You are on page 1of 16

Circulation Research

HEART FAILURE COMPENDIUM

Mechanisms and Models in Heart Failure


A Translational Approach
Douglas L. Mann , G. Michael Felker

ABSTRACT: Despite multiple attempts to develop a unifying hypothesis that explains the pathophysiology of heart failure
with a reduced ejection fraction (HFrEF), no single conceptual model has withstood the test of time. In the present
review, we discuss how the results of recent successful phase III clinical development programs in HFrEF are built
upon existing conceptual models for drug development. We will also discuss where recent successes in clinical trials
do not fit existing models to identify areas where further refinement of current paradigms may be needed. To provide
the necessary structure for this review, we will begin with a brief overview of the pathophysiology of HFrEF, followed
by an overview of the current conceptual models for HFrEF, and end with an analysis of the scientific rationale
and clinical development programs for 4 new therapeutic classes of drugs that have improved clinical outcomes in
HFrEF. The 4 new therapeutic classes discussed are ARNIs, SGLT2 (sodium-glucose cotransporter 2) inhibitors,
soluble guanylate cyclase stimulators, and myosin activators. With the exception of SGLT2 inhibitors, each of these
therapeutic advances was informed by the insights provided by existing conceptual models of heart failure. Although
the quest to determine the mechanism of action of SGLT2 inhibitors is ongoing, this therapeutic class of drugs may
represent the most important advance in cardiovascular therapeutics of recent decades and may lead to rethinking or
expanding our current conceptual models for HFrEF.

Key Words: angiotensin ◼ clinical trial ◼ drug therapy ◼ heart failure ◼ neprilysin ◼ soluble guanylate cyclase
Downloaded from http://ahajournals.org by on May 17, 2021

D
espite repeated attempts to develop a unifying emphasis on molecular signaling pathways as a means
hypothesis that explains the pathophysiology of to identify new drug targets, in favor of discussing how
heart failure with reduced ejection fraction (HFrEF), the results of 4 recent successful phase III clinical devel-
no single conceptual model has withstood the test of opment programs in HFrEF fit within our existing models
time. One reason for this shortcoming is that the devel- for drug development, with the hope that the recent past
opment of clinical HF in patients with a reduced EF will provide prologue for future drug discoveries. More
represents the complex interplay between structural importantly, we will discuss where recent successes in
and functional biological changes that are occurring in clinical trials do not fit existing models to identify areas
the heart, the autonomic nervous system, the kidney, where further refinement of current paradigms may be
the peripheral vasculature, and skeletal muscle. These needed. To provide the structure for this type of review,
biological changes are influenced by aging, genetic we will begin with a brief overview of the pathophysiology
background, comorbidities, and nutrition, as well as non- of HFrEF. This subject has been the topic of extensive
biological environmental factors, all of which add to the reviews1,2 and will be discussed here briefly to provide
complexity of understanding the pathophysiology of HF. context for understanding the clinical models for thera-
The current review will discuss the translational frame- peutic drug development. Following the review of the dif-
work that has provided a platform for developing drugs to ferent clinical models, we will conclude with a discussion
treat patients with HFrEF for the past 30 years. Here, we of what we have learned from recent successful phase
will depart from the traditional approach that places an III clinical trials.


Correspondence to: G. Michael Felker, MD, MHS, Duke Clinical Research Institute, 200 Morris St, Durham, NC 27705, Email michael.felker@duke.edu; or Douglas L.
Mann, MD, Center for Cardiovascular Research, 660 S. Euclid Ave, Campus Box 8086, St. Louis, MO 63110, Email dmann@dom.wustl.edu
For Disclosures, see page 1448.
© 2021 American Heart Association, Inc.
Circulation Research is available at www.ahajournals.org/journal/res

Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158 May 14, 2021   1435


Mann and Felker Translational Advances in HFrEF
Heart Failure Compendium

Nonstandard Abbreviations and Acronyms Ventricular Remodeling During


Sacubitril/Valsartan Therapy for
ACE angiotensin-converting enzyme Heart Failure
ANP atrial natriuretic peptide RAAS renin-angiotensin aldosterone
system
ARNI angiotensin receptor–neprilysin
inhibitors sGC soluble guanylate cyclase
ATOMIC-HF Acute Treatment With Omecamtiv SGLT2i sodium-glucose cotransporter 2
Mecarbil to Increase Contractility in inhibitors
Acute Heart Failure SNS sympathetic nervous system
BNP B-type natriuretic peptide SOCRATES-
CANVAS Canagliflozin Cardiovascular REDUCED Soluble Guanylate Cyclase Stimu-
Assessment Study lator in Heart Failure With Reduced
Ejection Fraction
COSMIC-HF The Chronic Oral Study of Myosin
Activation to Increase Contractility
in Heart Failure
CREDENCE Canagliflozin and Renal Events PATHOPHYSIOLOGY OF HFREF
in Diabetes With Established
Nephropathy Clinical Evaluation HFrEF arises secondary to a series of complex changes
DECLARE– in the molecular and cellular composition of the heart
TIMI 58 Dapagliflozin Effect on Cardio- (reviewed in Mann et al2,3). These changes lead to the
vascular Events–Thrombolysis in phenotypic changes in the size, shape, and function of the
Myocardial Infarction-58 failing heart that ultimately result in a decreased pumping
EMPEROR- capacity of the heart with a subsequent decrease in car-
Reduced Empagliflozin Outcome Trial in diac output (Figure 1). The molecular and cellular changes
Patients with Chronic Heart Failure may occur suddenly (eg, following a myocardial infarction),
and a Reduced Ejection Fraction or may arise more gradually (eg, following exposure to toxic
eNOS endothelial-constitutive NO chemotherapies or sustained hemodynamic overload), or
synthase may develop secondary to inherited mutations of genes
Downloaded from http://ahajournals.org by on May 17, 2021

GALACTIC-HF Global Approach to Lower- that affect sarcomere function. The initial decline in cardiac
ing Adverse Cardiac Outcomes output is perceived as arterial underfilling by the peripheral
Through Improving Contractility in arterial baroreceptors that regulate autonomic nervous sys-
Heart Failure tem parasympathetic and sympathetic signaling. When the
GC guanylate cyclase baroreceptor mechanoreceptors detect a decrease in arte-
HFrEF heart failure with reduced ejection rial filling, they trigger a series of homeostatic reflexes that
fraction are initiated by a withdrawal of parasympathetic tone that is
HR hazard ratio followed by a reciprocal increase in sympathetic (adrener-
iNOS inducible NO synthase gic) nervous system signaling (reviewed in Floras and Poni-
LV left ventricle kowski5 and van Bilsen et al6). The baroreceptor-mediated
NEP neutral endopeptidase increase in sympathetic nervous system (SNS) signaling
nNOS neuronal NO synthase triggers increased renin production by the kidneys, with
NOS NO synthase resultant activation of the renin-angiotensin-aldosterone
NPR A natriuretic peptide A receptor system (RAAS). Because SNS and RAAS signaling is highly
synergistic, they are referred to as the neurohumoral axis.7
NPR-B natriuretic peptide B receptor
The initial activation of the SNS and RAAS restores cir-
NT-proBNP N-terminal pro-B-type natriuretic
culatory homeostasis by increasing contractility, increas-
peptide
ing retention of sodium and water by the kidney, and by
OVERTURE Omapatrilat Versus Enalapril Ran-
increasing peripheral arterial vasoconstriction.1,7 In some
domized Trial of Utility in Reducing
Events patients, pumping capacity of the heart will return to nor-
mal once the tissue injury is resolved or the inciting stress
PARADIGM-HF Prospective Comparison of ARNI
With ACE Inhibition to Determine is removed. In this setting, the normalization of left ven-
Impact on Global Mortality and tricular (LV) function results in restoration of circulatory
Morbidity in Heart Failure homeostasis. However, if LV function remains depressed,
PROVE-HF Prospective Study of Biomark- the SNS and RAAS remain persistently activated in
ers, Symptom Improvement, and an ongoing attempt to maintain circulatory homeo-
stasis. Some patients with LV dysfunction will remain

