Abstract

For decades, heart failure with preserved ejection fraction (HFpEF) proved an elusive entity to treat. Sodium-glucose cotransporter 2 (SGLT2) inhibitors have recently been shown to reduce the composite of heart failure hospitalization or cardiovascular death in patients with HFpEF in the landmark DELIVER and EMPEROR-Preserved trials. While improvements in blood sugar, blood pressure, and attenuation of kidney disease progression all may play some role, preclinical and translational research have identified additional mechanisms of these agents. The SGLT2 inhibitors have intriguingly been shown to induce a nutrient-deprivation and hypoxic-like transcriptional paradigm, with increased ketosis, erythropoietin, and autophagic flux in addition to altering iron homeostasis, which may contribute to improved cardiac energetics and function. These agents also reduce epicardial adipose tissue and alter adipokine signalling, which may play a role in the reductions in inflammation and oxidative stress observed with SGLT2 inhibition. Emerging evidence also indicates that these drugs impact cardiomyocyte ionic homeostasis although whether this is through indirect mechanisms or via direct, off-target effects on other ion channels has yet to be clearly characterized. Finally, SGLT2 inhibitors have been shown to reduce myofilament stiffness as well as extracellular matrix remodelling/fibrosis in the heart, improving diastolic function. The SGLT2 inhibitors have established themselves as robust, disease-modifying therapies and as recent trial results are incorporated into clinical guidelines, will likely become foundational in the therapy of HFpEF.

Pathophysiology of heart failure with preserved ejection fraction (HFpEF) and mechanisms of sodium-glucose cotransporter 2 (SGLT2) inhibitors. cGMP, cyclic guanosine monophosphate; NO, nitric oxide; PKG, protein kinase G; sGC, soluble guanylate cyclase.
Graphical Abstract

Pathophysiology of heart failure with preserved ejection fraction (HFpEF) and mechanisms of sodium-glucose cotransporter 2 (SGLT2) inhibitors. cGMP, cyclic guanosine monophosphate; NO, nitric oxide; PKG, protein kinase G; sGC, soluble guanylate cyclase.

Heart failure is associated with an impaired quality of life, frequent hospitalizations, and increased mortality.1 Disease-modifying therapies for heart failure have traditionally consisted of neurohormonal modulators, which inhibit the renin–angiotensin–aldosterone (RAAS) or adrenergic systems.2,3 In patients with heart failure and a reduced ejection fraction (HFrEF), these therapies have proved unambiguously beneficial, resulting in striking reductions in mortality.3 The benefit of such therapies in patients who have heart failure and a preserved ejection fraction (HFpEF) has been less robust.3,4 Prognosis in these patients has remained poor and effective; disease-modifying therapies have remained elusive.3,4 Over the past decade, sodium-glucose cotransporter 2 (SGLT2) inhibitors have shown clinical benefit in patients with heart failure across the entire spectrum of ejection fraction, and among patients with worsening heart failure and acute heart failure.5–10 Several landmark trials, DELIVER, EMPEROR-Preserved, and SOLOIST-WHF, showed significant reductions in the composite of heart failure hospitalization/cardiovascular death with these agents in patients with HFpEF.6,8,9 In this state-of-the-art review, we will summarize the recent results of SGLT2 inhibitors, discuss mechanisms of these agents in HFpEF, as well as future directions.

Mechanisms of heart failure with preserved ejection fraction

The mechanisms and pathophysiology underlying HFpEF have been reviewed in detail previously and will only be briefly summarized here.4,11–13 HFpEF traditionally described patients with an ejection fraction greater than 40%, although heart failure with mildly reduced (∼40%–49%) and improved ejection have recently been characterized as clinically distinct phenotypes.14 Some patients with an ejection fraction >40% have clinical signs and symptoms of heart failure secondary to another disorder such as infiltrative cardiomyopathies (including hemochromatosis or amyloidosis) or other cardiac conditions such as valvular heart disease, hypertrophic cardiomyopathy, or pericardial disease.15 In patients with such conditions, the mechanisms described below may not be applicable.

There are a variety of pathophysiological processes, which contribute to the syndrome of HFpEF (Figure 1); there is likely heterogeneity in the degree to which specific mechanisms contribute to disease in individual patients. Diastolic dysfunction is a hallmark of HFpEF; the landmark invasive study of Zile et al.16 showed impaired active relaxation and increased passive stiffness in patients with HFpEF. Adverse remodelling and dysfunction are not just seen in the left ventricle but also in the atria and vasculature.4,11,12,17,18 This may be accompanied by other processes, which contribute to the progression of HFpEF including neurohormonal activation, chronotropic incompetence, atrial arrythmias, and renal dysfunction.4,11–13,18,19

Risk factors and pathophysiological mechanisms of heart failure with preserved ejection fraction. DM, diabetes mellitus; HFpEF, heart failure with preserved ejection fraction; MI, myocardial infarction; OSA, obstructive sleep apnea; RAAS, renin–angiotensin–aldosterone system
Figure 1

Risk factors and pathophysiological mechanisms of heart failure with preserved ejection fraction. DM, diabetes mellitus; HFpEF, heart failure with preserved ejection fraction; MI, myocardial infarction; OSA, obstructive sleep apnea; RAAS, renin–angiotensin–aldosterone system

Cellular and molecular mechanisms underlying HFpEF have been thoroughly reviewed in the past.11,13,20–22 Adverse changes to cardiomyocyte structure, function, and energetics in addition to changes in the extracellular matrix contribute to diastolic dysfunction in HFpEF.23 One key mediator of intrinsic cardiomyocyte stiffness in HFpEF is the giant elastic, cytoskeletal protein titin; dysregulation of the NO-sGC-cGMP-PKG pathway contributes to titin hypophosphorylation and isoform switching between the compliant N2BA and stiff N2B states.13,22 Increased collagen-dependent stiffness secondary to inflammatory and fibrotic processes also contribute to diastolic dysfunction in HFpEF.13,22 Finally, alterations in cardiomyocyte energetics and ionic homeostasis are other key mechanisms that may also contribute to HFpEF.13,24

Sodium-glucose cotransporter 2 inhibitors in HFpEF: a serendipitous discovery

The SGLT2 inhibitors were originally investigated in patients with type 2 diabetes mellitus. These agents were found to improve key outcomes including cardiovascular death, and unexpectedly reduced hospitalizations for heart failure as well.25,26 Subgroup analyses of patients with heart failure, including HFpEF specifically, suggested that these agents may improve outcomes in these settings.27,28 Clinical evaluation of these agents proceeded to patients with HFrEF as well as those with chronic kidney disease (CKD). The SGLT2 inhibitors proved beneficial in HFrEF, reducing the composite of heart failure hospitalization/cardiovascular death in the EMPEROR-Reduced and DAPA-HF trials.5,7 These agents also slowed the rate of kidney function deterioration as well as improved heart failure outcomes in patients with CKD.29–31 In the SOLOIST-WHF and EMPULSE trials, which included patients across the spectrum of ejection fraction, SGLT2 inhibitors improved outcomes in patients with worsening heart failure or acute heart failure, respectively.9,10 Finally, the landmark EMPEROR-Preserved and DELIVER trials both demonstrated significant reductions in their primary outcomes (cardiovascular death or hospitalization [+/− urgent visit] for heart failure) with SGLT2 inhibitors in HFpEF in addition to modest improvements in symptom burden measured by the Kansas City Cardiomyopathy Questionnaire score.6,8 These results appear to be consistent across the spectrum of mildly reduced or preserved ejection fraction (41%–49%, 50%–60%, and >60%) as well as in a variety of sub-groups based on sex, race, and presence of comorbidities such as diabetes mellitus or atrial fibrillation.6,8,32

While a five-trial meta-analysis of patients with heart failure across the entire ejection fraction spectrum did show a reduction in all-cause mortality with SGLT2 inhibitors, in a two-trial meta-analysis of only patients with HFpEF (EMPEROR-Preserved and DELIVER), no significant reduction in all-cause mortality and only a statistically borderline effect on cardiovascular death was observed.32 The magnitude of effect and absolute reduction in heart failure event rates with SGLT2 inhibitors was lower than that seen in trials of RAAS inhibitors or beta-blockers in HFrEF patients, many of which were stopped early due to clear evidence of benefit with treatment.33–35 The clinical benefits with SGLT2 inhibitors in heart failure were notably, however, observed in trials in which the vast majority of patients were already being treated with both an angiotensin-converting enzyme inhibitor/angiotensin receptor blocker/angiotensin receptor–neprilysin inhibitor (∼80%–95%) and a beta-blocker (∼80%–95%) with a significant proportion of patients treated with a mineralocorticoid receptor antagonist (∼35%–70%) as well.32 Heart failure hospitalization/cardiovascular death or a similar composite has been the primary outcome of many large trials evaluating other classes of heart failure therapies in HFpEF (including CHARM-Preserved, TOPCAT, and PARAGON-HF).36–38 Hospitalization due to heart failure was, however, only one cause for hospital admission in patients with HFpEF, a population of patients often with many comorbidities and risk factors for other cardiac and non-cardiac events.39

In summary, over the past decade, SGLT2 inhibitors have emerged unexpectedly as a therapy for heart failure. These drugs were subsequently prospectively evaluated in over 12 000 patients with HFpEF in two landmark Phase III trials where they proved efficacious. As these trial results are incorporated into clinical guidelines around the world, SGLT2 inhibitors are likely to become important mainstays of therapy in HFpEF.

Mechanisms of sodium-glucose cotransporter 2 inhibitors in heart failure with preserved ejection fraction

The mechanisms underlying clinical benefit with SGLT2 inhibitors in HFpEF are complex and likely multifactorial (Graphical Abstract). Attempts to tease apart the mechanisms of these agents have involved investigations in preclinical models, mechanistic trials including evaluation of proteomics and cardiac remodelling in human patients, in addition to extrapolation from trials conducted in other populations (such as HFrEF and diabetes mellitus/cardiovascular disease). Given the remarkable consistency in benefits of SGLT2 inhibitors observed in patients with heart failure across the entire spectrum of reduced to preserved ejection fraction, there is likely significant overlap in the underlying mechanisms of SGLT2 inhibitors in patients with HFrEF and those with HFpEF.32,40 These two entities share many common risk factors (including hypertension, diabetes mellitus, and CKD), and while amelioration of these traditional risk factors may play some role in the robust effect of these medications, translational research as well as the rapid time course of clinical benefit and remodelling seen with these drugs suggest there are additional mechanisms at play.

Here, we will summarize mechanisms, which may contribute to the benefit of SGLT2 inhibitors in HFpEF with a focus on direct and indirect myocardial effects related to metabolism, energetics, autophagic flux, ionic homeostasis, inflammation, oxidative stress, and cardiac remodelling (Figure 2).

Mechanisms of SGLT2 inhibitors in heart failure with preserved ejection fraction. HFpEF, heart failure with preserved ejection fraction.
Figure 2

Mechanisms of SGLT2 inhibitors in heart failure with preserved ejection fraction. HFpEF, heart failure with preserved ejection fraction.

