Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

European Heart Journal (2023) 00, 1–12 STATE OF THE ART REVIEW

https://doi.org/10.1093/eurheartj/ehad389 Heart failure and cardiomyopathies

Mechanisms of benefits of sodium-glucose


cotransporter 2 inhibitors in heart failure with
preserved ejection fraction
Arjun K. Pandey1, Deepak L. Bhatt 2, Avinash Pandey3, Nikolaus Marx 4
,
Francesco Cosentino 5,6, Ambarish Pandey7, and Subodh Verma 8*
1
Michael G. DeGroote School of Medicine, McMaster University, 90 Main Street West, Hamilton, Ontario L8P 1H6, Canada; 2Mount Sinai Heart, Icahn School of Medicine at Mount Sinai
Health System, 1 Gustave L. Levy Place, New York, NY 10029, USA; 3Department of Medicine, University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, Ontario K1Y 4W7, Canada;
4
Department of Internal Medicine, University Hospital Aachen, RWTH Aachen University, Templergraben 55, 52062 Aachen, Germany; 5Division of Cardiology, Department of Medicine,
Solna, Karolinska Institutet, Norrbacka S1:02, Stockholm, SE 17177, Sweden; 6Heart, Vascular and Neuro Theme, Department of Cardiology, Karolinska University Hospital, Anna Steckséns
gata 41, 171 64 Solna, Sweden; 7Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; and
8
Division of Cardiac Surgery, Li Ka Shing Knowledge Institute of St. Michael’s Hospital, University of Toronto, 30 Bond Street, Toronto, ON, Canada

Received 9 December 2022; revised 7 March 2023; accepted 29 May 2023

Abstract

For decades, heart failure with preserved ejection fraction (HFpEF) proved an elusive entity to treat. Sodium-glucose cotransporter 2 (SGLT2) in-
hibitors 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 pro-
gression 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 in-
direct 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 in-
hibitors 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.

* Corresponding author. Tel: +4168645997, Fax: +4168645881, Email: subodh.verma@unityhealth.to


© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com
2 Pandey et al.

Graphical Abstract

HFpEF and diastolic Mechanisms of SGLT2 inhibitors


dysfunction in HFpEF

Oxidative stress, inflammation, fibrosis Improved NO-sGC-cGMP-PKG signaling

Improved metabolism and energetics


Cardiac remodelling and hypertrophy

Reduced renal deterioration


Myofilament stiffness
Increased autophagic flux

Impaired energetics
Improved diastolic function

Ionic imbalances Reduced oxidative stress and inflammation

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.
.............................................................................................................................................................................................
Keywords Heart Failure • SGLT2 Inhibitor • HFpEF • Diabetes Mellitus

Heart failure is associated with an impaired quality of life, frequent hos- as clinically distinct phenotypes.14 Some patients with an ejection frac-
pitalizations, and increased mortality.1 Disease-modifying therapies for tion >40% have clinical signs and symptoms of heart failure secondary
heart failure have traditionally consisted of neurohormonal modulators, to another disorder such as infiltrative cardiomyopathies (including
which inhibit the renin–angiotensin–aldosterone (RAAS) or adrenergic hemochromatosis or amyloidosis) or other cardiac conditions such
systems.2,3 In patients with heart failure and a reduced ejection fraction as valvular heart disease, hypertrophic cardiomyopathy, or pericardial
(HFrEF), these therapies have proved unambiguously beneficial, result- disease.15 In patients with such conditions, the mechanisms described
ing in striking reductions in mortality.3 The benefit of such therapies in below may not be applicable.
patients who have heart failure and a preserved ejection fraction There are a variety of pathophysiological processes, which contrib-
(HFpEF) has been less robust.3,4 Prognosis in these patients has re- ute to the syndrome of HFpEF (Figure 1); there is likely heterogeneity
mained poor and effective; disease-modifying therapies have remained in the degree to which specific mechanisms contribute to disease in in-
elusive.3,4 Over the past decade, sodium-glucose cotransporter 2 dividual patients. Diastolic dysfunction is a hallmark of HFpEF; the land-
(SGLT2) inhibitors have shown clinical benefit in patients with heart fail- mark invasive study of Zile et al.16 showed impaired active relaxation
ure across the entire spectrum of ejection fraction, and among patients and increased passive stiffness in patients with HFpEF. Adverse remod-
with worsening heart failure and acute heart failure.5–10 Several land- elling and dysfunction are not just seen in the left ventricle but also in
mark trials, DELIVER, EMPEROR-Preserved, and SOLOIST-WHF, the atria and vasculature.4,11,12,17,18 This may be accompanied by other
showed significant reductions in the composite of heart failure hospital- processes, which contribute to the progression of HFpEF including neu-
ization/cardiovascular death with these agents in patients with rohormonal activation, chronotropic incompetence, atrial arrythmias,
HFpEF.6,8,9 In this state-of-the-art review, we will summarize the recent and renal dysfunction.4,11–13,18,19
results of SGLT2 inhibitors, discuss mechanisms of these agents in Cellular and molecular mechanisms underlying HFpEF have been
HFpEF, as well as future directions. thoroughly reviewed in the past.11,13,20–22 Adverse changes to cardio-
myocyte structure, function, and energetics in addition to changes in the
extracellular matrix contribute to diastolic dysfunction in HFpEF.23 One
Mechanisms of heart failure with key mediator of intrinsic cardiomyocyte stiffness in HFpEF is the giant elas-
tic, cytoskeletal protein titin; dysregulation of the NO-sGC-cGMP-PKG
preserved ejection fraction pathway contributes to titin hypophosphorylation and isoform switching
The mechanisms and pathophysiology underlying HFpEF have been between the compliant N2BA and stiff N2B states.13,22 Increased collagen-
reviewed in detail previously and will only be briefly summarized dependent stiffness secondary to inflammatory and fibrotic processes
here.4,11–13 HFpEF traditionally described patients with an ejection frac- also contribute to diastolic dysfunction in HFpEF.13,22 Finally, alterations
tion greater than 40%, although heart failure with mildly reduced in cardiomyocyte energetics and ionic homeostasis are other key mechan-
(∼40%–49%) and improved ejection have recently been characterized isms that may also contribute to HFpEF.13,24
Mechanisms of benefits of SGLT2 inhibitors in heart failure with preserved ejection fraction 3

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

Sodium-glucose cotransporter 2 to be consistent across the spectrum of mildly reduced or preserved


ejection fraction (41%–49%, 50%–60%, and >60%) as well as in a var-
inhibitors in HFpEF: a serendipitous iety of sub-groups based on sex, race, and presence of comorbidities
discovery such as diabetes mellitus or atrial fibrillation.6,8,32
While a five-trial meta-analysis of patients with heart failure across
The SGLT2 inhibitors were originally investigated in patients with type the entire ejection fraction spectrum did show a reduction in all-cause
2 diabetes mellitus. These agents were found to improve key outcomes mortality with SGLT2 inhibitors, in a two-trial meta-analysis of only pa-
including cardiovascular death, and unexpectedly reduced hospitaliza- tients with HFpEF (EMPEROR-Preserved and DELIVER), no significant
tions for heart failure as well.25,26 Subgroup analyses of patients with reduction in all-cause mortality and only a statistically borderline effect
heart failure, including HFpEF specifically, suggested that these agents on cardiovascular death was observed.32 The magnitude of effect and
may improve outcomes in these settings.27,28 Clinical evaluation of absolute reduction in heart failure event rates with SGLT2 inhibitors
these agents proceeded to patients with HFrEF as well as those with was lower than that seen in trials of RAAS inhibitors or beta-blockers
chronic kidney disease (CKD). The SGLT2 inhibitors proved beneficial in HFrEF patients, many of which were stopped early due to clear evi-
in HFrEF, reducing the composite of heart failure hospitalization/cardio- dence of benefit with treatment.33–35 The clinical benefits with SGLT2
vascular death in the EMPEROR-Reduced and DAPA-HF trials.5,7 These inhibitors in heart failure were notably, however, observed in trials in
agents also slowed the rate of kidney function deterioration as well as which the vast majority of patients were already being treated with
improved heart failure outcomes in patients with CKD.29–31 In the both an angiotensin-converting enzyme inhibitor/angiotensin receptor
SOLOIST-WHF and EMPULSE trials, which included patients across blocker/angiotensin receptor–neprilysin inhibitor (∼80%–95%) and a
the spectrum of ejection fraction, SGLT2 inhibitors improved out- beta-blocker (∼80%–95%) with a significant proportion of patients
comes in patients with worsening heart failure or acute heart failure, treated with a mineralocorticoid receptor antagonist (∼35%–70%) as
respectively.9,10 Finally, the landmark EMPEROR-Preserved and well.32 Heart failure hospitalization/cardiovascular death or a similar
DELIVER trials both demonstrated significant reductions in their pri- composite has been the primary outcome of many large trials evaluating
mary outcomes (cardiovascular death or hospitalization [+/− urgent other classes of heart failure therapies in HFpEF (including
visit] for heart failure) with SGLT2 inhibitors in HFpEF in addition to CHARM-Preserved, TOPCAT, and PARAGON-HF).36–38
modest improvements in symptom burden measured by the Kansas Hospitalization due to heart failure was, however, only one cause for
City Cardiomyopathy Questionnaire score.6,8 These results appear hospital admission in patients with HFpEF, a population of patients
4 Pandey et al.