1436   May 14, 2021 Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158


Mann and Felker Translational Advances in HFrEF

Heart Failure Compendium


Figure 1. Pathogenesis of heart failure with a reduced ejection fraction.
A, Heart failure begins after a so-called index event produces an initial decline in pumping capacity of the heart. B, The initial decline in cardiac
output is perceived as arterial underfilling by peripheral arterial baroreceptors, which leads to a withdrawal of parasympathetic tone that is
accompanied by a reciprocal increase in sympathetic (adrenergic) nervous activity (sympathovagal imbalance). The increase in sympathetic
Downloaded from http://ahajournals.org by on May 17, 2021

nervous signaling leads to activation of the renin-angiotensin-aldosterone system in the kidney, increased contractile force of the heart, and
peripheral arterial vasoconstriction. In the short term, these changes restore cardiovascular function to a normal homeostatic range, with the result
that the patient remains asymptomatic. With time, however, the sustained activation of these systems can lead to secondary end-organ damage
within the ventricle, with worsening left ventricular (LV) remodeling and subsequent cardiac decompensation. β-AR-responsiveness indicates beta-
adrenergic responsiveness. Reprinted from Mann.4 Copyright ©1999.

asymptomatic, despite RAAS-induced expansion of the Figure 2 illustrates the signaling pathway of the natri-
circulatory blood volume. The precise mechanism(s) that uretic peptide system. As shown, natriuretic peptides stim-
explains why certain patients remain asymptomatic is not ulate the production of the intracellular second-messenger
known. One plausible explanation is that the compensa- cGMP, via binding to the NPR-A (natriuretic peptide A
tory mechanisms that become activated are sufficient to receptor), which binds ANP and BNP preferentially, and
modulate cardiovascular and renal function within a phys- the NPR-B (natriuretic peptide B receptor), which binds
iological/homeostatic range, such that the patient’s exer- C-type natriuretic peptide preferentially. Both NPR-A and
cise capacity is preserved or is depressed only minimally.1 NPR-B are coupled to membrane-bound (particulate) GC
Several counter-regulatory biological systems are (guanylate cyclase), which activates the downstream sig-
upregulated in the setting of HF to offset the deleterious naling pathways of cGMP, including cGMP-dependent
effects of SNS and RAAS signaling on the cardiovas- protein kinases I and II, cGMP-gated ion channels, and
cular system. Principal among these are the natriuretic cGMP-regulated phosphodiesterases.9 Natriuretic pep-
peptides, including ANP (atrial natriuretic peptide) and tides are degraded by NEP (neutral endopeptidase) 24.11
BNP (B-type natriuretic peptide).8 Under physiologi- (neprilysin) or clearance via natriuretic peptide receptor-C
cal conditions, ANP and BNP function as natriuretic mediated internalization into cells followed by lysosomal
hormones that are released in response to increases degradation.9,10 The biologic importance of the natriuretic
in atrial and myocardial stretch. Once released, these peptides in renal sodium handling has been demonstrated
cardiac-derived peptides act on the kidney and periph- in multiple studies employing NPR antagonists, as well as
eral circulation to unload the heart by increasing renal overexpression of ANP or BNP. Moreover, the concentra-
excretion of sodium and water and arterial vasodilation, tions of natriuretic peptides that are detected in the circu-
as well as inhibiting the release of renin and aldoste- lation have provided important diagnostic and prognostic
rone by the kidney.8 information in HF.8 For reasons that are not entirely clear,

Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158 May 14, 2021   1437


Mann and Felker Translational Advances in HFrEF
Heart Failure Compendium
Downloaded from http://ahajournals.org by on May 17, 2021