Blood pressure, blood sugar, and kidney function

The SGLT2 inhibitors improve many traditional risk factors for HFpEF including blood pressure and blood sugar.41 However, these properties alone likely do not explain the benefits of this class of drugs in HFpEF.42 The SGLT2 inhibitors have been shown to reduce systolic blood pressure by an average 4 mmHg as assessed using 24-h ambulatory monitoring.43 In the EMPEROR-Preserved trial, empagliflozin had similar benefits in patients with baseline systolic blood pressure below and above the median.8 With regard to blood sugar, other agents that lower glucose to a similar or even greater degree do not have such potent benefits in heart failure.42 Relative effects of SGLT2 inhibitors in HFpEF in EMPEROR-Preserved and DELIVER were near identical in patients with and without diabetes mellitus.6,8

The cardiorenal effects of these drugs are an important consideration given the strong association between CKD and HFpEF. In Phase III trials, SGLT2 inhibitors have been shown to reduce the rate of decline in kidney function as well as to reduce kidney failure/death in patients with CKD with or without diabetes; the underlying mechanisms here are believed to be a reduction in trans-glomerular pressure and regulation of tubule-glomerular feedback to protect kidney function.29,31,44–47 This is distinct and may be synergistic with the mechanism of RAAS inhibitors, which reduce hyperfiltration through efferent vasodilation.45,48 Cardiorenal effects play a major role in heart failure: kidney dysfunction worsens heart failure, and impaired cardiovascular function impairs kidney perfusion and function.47,49 This interplay is mediated through several important factors, many of which are key markers and therapeutic targets in heart failure including natriuretic peptides and the adrenergic and RAAS systems.49 Improvement of kidney function is likely both a contributory mechanism and beneficial by-product of these drugs in heart failure but does not solely explain their benefits in HFpEF. The SGLT2 inhibitors were equally beneficial in patients with estimated glomerular filtration rate below and above 60 mL/min/1.73 m2 in EMPEROR-Preserved and DELIVER, and have been shown to improve cardiovascular outcomes throughout the spectrum of kidney function.5,8

Natriuresis/diuresis

Natriuresis/diuresis may be an important contributor to the clinical improvement observed with SGLT2 inhibitors in patients with acute heart failure.50–52 In the EMPA-RESPONSE-AHF trial, empagliflozin significantly increased urine output and net fluids loss in patients with acutely decompensated heart failure over the first 4 days of hospital admission compared with placebo.53 In the EMPULSE trial of patients admitted to hospital for acute heart failure, empagliflozin was associated with significant weight loss by Day 15, which was sustained to Day 90 (including after adjustment for daily loop diuretic dose).54 Significant improvements compared with placebo in both clinical scores and markers (haematocrit and N-terminal pro-B-type natriuretic peptide) of congestion were also observed with empagliflozin by Day 15.54 The acute decongestion observed with SGLT2 inhibitors has led to proposed frameworks for incorporating SGLT2 inhibitors into the management of patients presenting with acutely decompensated heart failure alongside other diuretics like furosemide and acetazolamide.51

While natriuresis/diuresis may explain the effect of SGLT2 inhibitors in patients with acute heart failure and may contribute to the rapid time-course benefit observed with these agents, the available evidence suggests against diuresis as the primary driving mechanism by SGLT2 inhibitors improves outcomes in chronic heart failure. In the EMPEROR-Reduced trial, empagliflozin was not more effective in patients initially identified as having clinical evidence of volume overload (n = 1477) compared with those initially characterized as euvolemic, and decline in weight with empagliflozin was similar in patients with and without volume overload.42,55 In the DELIVER trial, which evaluated dapagliflozin, there was a similar relative reduction (but greater absolute reduction) in the primary outcome of worsening heart failure event/cardiovascular death in patients with recent heart failure hospitalization compared with those without.56 There was no relation between changes in body weight and natriuretic peptides in EMPEROR-Reduced suggesting against diuresis-induced reduction in volume overload as the underlying cause of weight loss.55 Sustained weight reduction with SGLT2 inhibitors may instead be related to loss of calories through glycosuria and reduction in visceral fat.55,57,58

Metabolism, energetics, and autophagic flux

The SGLT2 membrane protein resides in the proximal convoluted tubule and is responsible for 90% of glucose reabsorption from the filtrate under physiological conditions.59 Renal glycosuria through SGLT2 inhibition represents a net loss of calories from the body, and some have suggested that the SGLT2 protein may also function as a physiological sensor of nutrient surplus.42,60–62 Inhibition of SGLT2 is therefore proposed to induce a state of perceived starvation and hypoxia, up-regulating signals to induce a ‘fasting’ and ‘hypoxic’-like transcriptional paradigm.42,60–65 This results in adaptions such as increased erythropoietin (EPO) levels and ketogenesis.42,60–62 The majority of cardiac energy is normally generated from fatty acid oxidation; in the failing heart, fatty acid oxidation is reduced, and alternative pathways including glucose metabolism and anaerobic glycolysis are relied upon.66,67 The SGLT2 inhibitors increase serum ketone bodies, which represent additional source of energy for the failing heart.66,68 Ketone bodies have been shown to increase cardiac contractility and have important signalling roles as well, triggering cellular processes like autophagy.66,68

The downstream effects of the fasting and hypoxic-like transcriptional paradigms have been suggested to be a key aspect of SGLT2 inhibitors, which may be mediated at a cellular level through autophagic flux.60,62,63 This is a cellular degradation process mediated by lysosomes by which cells recycle organelles, proteins, and debris to generate energy, and is triggered in response to cellular stress including perceived nutritional depletion and hypoxia. Induction of autophagy can also contribute to degradation of dysfunctional mitochondria and subsequent reduction in oxidative stress.60,62,63 This cellular process is impaired in the failing myocardium, and induction of autophagy by SGLT2 inhibitors may contribute to the improvements in cardiac function observed with these drugs.69–71 Several of the most widely discussed cellular mediators thought to mediate this effect are the nutrient-deprivation sensors AMPK and sirtuins (SIRT1, SIRT3, and SIRT6) as well as hypoxia-inducible factors (HIF) and mammalian target of rapamycin (mTOR). Regulation of these mediators, which are all involved in maintaining homeostasis of energy, metabolism, and oxygen, is dysfunctional in heart failure, which contributes to the development of cardiomyopathy.65,69 Mechanistic studies demonstrate that SGLT2 inhibitors increase the activities of AMPK, sirtuins, and HIFs and decrease phosphorylation/activation of mTOR in the myocardium as well as in other tissues, which may at least partially underlie their benefit on organ function.72–80 The totality of evidence on this topic has recently been reviewed in great detail.80 The mechanisms by which SGLT2 inhibitors promote autophagy in tissues that do not express the SGLT2 protein have yet to definitively established; proposed but unproven mechanisms include nutrient-deprivation signalling secondary to caloric loss, or direct effects of these drugs on other cellular transporters/proteins such as glucose transporters or sirtuins.80 In summary, the proposed paradigm is as follows: SGLT2 inhibition enhances autophagic flux and induces a fasting/hypoxic mimicry state resulting in activation of mediators including AMPK, sirtuins, and HIF, which promote ketosis and erythropoiesis that lead to reductions in oxidative stress and inflammation as well as improved cellular function. This emerging mechanism may explain the beneficial effects of SGLT2 inhibitors on metabolism and cellular function in tissues/organs (such as the heart) that do not express the SGLT2 protein and could be a contributing factor to the reduction in oxidative stress and inflammation observed with these agents.

Recent proteomics analysis from the EMPEROR-Pooled analysis, which included 535 patients from the EMPEROR-Preserved trial, assessed differential levels of circulating proteins in patients randomized to empagliflozin compared with placebo at baseline, Week 12 and Week 52.81 This analysis showed that empagliflozin had substantial effects on circulating levels of proteins previously shown to be mediators of cardiac autophagy and apoptosis (including IGFBP1, TfRI, FST, RBP2, and Mdk) in addition to EPO and other circulating proteins related to fibrosis, oxidative stress, hypertrophy, and energetics.81

The effect of SGLT2 inhibitors on iron homeostasis and erythrocytosis has proved to be an intriguing development and has recently been reviewed in detail.82 Iron deficiency is a common comorbidity among patients with heart failure and is associated with reduced functional status and poor outcomes.83,84 Increasingly, evidence suggests that iron deficiency in patients with heart failure may be more likely to be functional (i.e. related to inflammatory-mediated increases in hepcidin, resulting in suppression of duodenal iron absorption and release from hepatocytes and the reticulocyte-endothelial system) rather than absolute in nature.82 In either case, reduced circulating and bioreactive cytosolic iron contributes to both anaemia and may impair synthesis of iron-containing proteins involved in ATP production in cardiomyocytes.82,85 In a preclinical study, iron deficiency has been directly shown to impair contractility, cellular ATP levels, and ATP-linked respiration in isolated human cardiomyocytes.85 In several placebo-controlled trials including analyses from DAPA-HF and the EMPEROR programme, SGLT2 inhibitors have been consistently shown to decrease circulating levels of both hepcidin and ferritin, while increasing levels of transferrin receptor protein 1.81,82,86–89 This combination of findings suggests SGLT2 inhibitors may alleviate functional iron deficiency, enabling increased iron mobilization and augmenting bioreactive cytosolic iron levels in erythroid precursors and cardiomyocytes.82 While further research is needed to clarify the mechanisms by which SGLT2 inhibitors alter iron homeostasis, possible contributors include the reductions in inflammatory mediators (which may impact ferritin and hepcidin levels) as well as downstream effects of nutrient-deprivation signalling (including SIRT1) induced by SGLT2 inhibition.82 The SGLT2 inhibitors have also been consistently shown to increase EPO levels, doing so in as little as 1 month of treatment initiation in the EMPA-HEART CardioLink-6 trial.90 In the DAPA-HF trial, treatment with dapagliflozin led to rises in haematocrit to the point of correction into the non-anemic range in 62.2% of patients with anaemia at baseline (compared with 41.1% in the placebo arm).86 Intriguingly, while treatment with EPO analogues has not been shown to improve outcomes in heart failure or patients with diabetes mellitus and CKD, the degree of erythrocytosis/increase in haematocrit has been strongly associated with improved heart failure outcomes in the CANVAS program as well as the VERTIS-CV and EMPA-REG OUTCOME trials.91–95 While correction of anaemia may result in improved oxygen delivery to the failing myocardium, the association between changes in haematocrit and improved outcomes could also stem from common underlying pathways (i.e. autophagy signalling pathways and increased cytosolic bioreactive iron available for iron-dependent proteins in cardiomyocytes) as opposed to representing a direct causal mechanism.