often with many comorbidities and risk factors for other cardiac and here are believed to be a reduction in trans-glomerular pressure and
non-cardiac events.39 regulation of tubule-glomerular feedback to protect kidney func-
In summary, over the past decade, SGLT2 inhibitors have emerged un- tion.29,31,44–47 This is distinct and may be synergistic with the mechan-
expectedly as a therapy for heart failure. These drugs were subsequently ism of RAAS inhibitors, which reduce hyperfiltration through efferent
prospectively evaluated in over 12 000 patients with HFpEF in two land- vasodilation.45,48 Cardiorenal effects play a major role in heart failure:
mark Phase III trials where they proved efficacious. As these trial results kidney dysfunction worsens heart failure, and impaired cardiovascular
are incorporated into clinical guidelines around the world, SGLT2 inhibi- function impairs kidney perfusion and function.47,49 This interplay is
tors are likely to become important mainstays of therapy in HFpEF. mediated through several important factors, many of which are key mar-
kers and therapeutic targets in heart failure including natriuretic peptides
and the adrenergic and RAAS systems.49 Improvement of kidney function
Mechanisms of sodium-glucose 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.
cotransporter 2 inhibitors in heart The SGLT2 inhibitors were equally beneficial in patients with estimated
failure with preserved ejection glomerular filtration rate below and above 60 mL/min/1.73 m2 in
EMPEROR-Preserved and DELIVER, and have been shown to improve
fraction cardiovascular outcomes throughout the spectrum of kidney function.5,8
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 Natriuresis/diuresis
investigations in preclinical models, mechanistic trials including evalu- Natriuresis/diuresis may be an important contributor to the clinical im-
ation of proteomics and cardiac remodelling in human patients, in add- provement observed with SGLT2 inhibitors in patients with acute heart
ition to extrapolation from trials conducted in other populations (such failure.50–52 In the EMPA-RESPONSE-AHF trial, empagliflozin signifi-
as HFrEF and diabetes mellitus/cardiovascular disease). Given the re- cantly increased urine output and net fluids loss in patients with acutely
markable consistency in benefits of SGLT2 inhibitors observed in pa- decompensated heart failure over the first 4 days of hospital admission
tients with heart failure across the entire spectrum of reduced to compared with placebo.53 In the EMPULSE trial of patients admitted to
preserved ejection fraction, there is likely significant overlap in the hospital for acute heart failure, empagliflozin was associated with signifi-
underlying mechanisms of SGLT2 inhibitors in patients with HFrEF cant weight loss by Day 15, which was sustained to Day 90 (including
and those with HFpEF.32,40 These two entities share many common after adjustment for daily loop diuretic dose).54 Significant improve-
risk factors (including hypertension, diabetes mellitus, and CKD), and ments compared with placebo in both clinical scores and markers
while amelioration of these traditional risk factors may play some (haematocrit and N-terminal pro-B-type natriuretic peptide) of con-
role in the robust effect of these medications, translational research gestion were also observed with empagliflozin by Day 15.54 The acute
as well as the rapid time course of clinical benefit and remodelling decongestion observed with SGLT2 inhibitors has led to proposed fra-
seen with these drugs suggest there are additional mechanisms at play. meworks for incorporating SGLT2 inhibitors into the management of
Here, we will summarize mechanisms, which may contribute to the patients presenting with acutely decompensated heart failure alongside
benefit of SGLT2 inhibitors in HFpEF with a focus on direct and indirect other diuretics like furosemide and acetazolamide.51
myocardial effects related to metabolism, energetics, autophagic flux, While natriuresis/diuresis may explain the effect of SGLT2 inhibitors
ionic homeostasis, inflammation, oxidative stress, and cardiac remodel- in patients with acute heart failure and may contribute to the rapid
ling (Figure 2). 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
Blood pressure, blood sugar, and EMPEROR-Reduced trial, empagliflozin was not more effective in patients
kidney function initially identified as having clinical evidence of volume overload (n = 1477)
compared with those initially characterized as euvolemic, and decline in
The SGLT2 inhibitors improve many traditional risk factors for HFpEF weight with empagliflozin was similar in patients with and without volume
including blood pressure and blood sugar.41 However, these properties overload.42,55 In the DELIVER trial, which evaluated dapagliflozin, there was
alone likely do not explain the benefits of this class of drugs in HFpEF.42 a similar relative reduction (but greater absolute reduction) in the primary
The SGLT2 inhibitors have been shown to reduce systolic blood pressure outcome of worsening heart failure event/cardiovascular death in patients
by an average 4 mmHg as assessed using 24-h ambulatory monitoring.43 In with recent heart failure hospitalization compared with those without.56
the EMPEROR-Preserved trial, empagliflozin had similar benefits in pa- There was no relation between changes in body weight and natriuretic
tients with baseline systolic blood pressure below and above the median.8 peptides in EMPEROR-Reduced suggesting against diuresis-induced reduc-
With regard to blood sugar, other agents that lower glucose to a similar or tion in volume overload as the underlying cause of weight loss.55 Sustained
even greater degree do not have such potent benefits in heart failure.42 weight reduction with SGLT2 inhibitors may instead be related to loss of
Relative effects of SGLT2 inhibitors in HFpEF in EMPEROR-Preserved calories through glycosuria and reduction in visceral fat.55,57,58
and DELIVER were near identical in patients with and without diabetes
mellitus.6,8
The cardiorenal effects of these drugs are an important consider- Metabolism, energetics, and
ation given the strong association between CKD and HFpEF. In Phase
III trials, SGLT2 inhibitors have been shown to reduce the rate of de-
autophagic flux
cline in kidney function as well as to reduce kidney failure/death in pa- The SGLT2 membrane protein resides in the proximal convoluted tu-
tients with CKD with or without diabetes; the underlying mechanisms bule and is responsible for 90% of glucose reabsorption from the filtrate
Mechanisms of benefits of SGLT2 inhibitors in heart failure with preserved ejection fraction 5

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

under physiological conditions.59 Renal glycosuria through SGLT2 in- reduction in oxidative stress.60,62,63 This cellular process is impaired
hibition represents a net loss of calories from the body, and some in the failing myocardium, and induction of autophagy by SGLT2 inhibi-
have suggested that the SGLT2 protein may also function as a physio- tors may contribute to the improvements in cardiac function observed
logical sensor of nutrient surplus.42,60–62 Inhibition of SGLT2 is there- with these drugs.69–71 Several of the most widely discussed cellular
fore proposed to induce a state of perceived starvation and hypoxia, mediators thought to mediate this effect are the nutrient-deprivation
up-regulating signals to induce a ‘fasting’ and ‘hypoxic’-like transcrip- sensors AMPK and sirtuins (SIRT1, SIRT3, and SIRT6) as well as
tional paradigm.42,60–65 This results in adaptions such as increased hypoxia-inducible factors (HIF) and mammalian target of rapamycin
erythropoietin (EPO) levels and ketogenesis.42,60–62 The majority of (mTOR). Regulation of these mediators, which are all involved in maintain-
cardiac energy is normally generated from fatty acid oxidation; in the ing homeostasis of energy, metabolism, and oxygen, is dysfunctional in
failing heart, fatty acid oxidation is reduced, and alternative pathways heart failure, which contributes to the development of cardiomyop-
including glucose metabolism and anaerobic glycolysis are relied athy.65,69 Mechanistic studies demonstrate that SGLT2 inhibitors increase
upon.66,67 The SGLT2 inhibitors increase serum ketone bodies, which the activities of AMPK, sirtuins, and HIFs and decrease phosphorylation/ac-
represent additional source of energy for the failing heart.66,68 Ketone tivation of mTOR in the myocardium as well as in other tissues, which may
bodies have been shown to increase cardiac contractility and have im- at least partially underlie their benefit on organ function.72–80 The totality
portant signalling roles as well, triggering cellular processes like of evidence on this topic has recently been reviewed in great detail.80 The
autophagy.66,68 mechanisms by which SGLT2 inhibitors promote autophagy in tissues that
The downstream effects of the fasting and hypoxic-like transcription- do not express the SGLT2 protein have yet to definitively established; pro-
al paradigms have been suggested to be a key aspect of SGLT2 inhibi- posed but unproven mechanisms include nutrient-deprivation signalling
tors, which may be mediated at a cellular level through autophagic secondary to caloric loss, or direct effects of these drugs on other cellular
flux.60,62,63 This is a cellular degradation process mediated by lysosomes transporters/proteins such as glucose transporters or sirtuins.80 In sum-
by which cells recycle organelles, proteins, and debris to generate en- mary, the proposed paradigm is as follows: SGLT2 inhibition enhances au-
ergy, and is triggered in response to cellular stress including perceived tophagic flux and induces a fasting/hypoxic mimicry state resulting in
nutritional depletion and hypoxia. Induction of autophagy can also con- activation of mediators including AMPK, sirtuins, and HIF, which promote
tribute to degradation of dysfunctional mitochondria and subsequent ketosis and erythropoiesis that lead to reductions in oxidative stress and
6 Pandey et al.