Figure 2. Natriuretic peptides and pGC (particulate guanylyl cyclase) and sGC (soluble guanylyl cyclase) signaling pathways.
The natriuretic peptide system consists of 5 structurally similar peptides: ANP (atrial natriuretic peptide), urodilatin (an isoform of ANP), BNP
(B-type natriuretic peptide), CNP (C-type natriuretic peptide), and DNP (dendroaspis natriuretic peptide). Natriuretic peptides bind to GC-A
(particulate guanylyl cyclase A)/NPR-A (natriuretic peptide A receptor) or GC-B (particulate guanylyl cyclase B)/NPR-B (natriuretic peptide B
receptor; membrane-bound/pGCs) and activate the signaling pathways of cyclic guanosine monophosphate (cGMP). The GC-A/NPR-A receptor
preferentially binds ANP and BNP, and the GC-B/NPR-B receptor preferentially binds CNP. Both NPR-A and NPR-B are coupled to pGC. NO
binds to sGC in the cytoplasm, inducing cGMP production and activation of cGMP signaling pathways. Once the intracellular concentration of
cGMP increases, cGMP-gated cation channels, cGMP-dependent protein kinases generate important biological responses in different tissues.
Cyclic nucleotide PDEs (phosphodiesterases) hydrolyze cGMP thus inhibiting signal transduction. The PDEs are comprised of a superfamily of
11 diverse isozymes (numbered PDE1, PDE2, etc) that are compartmentalized within the cell. PDE5 is primarily expressed in the vascular smooth
muscle cells and catabolizes cGMP generated by soluble guanylate cyclase. PDE9 also catabolizes cGMP but is primarily expressed in the
gastrointestinal tract, kidney, and brain and catabolizes cGMP generated by particulate guanylate cyclase. The natriuretic peptides are degraded
by two major mechanisms: NPRC (natriuretic peptide C receptor)-mediated internalization, followed by lysosomal degradation and enzymatic
degradation by NEP (neutral endopeptidase) 24.11 (neprilysin), which is widely expressed in multiple tissues, where it often is colocalized with
angiotensin-converting enzyme. NOS indicates NO synthase; and PKG, protein kinase.

1438   May 14, 2021 Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158


Mann and Felker Translational Advances in HFrEF

Heart Failure Compendium


the effects of the natriuretic peptides on the kidney are shifts of the LV end-diastolic pressure-volume relationship
blunted with advancing HF, leaving SNS and RAAS sig- toward normal. Collectively, these changes are referred to
naling unopposed.11–13 The release of NO by endothelial as reverse LV remodeling.14,15 Relevant to this discussion,
cells is a second biological system that plays a central role the assessment of LV remodeling is a potential surrogate
in mediating vascular tone in HF. NO elicits physiological end point for drug or device effects on HF outcomes.16,17
effects in cells by binding to cytosolic GC (sGC [soluble
GC]), leading to cGMP accumulation (Figure 2).
Sustained SNS and RAAS signaling leads, respec- CONCEPTUAL THERAPEUTIC MODELS
tively, to increased circulating levels of norepinephrine and FOR HFREF
angiotensin II. These molecules have been referred to as
Investigators and clinicians have used different conceptual
neurohormones in the literature, which is a historical term
therapeutic models to develop strategies for treating heart
that reflects the observation that that many of the mole-
HFrEF. These models are described in detail elsewhere18,19
cules detected in the circulation of patients with HFrEF are
and will be discussed here in brief to provide context for
produced by the neuroendocrine system and thus act on
understanding more contemporary HF therapies.
the heart in an endocrine manner. However, studies have
shown that many neurohormones (eg, angiotensin II) are
synthesized by cell types residing within the myocardium
and thus act in an autocrine and paracrine manner. None- CARDIORENAL MODEL
theless, the important conceptual theme of the neurohor- The first conceptual therapeutic model was derived from
monal model is that the overexpression of portfolios of clinical observations dating as far back as 1680 that
biologically active molecules can contribute to the patho- linked injury of the heart to the development of periph-
genesis of HFrEF because of the toxic effects of these eral or pulmonary edema, which was described histori-
molecules on the heart and circulation (reviewed in Hartu- cally as hydrops or dropsy. The cardiorenal model posited
pee et al1). At some point, patients with LV dysfunction will that the formation of peripheral or pulmonary edema was
develop classic signs and symptoms of HF. The transition the result of an injury to the heart that either reduced
to symptomatic HF is accompanied by further activation of the ability of the heart to eject blood (forward failure)
the neurohormonal and inflammatory signaling pathways, or impaired the ability of the heart to receive blood from
as well as a series of adaptive changes within the myocar- peripheral organs (backward failure).18 Increases in left
dium referred to as LV remodeling (reviewed in Mann et and right-sided venous pressures led, respectively, to
Downloaded from http://ahajournals.org by on May 17, 2021