Other metabolic pathways may contribute to the effects of SGLT2 inhibitors as well. The SGLT2 inhibitors attenuate insulin resistance, which is associated with poor cardiac function and future risk of HFpEF.96–100 The SGLT2 inhibitors are also associated with reductions in serum uric acid, although the degree to which this has any clinical significance is unclear.41,101 Uric acid may increase oxidative stress, inflammation, and endothelial dysfunction and is associated with a poor prognosis in heart failure.41,101

Adipose tissue

Epicardial adipose tissue (EAT) is a form of white, visceral adipose tissue around the heart with important paracrine/vasocrine signalling properties.102,103 Expansion of this tissue in patients with diabetes mellitus and/or obesity induces states of insulin resistance, oxidative stress, inflammation, and fibrosis and alters calcium homeostasis, all of which may contribute to diastolic dysfunction.102,103 Expansion of EAT has been associated with a poor prognosis and worse haemodynamic profiles in patients with HFpEF.104,105 In meta-analyses, SGLT2 inhibitors have been associated with reductions in EAT despite having no significant effects on total body mass index.57,106 The reduction in EAT may contribute to the attenuation of myocardial inflammation and fibrosis observed with SGLT2 inhibitors. In addition to reducing visceral fat, SGLT2 inhibitors also modulate adipokines levels, increasing levels of adiponectin, an insulin-sensitizing and anti-inflammatory hormone, and reducing levels of leptin, a hormone released by adipose tissue in states of nutritional excess.107 Paracrine leptin release from epicardial tissue impairs calcium homeostasis and induces cardiac fibrosis and microcirculatory dysfunction.108

Inflammation and oxidative stress

The SGLT2 inhibitors are known to decrease oxidative stress and inflammation.109,110 The SGLT2 inhibitors reduce levels of circulating pro-inflammatory factors such as C-reactive protein, tumour necrosis factor-α, and interleukin-6.111 They also reduce markers of oxidative stress and reactive oxygen species including hydrogen peroxide in the myocardium.112,113 The attenuation of oxidative stress and inflammation associated with SGLT2 inhibitors is likely multifactorial, and a number of mechanisms including decreased uric acid, reduced epicardial fat, alterations in adipokine levels and up-regulation of autophagy all may play a role.114 Another important dimension is the NLRP3 inflammasome, a multimeric cytoplasmic protein complex in macrophages, which plays an important role in the chronic inflammation of heart failure and atherosclerosis.115–117 Treatment with SGLT2 inhibitors inhibits activation of the NLRP3 inflammasome, an effect which may be in part mediated by the increased ketosis and decreased uric acid associated with these agents.91,92 Reduced NLRP3 activity results in decreased macrophage infiltration and pro-inflammatory cytokine release.115–117 This effect may not be limited to the heart, with data showing inhibition of the NLRP3 inflammasome by SGLT2 inhibitors in the kidney, liver, and vasculature.117–119 Within the vasculature, SGLT2 inhibitors have been shown to increase nitric oxide bioavailability, attenuate endothelial dysfunction, and increase circulating levels of pro-angiogenic progenitor cells.120,121 In a head-to-head comparison, treatment with empagliflozin compared with insulin or metformin was recently shown to be associated with with improvement in microRNA profiles associated with endothelial dysfunction in patients with HFpEF and diabetes mellitus.122

Ion handling/homeostasis: Na+ and Ca2+

Features of ionic imbalance in heart failure include a state of calcium overload as well as increased intracellular sodium.123,124 The SGLT2 inhibitors reduce intracellular sodium and cytosolic calcium, although the mechanism underlying these changes has not been definitively established.125–127 Sodium–hydrogen antiporters (NHE) are expressed in various sites including the heart and the kidneys.128,129 The SGLT2 protein in the kidney interacts closely with NHE-3, which is a transporter primarily responsible for sodium reabsorption from the filtrate.62 Increased NHE-3 activity increases oxidative stress and adrenergic activation, and increased sodium reabsorption by this transporter decreases distal sodium delivery to the macula densa, leading to glomerular hyperfiltration and destruction of nephrons.62 The NHE-3 is functionally intertwined with SGLT2 in the nephron, and knockout/inhibition of one impairs the activity of the other.62 This may in part explain the nephroprotective effects of SGLT2 inhibitors. The NHE-1 is expressed in cardiomyocytes and is up-regulated in the failing heart, increasing intracellular sodium and calcium.130,131 The SGLT2 inhibitors reduce intracellular sodium despite the SGLT2 protein not being expressed in the heart.62,132 It has been postulated that direct, off-target inhibition of NHE-1 by SGLT2 inhibitors could be responsible for improved cardiac remodelling and reduced fibrosis with these agents. There is conflicting evidence about whether SGLT2 inhibitors can directly inhibit the NHE-1 transporter in cardiomyocytes.62,133,134 The reduction in intracellular sodium with SGLT2 inhibitors observed could instead be a downstream effect of other signalling pathways, and there is some evidence that SGLT2 inhibitor may reduce NHE-1 mRNA expression, which may be mediated through an AMPK-dependent pathway.62,135 Another potential off-target effect of SGLT2 inhibitors is interaction with the cardiac sodium channel Nav1.5 (also known as SCN5A), which mediates late sodium current.136,137 A molecular study showed that SGLT2 inhibitors may bind to the same site on Nav1.5 as known Class 1 antiarrhythmics such as lidocaine and inhibit late sodium current; whether this interaction has any clinical significance has yet to be determined.137 These effects on ionic homeostasis could in part explain the rapid time course of benefits observed with SGLT2 inhibitors.

Diastolic function and remodelling

Treatment with empagliflozin has been shown to improve diastolic function after only 3 months in a small cohort of patients with diabetes and high cardiovascular risk.138 The IDDIA trial of patients with type 2 diabetes mellitus and at least grade 1 left ventricular diastolic dysfunction showed improvements in left ventricular diastolic function, including diastolic reserve, assessed by diastolic stress echocardiography after 24 weeks of treatment with dapagliflozin compared with placebo.139 As discussed above, diastolic dysfunction in HFpEF has several components including intrinsic myofilament stiffness (related to titin regulation and NO-sGC-cGMP-PKG signalling) as well as extracellular matrix-related stiffness/myocardial fibrosis.23 It may be here where the various pathways involved in SGLT2-mediated reductions in oxidative stress and inflammation converge. In animal models, SGLT2 inhibitors increase NO bioavailability, and restore the pathologically altered phosphorylation of titin mediated through downstream cGMP-PKG signalling; this translates to improved diastolic function.140 In a mechanistic analysis of isolated human and murine myocardial tissue, SGLT2 inhibitors were shown to reduce diastolic tension and passive myofilament stiffness as well as alter phosphorylation of myofilament proteins including titin.141,142 Beyond direct cardiomyocyte effects, SGLT2 inhibitors have been shown to reduce macrophage infiltration, regulate macrophage polarization, inhibit cardiac fibroblast differentiation, and suppress fibrotic markers such as type 1 collagen, resulting in attenuated fibrosis and adverse extracellular remodelling in the heart.66,78,109,113,118,143–145 In addition to reducing myofilament stiffness and fibrosis, SGLT2 inhibitors have been shown to reduce hypertrophy measured by left ventricular mass index within only 6 months in patients with type 2 diabetes mellitus and coronary artery disease in the EMPA-HEART CardioLink-6 trial.146 A systematic review of five randomized controlled trials showed that SGLT2 inhibitors were associated with left ventricular mass regression compared with placebo, although none of the included trials were conducted in patients with HFpEF.147

Future directions

While SGLT2 inhibitors have already proved to improve outcomes in patients with HFpEF, there are several avenues of research that are ongoing (Figure 3).

Ongoing and completed trials of SGLT2 inhibitors in patients with cardiovascular or renal disease. CKD, chronic kidney disease; CV, cardiovascular; CVD, cardiovascular disease; DM, diabetes mellitus; HFH, heart failure hospitalization; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction, HR, hazard ratio; nsMRA, non-steroidal mineralocorticoid receptor antagonist.
Figure 3

Ongoing and completed trials of SGLT2 inhibitors in patients with cardiovascular or renal disease. CKD, chronic kidney disease; CV, cardiovascular; CVD, cardiovascular disease; DM, diabetes mellitus; HFH, heart failure hospitalization; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction, HR, hazard ratio; nsMRA, non-steroidal mineralocorticoid receptor antagonist.

The DAPA ACT HF-TIMI 68 trial is evaluating the effects of in-hospital initiation of dapagliflozin on cardiovascular death or worsening heart in patients hospitalized for acute heart failure, irrespective of ejection fraction.148 This trial will help inform the most appropriate time point for initiation of these agents in patients with HFpEF. Atrial fibrillation is known to be associated with HFpEF: each condition independently increases the risk factor for the other.15 The relative benefit of SGLT2 inhibitors in patients with HFpEF is similar in those with and without atrial fibrillation; however, given that patients with atrial fibrillation suffer from a higher rate of adverse heart failure events at baseline, the absolute benefit may likely be greater.149 The SGLT2 inhibitors have also been shown to reduce the rate of atrial fibrillation events in analyses of adverse event reporting from randomized controlled trials.149 DAPA-AF and EMPA-AF are two ongoing randomized trials, which will quantify the effects of SGLT2 inhibitors on atrial fibrillation burden.150,151 Finally, there are also ongoing trials (DAPA-MI and EMPACT-MI) evaluating the effects of SGLT2 inhibitors following myocardial infarction.152,153

Two other classes of medications, which have emerged over the past decade and have also shown benefit in patients with diabetes and/or kidney disease, are glucagon-like peptide-1 receptor agonists and non-steroidal mineralocorticoid receptor antagonists.154,155 Evaluation of potential synergy of these classes of agents with SGLT2 inhibitors is an area of interest. Two trials, MIRACLE and CONFIDENCE, will evaluate the effects of the combination of SGLT2 inhibitors with non-steroidal mineralocorticoid receptor antagonists in patients with cardiorenal disease.156,157

Finally, SGLT1 inhibition, through molecules such as the SGLT1/2 inhibitor sotagliflozin, has been hypothesized to also have possible supplementary benefits in heart failure as well.158 The SGLT1 is expressed in the small intestine where it contributes to glucose absorption, as well as in other sites including the kidney.158 Decreases in SGLT1-mediated glucose absorption increase serum glucagon-like peptide-1 levels, and SGLT1 inhibition may also inhibit hypertrophy and fibrosis within the myocardium.158,159 The SGLT1/2 inhibitor sotagliflozin reduced myocardial infarction and stroke in the SCORED trial, a finding that has not been seen with traditional SGLT2 inhibitors, and which has been postulated to be mediated in part through increased endogenous incretin levels.31 There are no direct data comparing dual SGLT1/2 inhibitors with SGLT2 inhibitors, but this may prove to be an area of interest in the future.

Conclusion

In summary, HFpEF has proved an elusive entity to treat. With the results of DELIVER, SOLOIST-WHF, and EMPEROR-Preserved, SGLT2 inhibitors have solidified themselves as robust therapies to improve outcomes in patients with HFpEF. The underlying mechanism of these agents cannot be wholly ascribed to improvements in traditional risk factors such as blood pressure, blood sugar, volume status, and renal function. Rather, the rapid time course of improvement as well as experimental evidence suggests that a combination of mechanisms for SGLT2 inhibitors in HFpEF including induction of autophagy improved ionic homeostasis and reduced inflammation and oxidative stress, which contribute to reduced adverse remodelling and diastolic dysfunction. As recent trial results are incorporated into clinical guidelines, SGLT2 inhibitors will become foundational in the therapy of HFpEF.

Acknowledgements

The authors would like to thank Sana Khan for assistance with preparation of the figures.

Declarations

Disclosure of Interest

A.P. has no disclosures.