inflammation as well as improved cellular function. This emerging mechan- is associated with poor cardiac function and future risk of HFpEF.96–100 The
ism may explain the beneficial effects of SGLT2 inhibitors on metabolism SGLT2 inhibitors are also associated with reductions in serum uric acid, al-
and cellular function in tissues/organs (such as the heart) that do not ex- though the degree to which this has any clinical significance is unclear.41,101
press the SGLT2 protein and could be a contributing factor to the reduc- Uric acid may increase oxidative stress, inflammation, and endothelial dys-
tion in oxidative stress and inflammation observed with these agents. function and is associated with a poor prognosis in heart failure.41,101
Recent proteomics analysis from the EMPEROR-Pooled analysis,
which included 535 patients from the EMPEROR-Preserved trial, as-
sessed differential levels of circulating proteins in patients randomized Adipose tissue
to empagliflozin compared with placebo at baseline, Week 12 and Epicardial adipose tissue (EAT) is a form of white, visceral adipose tissue
Week 52.81 This analysis showed that empagliflozin had substantial ef- around the heart with important paracrine/vasocrine signalling proper-
fects on circulating levels of proteins previously shown to be mediators ties.102,103 Expansion of this tissue in patients with diabetes mellitus and/
of cardiac autophagy and apoptosis (including IGFBP1, TfRI, FST, RBP2, or obesity induces states of insulin resistance, oxidative stress, inflamma-
and Mdk) in addition to EPO and other circulating proteins related to tion, and fibrosis and alters calcium homeostasis, all of which may contrib-
fibrosis, oxidative stress, hypertrophy, and energetics.81 ute to diastolic dysfunction.102,103 Expansion of EAT has been associated
The effect of SGLT2 inhibitors on iron homeostasis and erythrocy- with a poor prognosis and worse haemodynamic profiles in patients
tosis has proved to be an intriguing development and has recently with HFpEF.104,105 In meta-analyses, SGLT2 inhibitors have been asso-
been reviewed in detail.82 Iron deficiency is a common comorbidity ciated with reductions in EAT despite having no significant effects on total
among patients with heart failure and is associated with reduced func- body mass index.57,106 The reduction in EAT may contribute to the attenu-
tional status and poor outcomes.83,84 Increasingly, evidence suggests ation of myocardial inflammation and fibrosis observed with SGLT2 inhi-
that iron deficiency in patients with heart failure may be more likely bitors. In addition to reducing visceral fat, SGLT2 inhibitors also
to be functional (i.e. related to inflammatory-mediated increases in hep- modulate adipokines levels, increasing levels of adiponectin, an insulin-
cidin, resulting in suppression of duodenal iron absorption and release sensitizing and anti-inflammatory hormone, and reducing levels of leptin,
from hepatocytes and the reticulocyte-endothelial system) rather than a hormone released by adipose tissue in states of nutritional excess.107
absolute in nature.82 In either case, reduced circulating and bioreactive Paracrine leptin release from epicardial tissue impairs calcium homeostasis
cytosolic iron contributes to both anaemia and may impair synthesis of and induces cardiac fibrosis and microcirculatory dysfunction.108
iron-containing proteins involved in ATP production in cardiomyo-
cytes.82,85 In a preclinical study, iron deficiency has been directly shown
to impair contractility, cellular ATP levels, and ATP-linked respiration in Inflammation and oxidative stress
isolated human cardiomyocytes.85 In several placebo-controlled trials The SGLT2 inhibitors are known to decrease oxidative stress and inflamma-
including analyses from DAPA-HF and the EMPEROR programme, tion.109,110 The SGLT2 inhibitors reduce levels of circulating pro-inflammatory
SGLT2 inhibitors have been consistently shown to decrease circulating factors such as C-reactive protein, tumour necrosis factor-α, and
levels of both hepcidin and ferritin, while increasing levels of transferrin interleukin-6.111 They also reduce markers of oxidative stress and reactive
receptor protein 1.81,82,86–89 This combination of findings suggests oxygen species including hydrogen peroxide in the myocardium.112,113 The at-
SGLT2 inhibitors may alleviate functional iron deficiency, enabling in- tenuation of oxidative stress and inflammation associated with SGLT2 inhibi-
creased iron mobilization and augmenting bioreactive cytosolic iron le- tors is likely multifactorial, and a number of mechanisms including decreased
vels in erythroid precursors and cardiomyocytes.82 While further uric acid, reduced epicardial fat, alterations in adipokine levels and up-regulation
research is needed to clarify the mechanisms by which SGLT2 inhibitors of autophagy all may play a role.114 Another important dimension is the
alter iron homeostasis, possible contributors include the reductions in NLRP3 inflammasome, a multimeric cytoplasmic protein complex in macro-
inflammatory mediators (which may impact ferritin and hepcidin levels) phages, which plays an important role in the chronic inflammation of heart fail-
as well as downstream effects of nutrient-deprivation signalling (includ- ure and atherosclerosis.115–117 Treatment with SGLT2 inhibitors inhibits
ing SIRT1) induced by SGLT2 inhibition.82 The SGLT2 inhibitors have activation of the NLRP3 inflammasome, an effect which may be in part
also been consistently shown to increase EPO levels, doing so in as little mediated by the increased ketosis and decreased uric acid associated with
as 1 month of treatment initiation in the EMPA-HEART CardioLink-6 these agents.91,92 Reduced NLRP3 activity results in decreased macrophage
trial.90 In the DAPA-HF trial, treatment with dapagliflozin led to rises infiltration and pro-inflammatory cytokine release.115–117 This effect may
in haematocrit to the point of correction into the non-anemic range not be limited to the heart, with data showing inhibition of the NLRP3 inflam-
in 62.2% of patients with anaemia at baseline (compared with 41.1% masome by SGLT2 inhibitors in the kidney, liver, and vasculature.117–119
in the placebo arm).86 Intriguingly, while treatment with EPO analogues Within the vasculature, SGLT2 inhibitors have been shown to increase nitric
has not been shown to improve outcomes in heart failure or patients oxide bioavailability, attenuate endothelial dysfunction, and increase circulating
with diabetes mellitus and CKD, the degree of erythrocytosis/increase levels of pro-angiogenic progenitor cells.120,121 In a head-to-head comparison,
in haematocrit has been strongly associated with improved heart fail- treatment with empagliflozin compared with insulin or metformin was recent-
ure outcomes in the CANVAS program as well as the VERTIS-CV and ly shown to be associated with with improvement in microRNA profiles as-
EMPA-REG OUTCOME trials.91–95 While correction of anaemia may sociated with endothelial dysfunction in patients with HFpEF and diabetes
result in improved oxygen delivery to the failing myocardium, the as- mellitus.122
sociation 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 Ion handling/homeostasis: Na+
for iron-dependent proteins in cardiomyocytes) as opposed to repre-
senting a direct causal mechanism.
and Ca2+
Other metabolic pathways may contribute to the effects of SGLT2 Features of ionic imbalance in heart failure include a state of calcium
inhibitors as well. The SGLT2 inhibitors attenuate insulin resistance, which overload as well as increased intracellular sodium.123,124 The SGLT2
Mechanisms of benefits of SGLT2 inhibitors in heart failure with preserved ejection fraction 7

inhibitors reduce intracellular sodium and cytosolic calcium, although


the mechanism underlying these changes has not been definitively es-
Diastolic function and remodelling
tablished.125–127 Sodium–hydrogen antiporters (NHE) are expressed Treatment with empagliflozin has been shown to improve diastolic
in various sites including the heart and the kidneys.128,129 The SGLT2 function after only 3 months in a small cohort of patients with diabetes
protein in the kidney interacts closely with NHE-3, which is a transport- and high cardiovascular risk.138 The IDDIA trial of patients with type 2
er primarily responsible for sodium reabsorption from the filtrate.62 diabetes mellitus and at least grade 1 left ventricular diastolic dysfunc-
Increased NHE-3 activity increases oxidative stress and adrenergic ac- tion showed improvements in left ventricular diastolic function, includ-
tivation, and increased sodium reabsorption by this transporter de- ing diastolic reserve, assessed by diastolic stress echocardiography after
creases distal sodium delivery to the macula densa, leading to 24 weeks of treatment with dapagliflozin compared with placebo.139 As
glomerular hyperfiltration and destruction of nephrons.62 The NHE-3 discussed above, diastolic dysfunction in HFpEF has several compo-
is functionally intertwined with SGLT2 in the nephron, and knockout/ nents including intrinsic myofilament stiffness (related to titin regulation
inhibition of one impairs the activity of the other.62 This may in part ex- and NO-sGC-cGMP-PKG signalling) as well as extracellular matrix-
plain the nephroprotective effects of SGLT2 inhibitors. The NHE-1 is related stiffness/myocardial fibrosis.23 It may be here where the various
expressed in cardiomyocytes and is up-regulated in the failing heart, in- pathways involved in SGLT2-mediated reductions in oxidative stress
creasing intracellular sodium and calcium.130,131 The SGLT2 inhibitors and inflammation converge. In animal models, SGLT2 inhibitors in-
reduce intracellular sodium despite the SGLT2 protein not being ex- crease NO bioavailability, and restore the pathologically altered
pressed in the heart.62,132 It has been postulated that direct, off-target phosphorylation of titin mediated through downstream cGMP-PKG
inhibition of NHE-1 by SGLT2 inhibitors could be responsible for im- signalling; this translates to improved diastolic function.140 In a mechan-
proved cardiac remodelling and reduced fibrosis with these agents. istic analysis of isolated human and murine myocardial tissue, SGLT2 in-
There is conflicting evidence about whether SGLT2 inhibitors can dir- hibitors were shown to reduce diastolic tension and passive
ectly inhibit the NHE-1 transporter in cardiomyocytes.62,133,134 The re- myofilament stiffness as well as alter phosphorylation of myofilament
duction in intracellular sodium with SGLT2 inhibitors observed could proteins including titin.141,142 Beyond direct cardiomyocyte effects,
instead be a downstream effect of other signalling pathways, and there SGLT2 inhibitors have been shown to reduce macrophage infiltration,
is some evidence that SGLT2 inhibitor may reduce NHE-1 mRNA ex- regulate macrophage polarization, inhibit cardiac fibroblast differenti-
pression, which may be mediated through an AMPK-dependent path- ation, and suppress fibrotic markers such as type 1 collagen, resulting
way.62,135 Another potential off-target effect of SGLT2 inhibitors is in attenuated fibrosis and adverse extracellular remodelling in the
interaction with the cardiac sodium channel Nav1.5 (also known as heart.66,78,109,113,118,143–145 In addition to reducing myofilament stiff-
SCN5A), which mediates late sodium current.136,137 A molecular study ness and fibrosis, SGLT2 inhibitors have been shown to reduce
showed that SGLT2 inhibitors may bind to the same site on Nav1.5 as hypertrophy measured by left ventricular mass index within only 6
known Class 1 antiarrhythmics such as lidocaine and inhibit late sodium months in patients with type 2 diabetes mellitus and coronary artery
current; whether this interaction has any clinical significance has yet to disease in the EMPA-HEART CardioLink-6 trial.146 A systematic review
be determined.137 These effects on ionic homeostasis could in part ex- of five randomized controlled trials showed that SGLT2 inhibitors were
plain the rapid time course of benefits observed with SGLT2 inhibitors. associated with left ventricular mass regression compared with

Figure 3 Ongoing and completed trials of SGLT2 inhibitors in patients with cardiovascular or renal disease. CKD, chronic kidney disease; CV, car-
diovascular; 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.
8 Pandey et al.

placebo, although none of the included trials were conducted in pa- which contribute to reduced adverse remodelling and diastolic dysfunc-
tients with HFpEF.147 tion. As recent trial results are incorporated into clinical guidelines,
SGLT2 inhibitors will become foundational in the therapy of HFpEF.