al2,3). Medical and device therapies that lead to improved pulmonary edema or peripheral edema by altering out-
clinical outcomes in HFrEF patients almost always lead ward starling forces thereby increasing the flow of fluid
to decreased LV volume and mass and restore a more from the microvasculature into the interstitium. As shown
normal elliptical shape to the ventricle. These beneficial in Table 1, both interpretations of the cardiorenal model
changes represent the summation of biologic changes in have merit, insofar as injury to the heart often leads to
cardiac myocyte size and function, as well as modifications inappropriate retention of sodium and water by the kid-
in LV structure and organization that are accompanied by ney, with resulting expansion of intravascular volume and

Table 1.  Conceptual Therapeutic Models for Heart Failure With a Reduced Ejection Fraction

Heart failure models Conceptual advances Conceptual disadvantages Therapeutic advances


Cardiorenal Recognized the contribution of cardiac Did not explain disease progression Led to the use of diuretics
injury to the pathogenesis of heart failure
Recognized the importance of inappro- Impeded progress with vasodilators
priate sodium retention by the kidney
Impeded progress with β-blockers
Cardiocirculatory model (he- Recognized the importance of the pe- Did not explain disease progression Led to the use of vasodilators
modynamic) ripheral circulation
Fostered widespread use of inotropes
Impeded progress with β-blockers
Neurohormonal Heart failure viewed as a biological prob- Does not explain disease progression
lem, not as a hemodynamic problem completely
Does not predict success for all “neuro- Led to the use of ACE inhibitors, β-
hormonal” antagonists blockers, aldosterone antagonists, and
ARBs
Does not explain the benefit of emerging
device therapies

ACE indicates angiotensin-converting enzyme; and ARB, angiotensin receptor blockers.


Adapted from Mann19 with permission. Copyright ©2014, Elsevier.

Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158 May 14, 2021   1439


Mann and Felker Translational Advances in HFrEF
Heart Failure Compendium

interstitial edema. However, as discussed above studies of the neurohormonal model is that it focused on HF
have shown that the inappropriate retention of sodium fundamentally as a biological problem rather than as a
and water by the kidney is mediated by SNS and RAAS hemodynamic problem. The neurohormonal model led to
signaling, rather than decreased cardiac output, per se. the use of beta-adrenergic blocking agents to block the
One of the major therapeutic advances of the cardiore- deleterious effects of the sympathetic nervous system in
nal model is that it led to the use of diuretics to manage the heart, and angiotensin-converting enzyme inhibitors,
the volume overload in HF, which remains a mainstay of angiotensin receptor blockers, and aldosterone antago-
therapy in the current era (Table 1).20 nists to block the deleterious effects of RAAS. These
therapies were shown to clearly improve long-term mor-
bidity and mortality in a broad population of patients
with HFrEF, and collectively, they represent the corner-
CARDIOCIRCULATORY MODEL
stone of modern guideline-directed medical therapy for
The second conceptual therapeutic model for treating HF HF (Table 1).27,28 Although the neurohormonal model
was based on physiological concepts derived from stud- has provided several important insights with respect
ies of ventricular mechanics, which demonstrated that car- to explaining disease progression, as well as provided
diac function was regulated by inotropy, as well as cardiac important insights in terms of drug development for HF,
preload and afterload. The cardiocirculatory or hemody- many HFrEF patients continue to have progressive dis-
namic model proposed that HF was the result of abnor- ease despite optimal guideline-directed medical therapy.
mal pumping capacity of the heart that was exacerbated Moreover, as HF progresses, many HFrEF patients
by the increased afterload imposed by peripheral arterial become refractory or intolerant to conventional medical
vasoconstriction.18 Although peripheral vasoconstriction therapy, often requiring the withdrawal of conventional
maintained perfusion to vital organs, it also increased the medical therapies.29 Thus, there remains a clear need
afterload on the heart, decreased renal blood flow, which to develop additional treatments for this population, in
leads to increased sodium retention by the kidney, and particular, treatments that do not have overlapping side
reduced blood flow to exercising skeletal muscle, which effects or intolerances (hypotension, renal dysfunction,
was believed to contribute to exercise intolerance. The hyperkalemia) with existing treatments.
cardiocirculatory model led to the use of orally active
vasodilators to unload the failing heart and provided the
scientific rationale for the first large scale morbidity mor- RECENT THERAPEUTIC ADVANCES IN
Downloaded from http://ahajournals.org by on May 17, 2021

tality trial in HF.21 Additionally, the cardiocirculatory model


HFREF
also led to the use of inotropic agents to improve pump-
ing capacity of the heart to stabilize hemodynamics and In the preceding section, we have described 3 concep-
improve symptoms. Although inotropes produced dra- tual models that were used to develop HF therapeutics
matic immediate short-term hemodynamic effects, the (see Table 1). In the section that follows, we will discuss
long-term use of inotropes was associated with a dra- 4 new classes of HF therapeutics that have improved
matic and unexpected increase in patient morbidity and clinical outcomes in large phase III trials in patients
mortality.19,22 As shown in Table 1, the cardiocirculatory with HFrEF (see data summarized in Table 2). As will
model forms the basis for the current treatment of acute be discussed 3 of these new therapeutic classes were
decompensated HF, wherein diuretics, vasodilators, and developed based on the insights provided by existing
intravenous inotropic agents remain the primary available
therapies, despite multiple attempts to further expand the Table 2.  Summary of Benefits of Novel Therapies for HF
armamentarium.23–26 Unfortunately, the cardiocirculatory With Reduced Ejection Fraction on Clinical End Points
model impeded progress in the field of HF conceptually, Sacubitril/ SGLT2 Omecamtiv
insofar as it prohibited the use of beta-adrenergic blocking valsartan inhibitors Vericiguat mecarbil
agents, which appeared counterintuitive because of their Improves LV remod- Yes Yes No Yes
eling
negative inotropic effects. Moreover, the cardiocirculatory
model did not recognize the importance of cardiac remod- Decreases natri- Yes Yes Yes Yes
uretic peptides
eling as a mechanism for disease progression in HF.
Improves symp- Yes Yes unknown unknown
toms/quality of life

NEUROHORMONAL MODEL CV death or HF


hospitalization
Yes Yes Yes Yes

The neurohormonal model was based on the observation Improves CV mor- Yes Yes No No
that many of the biologically active molecules elaborated tality
by the SNS and RAAS (eg, norepinephrine, angiotensin Improves HF hospi- Yes Yes Yes Yes
II, aldosterone) were overtly toxic to the heart and cir- talizations
culation when expressed at levels that were observed CV indicates cardiovascular; HF, heart failure; LV, left ventricular; and SGLT2,
in the failing heart.4 The important conceptual advance sodium-glucose cotransporter 2.