D.L.B. discloses the following relationships—advisory board: AngioWave, Bayer, Boehringer Ingelheim, Cardax, CellProthera, Cereno Scientific, Elsevier Practice Update Cardiology, High Enroll, Janssen, Level Ex, McKinsey, Medscape Cardiology, Merck, MyoKardia, NirvaMed, Novo Nordisk, PhaseBio, PLx Pharma, Regado Biosciences, Stasys; board of directors: AngioWave (stock options), Boston VA Research Institute, Bristol Myers Squibb (stock), DRS.LINQ (stock options), High Enroll (stock), Society of Cardiovascular Patient Care, TobeSoft; chair: Inaugural Chair, American Heart Association Quality Oversight Committee; consultant: Broadview Ventures; data monitoring committees: Acesion Pharma, Assistance Publique-Hôpitaux de Paris, Baim Institute for Clinical Research (formerly Harvard Clinical Research Institute, for the PORTICO trial, funded by St. Jude Medical, now Abbott), Boston Scientific (chair, PEITHO trial), Cleveland Clinic (including for the ExCEED trial, funded by Edwards), Contego Medical (chair, PERFORMANCE 2), Duke Clinical Research Institute, Mayo Clinic, Mount Sinai School of Medicine (for the ENVISAGE trial, funded by Daiichi Sankyo; for the ABILITY-DM trial, funded by Concept Medical), Novartis, Population Health Research Institute; Rutgers University (for the NIH-funded MINT Trial); honoraria: American College of Cardiology (senior associate editor, Clinical Trials and News, ACC.org; chair, ACC Accreditation Oversight Committee), Arnold and Porter law firm (work related to Sanofi/Bristol Myers Squibb clopidogrel litigation), Baim Institute for Clinical Research (formerly Harvard Clinical Research Institute; RE-DUAL PCI clinical trial steering committee funded by Boehringer Ingelheim; AEGIS-II executive committee funded by CSL Behring), Belvoir Publications (editor in chief, Harvard Heart Letter), Canadian Medical and Surgical Knowledge Translation Research Group (clinical trial steering committees), Cowen and Company, Duke Clinical Research Institute (clinical trial steering committees, including the PRONOUNCE trial, funded by Ferring Pharmaceuticals), HMP Global (editor in chief, Journal of Invasive Cardiology), Journal of the American College of Cardiology (guest editor; associate editor), K2P (co-chair, interdisciplinary curriculum), Level Ex, Medtelligence/ReachMD (CME steering committees), MJH Life Sciences, Oakstone CME (course director, Comprehensive Review of Interventional Cardiology), Piper Sandler, Population Health Research Institute (for the COMPASS operations committee, publications committee, steering committee, and USA national co-leader, funded by Bayer), Slack Publications (chief medical editor, Cardiology Today’s Intervention), Society of Cardiovascular Patient Care (secretary/treasurer), WebMD (CME steering committees), Wiley (steering committee); Other: Clinical Cardiology (deputy editor), NCDR-ACTION Registry Steering Committee (chair), VA CART Research and Publications Committee (chair); Patent: Sotagliflozin (named on a patent for sotagliflozin assigned to Brigham and Women's Hospital who assigned to Lexicon; neither I nor Brigham and Women's Hospital receive any income from this patent); Research Funding: Abbott, Acesion Pharma, Afimmune, Aker Biomarine, Amarin, Amgen, AstraZeneca, Bayer, Beren, Boehringer Ingelheim, Boston Scientific, Bristol Myers Squibb, Cardax, CellProthera, Cereno Scientific, Chiesi, CinCor, CSL Behring, Eisai, Ethicon, Faraday Pharmaceuticals, Ferring Pharmaceuticals, Forest Laboratories, Fractyl, Garmin, HLS Therapeutics, Idorsia, Ironwood, Ischemix, Janssen, Javelin, Lexicon, Lilly, Medtronic, Merck, Moderna, MyoKardia, NirvaMed, Novartis, Novo Nordisk, Owkin, Pfizer, PhaseBio, PLx Pharma, Recardio, Regeneron, Reid Hoffman Foundation, Roche, Sanofi, Stasys, Synaptic, The Medicines Company, Youngene, 89Bio; Royalties: Elsevier (editor, Braunwald’s Heart Disease); site co-investigator: Abbott, Biotronik, Boston Scientific, CSI, Endotronix, St. Jude Medical (now Abbott), Philips, SpectraWAVE, Svelte, Vascular Solutions; trustee: American College of Cardiology; Unfunded Research: FlowCo, Takeda.

A.P. has no disclosures.

N.M. has received support for clinical trial leadership from Boehringer Ingelheim, Novo Nordisk, served as a consultant to Boehringer Ingelheim, Merck, Novo Nordisk, AstraZeneca, BMS, received grant support from Boehringer Ingelheim, Merck, Novo Nordisk, and served as a speaker for Boehringer Ingelheim, Merck, Novo Nordisk, Lilly, BMS, and AstraZeneca. He declines all personal compensation from pharma or device companies.

F.C. has served as a speaker and advisory board members for AstraZeneca, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Lilly, Merck Sharp & Dohme, Novo Nordisk, and Pfizer; and has received research grants from the City Pharmacy, Abu Dhabi, UAE, King Gustav V and Queen Victoria Foundation, the Swedish Heart & Lung Foundation, and the Swedish Research Council.

A.P. has received research grant support from the Gilead Sciences Research Scholar Program and Applied Therapeutics; honoraria outside the present study as an advisor/consultant for Tricog Health Inc and Lilly, USA, Rivus, and Roche Diagnostics, and nonfinancial support from Pfizer and Merck.

S.V. holds a Tier 1 Canada Research Chair in Cardiovascular Surgery, and has received research grants and/or speaking honoraria from Amarin, Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly, EOCI Pharmacomm Ltd, HLS Therapeutics, Janssen, Merck, Novartis, Novo Nordisk, Pfizer, PhaseBio, Sanofi, Sun Pharmaceuticals, and the Toronto Knowledge Translation Working Group. He is the President of the Canadian Medical and Surgical Knowledge Translation Research Group, a federally incorporated not-for-profit physician organization.

Data Availability

All data described were obtained from published sources that are cited within the manuscript.

Funding

All authors declare no funding for this contribution.

References

1

Mudd
JO
,
Kass
DA
.
Tackling heart failure in the twenty-first century
.
Nature
2008
;
451
:
919
928
. https://doi.org/10.1038/nature06798

2

Rossignol
P
,
Hernandez
AF
,
Solomon
SD
,
Zannad
F
.
Heart failure drug treatment
.
Lancet
2019
;
393
:
1034
1044
. https://doi.org/10.1016/S0140-6736(18)31808-7

3

Lewis
KS
,
Butler
J
,
Bauersachs
J
,
Sandner
P
. The three-decade long journey in heart failure drug development. In:
Bauersachs
J
,
Butler
J
,
Sandner
P
, editors.
Heart Failure [Internet]
.:
Springer International Publishing
;
2017
,
1
14
.
[cited 2022 Oct 30]. (Handbook of Experimental Pharmacology). Available from:
https://doi.org/10.1007/164_2016_101

4

Borlaug
BA
,
Paulus
WJ
.
Heart failure with preserved ejection fraction: pathophysiology, diagnosis, and treatment
.
Eur Heart J
2011
;
32
:
670
679
. https://doi.org/10.1093/eurheartj/ehq426

5

McMurray
JJV
,
Solomon
SD
,
Inzucchi
SE
,
Køber
L
,
Kosiborod
MN
,
Martinez
FA
, et al.
Dapagliflozin in patients with heart failure and reduced ejection fraction
.
N Engl J Med
2019
;
381
:
1995
2008
. https://doi.org/10.1056/NEJMoa1911303

6

Solomon
SD
,
de Boer
RA
,
DeMets
D
,
Hernandez
AF
,
Inzucchi
SE
,
Kosiborod
MN
, et al.
Dapagliflozin in heart failure with preserved and mildly reduced ejection fraction: rationale and design of the DELIVER trial
.
Eur J Heart Fail
2021
;
23
:
1217
1225
. https://doi.org/10.1002/ejhf.2249

7

Packer
M
,
Anker
SD
,
Butler
J
,
Filippatos
G
,
Pocock
SJ
,
Carson
P
, et al.
Cardiovascular and renal outcomes with empagliflozin in heart failure
.
N Engl J Med
2020
;
383
:
1413
1424
. https://doi.org/10.1056/NEJMoa2022190

8

Anker
SD
,
Butler
J
,
Filippatos
G
,
Ferreira
JP
,
Bocchi
E
,
Böhm
M
, et al.
Empagliflozin in heart failure with a preserved ejection fraction
.
N Engl J Med
2021
;
385
:
1451
1461
. https://doi.org/10.1056/NEJMoa2107038

9

Bhatt
DL
,
Szarek
M
,
Steg
PG
,
Cannon
CP
,
Leiter
LA
,
McGuire
DK
, et al.
Sotagliflozin in patients with diabetes and recent worsening heart failure
.
N Engl J Med
2021
;
384
:
117
128
. https://doi.org/10.1056/NEJMoa2030183

10

Voors
AA
,
Angermann
CE
,
Teerlink
JR
,
Collins
SP
,
Kosiborod
M
,
Biegus
J
, et al.
The SGLT2 inhibitor empagliflozin in patients hospitalized for acute heart failure: a multinational randomized trial
.
Nat Med
2022
;
28
:
568
574
. https://doi.org/10.1038/s41591-021-01659-1

11

Mishra
S
,
Kass
DA
.
Cellular and molecular pathobiology of heart failure with preserved ejection fraction
.
Nat Rev Cardiol
2021
;
18
:
400
423
. https://doi.org/10.1038/s41569-020-00480-6

12

Ouzounian
M
,
Lee
DS
,
Liu
PP
.
Diastolic heart failure: mechanisms and controversies
.
Nat Rev Cardiol
2008
;
5
:
375
386
. https://doi.org/10.1038/ncpcardio1245

13

Lam
CSP
,
Voors
AA
,
de Boer
RA
,
Solomon
SD
,
van Veldhuisen
DJ
.
Heart failure with preserved ejection fraction: from mechanisms to therapies
.
Eur Heart J
2018
;
39
:
2780
2792
. https://doi.org/10.1093/eurheartj/ehy301

14

McDonagh
TA
,
Metra
M
,
Adamo
M
,
Gardner
RS
,
Baumbach
A
,
Böhm
M
, et al.
2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: developed by the task force for the diagnosis and treatment of acute and chronic heart failure of the European society of cardiology (ESC) with the special contribution of the heart failure association (HFA) of the ESC
.
Eur Heart J
2021
;
42
:
3599
3726
.

15

Dunlay
SM
,
Roger
VL
,
Redfield
MM
.
Epidemiology of heart failure with preserved ejection fraction
.
Nat Rev Cardiol
2017
;
14
:
591
602
. https://doi.org/10.1038/nrcardio.2017.65

16

Zile
MR
,
Baicu
CF
,
Gaasch
WH
.
Diastolic heart failure—abnormalities in active relaxation and passive stiffness of the left ventricle
.
N Engl J Med
2004
;
350
:
1953
1959
. https://doi.org/10.1056/NEJMoa032566

17

Lam
CSP
,
Donal
E
,
Kraigher-Krainer
E
,
Vasan
RS
.
Epidemiology and clinical course of heart failure with preserved ejection fraction
.
Eur J Heart Fail
2011
;
13
:
18
28
. https://doi.org/10.1093/eurjhf/hfq121

18

Rossi
A
,
Gheorghiade
M
,
Triposkiadis
F
,
Solomon
SD
,
Pieske
B
,
Butler
J
.
Left atrium in heart failure with preserved ejection fraction
.
Circulation Heart Failure
2014
;
7
:
1042
1049
. https://doi.org/10.1161/CIRCHEARTFAILURE.114.001276

19

Sarma
S
,
Stoller
D
,
Hendrix
J
,
Howden
E
,
Lawley
J
,
Livingston
S
, et al.
Mechanisms of chronotropic incompetence in heart failure with preserved ejection fraction
.
Circ Heart Fail
2020
;
13
:
e006331
. https://doi.org/10.1161/CIRCHEARTFAILURE.119.006331

20

Simmonds
SJ
,
Cuijpers
I
,
Heymans
S
,
Jones
EAV
.
Cellular and molecular differences between HFpEF and HFrEF: a step ahead in an improved pathological understanding
.
Cells
2020
;
9
:
242
. https://doi.org/10.3390/cells9010242