Future directions
Acknowledgements
While SGLT2 inhibitors have already proved to improve outcomes in
patients with HFpEF, there are several avenues of research that are on- The authors would like to thank Sana Khan for assistance with prepar-
going (Figure 3). ation of the figures.
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 ejec-
Declarations
tion fraction.148 This trial will help inform the most appropriate time Disclosure of Interest
point for initiation of these agents in patients with HFpEF. Atrial fibrillation
A.P. has no disclosures.
is known to be associated with HFpEF: each condition independently in-
D.L.B. discloses the following relationships—advisory board:
creases the risk factor for the other.15 The relative benefit of SGLT2 inhi-
AngioWave, Bayer, Boehringer Ingelheim, Cardax, CellProthera, Cereno
bitors in patients with HFpEF is similar in those with and without atrial
Scientific, Elsevier Practice Update Cardiology, High Enroll, Janssen, Level
fibrillation; however, given that patients with atrial fibrillation suffer from
Ex, McKinsey, Medscape Cardiology, Merck, MyoKardia, NirvaMed,
a higher rate of adverse heart failure events at baseline, the absolute benefit
Novo Nordisk, PhaseBio, PLx Pharma, Regado Biosciences, Stasys; board
may likely be greater.149 The SGLT2 inhibitors have also been shown to
of directors: AngioWave (stock options), Boston VA Research Institute,
reduce the rate of atrial fibrillation events in analyses of adverse event re-
Bristol Myers Squibb (stock), DRS.LINQ (stock options), High Enroll
porting from randomized controlled trials.149 DAPA-AF and EMPA-AF are
(stock), Society of Cardiovascular Patient Care, TobeSoft; chair:
two ongoing randomized trials, which will quantify the effects of SGLT2
Inaugural Chair, American Heart Association Quality Oversight Committee;
inhibitors on atrial fibrillation burden.150,151 Finally, there are also ongoing
consultant: Broadview Ventures; data monitoring committees:
trials (DAPA-MI and EMPACT-MI) evaluating the effects of SGLT2 inhibi-
Acesion Pharma, Assistance Publique-Hôpitaux de Paris, Baim
tors following myocardial infarction.152,153
Institute for Clinical Research (formerly Harvard Clinical Research
Two other classes of medications, which have emerged over the past
Institute, for the PORTICO trial, funded by St. Jude Medical, now
decade and have also shown benefit in patients with diabetes and/or kidney
Abbott), Boston Scientific (chair, PEITHO trial), Cleveland Clinic (in-
disease, are glucagon-like peptide-1 receptor agonists and non-steroidal
cluding for the ExCEED trial, funded by Edwards), Contego Medical
mineralocorticoid receptor antagonists.154,155 Evaluation of potential syn-
(chair, PERFORMANCE 2), Duke Clinical Research Institute, Mayo
ergy of these classes of agents with SGLT2 inhibitors is an area of interest.
Clinic, Mount Sinai School of Medicine (for the ENVISAGE trial, funded
Two trials, MIRACLE and CONFIDENCE, will evaluate the effects of the
by Daiichi Sankyo; for the ABILITY-DM trial, funded by Concept
combination of SGLT2 inhibitors with non-steroidal mineralocorticoid re-
Medical), Novartis, Population Health Research Institute; Rutgers
ceptor antagonists in patients with cardiorenal disease.156,157
University (for the NIH-funded MINT Trial); honoraria: American
Finally, SGLT1 inhibition, through molecules such as the SGLT1/2 in-
College of Cardiology (senior associate editor, Clinical Trials and
hibitor sotagliflozin, has been hypothesized to also have possible supple-
News, ACC.org; chair, ACC Accreditation Oversight Committee),
mentary benefits in heart failure as well.158 The SGLT1 is expressed in
Arnold and Porter law firm (work related to Sanofi/Bristol Myers
the small intestine where it contributes to glucose absorption, as well as
Squibb clopidogrel litigation), Baim Institute for Clinical Research (for-
in other sites including the kidney.158 Decreases in SGLT1-mediated glu-
merly Harvard Clinical Research Institute; RE-DUAL PCI clinical trial
cose absorption increase serum glucagon-like peptide-1 levels, and
steering committee funded by Boehringer Ingelheim; AEGIS-II execu-
SGLT1 inhibition may also inhibit hypertrophy and fibrosis within the myo-
tive committee funded by CSL Behring), Belvoir Publications (editor
cardium.158,159 The SGLT1/2 inhibitor sotagliflozin reduced myocardial in-
in chief, Harvard Heart Letter), Canadian Medical and Surgical
farction and stroke in the SCORED trial, a finding that has not been seen
Knowledge Translation Research Group (clinical trial steering commit-
with traditional SGLT2 inhibitors, and which has been postulated to be
tees), Cowen and Company, Duke Clinical Research Institute (clinical
mediated in part through increased endogenous incretin levels.31 There
trial steering committees, including the PRONOUNCE trial, funded
are no direct data comparing dual SGLT1/2 inhibitors with SGLT2 inhibi-
by Ferring Pharmaceuticals), HMP Global (editor in chief, Journal of
tors, but this may prove to be an area of interest in the future.
Invasive Cardiology), Journal of the American College of Cardiology
(guest editor; associate editor), K2P (co-chair, interdisciplinary curric-
ulum), Level Ex, Medtelligence/ReachMD (CME steering committees),
Conclusion MJH Life Sciences, Oakstone CME (course director, Comprehensive
In summary, HFpEF has proved an elusive entity to treat. With the re- Review of Interventional Cardiology), Piper Sandler, Population
sults of DELIVER, SOLOIST-WHF, and EMPEROR-Preserved, SGLT2 Health Research Institute (for the COMPASS operations committee,
inhibitors have solidified themselves as robust therapies to improve publications committee, steering committee, and USA national co-
outcomes in patients with HFpEF. The underlying mechanism of these leader, funded by Bayer), Slack Publications (chief medical editor,
agents cannot be wholly ascribed to improvements in traditional risk Cardiology Today’s Intervention), Society of Cardiovascular Patient
factors such as blood pressure, blood sugar, volume status, and renal Care (secretary/treasurer), WebMD (CME steering committees),
function. Rather, the rapid time course of improvement as well as ex- Wiley (steering committee); Other: Clinical Cardiology (deputy editor),
perimental evidence suggests that a combination of mechanisms for NCDR-ACTION Registry Steering Committee (chair), VA CART
SGLT2 inhibitors in HFpEF including induction of autophagy improved Research and Publications Committee (chair); Patent: Sotagliflozin
ionic homeostasis and reduced inflammation and oxidative stress, (named on a patent for sotagliflozin assigned to Brigham and Women’s
Mechanisms of benefits of SGLT2 inhibitors in heart failure with preserved ejection fraction 9

Hospital who assigned to Lexicon; neither I nor Brigham and Women’s 4. Borlaug BA, Paulus WJ. Heart failure with preserved ejection fraction: pathophysi-
Hospital receive any income from this patent); Research Funding: ology, diagnosis, and treatment. Eur Heart J 2011;32:670–679. https://doi.org/10.
1093/eurheartj/ehq426
Abbott, Acesion Pharma, Afimmune, Aker Biomarine, Amarin, Amgen, 5. McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod MN, Martinez FA, et al.
AstraZeneca, Bayer, Beren, Boehringer Ingelheim, Boston Scientific, Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med
