of ≤45 mL/min/1.73 m2, initiation of RRT, or death from renal causes. d Defined as an eGFR of between 30–90 mL/min/1.73 m2 and a UACR of between 300–5000 mg/g. e Defined as an eGFR of between 25–75 mL/min/1.73 m2 and a UACR of between 200–5000 mg/g. fDefined as an eGFR of between 20–45 mL/min/1.73 m2 irrespective of albuminuria or between 45–90 mL/min/1.73 m2 and a UACR ≥200 mg/g. gDefined as an eGFR of between 25–60 mL/min/1.73 m2. h≥1 major CV risk factor in participants aged ≥18 y or ≥2 minor CV risk factors in participants aged ≥55 y. i Defined as hospitalization or an urgent visit resulting in intravenous therapy for HF. j Defined as the composite outcome of ≥50% reduction in eGFR sustained for ≥28 days, ESKD, or death from renal causes. k Defined as the composite outcome of ≥50% reduction in eGFR, ESKD, or death from renal causes. lDefined as chronic dialysis or renal transplantation, a sustained reduction of ≥40% in eGFR, or a sustained eGFR of <15 mL/min/1.73 m2 (baseline eGFR of ≥30 mL/min/1.73 m2) or <10 mL/min/1.73 m2 (baseline eGFR of <30 mL/min/1.73 m2). CI, confidence interval; CKD, chronic kidney disease; CV, cardiovascular; CVD, cardiovascular disease; CVOT, cardiovascular outcomes trial; d, days; eGFR, estimated glomerular filtration rate; EF, ejection fraction; ESKD, end-stage kidney disease; HF, heart failure; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction; HHF, hospitalization for heart failure; HR, hazard ratio; ΔKCCQ, change in Kansas City Cardiomyopathy Questionnaire score at 8 months; LS, least-squares; MACE, major adverse cardiovascular event; NYHA, New York Heart Association; RRT, renal replacement therapy; sCr, serum creatinine; SGLT2i, sodium-glucose cotransporter-2 inhibitor; T2D, type 2 diabetes; UA, unstable angina; UACR, urinary albumin-to-creatinine ratio; y, year.
Table 2. Changes in inflammation and fibrosis markers in patients with T2D treated with SGLT2is [145,146]. Increase in level is marked as “↑”, decrease as “↓”, and significant changes (p < 0.05) as “*”.
Table 2. Changes in inflammation and fibrosis markers in patients with T2D treated with SGLT2is [145,146]. Increase in level is marked as “↑”, decrease as “↓”, and significant changes (p < 0.05) as “*”.
Inflammatory MarkerCanagliflozinDapagliflozinEmpagliflozin
Adiponectin a↑ *↑ *
Leptin
hsCRP↓ *
TNF-α↓ *↓ *
TNFR1↓ *
IL-6↓ * ↓ *
IFN-γ ↓ *
MMP7↓ *
FN1 ↓ *
CCL5No change
HGF
VEGFA↓300 mg (but not 100 mg)
FABP1
a Only adiponectin levels were increased with SGLT2is. CCL, C-C motif chemokine ligand, FABP1, fatty acid binding protein 1; FN1, fibronectin 1; HGF, hepatocyte growth factor; hsCRP, high-sensitivity C-reactive protein; IFN-γ: interferon-gamma; IL-6, interleukin-6; MMP, matrix metalloproteinase; SGLT2i, sodium-glucose cotransporter 2 inhibitor; T2D, type 2 diabetes; TNF-α, tumor necrosis factor-alpha; TNFR1, TNF receptor 1; VEGFA, vascular endothelial growth factor A.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rastogi, A.; Januzzi, J.L., Jr. Pleiotropic Effects of Sodium-Glucose Cotransporter-2 Inhibitors in Cardiovascular Disease and Chronic Kidney Disease. J. Clin. Med. 2023, 12, 2824. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm12082824

AMA Style

Rastogi A, Januzzi JL Jr. Pleiotropic Effects of Sodium-Glucose Cotransporter-2 Inhibitors in Cardiovascular Disease and Chronic Kidney Disease. Journal of Clinical Medicine. 2023; 12(8):2824. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm12082824

Chicago/Turabian Style

Rastogi, Anjay, and James L. Januzzi, Jr. 2023. "Pleiotropic Effects of Sodium-Glucose Cotransporter-2 Inhibitors in Cardiovascular Disease and Chronic Kidney Disease" Journal of Clinical Medicine 12, no. 8: 2824. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm12082824

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