21

Murphy
SP
,
Kakkar
R
,
McCarthy
CP
,
Januzzi
JL
.
Inflammation in heart failure
.
J Am Coll Cardiol
2020
;
75
:
1324
1340
. https://doi.org/10.1016/j.jacc.2020.01.014

22

Gevaert
AB
,
Boen
JRA
,
Segers
VF
,
Van Craenenbroeck
EM
.
Heart failure with preserved ejection fraction: a review of cardiac and noncardiac pathophysiology
.
Front Physiol
2019
;
10
:
638
. https://doi.org/10.3389/fphys.2019.00638

23

Zile
MR
,
Baicu
CF
,
Ikonomidis
JS
,
Stroud
RE
,
Nietert
PJ
,
Bradshaw
AD
, et al.
Myocardial stiffness in patients with heart failure and a preserved ejection fraction: contributions of collagen and titin
.
Circulation
2015
;
131
:
1247
1259
. https://doi.org/10.1161/CIRCULATIONAHA.114.013215

24

Miranda-Silva
D
,
Lima
T
,
Rodrigues
P
,
Leite-Moreira
A
,
Falcão-Pires
I
.
Mechanisms underlying the pathophysiology of heart failure with preserved ejection fraction: the tip of the iceberg
.
Heart Fail Rev
2021
;
26
:
453
478
. https://doi.org/10.1007/s10741-020-10042-0

25

Verma
S
,
McMurray
JJV
.
The serendipitous story of SGLT2 inhibitors in heart failure
.
Circulation
2019
;
139
:
2537
2541
. https://doi.org/10.1161/CIRCULATIONAHA.119.040514

26

Zinman
B
,
Wanner
C
,
Lachin
JM
,
Fitchett
D
,
Bluhmki
E
,
Hantel
S
, et al.
Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes
.
N Engl J Med
2015
;
373
:
2117
2128
. https://doi.org/10.1056/NEJMoa1504720

27

Fukuta
H
,
Hagiwara
H
,
Kamiya
T
.
Sodium-glucose cotransporter 2 inhibitors in heart failure with preserved ejection fraction: a meta-analysis of randomized controlled trials
.
Int J Cardiol Heart Vasc
2022
;
42
:
101103
.

28

Cardoso
R
,
Graffunder
FP
,
Ternes
CMP
,
Fernandes
A
,
Rocha
AV
,
Fernandes
G
, et al.
SGLT2 inhibitors decrease cardiovascular death and heart failure hospitalizations in patients with heart failure: a systematic review and meta-analysis
.
EClinicalMedicine
2021
;
36
:
100933
. https://doi.org/10.1016/j.eclinm.2021.100933

29

Heerspink
HJL
,
Stefánsson
BV
,
Correa-Rotter
R
,
Chertow
GM
,
Greene
T
,
Hou
FF
, et al.
Dapagliflozin in patients with chronic kidney disease
.
N Engl J Med
2020
;
383
:
1436
1446
. https://doi.org/10.1056/NEJMoa2024816

30

Mahaffey
KW
,
Jardine
MJ
,
Bompoint
S
,
Cannon
CP
,
Neal
B
,
Heerspink
HJL
, et al.
Canagliflozin and cardiovascular and renal outcomes in type 2 diabetes mellitus and chronic kidney disease in primary and secondary cardiovascular prevention groups
.
Circulation
2019
;
140
:
739
750
. https://doi.org/10.1161/CIRCULATIONAHA.119.042007

31

Bhatt
DL
,
Szarek
M
,
Pitt
B
,
Cannon
CP
,
Leiter
LA
,
McGuire
DK
, et al.
Sotagliflozin in patients with diabetes and chronic kidney disease
.
N Engl J Med
2021
;
384
:
129
139
. https://doi.org/10.1056/NEJMoa2030186

32

Vaduganathan
M
,
Docherty
KF
,
Claggett
BL
,
Jhund
PS
,
de Boer
RA
,
Hernandez
AF
, et al.
SGLT-2 inhibitors in patients with heart failure: a comprehensive meta-analysis of five randomised controlled trials
.
Lancet
2022
;
400
:
757
767
. https://doi.org/10.1016/S0140-6736(22)01429-5

33

The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial
.
Lancet
1999
;
353
:
9
13
. https://doi.org/10.1016/S0140-6736(98)11181-9

34

Pitt
B
,
Zannad
F
,
Remme
WJ
,
Cody
R
,
Castaigne
A
,
Perez
A
, et al.
The effect of spironolactone on morbidity and mortality in patients with severe heart failure
.
N Engl J Med
1999
;
341
:
709
717
. https://doi.org/10.1056/NEJM199909023411001

35

Shah
A
,
Gandhi
D
,
Srivastava
S
,
Shah
KJ
,
Mansukhani
R
.
Heart failure: a class review of pharmacotherapy
.
P T
2017
;
42
:
464
472
.

36

Solomon
SD
,
McMurray
JJV
,
Anand
IS
,
Ge
J
,
Lam
CSP
,
Maggioni
AP
, et al.
Angiotensin–neprilysin inhibition in heart failure with preserved ejection fraction
.
N Engl J Med
2019
;
381
:
1609
1620
. https://doi.org/10.1056/NEJMoa1908655

37

Yusuf
S
,
Pfeffer
MA
,
Swedberg
K
,
Granger
CB
,
Held
P
,
McMurray
JJV
, et al.
Effects of candesartan in patients with chronic heart failure and preserved left-ventricular ejection fraction: the CHARM-preserved trial
.
Lancet
2003
:
362
:
777
781
. https://doi.org/10.1016/S0140-6736(03)14285-7

38

Pitt
B
,
Pfeffer
MA
,
Assmann
SF
,
Boineau
R
,
Anand
IS
,
Claggett
B
, et al.
Spironolactone for heart failure with preserved ejection fraction
.
N Engl J Med
2014
;
370
:
1383
1392
. https://doi.org/10.1056/NEJMoa1313731

39

Packer
M
,
Zannad
F
.
Do the favorable effects of digoxin and SGLT2 inhibitors really differ in patients with heart failure and a reduced ejection fraction? A provocative side-by-side examination of trial outcomes
.
J Card Fail
2022
;
28
:
682
683
. https://doi.org/10.1016/j.cardfail.2022.01.001

40

Jhund
PS
,
Kondo
T
,
Butt
JH
,
Docherty
KF
,
Claggett
BL
,
Desai
AS
, 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
. https://doi.org/10.1038/s41591-022-01971-4

41

Bonora
BM
,
Avogaro
A
,
Fadini
GP
.
Extraglycemic effects of SGLT2 inhibitors: a review of the evidence
.
Diabetes Metab Syndr Obes
2020
;
13
:
161
174
. https://doi.org/10.2147/DMSO.S233538

42

Packer
M
.
Critical examination of mechanisms underlying the reduction in heart failure events with SGLT2 inhibitors: identification of a molecular link between their actions to stimulate erythrocytosis and to alleviate cellular stress
.
Cardiovasc Res
2021
;
117
:
74
84
. https://doi.org/10.1093/cvr/cvaa064

43

Baker
WL
,
Buckley
LF
,
Kelly
MS
,
Bucheit
JD
,
Parod
ED
,
Brown
R
, et al.
Effects of sodium-glucose cotransporter 2 inhibitors on 24-hour ambulatory blood pressure: a systematic review and meta-analysis
.
J Am Heart Assoc
2017
;
6
:
e005686
. https://doi.org/10.1161/JAHA.117.005686

44

Miyata
KN
,
Zhang
SL
,
Chan
JSD
.
The rationale and evidence for SGLT2 inhibitors as a treatment for nondiabetic glomerular disease
.
Glomerular Dis
2021
;
1
:
21
33
. https://doi.org/10.1159/000513659

45

Cherney
DZ
,
Odutayo
A
,
Aronson
R
,
Ezekowitz
J
,
Parker
JD
.
Sodium glucose cotransporter-2 inhibition and cardiorenal protection: JACC review topic of the week
.
J Am Coll Cardiol
2019
;
74
:
2511
2524
. https://doi.org/10.1016/j.jacc.2019.09.022

46

Perkovic
V
,
Jardine
MJ
,
Neal
B
,
Bompoint
S
,
Heerspink
HJL
,
Charytan
DM
, et al.
Canagliflozin and renal outcomes in type 2 diabetes and nephropathy
.
N Engl J Med
2019
;
380
:
2295
2306
. https://doi.org/10.1056/NEJMoa1811744

47

Zelniker
TA
,
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
. https://doi.org/10.1016/j.jacc.2019.11.031

48

Zou
H
,
Zhou
B
,
Xu
G
.
SGLT2 inhibitors: a novel choice for the combination therapy in diabetic kidney disease
.
Cardiovasc Diabetol
2017
;
16
:
65
. https://doi.org/10.1186/s12933-017-0547-1

49

Ronco
C
,
Haapio
M
,
House
AA
,
Anavekar
N
,
Bellomo
R
.
Cardiorenal syndrome
.
J Am Coll Cardiol
2008
;
52
:
1527
1539
. https://doi.org/10.1016/j.jacc.2008.07.051

50

Verma
A
,
Patel
AB
,
Waikar
SS
.
SGLT2 inhibitor: not a traditional diuretic for heart failure
.
Cell Metab
2020
;
32
:
13
14
. https://doi.org/10.1016/j.cmet.2020.06.014

51

Mebazaa
A
,
Solal
AC
,
Colombo
PC
.
Assessing and treating congestion in acute decompensated heart failure: are we seeing the light at the end of the tunnel?
Eur Heart J
2023
;
44
:
51
53
. https://doi.org/10.1093/eurheartj/ehac680

52

Griffin
M
,
Rao
VS
,
Ivey-Miranda
J
,
Fleming
J
,
Mahoney
D
,
Maulion
C
, et al.
Empagliflozin in heart failure
.
Circulation
2020
;
142
:
1028
1039
. https://doi.org/10.1161/CIRCULATIONAHA.120.045691

53

Damman
K
,
Beusekamp
JC
,
Boorsma
EM
,
Swart
HP
,
Smilde
TDJ
,
Elvan
A
, et al.
Randomized, double-blind, placebo-controlled, multicentre pilot study on the effects of empagliflozin on clinical outcomes in patients with acute decompensated heart failure (EMPA-RESPONSE-AHF)
.
Eur J Heart Fail
2020
;
22
:
713
722
. https://doi.org/10.1002/ejhf.1713

54

Biegus
J
,
Voors
AA
,
Collins
SP
,
Kosiborod
MN
,
Teerlink
JR
,
Angermann
CE
, et al.
Impact of empagliflozin on decongestion in acute heart failure: the EMPULSE trial
.
Eur Heart J
2023
;
44
:
41
50
. https://doi.org/10.1093/eurheartj/ehac530

55

Packer
M
,
Anker
SD
,
Butler
J
,
Filippatos
G
,
Ferreira
JP
,
Pocock
SJ
, et al.
Empagliflozin in patients with heart failure, reduced ejection fraction, and volume overload
.
J Am Coll Cardiol
2021
;
77
:
1381
1392
. https://doi.org/10.1016/j.jacc.2021.01.033

56

Cunningham
JW
,
Vaduganathan
M
,
Claggett
BL
,
Kulac
IJ
,
Desai
AS
,
Jhund
PS
, et al.
Dapagliflozin in patients recently hospitalized with heart failure and mildly reduced or preserved ejection fraction
.
J Am Coll Cardiol
2022
;
80
:
1302
1310
. https://doi.org/10.1016/j.jacc.2022.07.021