Bristol Myers Squibb, Cardax, CellProthera, Cereno Scientific, Chiesi, 2019;381:1995–2008. https://doi.org/10.1056/NEJMoa1911303
CinCor, CSL Behring, Eisai, Ethicon, Faraday Pharmaceuticals, Ferring 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: ra-
Pharmaceuticals, Forest Laboratories, Fractyl, Garmin, HLS Therapeutics, tionale and design of the DELIVER trial. Eur J Heart Fail 2021;23:1217–1225. https://
Idorsia, Ironwood, Ischemix, Janssen, Javelin, Lexicon, Lilly, Medtronic, Merck, doi.org/10.1002/ejhf.2249
Moderna, MyoKardia, NirvaMed, Novartis, Novo Nordisk, Owkin, Pfizer, 7. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, et al. Cardiovascular
PhaseBio, PLx Pharma, Recardio, Regeneron, Reid Hoffman Foundation, and renal outcomes with empagliflozin in heart failure. N Engl J Med 2020;383:
1413–1424. https://doi.org/10.1056/NEJMoa2022190
Roche, Sanofi, Stasys, Synaptic, The Medicines Company, Youngene, 89Bio;
8. Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Böhm M, et al. Empagliflozin in
Royalties: Elsevier (editor, Braunwald’s Heart Disease); site co-investigator: heart failure with a preserved ejection fraction. N Engl J Med 2021;385:1451–1461.
Abbott, Biotronik, Boston Scientific, CSI, Endotronix, St. Jude Medical https://doi.org/10.1056/NEJMoa2107038
(now Abbott), Philips, SpectraWAVE, Svelte, Vascular Solutions; trustee: 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:
American College of Cardiology; Unfunded Research: FlowCo, Takeda.
117–128. https://doi.org/10.1056/NEJMoa2030183
A.P. has no disclosures. 10. Voors AA, Angermann CE, Teerlink JR, Collins SP, Kosiborod M, Biegus J, et al. The
N.M. has received support for clinical trial leadership from SGLT2 inhibitor empagliflozin in patients hospitalized for acute heart failure: a multi-
Boehringer Ingelheim, Novo Nordisk, served as a consultant to national randomized trial. Nat Med 2022;28:568–574. https://doi.org/10.1038/s41591-
Boehringer Ingelheim, Merck, Novo Nordisk, AstraZeneca, BMS, re- 021-01659-1
11. Mishra S, Kass DA. Cellular and molecular pathobiology of heart failure with preserved
ceived grant support from Boehringer Ingelheim, Merck, Novo ejection fraction. Nat Rev Cardiol 2021;18:400–423. https://doi.org/10.1038/s41569-
Nordisk, and served as a speaker for Boehringer Ingelheim, Merck, 020-00480-6
Novo Nordisk, Lilly, BMS, and AstraZeneca. He declines all personal 12. Ouzounian M, Lee DS, Liu PP. Diastolic heart failure: mechanisms and controversies.
compensation from pharma or device companies. 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
F.C. has served as a speaker and advisory board members for preserved ejection fraction: from mechanisms to therapies. Eur Heart J 2018;39:
AstraZeneca, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Lilly, 2780–2792. https://doi.org/10.1093/eurheartj/ehy301
Merck Sharp & Dohme, Novo Nordisk, and Pfizer; and has received re- 14. McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M, et al. 2021
search grants from the City Pharmacy, Abu Dhabi, UAE, King Gustav V ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: de-
veloped by the task force for the diagnosis and treatment of acute and chronic heart
and Queen Victoria Foundation, the Swedish Heart & Lung Foundation,
failure of the European society of cardiology (ESC) with the special contribution of the
and the Swedish Research Council. heart failure association (HFA) of the ESC. Eur Heart J 2021;42:3599–3726.
A.P. has received research grant support from the Gilead Sciences 15. Dunlay SM, Roger VL, Redfield MM. Epidemiology of heart failure with preserved ejec-
Research Scholar Program and Applied Therapeutics; honoraria out- tion fraction. Nat Rev Cardiol 2017;14:591–602. https://doi.org/10.1038/nrcardio.2017.
65
side the present study as an advisor/consultant for Tricog Health Inc
16. Zile MR, Baicu CF, Gaasch WH. Diastolic heart failure—abnormalities in active relax-
and Lilly, USA, Rivus, and Roche Diagnostics, and nonfinancial support ation and passive stiffness of the left ventricle. N Engl J Med 2004;350:1953–1959.
from Pfizer and Merck. https://doi.org/10.1056/NEJMoa032566
S.V. holds a Tier 1 Canada Research Chair in Cardiovascular Surgery, 17. Lam CSP, Donal E, Kraigher-Krainer E, Vasan RS. Epidemiology and clinical course of
and has received research grants and/or speaking honoraria from heart failure with preserved ejection fraction. Eur J Heart Fail 2011;13:18–28. https://
doi.org/10.1093/eurjhf/hfq121
Amarin, Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol 18. Rossi A, Gheorghiade M, Triposkiadis F, Solomon SD, Pieske B, Butler J. Left atrium in
Myers Squibb, Eli Lilly, EOCI Pharmacomm Ltd, HLS Therapeutics, heart failure with preserved ejection fraction. Circulation Heart Failure 2014;7:
Janssen, Merck, Novartis, Novo Nordisk, Pfizer, PhaseBio, Sanofi, Sun 1042–1049. https://doi.org/10.1161/CIRCHEARTFAILURE.114.001276
Pharmaceuticals, and the Toronto Knowledge Translation Working 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
Group. He is the President of the Canadian Medical and Surgical Heart Fail 2020;13:e006331. https://doi.org/10.1161/CIRCHEARTFAILURE.119.
Knowledge Translation Research Group, a federally incorporated 006331
not-for-profit physician organization. 20. Simmonds SJ, Cuijpers I, Heymans S, Jones EAV. Cellular and molecular differences be-
tween HFpEF and HFrEF: a step ahead in an improved pathological understanding. Cells
2020;9:242. https://doi.org/10.3390/cells9010242
Data Availability 21. Murphy SP, Kakkar R, McCarthy CP, Januzzi JL. Inflammation in heart failure. J Am Coll
All data described were obtained from published sources that are cited 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 pre-
within the manuscript.
served ejection fraction: a review of cardiac and noncardiac pathophysiology. Front
Physiol 2019;10:638. https://doi.org/10.3389/fphys.2019.00638
Funding 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
All authors declare no funding for this contribution.
of collagen and titin. Circulation 2015;131:1247–1259. https://doi.org/10.1161/
CIRCULATIONAHA.114.013215
References 24. Miranda-Silva D, Lima T, Rodrigues P, Leite-Moreira A, Falcão-Pires I. Mechanisms
1. Mudd JO, Kass DA. Tackling heart failure in the twenty-first century. Nature 2008;451: underlying the pathophysiology of heart failure with preserved ejection fraction: the
919–928. https://doi.org/10.1038/nature06798 tip of the iceberg. Heart Fail Rev 2021;26:453–478. https://doi.org/10.1007/s10741-
2. Rossignol P, Hernandez AF, Solomon SD, Zannad F. Heart failure drug treatment. 020-10042-0
Lancet 2019;393:1034–1044. https://doi.org/10.1016/S0140-6736(18)31808-7 25. Verma S, McMurray JJV. The serendipitous story of SGLT2 inhibitors in heart failure.
3. Lewis KS, Butler J, Bauersachs J, Sandner P. The three-decade long journey in heart fail- Circulation 2019;139:2537–2541. https://doi.org/10.1161/CIRCULATIONAHA.119.
ure drug development. In: Bauersachs J, Butler J, Sandner P, editors. Heart Failure 040514
[Internet].: Springer International Publishing; 2017, 1–14. [cited 2022 Oct 30]. 26. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin,
(Handbook of Experimental Pharmacology). Available from: https://doi.org/10.1007/ cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 2015;373:
164_2016_101 2117–2128. https://doi.org/10.1056/NEJMoa1504720
10 Pandey et al.