57

Masson
W
,
Lavalle-Cobo
A
,
Nogueira
JP
.
Effect of SGLT2-inhibitors on epicardial adipose tissue: a meta-analysis
.
Cells
2021
;
10
:
2150
. https://doi.org/10.3390/cells10082150

58

Tosaki
T
,
Kamiya
H
,
Himeno
T
,
Kato
Y
,
Kondo
M
,
Toyota
K
, et al.
Sodium-glucose co-transporter 2 inhibitors reduce the abdominal visceral fat area and may influence the renal function in patients with type 2 diabetes
.
Intern Med
2017
;
56
:
597
604
. https://doi.org/10.2169/internalmedicine.56.7196

59

DeFronzo
RA
,
Norton
L
,
Abdul-Ghani
M
.
Renal, metabolic and cardiovascular considerations of SGLT2 inhibition
.
Nat Rev Nephrol
2017
;
13
:
11
26
. https://doi.org/10.1038/nrneph.2016.170

60

Gao
YM
,
Feng
ST
,
Wen
Y
,
Tang
TT
,
Wang
B
,
Liu
BC
.
Cardiorenal protection of SGLT2 inhibitors—perspectives from metabolic reprogramming
.
eBioMedicine
2022
;
83
:
104215
. https://doi.org/10.1016/j.ebiom.2022.104215

61

Hoong
CWS
,
Chua
MWJ
.
SGLT2 inhibitors as calorie restriction mimetics: insights on longevity pathways and age-related diseases
.
Endocrinology
2021
;
162
:
bqab079
. https://doi.org/10.1210/endocr/bqab079

62

Packer
M
.
Autophagy stimulation and intracellular sodium reduction as mediators of the cardioprotective effect of sodium–glucose cotransporter 2 inhibitors
.
Eur J Heart Fail
2020
;
22
:
618
628
. https://doi.org/10.1002/ejhf.1732

63

Esterline
RL
,
Vaag
A
,
Oscarsson
J
,
Vora
J
.
MECHANISMS IN ENDOCRINOLOGY: SGLT2 inhibitors: clinical benefits by restoration of normal diurnal metabolism?
Eur J Endocrinol
2018
;
178
:
R113
R125
. https://doi.org/10.1530/EJE-17-0832

64

Osataphan
S
,
Macchi
C
,
Singhal
G
,
Chimene-Weiss
J
,
Sales
V
,
Kozuka
C
, et al.
SGLT2 inhibition reprograms systemic metabolism via FGF21-dependent and -independent mechanisms
.
JCI Insight
2019
;
4
:
e123130
. https://doi.org/10.1172/jci.insight.123130

65

Packer
M
.
Role of deranged energy deprivation signaling in the pathogenesis of cardiac and renal disease in states of perceived nutrient overabundance
.
Circulation
2020
;
141
:
2095
2105
. https://doi.org/10.1161/CIRCULATIONAHA.119.045561

66

Lopaschuk
GD
,
Verma
S
.
Mechanisms of cardiovascular benefits of sodium glucose co-transporter 2 (SGLT2) inhibitors
.
JACC Basic Transl Sci
2020
;
5
:
632
644
. https://doi.org/10.1016/j.jacbts.2020.02.004

67

Verma
S
.
Potential mechanisms of sodium-glucose co-transporter 2 inhibitor-related cardiovascular benefits
.
Am J Med
2019
;
132
:
S39
S48
. https://doi.org/10.1016/j.amjmed.2019.08.007

68

Saucedo-Orozco
H
,
Voorrips
SN
,
Yurista
SR
,
de Boer
RA
,
Westenbrink
BD
.
SGLT2 inhibitors and ketone metabolism in heart failure
.
J Lipid Atheroscler
2022
;
11
:
1
19
. https://doi.org/10.12997/jla.2022.11.1.1

69

De Meyer
GRY
,
De Keulenaer
GW
,
Martinet
W
.
Role of autophagy in heart failure associated with aging
.
Heart Fail Rev
2010
;
15
:
423
430
. https://doi.org/10.1007/s10741-010-9166-6

70

Du
J
,
Liu
Y
,
Fu
J.
Autophagy and heart failure. In:
Le
W
, editor.
Autophagy: Biology and Diseases: Clinical Science [Internet]
.
Singapore
:
Springer
;
2020
,
223
7
.
[cited 2022 Nov 1]. (Advances in Experimental Medicine and Biology). Available from:
https://doi.org/10.1007/978-981-15-4272-5_16

71

Warbrick
I
,
Rabkin
SW
.
Hypoxia-inducible factor 1-alpha (HIF-1α) as a factor mediating the relationship between obesity and heart failure with preserved ejection fraction
.
Obes Rev
2019
;
20
:
701
712
. https://doi.org/10.1111/obr.12828

72

Koyani
CN
,
Plastira
I
,
Sourij
H
,
Hallström
S
,
Schmidt
A
,
Rainer
PP
, et al.
Empagliflozin protects heart from inflammation and energy depletion via AMPK activation
.
Pharmacol Res
2020
;
158
:
104870
. https://doi.org/10.1016/j.phrs.2020.104870

73

Zhou
H
,
Wang
S
,
Zhu
P
,
Hu
S
,
Chen
Y
,
Ren
J
.
Empagliflozin rescues diabetic myocardial microvascular injury via AMPK-mediated inhibition of mitochondrial fission
.
Redox Biol
2018
;
15
:
335
346
. https://doi.org/10.1016/j.redox.2017.12.019

74

Hawley
SA
,
Ford
RJ
,
Smith
BK
,
Gowans
GJ
,
Mancini
SJ
,
Pitt
RD
, et al.
The Na+/glucose cotransporter inhibitor canagliflozin activates AMPK by inhibiting mitochondrial function and increasing cellular AMP levels
.
Diabetes
2016
;
65
:
2784
2794
. https://doi.org/10.2337/db16-0058

75

Yang
L
,
Liang
B
,
Li
J
,
Zhang
X
,
Chen
H
,
Sun
J
, et al.
Dapagliflozin alleviates advanced glycation end product induced podocyte injury through AMPK/mTOR mediated autophagy pathway
.
Cell Signal
2022
;
90
:
110206
. https://doi.org/10.1016/j.cellsig.2021.110206

76

Ren
FF
,
Xie
ZY
,
Jiang
YN
,
Guan
X
,
Chen
QY
,
Lai
TF
, et al.
Dapagliflozin attenuates pressure overload-induced myocardial remodeling in mice via activating SIRT1 and inhibiting endoplasmic reticulum stress
.
Acta Pharmacol Sin
2022
;
43
:
1721
1732
. https://doi.org/10.1038/s41401-021-00805-2

77

Packer
M
.
Cardioprotective effects of sirtuin-1 and its downstream effectors
.
Circ Heart Fail
2020
;
13
:
e007197
. https://doi.org/10.1161/CIRCHEARTFAILURE.120.007197

78

Yang
Z
,
Li
T
,
Xian
J
,
Chen
J
,
Huang
Y
,
Zhang
Q
, et al.
SGLT2 inhibitor dapagliflozin attenuates cardiac fibrosis and inflammation by reverting the HIF-2α signaling pathway in arrhythmogenic cardiomyopathy
.
FASEB J
2022
;
36
:
e22410
. https://doi.org/10.1096/fj.202200243R

79

Bessho
R
,
Takiyama
Y
,
Takiyama
T
,
Kitsunai
H
,
Takeda
Y
,
Sakagami
H
, et al.
Hypoxia-inducible factor-1α is the therapeutic target of the SGLT2 inhibitor for diabetic nephropathy
.
Sci Rep
2019
;
9
:
14754
. https://doi.org/10.1038/s41598-019-51343-1

80

Packer
M
.
Critical reanalysis of the mechanisms underlying the cardiorenal benefits of SGLT2 inhibitors and reaffirmation of the nutrient deprivation signaling/autophagy hypothesis
.
Circulation
2022
;
146
:
1383
1405
. https://doi.org/10.1161/CIRCULATIONAHA.122.061732

81

Zannad
F
,
Ferreira
JP
,
Butler
J
,
Filippatos
G
,
Januzzi
JL
,
Sumin
M
, et al.
Effect of empagliflozin on circulating proteomics in heart failure: mechanistic insights from the EMPEROR program
.
Eur Heart J
2022
;
43
:
4991
5002
. https://doi.org/10.1093/eurheartj/ehac495

82

Packer
M
.
How can sodium–glucose cotransporter 2 inhibitors stimulate erythrocytosis in patients who are iron-deficient? Implications for understanding iron homeostasis in heart failure
.
Eur J Heart Fail
2022
;
24
:
2287
2296
. https://doi.org/10.1002/ejhf.2731

83

Anand
IS
,
Gupta
P
.
Anemia and iron deficiency in heart failure
.
Circulation
2018
;
138
:
80
98
. https://doi.org/10.1161/CIRCULATIONAHA.118.030099

84

Alnuwaysir
RIS
,
Hoes
MF
,
van Veldhuisen
DJ
,
van der Meer
P
,
Beverborg
NG
.
Iron deficiency in heart failure: mechanisms and pathophysiology
.
J Clin Med
2021
;
11
:
125
. https://doi.org/10.3390/jcm11010125

85

Hoes
MF
,
Grote Beverborg
N
,
Kijlstra
JD
,
Kuipers
J
,
Swinkels
DW
,
Giepmans
BNG
, et al.
Iron deficiency impairs contractility of human cardiomyocytes through decreased mitochondrial function
.
Eur J Heart Fail
2018
;
20
:
910
919
. https://doi.org/10.1002/ejhf.1154

86

Docherty
KF
,
Curtain
JP
,
Anand
IS
,
Bengtsson
O
,
Inzucchi
SE
,
Køber
L
, et al.
Effect of dapagliflozin on anaemia in DAPA-HF
.
Eur J Heart Fail
2021
;
23
:
617
628
. https://doi.org/10.1002/ejhf.2132

87

Docherty
KF
,
Welsh
P
,
Verma
S
,
De Boer
RA
,
O’Meara
E
,
Bengtsson
O
, et al.
Iron deficiency in heart failure and effect of dapagliflozin: findings from DAPA-HF
.
Circulation
2022
;
146
:
980
994
. https://doi.org/10.1161/CIRCULATIONAHA.122.060511

88

Fuchs Andersen
C
,
Omar
M
,
Glenthøj
A
,
El Fassi
D
,
Møller
HJ
,
Lindholm Kurtzhals
JA
, et al.
Effects of empagliflozin on erythropoiesis in heart failure: data from the Empire HF trial
.
Eur J Heart Fail
2023
;
25
:
226
234
. https://doi.org/10.1002/ejhf.2735

89

von Haehling
S
,
Sato
R
,
Butler
J
,
Anker
SD
.
Little at a time: trying to understand the battery of benefits of sodium–glucose contransporter 2 inhibitors in heart failure
.
Eur J Heart Fail
2023
;
25
:
235
237
. https://doi.org/10.1002/ejhf.2766

90

Mazer
CD
,
Hare
GMT
,
Connelly
PW
,
Gilbert
RE
,
Shehata
N
,
Quan
A
, 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
. https://doi.org/10.1161/CIRCULATIONAHA.119.044235

91

Swedberg
K
,
Young
JB
,
Anand
IS
,
Cheng
S
,
Desai
AS
,
Diaz
R
, et al.
Treatment of anemia with darbepoetin alfa in systolic heart failure
.
N Engl J Med
2013
;
368
:
1210
1219
. https://doi.org/10.1056/NEJMoa1214865