27. Fukuta H, Hagiwara H, Kamiya T. Sodium-glucose cotransporter 2 inhibitors in heart 50. Verma A, Patel AB, Waikar SS. SGLT2 inhibitor: not a traditional diuretic for heart fail-
failure with preserved ejection fraction: a meta-analysis of randomized controlled ure. Cell Metab 2020;32:13–14. https://doi.org/10.1016/j.cmet.2020.06.014
trials. Int J Cardiol Heart Vasc 2022;42:101103. 51. Mebazaa A, Solal AC, Colombo PC. Assessing and treating congestion in acute decom-
28. Cardoso R, Graffunder FP, Ternes CMP, Fernandes A, Rocha AV, Fernandes G, et al. pensated heart failure: are we seeing the light at the end of the tunnel? Eur Heart J 2023;
SGLT2 inhibitors decrease cardiovascular death and heart failure hospitalizations in pa- 44:51–53. https://doi.org/10.1093/eurheartj/ehac680
tients with heart failure: a systematic review and meta-analysis. EClinicalMedicine 2021; 52. Griffin M, Rao VS, Ivey-Miranda J, Fleming J, Mahoney D, Maulion C, et al.
36:100933. https://doi.org/10.1016/j.eclinm.2021.100933 Empagliflozin in heart failure. Circulation 2020;142:1028–1039. https://doi.org/10.1161/
29. Heerspink HJL, Stefánsson BV, Correa-Rotter R, Chertow GM, Greene T, Hou FF, CIRCULATIONAHA.120.045691
et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med 2020;383: 53. Damman K, Beusekamp JC, Boorsma EM, Swart HP, Smilde TDJ, Elvan A, et al.
1436–1446. https://doi.org/10.1056/NEJMoa2024816 Randomized, double-blind, placebo-controlled, multicentre pilot study on the effects
30. Mahaffey KW, Jardine MJ, Bompoint S, Cannon CP, Neal B, Heerspink HJL, et al. of empagliflozin on clinical outcomes in patients with acute decompensated heart fail-
Canagliflozin and cardiovascular and renal outcomes in type 2 diabetes mellitus and ure (EMPA-RESPONSE-AHF). Eur J Heart Fail 2020;22:713–722. https://doi.org/10.
chronic kidney disease in primary and secondary cardiovascular prevention groups. 1002/ejhf.1713
Circulation 2019;140:739–750. https://doi.org/10.1161/CIRCULATIONAHA.119. 54. Biegus J, Voors AA, Collins SP, Kosiborod MN, Teerlink JR, Angermann CE, et al.
042007 Impact of empagliflozin on decongestion in acute heart failure: the EMPULSE trial.
31. Bhatt DL, Szarek M, Pitt B, Cannon CP, Leiter LA, McGuire DK, et al. Sotagliflozin in Eur Heart J 2023;44:41–50. https://doi.org/10.1093/eurheartj/ehac530
patients with diabetes and chronic kidney disease. N Engl J Med 2021;384:129–139. 55. Packer M, Anker SD, Butler J, Filippatos G, Ferreira JP, Pocock SJ, et al. Empagliflozin in
https://doi.org/10.1056/NEJMoa2030186 patients with heart failure, reduced ejection fraction, and volume overload. J Am Coll
32. Vaduganathan M, Docherty KF, Claggett BL, Jhund PS, de Boer RA, Hernandez AF, Cardiol 2021;77:1381–1392. https://doi.org/10.1016/j.jacc.2021.01.033
et al. SGLT-2 inhibitors in patients with heart failure: a comprehensive meta-analysis 56. Cunningham JW, Vaduganathan M, Claggett BL, Kulac IJ, Desai AS, Jhund PS, et al.
of five randomised controlled trials. Lancet 2022;400:757–767. https://doi.org/10. Dapagliflozin in patients recently hospitalized with heart failure and mildly reduced
1016/S0140-6736(22)01429-5 or preserved ejection fraction. J Am Coll Cardiol 2022;80:1302–1310. https://doi.org/
33. The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial. Lancet 1999; 10.1016/j.jacc.2022.07.021
353:9–13. https://doi.org/10.1016/S0140-6736(98)11181-9 57. Masson W, Lavalle-Cobo A, Nogueira JP. Effect of SGLT2-inhibitors on epicardial adi-
34. Pitt B, Zannad F, Remme WJ, Cody R, Castaigne A, Perez A, et al. The effect of spir- pose tissue: a meta-analysis. Cells 2021;10:2150. https://doi.org/10.3390/
onolactone on morbidity and mortality in patients with severe heart failure. N Engl J cells10082150
Med 1999;341:709–717. https://doi.org/10.1056/NEJM199909023411001 58. Tosaki T, Kamiya H, Himeno T, Kato Y, Kondo M, Toyota K, et al. Sodium-glucose co-
35. Shah A, Gandhi D, Srivastava S, Shah KJ, Mansukhani R. Heart failure: a class review of transporter 2 inhibitors reduce the abdominal visceral fat area and may influence the
pharmacotherapy. P T 2017;42:464–472. renal function in patients with type 2 diabetes. Intern Med 2017;56:597–604. https://
36. Solomon SD, McMurray JJV, Anand IS, Ge J, Lam CSP, Maggioni AP, et al. Angiotensin– doi.org/10.2169/internalmedicine.56.7196
59. DeFronzo RA, Norton L, Abdul-Ghani M. Renal, metabolic and cardiovascular consid-
neprilysin inhibition in heart failure with preserved ejection fraction. N Engl J Med 2019;
erations of SGLT2 inhibition. Nat Rev Nephrol 2017;13:11–26. https://doi.org/10.1038/
381:1609–1620. https://doi.org/10.1056/NEJMoa1908655
nrneph.2016.170
37. Yusuf S, Pfeffer MA, Swedberg K, Granger CB, Held P, McMurray JJV, et al. Effects of
60. Gao YM, Feng ST, Wen Y, Tang TT, Wang B, Liu BC. Cardiorenal protection of SGLT2
candesartan in patients with chronic heart failure and preserved left-ventricular ejec-
inhibitors—perspectives from metabolic reprogramming. eBioMedicine 2022;83:
tion fraction: the CHARM-preserved trial. Lancet 2003:362:777–781. https://doi.org/
104215. https://doi.org/10.1016/j.ebiom.2022.104215
10.1016/S0140-6736(03)14285-7
61. Hoong CWS, Chua MWJ. SGLT2 inhibitors as calorie restriction mimetics: insights on
38. Pitt B, Pfeffer MA, Assmann SF, Boineau R, Anand IS, Claggett B, et al. Spironolactone
longevity pathways and age-related diseases. Endocrinology 2021;162:bqab079. https://
for heart failure with preserved ejection fraction. N Engl J Med 2014;370:1383–1392.
doi.org/10.1210/endocr/bqab079
https://doi.org/10.1056/NEJMoa1313731
62. Packer M. Autophagy stimulation and intracellular sodium reduction as mediators of
39. Packer M, Zannad F. Do the favorable effects of digoxin and SGLT2 inhibitors really
the cardioprotective effect of sodium–glucose cotransporter 2 inhibitors. Eur J
differ in patients with heart failure and a reduced ejection fraction? A provocative
Heart Fail 2020;22:618–628. https://doi.org/10.1002/ejhf.1732
side-by-side examination of trial outcomes. J Card Fail 2022;28:682–683. https://doi.
63. Esterline RL, Vaag A, Oscarsson J, Vora J. MECHANISMS IN ENDOCRINOLOGY:
org/10.1016/j.cardfail.2022.01.001
SGLT2 inhibitors: clinical benefits by restoration of normal diurnal metabolism? Eur J
40. Jhund PS, Kondo T, Butt JH, Docherty KF, Claggett BL, Desai AS, et al. Dapagliflozin
Endocrinol 2018;178:R113–R125. https://doi.org/10.1530/EJE-17-0832
across the range of ejection fraction in patients with heart failure: a patient-level,
64. Osataphan S, Macchi C, Singhal G, Chimene-Weiss J, Sales V, Kozuka C, et al. SGLT2
pooled meta-analysis of DAPA-HF and DELIVER. Nat Med 2022;28:1956–1964. inhibition reprograms systemic metabolism via FGF21-dependent and -independent
https://doi.org/10.1038/s41591-022-01971-4 mechanisms. JCI Insight 2019;4:e123130. https://doi.org/10.1172/jci.insight.123130
41. Bonora BM, Avogaro A, Fadini GP. Extraglycemic effects of SGLT2 inhibitors: a review 65. Packer M. Role of deranged energy deprivation signaling in the pathogenesis of cardiac
of the evidence. Diabetes Metab Syndr Obes 2020;13:161–174. https://doi.org/10.2147/ and renal disease in states of perceived nutrient overabundance. Circulation 2020;141:
DMSO.S233538 2095–2105. https://doi.org/10.1161/CIRCULATIONAHA.119.045561
42. Packer M. Critical examination of mechanisms underlying the reduction in heart failure 66. Lopaschuk GD, Verma S. Mechanisms of cardiovascular benefits of sodium glucose co-
events with SGLT2 inhibitors: identification of a molecular link between their actions transporter 2 (SGLT2) inhibitors. JACC Basic Transl Sci 2020;5:632–644. https://doi.org/
to stimulate erythrocytosis and to alleviate cellular stress. Cardiovasc Res 2021;117: 10.1016/j.jacbts.2020.02.004
74–84. https://doi.org/10.1093/cvr/cvaa064 67. Verma S. Potential mechanisms of sodium-glucose co-transporter 2 inhibitor-related
43. Baker WL, Buckley LF, Kelly MS, Bucheit JD, Parod ED, Brown R, et al. Effects of cardiovascular benefits. Am J Med 2019;132:S39–S48. https://doi.org/10.1016/j.
sodium-glucose cotransporter 2 inhibitors on 24-hour ambulatory blood pressure: amjmed.2019.08.007
a systematic review and meta-analysis. J Am Heart Assoc 2017;6:e005686. https://doi. 68. Saucedo-Orozco H, Voorrips SN, Yurista SR, de Boer RA, Westenbrink BD. SGLT2
org/10.1161/JAHA.117.005686 inhibitors and ketone metabolism in heart failure. J Lipid Atheroscler 2022;11:1–19.
44. Miyata KN, Zhang SL, Chan JSD. The rationale and evidence for SGLT2 inhibitors as a https://doi.org/10.12997/jla.2022.11.1.1
treatment for nondiabetic glomerular disease. Glomerular Dis 2021;1:21–33. https:// 69. De Meyer GRY, De Keulenaer GW, Martinet W. Role of autophagy in heart failure
doi.org/10.1159/000513659 associated with aging. Heart Fail Rev 2010;15:423–430. https://doi.org/10.1007/
45. Cherney DZ, Odutayo A, Aronson R, Ezekowitz J, Parker JD. Sodium glucose s10741-010-9166-6
cotransporter-2 inhibition and cardiorenal protection: JACC review topic of the 70. Du J, Liu Y, Fu J. Autophagy and heart failure. In: Le W, editor. Autophagy: Biology and
week. J Am Coll Cardiol 2019;74:2511–2524. https://doi.org/10.1016/j.jacc.2019.09.022 Diseases: Clinical Science [Internet]. Singapore: Springer; 2020, 223–7. [cited 2022 Nov
46. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al. 1]. (Advances in Experimental Medicine and Biology). Available from: https://doi.org/
Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med 10.1007/978-981-15-4272-5_16
2019;380:2295–2306. https://doi.org/10.1056/NEJMoa1811744 71. Warbrick I, Rabkin SW. Hypoxia-inducible factor 1-alpha (HIF-1α) as a factor mediat-
47. Zelniker TA, Braunwald E. Mechanisms of cardiorenal effects of sodium-glucose co- ing the relationship between obesity and heart failure with preserved ejection fraction.
transporter 2 inhibitors: JACC state-of-the-art review. J Am Coll Cardiol 2020;75: Obes Rev 2019;20:701–712. https://doi.org/10.1111/obr.12828
422–434. https://doi.org/10.1016/j.jacc.2019.11.031 72. Koyani CN, Plastira I, Sourij H, Hallström S, Schmidt A, Rainer PP, et al. Empagliflozin
48. Zou H, Zhou B, Xu G. SGLT2 inhibitors: a novel choice for the combination therapy in protects heart from inflammation and energy depletion via AMPK activation.
diabetic kidney disease. Cardiovasc Diabetol 2017;16:65. https://doi.org/10.1186/ Pharmacol Res 2020;158:104870. https://doi.org/10.1016/j.phrs.2020.104870
s12933-017-0547-1 73. Zhou H, Wang S, Zhu P, Hu S, Chen Y, Ren J. Empagliflozin rescues diabetic myocardial
49. Ronco C, Haapio M, House AA, Anavekar N, Bellomo R. Cardiorenal syndrome. J Am microvascular injury via AMPK-mediated inhibition of mitochondrial fission. Redox Biol
Coll Cardiol 2008;52:1527–1539. https://doi.org/10.1016/j.jacc.2008.07.051 2018;15:335–346. https://doi.org/10.1016/j.redox.2017.12.019
Mechanisms of benefits of SGLT2 inhibitors in heart failure with preserved ejection fraction 11

74. Hawley SA, Ford RJ, Smith BK, Gowans GJ, Mancini SJ, Pitt RD, et al. The Na+/glucose 96. Xu B, Li S, Kang B, Zhou J. The current role of sodium-glucose cotransporter 2 inhi-
cotransporter inhibitor canagliflozin activates AMPK by inhibiting mitochondrial func- bitors in type 2 diabetes mellitus management. Cardiovasc Diabetol 2022;21:83. https://
tion and increasing cellular AMP levels. Diabetes 2016;65:2784–2794. https://doi.org/ doi.org/10.1186/s12933-022-01512-w
10.2337/db16-0058 97. Yang Y, Zhao C, Ye Y, Yu M, Qu X. Prospect of sodium–glucose co-transporter 2 in-
75. Yang L, Liang B, Li J, Zhang X, Chen H, Sun J, et al. Dapagliflozin alleviates advanced hibitors combined with insulin for the treatment of type 2 diabetes. Front Endocrinol
glycation end product induced podocyte injury through AMPK/mTOR mediated au- (Lausanne) 2020;11:190. https://doi.org/10.3389/fendo.2020.00190
tophagy pathway. Cell Signal 2022;90:110206. https://doi.org/10.1016/j.cellsig.2021. 98. Yaribeygi H, Sathyapalan T, Maleki M, Jamialahmadi T, Sahebkar A. Molecular mechan-
110206 isms by which SGLT2 inhibitors can induce insulin sensitivity in diabetic milieu: a mech-
76. Ren FF, Xie ZY, Jiang YN, Guan X, Chen QY, Lai TF, et al. Dapagliflozin attenuates anistic review. Life Sci 2020;240:117090. https://doi.org/10.1016/j.lfs.2019.117090
pressure overload-induced myocardial remodeling in mice via activating SIRT1 and in- 99. Son TK, Toan NH, Thang N, Le Trong Tuong H, Tien HA, Thuy NH, et al. Prediabetes
hibiting endoplasmic reticulum stress. Acta Pharmacol Sin 2022;43:1721–1732. https:// and insulin resistance in a population of patients with heart failure and reduced or pre-
doi.org/10.1038/s41401-021-00805-2 served ejection fraction but without diabetes, overweight or hypertension. Cardiovasc
77. Packer M. Cardioprotective effects of sirtuin-1 and its downstream effectors. Circ Diabetol 2022;21:75. https://doi.org/10.1186/s12933-022-01509-5
Heart Fail 2020;13:e007197. https://doi.org/10.1161/CIRCHEARTFAILURE.120. 100. Savji N, Meijers WC, Bartz TM, Bhambhani V, Cushman M, Nayor M, et al. The asso-
007197 ciation of obesity and cardiometabolic traits with incident HFpEF and HFrEF. JACC
78. Yang Z, Li T, Xian J, Chen J, Huang Y, Zhang Q, et al. SGLT2 inhibitor dapagliflozin at- Heart Failure 2018;6:701–709. https://doi.org/10.1016/j.jchf.2018.05.018
tenuates cardiac fibrosis and inflammation by reverting the HIF-2α signaling pathway in 101. Bailey CJ. Uric acid and the cardio-renal effects of SGLT2 inhibitors. Diabetes Obes
arrhythmogenic cardiomyopathy. FASEB J 2022;36:e22410. https://doi.org/10.1096/fj. Metab 2019;21:1291–1298. https://doi.org/10.1111/dom.13670
202200243R 102. Iacobellis G. Epicardial adipose tissue in endocrine and metabolic diseases. Endocrine
79. Bessho R, Takiyama Y, Takiyama T, Kitsunai H, Takeda Y, Sakagami H, et al. 2014;46:8–15. https://doi.org/10.1007/s12020-013-0099-4
Hypoxia-inducible factor-1α is the therapeutic target of the SGLT2 inhibitor for dia- 103. Iacobellis G, Bianco AC. Epicardial adipose tissue: emerging physiological, pathophysio-
betic nephropathy. Sci Rep 2019;9:14754. https://doi.org/10.1038/s41598-019- logical and clinical features. Trends Endocrinol Metab 2011;22:450–457. https://doi.org/
51343-1 10.1016/j.tem.2011.07.003
80. Packer M. Critical reanalysis of the mechanisms underlying the cardiorenal benefits of 104. van Woerden G, van Veldhuisen DJ, Manintveld OC, van Empel VPM, Willems TP, de
SGLT2 inhibitors and reaffirmation of the nutrient deprivation signaling/autophagy hypoth- Boer RA, et al. Epicardial adipose tissue and outcome in heart failure with mid-range
esis. Circulation 2022;146:1383–1405. https://doi.org/10.1161/CIRCULATIONAHA.122. and preserved ejection fraction. Circ Heart Fail 2022;15:e009238. https://doi.org/10.
061732 1161/CIRCHEARTFAILURE.121.009238
81. Zannad F, Ferreira JP, Butler J, Filippatos G, Januzzi JL, Sumin M, et al. Effect of empagli- 105. Pugliese NR, Paneni F, Mazzola M, De Biase N, Del Punta L, Gargani L, et al. Impact of
flozin on circulating proteomics in heart failure: mechanistic insights from the epicardial adipose tissue on cardiovascular haemodynamics, metabolic profile, and
EMPEROR program. Eur Heart J 2022;43:4991–5002. https://doi.org/10.1093/ prognosis in heart failure. Eur J Heart Fail 2021;23:1858–1871. https://doi.org/10.
eurheartj/ehac495 1002/ejhf.2337
82. Packer M. How can sodium–glucose cotransporter 2 inhibitors stimulate erythrocyto- 106. Kalman E, Chan J, Goel V, Yeong C, Nelson A, Nerlekar N. SGLT2 inhibitors reduce
sis in patients who are iron-deficient? Implications for understanding iron homeostasis epicardial adipose tissue: a meta-analysis. Heart Lung Circ 2022;31:S179–S180. https://
in heart failure. Eur J Heart Fail 2022;24:2287–2296. https://doi.org/10.1002/ejhf.2731 doi.org/10.1016/j.hlc.2022.06.286
83. Anand IS, Gupta P. Anemia and iron deficiency in heart failure. Circulation 2018;138: 107. Wu P, Wen W, Li J, Xu J, Zhao M, Chen H, et al. Systematic review and meta-analysis of
80–98. https://doi.org/10.1161/CIRCULATIONAHA.118.030099 randomized controlled trials on the effect of SGLT2 inhibitor on blood leptin and adi-
84. Alnuwaysir RIS, Hoes MF, van Veldhuisen DJ, van der Meer P, Beverborg NG. Iron de- ponectin level in patients with type 2 diabetes. Horm Metab Res 2019;51:487–494.
ficiency in heart failure: mechanisms and pathophysiology. J Clin Med 2021;11:125. https://doi.org/10.1055/a-0958-2441
https://doi.org/10.3390/jcm11010125 108. Packer M. Do sodium-glucose co-transporter-2 inhibitors prevent heart failure with a
85. Hoes MF, Grote Beverborg N, Kijlstra JD, Kuipers J, Swinkels DW, Giepmans BNG, preserved ejection fraction by counterbalancing the effects of leptin? A novel hypoth-
et al. Iron deficiency impairs contractility of human cardiomyocytes through decreased esis. Diabetes Obes Metab 2018;20:1361–1366. https://doi.org/10.1111/dom.13229
mitochondrial function. Eur J Heart Fail 2018;20:910–919. https://doi.org/10.1002/ejhf. 109. Scisciola L, Cataldo V, Taktaz F, Fontanella RA, Pesapane A, Ghosh P, et al.
1154 Anti-inflammatory role of SGLT2 inhibitors as part of their anti-atherosclerotic activ-
86. Docherty KF, Curtain JP, Anand IS, Bengtsson O, Inzucchi SE, Køber L, et al. Effect of ity: data from basic science and clinical trials. Fron Cardiovasc Med 2022;9:1008922.
dapagliflozin on anaemia in DAPA-HF. Eur J Heart Fail 2021;23:617–628. https://doi. https://doi.org/10.3389/fcvm.2022.1008922
org/10.1002/ejhf.2132 110. Chen S, Coronel R, Hollmann MW, Weber NC, Zuurbier CJ. Direct cardiac effects of
87. Docherty KF, Welsh P, Verma S, De Boer RA, O’Meara E, Bengtsson O, et al. Iron de- SGLT2 inhibitors. Cardiovasc Diabetol 2022;21:45. https://doi.org/10.1186/s12933-
ficiency in heart failure and effect of dapagliflozin: findings from DAPA-HF. Circulation 022-01480-1
2022;146:980–994. https://doi.org/10.1161/CIRCULATIONAHA.122.060511 111. Theofilis P, Sagris M, Oikonomou E, Antonopoulos AS, Siasos G, Tsioufis K, et al.
88. Fuchs Andersen C, Omar M, Glenthøj A, El Fassi D, Møller HJ, Lindholm Kurtzhals JA, Anti-inflammatory potential of SGLT2 inhibitors: a systematic review and
et al. Effects of empagliflozin on erythropoiesis in heart failure: data from the Empire meta-analysis of preclinical studies in rodents. Eur Heart J 2022;43:2683. https://doi.
HF trial. Eur J Heart Fail 2023;25:226–234. https://doi.org/10.1002/ejhf.2735 org/10.1093/eurheartj/ehac544.2683
89. von Haehling S, Sato R, Butler J, Anker SD. Little at a time: trying to understand the 112. Xing YJ, Liu BH, Wan SJ, Cheng Y, Zhou SM, Sun Y, et al. A SGLT2 inhibitor dapagli-
battery of benefits of sodium–glucose contransporter 2 inhibitors in heart failure. flozin alleviates diabetic cardiomyopathy by suppressing high glucose-induced oxida-
Eur J Heart Fail 2023;25:235–237. https://doi.org/10.1002/ejhf.2766 tive stress in vivo and in vitro. Front Pharmacol 2021;12:708177. https://doi.org/10.
90. Mazer CD, Hare GMT, Connelly PW, Gilbert RE, Shehata N, Quan A, et al. Effect of 3389/fphar.2021.708177
empagliflozin on erythropoietin levels, iron stores, and red blood cell morphology in 113. Li C, Zhang J, Xue M, Li X, Han F, Liu X, et al. SGLT2 inhibition with empagliflozin at-
patients with type 2 diabetes mellitus and coronary artery disease. Circulation 2020; tenuates myocardial oxidative stress and fibrosis in diabetic mice heart. Cardiovasc
141:704–707. https://doi.org/10.1161/CIRCULATIONAHA.119.044235 Diabetol 2019;18:15. https://doi.org/10.1186/s12933-019-0816-2
91. Swedberg K, Young JB, Anand IS, Cheng S, Desai AS, Diaz R, et al. Treatment of anemia 114. Elrakaybi A, Laubner K, Zhou Q, Hug MJ, Seufert J. Cardiovascular protection by
with darbepoetin alfa in systolic heart failure. N Engl J Med 2013;368:1210–1219. SGLT2 inhibitors—do anti-inflammatory mechanisms play a role? Mol Metab 2022;
https://doi.org/10.1056/NEJMoa1214865 64:101549. https://doi.org/10.1016/j.molmet.2022.101549
92. Pfeffer MA, Burdmann EA, Chen CY, Cooper ME, de Zeeuw D, Eckardt KU, et al. A 115. Tong Y, Wang Z, Cai L, Lin L, Liu J, Cheng J. NLRP3 inflammasome and its central role
trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. N Engl J Med in the cardiovascular diseases. Oxid Med Cell Longev 2020;2020:4293206. https://doi.
2009;361:2019–2032. https://doi.org/10.1056/NEJMoa0907845 org/10.1155/2020/4293206
93. Segar MW, Kolkailah AA, Frederich R, Pong A, Cannon CP, Cosentino F, et al. 116. Kim SR, Lee SG, Kim SH, Kim JH, Choi E, Cho W, et al. SGLT2 inhibition modulates
Mediators of ertugliflozin effects on heart failure and kidney outcomes among patients NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular
with type 2 diabetes mellitus. Diabetes Obes Metab 2022;24:1829–1839. https://doi. disease. Nat Commun 2020;11:2127. https://doi.org/10.1038/s41467-020-15983-6
org/10.1111/dom.14769 117. Byrne NJ, Matsumura N, Maayah ZH, Ferdaoussi M, Takahara S, Darwesh AM, et al.
94. Fitchett D, Inzucchi SE, Zinman B, Wanner C, Schumacher M, Schmoor C, et al. Empagliflozin blunts worsening cardiac dysfunction associated with reduced NLRP3
Mediators of the improvement in heart failure outcomes with empagliflozin in the (nucleotide-binding domain-like receptor protein 3) inflammasome activation in heart
EMPA-REG OUTCOME trial. ESC Heart Fail 2021;8:4517–4527. https://doi.org/10. failure. Circ Heart Fail 2020;13:e006277. https://doi.org/10.1161/CIRCHEARTFAILURE.
1002/ehf2.13615 119.006277
95. Li J, Woodward M, Perkovic V, Figtree GA, Heerspink HJL, Mahaffey KW, et al. 118. Leng W, Wu M, Pan H, Lei X, Chen L, Wu Q, et al. The SGLT2 inhibitor dapagliflozin
Mediators of the effects of canagliflozin on heart failure in patients with type 2 diabetes. attenuates the activity of ROS-NLRP3 inflammasome axis in steatohepatitis with dia-
JACC Heart Fail 2020;8:57–66. https://doi.org/10.1016/j.jchf.2019.08.004 betes mellitus. Ann Transl Med 2019;7:429. https://doi.org/10.21037/atm.2019.09.03
12 Pandey et al.