92

Pfeffer
MA
,
Burdmann
EA
,
Chen
CY
,
Cooper
ME
,
de Zeeuw
D
,
Eckardt
KU
, et al.
A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease
.
N Engl J Med
2009
;
361
:
2019
2032
. https://doi.org/10.1056/NEJMoa0907845

93

Segar
MW
,
Kolkailah
AA
,
Frederich
R
,
Pong
A
,
Cannon
CP
,
Cosentino
F
, 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
. https://doi.org/10.1111/dom.14769

94

Fitchett
D
,
Inzucchi
SE
,
Zinman
B
,
Wanner
C
,
Schumacher
M
,
Schmoor
C
, et al.
Mediators of the improvement in heart failure outcomes with empagliflozin in the EMPA-REG OUTCOME trial
.
ESC Heart Fail
2021
;
8
:
4517
4527
. https://doi.org/10.1002/ehf2.13615

95

Li
J
,
Woodward
M
,
Perkovic
V
,
Figtree
GA
,
Heerspink
HJL
,
Mahaffey
KW
, et al.
Mediators of the effects of canagliflozin on heart failure in patients with type 2 diabetes
.
JACC Heart Fail
2020
;
8
:
57
66
. https://doi.org/10.1016/j.jchf.2019.08.004

96

Xu
B
,
Li
S
,
Kang
B
,
Zhou
J
.
The current role of sodium-glucose cotransporter 2 inhibitors in type 2 diabetes mellitus management
.
Cardiovasc Diabetol
2022
;
21
:
83
. https://doi.org/10.1186/s12933-022-01512-w

97

Yang
Y
,
Zhao
C
,
Ye
Y
,
Yu
M
,
Qu
X
.
Prospect of sodium–glucose co-transporter 2 inhibitors combined with insulin for the treatment of type 2 diabetes
.
Front Endocrinol (Lausanne)
2020
;
11
:
190
. https://doi.org/10.3389/fendo.2020.00190

98

Yaribeygi
H
,
Sathyapalan
T
,
Maleki
M
,
Jamialahmadi
T
,
Sahebkar
A
.
Molecular mechanisms by which SGLT2 inhibitors can induce insulin sensitivity in diabetic milieu: a mechanistic review
.
Life Sci
2020
;
240
:
117090
. https://doi.org/10.1016/j.lfs.2019.117090

99

Son
TK
,
Toan
NH
,
Thang
N
,
Le Trong Tuong
H
,
Tien
HA
,
Thuy
NH
, et al.
Prediabetes and insulin resistance in a population of patients with heart failure and reduced or preserved ejection fraction but without diabetes, overweight or hypertension
.
Cardiovasc Diabetol
2022
;
21
:
75
. https://doi.org/10.1186/s12933-022-01509-5

100

Savji
N
,
Meijers
WC
,
Bartz
TM
,
Bhambhani
V
,
Cushman
M
,
Nayor
M
, et al.
The association of obesity and cardiometabolic traits with incident HFpEF and HFrEF
.
JACC Heart Failure
2018
;
6
:
701
709
. https://doi.org/10.1016/j.jchf.2018.05.018

101

Bailey
CJ
.
Uric acid and the cardio-renal effects of SGLT2 inhibitors
.
Diabetes Obes Metab
2019
;
21
:
1291
1298
. https://doi.org/10.1111/dom.13670

102

Iacobellis
G
.
Epicardial adipose tissue in endocrine and metabolic diseases
.
Endocrine
2014
;
46
:
8
15
. https://doi.org/10.1007/s12020-013-0099-4

103

Iacobellis
G
,
Bianco
AC
.
Epicardial adipose tissue: emerging physiological, pathophysiological and clinical features
.
Trends Endocrinol Metab
2011
;
22
:
450
457
. https://doi.org/10.1016/j.tem.2011.07.003

104

van Woerden
G
,
van Veldhuisen
DJ
,
Manintveld
OC
,
van Empel
VPM
,
Willems
TP
,
de Boer
RA
, et al.
Epicardial adipose tissue and outcome in heart failure with mid-range and preserved ejection fraction
.
Circ Heart Fail
2022
;
15
:
e009238
. https://doi.org/10.1161/CIRCHEARTFAILURE.121.009238

105

Pugliese
NR
,
Paneni
F
,
Mazzola
M
,
De Biase
N
,
Del Punta
L
,
Gargani
L
, et al.
Impact of epicardial adipose tissue on cardiovascular haemodynamics, metabolic profile, and prognosis in heart failure
.
Eur J Heart Fail
2021
;
23
:
1858
1871
. https://doi.org/10.1002/ejhf.2337

106

Kalman
E
,
Chan
J
,
Goel
V
,
Yeong
C
,
Nelson
A
,
Nerlekar
N
.
SGLT2 inhibitors reduce epicardial adipose tissue: a meta-analysis
.
Heart Lung Circ
2022
;
31
:
S179
S180
. https://doi.org/10.1016/j.hlc.2022.06.286

107

Wu
P
,
Wen
W
,
Li
J
,
Xu
J
,
Zhao
M
,
Chen
H
, et al.
Systematic review and meta-analysis of randomized controlled trials on the effect of SGLT2 inhibitor on blood leptin and adiponectin level in patients with type 2 diabetes
.
Horm Metab Res
2019
;
51
:
487
494
. https://doi.org/10.1055/a-0958-2441

108

Packer
M
.
Do sodium-glucose co-transporter-2 inhibitors prevent heart failure with a preserved ejection fraction by counterbalancing the effects of leptin? A novel hypothesis
.
Diabetes Obes Metab
2018
;
20
:
1361
1366
. https://doi.org/10.1111/dom.13229

109

Scisciola
L
,
Cataldo
V
,
Taktaz
F
,
Fontanella
RA
,
Pesapane
A
,
Ghosh
P
, et al.
Anti-inflammatory role of SGLT2 inhibitors as part of their anti-atherosclerotic activity: data from basic science and clinical trials
.
Fron Cardiovasc Med
2022
;
9
:
1008922
. https://doi.org/10.3389/fcvm.2022.1008922

110

Chen
S
,
Coronel
R
,
Hollmann
MW
,
Weber
NC
,
Zuurbier
CJ
.
Direct cardiac effects of SGLT2 inhibitors
.
Cardiovasc Diabetol
2022
;
21
:
45
. https://doi.org/10.1186/s12933-022-01480-1

111

Theofilis
P
,
Sagris
M
,
Oikonomou
E
,
Antonopoulos
AS
,
Siasos
G
,
Tsioufis
K
, et al.
Anti-inflammatory potential of SGLT2 inhibitors: a systematic review and meta-analysis of preclinical studies in rodents
.
Eur Heart J
2022
;
43
:
2683
. https://doi.org/10.1093/eurheartj/ehac544.2683

112

Xing
YJ
,
Liu
BH
,
Wan
SJ
,
Cheng
Y
,
Zhou
SM
,
Sun
Y
, et al.
A SGLT2 inhibitor dapagliflozin alleviates diabetic cardiomyopathy by suppressing high glucose-induced oxidative stress in vivo and in vitro
.
Front Pharmacol
2021
;
12
:
708177
. https://doi.org/10.3389/fphar.2021.708177

113

Li
C
,
Zhang
J
,
Xue
M
,
Li
X
,
Han
F
,
Liu
X
, et al.
SGLT2 inhibition with empagliflozin attenuates myocardial oxidative stress and fibrosis in diabetic mice heart
.
Cardiovasc Diabetol
2019
;
18
:
15
. https://doi.org/10.1186/s12933-019-0816-2

114

Elrakaybi
A
,
Laubner
K
,
Zhou
Q
,
Hug
MJ
,
Seufert
J
.
Cardiovascular protection by SGLT2 inhibitors—do anti-inflammatory mechanisms play a role?
Mol Metab
2022
;
64
:
101549
. https://doi.org/10.1016/j.molmet.2022.101549

115

Tong
Y
,
Wang
Z
,
Cai
L
,
Lin
L
,
Liu
J
,
Cheng
J
.
NLRP3 inflammasome and its central role in the cardiovascular diseases
.
Oxid Med Cell Longev
2020
;
2020
:
4293206
. https://doi.org/10.1155/2020/4293206

116

Kim
SR
,
Lee
SG
,
Kim
SH
,
Kim
JH
,
Choi
E
,
Cho
W
, et al.
SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease
.
Nat Commun
2020
;
11
:
2127
. https://doi.org/10.1038/s41467-020-15983-6

117

Byrne
NJ
,
Matsumura
N
,
Maayah
ZH
,
Ferdaoussi
M
,
Takahara
S
,
Darwesh
AM
, et al.
Empagliflozin blunts worsening cardiac dysfunction associated with reduced NLRP3 (nucleotide-binding domain-like receptor protein 3) inflammasome activation in heart failure
.
Circ Heart Fail
2020
;
13
:
e006277
. https://doi.org/10.1161/CIRCHEARTFAILURE.119.006277

118

Leng
W
,
Wu
M
,
Pan
H
,
Lei
X
,
Chen
L
,
Wu
Q
, et al.
The SGLT2 inhibitor dapagliflozin attenuates the activity of ROS-NLRP3 inflammasome axis in steatohepatitis with diabetes mellitus
.
Ann Transl Med
2019
;
7
:
429
. https://doi.org/10.21037/atm.2019.09.03

119

Yaribeygi
H
,
Katsiki
N
,
Butler
AE
,
Sahebkar
A
.
Effects of antidiabetic drugs on NLRP3 inflammasome activity, with a focus on diabetic kidneys
.
Drug Discov Today
2019
;
24
:
256
262
. https://doi.org/10.1016/j.drudis.2018.08.005

120

Durante
W
,
Behnammanesh
G
,
Peyton
KJ
.
Effects of sodium-glucose co-transporter 2 inhibitors on vascular cell function and arterial remodeling
.
Int J Mol Sci
2021
;
22
:
8786
. https://doi.org/10.3390/ijms22168786

121

Hess
DA
,
Terenzi
DC
,
Trac
JZ
,
Quan
A
,
Mason
T
,
Al-Omran
M
, et al.
SGLT2 inhibition with empagliflozin increases circulating provascular progenitor cells in people with type 2 diabetes mellitus
.
Cell Metab
2019
;
30
:
609
613
. https://doi.org/10.1016/j.cmet.2019.08.015

122

Mone
P
,
Lombardi
A
,
Kansakar
U
,
Varzideh
F
,
Jankauskas
SS
,
Pansini
A
, et al.
Empagliflozin improves the microRNA signature of endothelial dysfunction in patients with heart failure with preserved ejection fraction and diabetes
.
J Pharmacol Exp Ther
2023
;
384
:
116
122
. https://doi.org/10.1124/jpet.121.001251

123

Despa
S
,
Islam
MA
,
Weber
CR
,
Pogwizd
SM
,
Bers
DM
.
Intracellular Na(+) concentration is elevated in heart failure but Na/K pump function is unchanged
.
Circulation
2002
;
105
:
2543
2548
. https://doi.org/10.1161/01.CIR.0000016701.85760.97

124

Luo
M
,
Anderson
ME
.
Mechanisms of altered Ca2+ handling in heart failure
.
Circ Res
2013
;
113
:
690
708
. https://doi.org/10.1161/CIRCRESAHA.113.301651

125

Kaplan
A
,
Abidi
E
,
El-Yazbi
A
,
Eid
A
,
Booz
GW
,
Zouein
FA
.
Direct cardiovascular impact of SGLT2 inhibitors: mechanisms and effects
.
Heart Fail Rev
2018
;
23
:
419
437
. https://doi.org/10.1007/s10741-017-9665-9

126

Baartscheer
A
,
Schumacher
CA
,
Wüst
RCI
,
Fiolet
JWT
,
Stienen
GJM
,
Coronel
R
, et al.
Empagliflozin decreases myocardial cytoplasmic Na+ through inhibition of the cardiac Na+/H+ exchanger in rats and rabbits
.
Diabetologia
2017
;
60
:
568
573
. https://doi.org/10.1007/s00125-016-4134-x

127

Verma
S
,
McMurray
JJV
.
SGLT2 inhibitors and mechanisms of cardiovascular benefit: a state-of-the-art review
.
Diabetologia
2018
;
61
:
2108
2117
. https://doi.org/10.1007/s00125-018-4670-7

128

Arjun
S
,
Bell
RM
.
SGLT2 inhibitors: reviving the sodium-hydrogen exchanger cardioprotection hypothesis?
Cardiovasc Res
2019
;
115
:
1454
1456
. https://doi.org/10.1093/cvr/cvz105

129

De Pascalis
A
,
Cianciolo
G
,
Capelli
I
,
Brunori
G
,
La Manna
G
.
SGLT2 inhibitors, sodium and off-target effects: an overview
.
J Nephrol
2021
;
34
:
673
680
. https://doi.org/10.1007/s40620-020-00845-7

130

Nakamura
TY
,
Iwata
Y
,
Arai
Y
,
Komamura
K
,
Wakabayashi
S
.
Activation of Na+/H+ exchanger 1 is sufficient to generate Ca2 + signals that induce cardiac hypertrophy and heart failure
.
Circ Res
2008
;
103
:
891
899
. https://doi.org/10.1161/CIRCRESAHA.108.175141

131

Karmazyn
M
.
Therapeutic potential of Na-H exchange inhibitors for the treatment of heart failure
.
Expert Opin Investig Drugs
2001
;
10
:
835
843
. https://doi.org/10.1517/13543784.10.5.835

132

Uthman
L
,
Baartscheer
A
,
Bleijlevens
B
,
Schumacher
CA
,
Fiolet
JWT
,
Koeman
A
, 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
. https://doi.org/10.1007/s00125-017-4509-7

133

Chung
YJ
,
Park
KC
,
Tokar
S
,
Eykyn
TR
,
Fuller
W
,
Pavlovic
D
, et al.
SGLT2 inhibitors and the cardiac Na+/H+ exchanger-1: the plot thickens
.
Cardiovasc Res
2021
;
117
:
2702
2704
. https://doi.org/10.1093/cvr/cvab184

134

Chung
YJ
,
Park
KC
,
Tokar
S
,
Eykyn
TR
,
Fuller
W
,
Pavlovic
D
, et al.
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
. https://doi.org/10.1093/cvr/cvaa323

135

Ye
Y
,
Jia
X
,
Bajaj
M
,
Birnbaum
Y
.
Dapagliflozin attenuates Na+/H+ exchanger-1 in cardiofibroblasts via AMPK activation
.
Cardiovasc Drugs Ther
2018
;
32
:
553
558
. https://doi.org/10.1007/s10557-018-6837-3

136

Jiang
D
,
Shi
H
,
Tonggu
L
,
Gamal El-Din
TM
,
Lenaeus
MJ
,
Zhao
Y
, et al.
Structure of the cardiac sodium channel
.
Cell
2020
;
180
:
122
134.e10
. https://doi.org/10.1016/j.cell.2019.11.041

137

Philippaert
K
,
Kalyaanamoorthy
S
,
Fatehi
M
,
Long
W
,
Soni
S
,
Byrne
NJ
, et al.
Cardiac late sodium channel current is a molecular target for the sodium/glucose cotransporter 2 inhibitor empagliflozin
.
Circulation
2021
;
143
:
2188
2204
. https://doi.org/10.1161/CIRCULATIONAHA.121.053350

138

Verma
S
,
Garg
A
,
Yan
AT
,
Gupta
AK
,
Al-Omran
M
,
Sabongui
A
, et al.
Effect of empagliflozin on left ventricular mass and diastolic function in individuals with diabetes: an important clue to the EMPA-REG OUTCOME trial?
Diabetes Care
2016
;
39
:
e212
e213
. https://doi.org/10.2337/dc16-1312

139

Shim
CY
,
Seo
J
,
Cho
I
,
Lee
CJ
,
Cho
IJ
,
Lhagvasuren
P
, et al.
Randomized, controlled trial to evaluate the effect of dapagliflozin on left ventricular diastolic function in patients with type 2 diabetes mellitus
.
Circulation
2021
;
143
:
510
512
. https://doi.org/10.1161/CIRCULATIONAHA.120.051992

140

Salah
HM
,
Verma
S
,
Santos-Gallego
CG
,
Bhatt
AS
,
Vaduganathan
M
,
Khan
MS
, et al.
Sodium-glucose cotransporter 2 inhibitors and cardiac remodeling
.
J Cardiovasc Trans Res
2022
;
15
:
944
956
. https://doi.org/10.1007/s12265-022-10220-5

141

Pabel
S
,
Wagner
S
,
Bollenberg
H
,
Bengel
P
,
Kovács
Á
,
Schach
C
, et al.
Empagliflozin directly improves diastolic function in human heart failure
.
Eur J Heart Fail
2018
;
20
:
1690
1700
. https://doi.org/10.1002/ejhf.1328

142

Pabel
S
,
Hamdani
N
,
Luedde
M
,
Sossalla
S
.
SGLT2 inhibitors and their mode of action in heart failure—has the mystery been unravelled?
Curr Heart Fail Rep
2021
;
18
:
315
328
. https://doi.org/10.1007/s11897-021-00529-8

143

Heerspink
HJL
,
Perco
P
,
Mulder
S
,
Leierer
J
,
Hansen
MK
,
Heinzel
A
, et al.
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
. https://doi.org/10.1007/s00125-019-4859-4

144

Quagliariello
V
,
De Laurentiis
M
,
Rea
D
,
Barbieri
A
,
Monti
MG
,
Carbone
A
, et al.
The SGLT-2 inhibitor empagliflozin improves myocardial strain, reduces cardiac fibrosis and pro-inflammatory cytokines in non-diabetic mice treated with doxorubicin
.
Cardiovasc Diabetol
2021
;
20
:
150
. https://doi.org/10.1186/s12933-021-01346-y

145

Xu
L
,
Nagata
N
,
Nagashimada
M
,
Zhuge
F
,
Ni
Y
,
Chen
G
, et al.
SGLT2 inhibition by empagliflozin promotes fat utilization and browning and attenuates inflammation and insulin resistance by polarizing m2 macrophages in diet-induced obese mice
.
EBioMedicine
2017
;
20
:
137
149
. https://doi.org/10.1016/j.ebiom.2017.05.028

146

Verma
S
,
Mazer
CD
,
Yan
AT
,
Mason
T
,
Garg
V
,
Teoh
H
, et al.
Effect of empagliflozin on left ventricular mass in patients with type 2 diabetes mellitus and coronary artery disease: the EMPA-HEART CardioLink-6 randomized clinical trial
.
Circulation
2019
;
140
:
1693
1702
. https://doi.org/10.1161/CIRCULATIONAHA.119.042375

147

Dhingra
NK
,
Mistry
N
,
Puar
P
,
Verma
R
,
Anker
S
,
Mazer
CD
, et al.
SGLT2 inhibitors and cardiac remodelling: a systematic review and meta-analysis of randomized cardiac magnetic resonance imaging trials
.
ESC Heart Failure
2021
;
8
:
4693
4700
. https://doi.org/10.1002/ehf2.13645

148

ClinicalTrials.gov
.
Dapagliflozin and Effect on Cardiovascular Events in Acute Heart Failure -Thrombolysis in Myocardial Infarction 68 (DAPA ACT HF-TIMI 68). NCT04363697. Available from
: https://clinicaltrials.gov/ct2/show/NCT04363697

149

Pandey
AK
,
Okaj
I
,
Kaur
H
,
Belley-Cote
EP
,
Wang
J
,
Oraii
A
, et al.
Sodium-glucose co-transporter inhibitors and atrial fibrillation: a systematic review and meta-analysis of randomized controlled trials
.
J Am Heart Assoc
2021
;
10
:
e022222
. https://doi.org/10.1161/JAHA.121.022222

150

ClinicalTrials.gov
.
Empagliflozin and Atrial Fibrillation Treatment (EMPA-AF). NCT04583813. Available from
: https://clinicaltrials.gov/ct2/show/NCT04583813

151

ClinicalTrials.gov
.
Use of Dapagliflozin to Reduce Burden of Atrial Fibrillation in Patients Undergoing Catheter Ablation of Symptomatic Atrial Fibrillation (DAPA-AF). NCT04792190. Available from
: https://clinicaltrials.gov/ct2/show/NCT04792190

152

ClinicalTrials.gov
.
EMPACT-MI: A Study to Test Whether Empagliflozin Can Lower the Risk of Heart Failure and Death in People Who Had a Heart Attack (Myocardial Infarction). NCT04509674. Available from
: https://clinicaltrials.gov/ct2/show/NCT04509674

153

ClinicalTrials.gov
.
Dapagliflozin Effects on Cardiovascular Events in Patients With an Acute Heart Attack (DAPA-MI). NCT04564742. Available from
: https://clinicaltrials.gov/ct2/show/NCT04564742

154

Agarwal
R
,
Filippatos
G
,
Pitt
B
,
Anker
SD
,
Rossing
P
,
Joseph
A
, et al.
Cardiovascular and kidney outcomes with finerenone in patients with type 2 diabetes and chronic kidney disease: the FIDELITY pooled analysis
.
Eur Heart J
2022
;
43
:
474
484
. https://doi.org/10.1093/eurheartj/ehab777

155

Brown
E
,
Heerspink
HJL
,
Cuthbertson
DJ
,
Wilding
JPH
.
SGLT2 inhibitors and GLP-1 receptor agonists: established and emerging indications
.
Lancet
2021
;
398
:
262
276
. https://doi.org/10.1016/S0140-6736(21)00536-5

156

ClinicalTrials.gov
.
Efficacy, Safety and Tolerability of AZD9977 and Dapagliflozin in Participants With Heart Failure and Chronic Kidney Disease (MIRACLE). NCT04595370. Available from
: https://clinicaltrials.gov/ct2/show/NCT04595370

157

ClinicalTrials.gov
.
A Study to Learn How Well the Treatment Combination of Finerenone and Empagliflozin Works and How Safe it is Compared to Each Treatment Alone in Adult Participants With Long-term Kidney Disease (Chronic Kidney Disease) and Type 2 Diabetes (CONFIDENCE). NCT05254002. Available from
: https://clinicaltrials.gov/ct2/show/NCT05254002

158

Pitt
B
,
Bhatt
DL
.
Does SGLT1 inhibition add benefit to SGLT2 inhibition in type 2 diabetes?
Circulation
2021
;
144
:
4
6
. https://doi.org/10.1161/CIRCULATIONAHA.121.054442

159

Takebayashi
K
,
Inukai
T
.
Effect of sodium glucose cotransporter 2 inhibitors with low SGLT2/SGLT1 selectivity on circulating glucagon-like peptide 1 levels in type 2 diabetes Mellitus
.
J Clin Med Res
2017
;
9
:
745
753
. https://doi.org/10.14740/jocmr3112w

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)