119. Yaribeygi H, Katsiki N, Butler AE, Sahebkar A. Effects of antidiabetic drugs on NLRP3 140. Salah HM, Verma S, Santos-Gallego CG, Bhatt AS, Vaduganathan M, Khan MS, et al.
inflammasome activity, with a focus on diabetic kidneys. Drug Discov Today 2019;24: Sodium-glucose cotransporter 2 inhibitors and cardiac remodeling. J Cardiovasc
256–262. https://doi.org/10.1016/j.drudis.2018.08.005 Trans Res 2022;15:944–956. https://doi.org/10.1007/s12265-022-10220-5
120. Durante W, Behnammanesh G, Peyton KJ. Effects of sodium-glucose co-transporter 2 141. Pabel S, Wagner S, Bollenberg H, Bengel P, Kovács Á, Schach C, et al. Empagliflozin dir-
inhibitors on vascular cell function and arterial remodeling. Int J Mol Sci 2021;22:8786. ectly improves diastolic function in human heart failure. Eur J Heart Fail 2018;20:
https://doi.org/10.3390/ijms22168786 1690–1700. https://doi.org/10.1002/ejhf.1328
121. Hess DA, Terenzi DC, Trac JZ, Quan A, Mason T, Al-Omran M, et al. SGLT2 inhibition 142. Pabel S, Hamdani N, Luedde M, Sossalla S. SGLT2 inhibitors and their mode of action in
with empagliflozin increases circulating provascular progenitor cells in people with heart failure—has the mystery been unravelled? Curr Heart Fail Rep 2021;18:315–328.
type 2 diabetes mellitus. Cell Metab 2019;30:609–613. https://doi.org/10.1016/j. https://doi.org/10.1007/s11897-021-00529-8
cmet.2019.08.015 143. Heerspink HJL, Perco P, Mulder S, Leierer J, Hansen MK, Heinzel A, et al. Canagliflozin
122. Mone P, Lombardi A, Kansakar U, Varzideh F, Jankauskas SS, Pansini A, et al. reduces inflammation and fibrosis biomarkers: a potential mechanism of action for
Empagliflozin improves the microRNA signature of endothelial dysfunction in patients beneficial effects of SGLT2 inhibitors in diabetic kidney disease. Diabetologia 2019;
with heart failure with preserved ejection fraction and diabetes. J Pharmacol Exp Ther 62:1154–1166. https://doi.org/10.1007/s00125-019-4859-4
2023;384:116–122. https://doi.org/10.1124/jpet.121.001251 144. Quagliariello V, De Laurentiis M, Rea D, Barbieri A, Monti MG, Carbone A, et al. The
123. Despa S, Islam MA, Weber CR, Pogwizd SM, Bers DM. Intracellular Na(+) concentra- SGLT-2 inhibitor empagliflozin improves myocardial strain, reduces cardiac fibrosis
tion is elevated in heart failure but Na/K pump function is unchanged. Circulation 2002; and pro-inflammatory cytokines in non-diabetic mice treated with doxorubicin.
105:2543–2548. https://doi.org/10.1161/01.CIR.0000016701.85760.97 Cardiovasc Diabetol 2021;20:150. https://doi.org/10.1186/s12933-021-01346-y
124. Luo M, Anderson ME. Mechanisms of altered Ca2+ handling in heart failure. Circ Res 145. Xu L, Nagata N, Nagashimada M, Zhuge F, Ni Y, Chen G, et al. SGLT2 inhibition by
2013;113:690–708. https://doi.org/10.1161/CIRCRESAHA.113.301651 empagliflozin promotes fat utilization and browning and attenuates inflammation
125. Kaplan A, Abidi E, El-Yazbi A, Eid A, Booz GW, Zouein FA. Direct cardiovascular im- and insulin resistance by polarizing m2 macrophages in diet-induced obese mice.
pact of SGLT2 inhibitors: mechanisms and effects. Heart Fail Rev 2018;23:419–437. EBioMedicine 2017;20:137–149. https://doi.org/10.1016/j.ebiom.2017.05.028
https://doi.org/10.1007/s10741-017-9665-9 146. Verma S, Mazer CD, Yan AT, Mason T, Garg V, Teoh H, et al. Effect of empagliflozin on
126. Baartscheer A, Schumacher CA, Wüst RCI, Fiolet JWT, Stienen GJM, Coronel R, et al. left ventricular mass in patients with type 2 diabetes mellitus and coronary artery dis-
Empagliflozin decreases myocardial cytoplasmic Na+ through inhibition of the cardiac ease: the EMPA-HEART CardioLink-6 randomized clinical trial. Circulation 2019;140:
Na+/H+ exchanger in rats and rabbits. Diabetologia 2017;60:568–573. https://doi.org/ 1693–1702. https://doi.org/10.1161/CIRCULATIONAHA.119.042375
10.1007/s00125-016-4134-x 147. Dhingra NK, Mistry N, Puar P, Verma R, Anker S, Mazer CD, et al. SGLT2 inhibitors
127. Verma S, McMurray JJV. SGLT2 inhibitors and mechanisms of cardiovascular benefit: a and cardiac remodelling: a systematic review and meta-analysis of randomized cardiac
state-of-the-art review. Diabetologia 2018;61:2108–2117. https://doi.org/10.1007/
magnetic resonance imaging trials. ESC Heart Failure 2021;8:4693–4700. https://doi.
s00125-018-4670-7
org/10.1002/ehf2.13645
128. Arjun S, Bell RM. SGLT2 inhibitors: reviving the sodium-hydrogen exchanger cardio-
148. ClinicalTrials.gov. Dapagliflozin and Effect on Cardiovascular Events in Acute Heart
protection hypothesis? Cardiovasc Res 2019;115:1454–1456. https://doi.org/10.1093/
Failure -Thrombolysis in Myocardial Infarction 68 (DAPA ACT HF-TIMI 68).
cvr/cvz105
NCT04363697. Available from: https://clinicaltrials.gov/ct2/show/NCT04363697
129. De Pascalis A, Cianciolo G, Capelli I, Brunori G, La Manna G. SGLT2 inhibitors, sodium
149. Pandey AK, Okaj I, Kaur H, Belley-Cote EP, Wang J, Oraii A, et al. Sodium-glucose co-
and off-target effects: an overview. J Nephrol 2021;34:673–680. https://doi.org/10.
transporter inhibitors and atrial fibrillation: a systematic review and meta-analysis of
1007/s40620-020-00845-7
randomized controlled trials. J Am Heart Assoc 2021;10:e022222. https://doi.org/10.
130. Nakamura TY, Iwata Y, Arai Y, Komamura K, Wakabayashi S. Activation of Na+/H+
1161/JAHA.121.022222
exchanger 1 is sufficient to generate Ca2 + signals that induce cardiac hypertrophy
150. ClinicalTrials.gov. Empagliflozin and Atrial Fibrillation Treatment (EMPA-AF).
and heart failure. Circ Res 2008;103:891–899. https://doi.org/10.1161/CIRCRESAHA.
NCT04583813. Available from: https://clinicaltrials.gov/ct2/show/NCT04583813
108.175141
151. ClinicalTrials.gov. Use of Dapagliflozin to Reduce Burden of Atrial Fibrillation in
131. Karmazyn M. Therapeutic potential of Na-H exchange inhibitors for the treatment of
Patients Undergoing Catheter Ablation of Symptomatic Atrial Fibrillation
heart failure. Expert Opin Investig Drugs 2001;10:835–843. https://doi.org/10.1517/
(DAPA-AF). NCT04792190. Available from: https://clinicaltrials.gov/ct2/show/
13543784.10.5.835
NCT04792190
132. Uthman L, Baartscheer A, Bleijlevens B, Schumacher CA, Fiolet JWT, Koeman A, et al.
152. ClinicalTrials.gov. EMPACT-MI: A Study to Test Whether Empagliflozin Can Lower
Class effects of SGLT2 inhibitors in mouse cardiomyocytes and hearts: inhibition of Na
+/H+ exchanger, lowering of cytosolic Na+ and vasodilation. Diabetologia 2018;61: the Risk of Heart Failure and Death in People Who Had a Heart Attack (Myocardial
722–726. https://doi.org/10.1007/s00125-017-4509-7 Infarction). NCT04509674. Available from: https://clinicaltrials.gov/ct2/show/
133. Chung YJ, Park KC, Tokar S, Eykyn TR, Fuller W, Pavlovic D, et al. SGLT2 inhibitors and NCT04509674
the cardiac Na+/H+ exchanger-1: the plot thickens. Cardiovasc Res 2021;117: 153. ClinicalTrials.gov. Dapagliflozin Effects on Cardiovascular Events in Patients With an
2702–2704. https://doi.org/10.1093/cvr/cvab184 Acute Heart Attack (DAPA-MI). NCT04564742. Available from: https://clinicaltrials.
134. Chung YJ, Park KC, Tokar S, Eykyn TR, Fuller W, Pavlovic D, et al. Off-target effects of gov/ct2/show/NCT04564742
sodium-glucose co-transporter 2 blockers: empagliflozin does not inhibit Na+/H+ 154. Agarwal R, Filippatos G, Pitt B, Anker SD, Rossing P, Joseph A, et al. Cardiovascular and
exchanger-1 or lower [Na + ]i in the heart. Cardiovasc Res 2021;117:2794–2806. kidney outcomes with finerenone in patients with type 2 diabetes and chronic kidney
https://doi.org/10.1093/cvr/cvaa323 disease: the FIDELITY pooled analysis. Eur Heart J 2022;43:474–484. https://doi.org/10.
135. Ye Y, Jia X, Bajaj M, Birnbaum Y. Dapagliflozin attenuates Na+/H+ exchanger-1 in car- 1093/eurheartj/ehab777
diofibroblasts via AMPK activation. Cardiovasc Drugs Ther 2018;32:553–558. https:// 155. Brown E, Heerspink HJL, Cuthbertson DJ, Wilding JPH. SGLT2 inhibitors and GLP-1
doi.org/10.1007/s10557-018-6837-3 receptor agonists: established and emerging indications. Lancet 2021;398:262–276.
136. Jiang D, Shi H, Tonggu L, Gamal El-Din TM, Lenaeus MJ, Zhao Y, et al. Structure of the https://doi.org/10.1016/S0140-6736(21)00536-5
cardiac sodium channel. Cell 2020;180:122–134.e10. https://doi.org/10.1016/j.cell. 156. ClinicalTrials.gov. Efficacy, Safety and Tolerability of AZD9977 and Dapagliflozin in
2019.11.041 Participants With Heart Failure and Chronic Kidney Disease (MIRACLE).
137. Philippaert K, Kalyaanamoorthy S, Fatehi M, Long W, Soni S, Byrne NJ, et al. Cardiac NCT04595370. Available from: https://clinicaltrials.gov/ct2/show/NCT04595370
late sodium channel current is a molecular target for the sodium/glucose cotranspor- 157. ClinicalTrials.gov. A Study to Learn How Well the Treatment Combination of
ter 2 inhibitor empagliflozin. Circulation 2021;143:2188–2204. https://doi.org/10.1161/ Finerenone and Empagliflozin Works and How Safe it is Compared to Each
CIRCULATIONAHA.121.053350 Treatment Alone in Adult Participants With Long-term Kidney Disease (Chronic
138. Verma S, Garg A, Yan AT, Gupta AK, Al-Omran M, Sabongui A, et al. Effect of empa- Kidney Disease) and Type 2 Diabetes (CONFIDENCE). NCT05254002. Available
gliflozin on left ventricular mass and diastolic function in individuals with diabetes: an from: https://clinicaltrials.gov/ct2/show/NCT05254002
important clue to the EMPA-REG OUTCOME trial? Diabetes Care 2016;39: 158. Pitt B, Bhatt DL. Does SGLT1 inhibition add benefit to SGLT2 inhibition in type 2 dia-
e212–e213. https://doi.org/10.2337/dc16-1312 betes? Circulation 2021;144:4–6. https://doi.org/10.1161/CIRCULATIONAHA.121.
139. Shim CY, Seo J, Cho I, Lee CJ, Cho IJ, Lhagvasuren P, et al. Randomized, controlled trial 054442
to evaluate the effect of dapagliflozin on left ventricular diastolic function in patients 159. Takebayashi K, Inukai T. Effect of sodium glucose cotransporter 2 inhibitors with low
with type 2 diabetes mellitus. Circulation 2021;143:510–512. https://doi.org/10.1161/ SGLT2/SGLT1 selectivity on circulating glucagon-like peptide 1 levels in type 2 diabetes
CIRCULATIONAHA.120.051992 Mellitus. J Clin Med Res 2017;9:745–753. https://doi.org/10.14740/jocmr3112w

You might also like