1440   May 14, 2021 Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158


Mann and Felker Translational Advances in HFrEF

Heart Failure Compendium


conceptual HF models, whereas one therapeutic class, the risk of angioedema secondary to elevated bradyki-
the SGLT2 (sodium-glucose cotransporter 2)-inhibi- nin levels would be reduced by combing a NEP inhibitor
tors (SGLT2i), has less well-understood mechanisms of with an angiotensin receptor blocker rather than an ACE
action, and may require refining, expanding or develop- inhibitor. Given that valsartan dosing in HF was relatively
ing new conceptual models of HF. well understood,33 and leveraging the experience with
omapatrilat, sacubitril/valsartan was developed as a novel
therapeutic agent for HFrEF without going through tradi-
ANGIOTENSIN-NEPRILYSIN INHIBITION tional testing in a phase II trial. An initial trial of sacubitril/
Mechanism of Action valsartan compared with valsartan alone in 1328 patients
with mild-moderate hypertension provide evidence of the
Angiotensin receptor–neprilysin inhibitors (ARNI) magnitude of blood pressure lowering (2–4 mm differ-
represent a novel second-generation neurohormonal ence in systolic blood pressure compared with valsartan),
antagonist that combines a RAAS antagonist with a as well as initial evidence of safety.34 The effect of sacu-
NEP inhibitor. NEP inhibitors attenuate RAAS signal- bitril/valsartan on morbidity and mortality in HFrEF was
ing by preventing degradation of peptides that serve evaluated in a large phase III outcomes study, the PAR-
as natural counter-regulatory antagonists of RAAS sig- ADIGM-HF trial (Prospective Comparison of ARNI With
naling. NEP is expressed in multiple tissues, including
ACE Inhibition to Determine Impact on Global Mortality
vascular endothelium, smooth muscle cells, myocytes,
and Morbidity in Heart Failure).35 PARADIGM-HF was a
fibroblasts, kidney tubule cells, and nerve cells. NEP
double-blind randomized controlled trial of enalapril ver-
degrades multiple peptides, including natriuretic pep-
sus sacubitril/valsartan in 8442 patients with New York
tides, angiotensin I, angiotensin II, endothelin-I, adre-
Heart Association (NYHA) class II to IV symptoms and
nomedullin, opioids, bradykinin, chemotactic peptides,
an LVEF ≤40%. The primary end point of PARADIGM
enkephalins, and Aβ (amyloid-β peptide).30 NEP-medi-
was the composite of time cardiovascular death or first
ated inhibition of the degradation of natriuretic peptides
HF hospitalization. For inclusion in the trial, participants
leads to vasorelaxation and vasodilation of vascular
had to be taking a stable dose of ACE or angiotensin
arteries, natriuresis, inhibition of hypertrophy, and fibro-
receptor blocker equivalent to at least 10 mg of enalapril
sis. However, inhibition of the degradation of other
daily for at least 4 weeks. In addition, before randomiza-
vasoactive peptides, such as angiotensin II, angiotensin
tion, patients had to tolerate a single-blind run-in period
1-7, and endothelin-I, opposes the vasodilatory effects
Downloaded from http://ahajournals.org by on May 17, 2021

of enalapril 10 mg twice-daily followed by sacubitril/val-


of natriuretic peptides. Accordingly, NEP inhibition has
sartan up-titrated to 97 out of 103 twice-daily. The trial
variable effects on blood pressure.
was stopped early by the Data Safety Monitoring Board,
given the overwhelming evidence for a clinically important
Clinical Trials benefit of sacubitril/valsartan on the primary end point
Because of the salutary effects of natriuretic peptides of cardiovascular death or HF hospitalization with sacu-
in HF, NEP inhibition has been pursued as a therapeutic bitril/valsartan compared with enalapril (HR, 0.80 [95%
strategy. The early use of omapatrilat, a dual vasopepti- CI, 0.73–0.87]; P<0.001). The benefits with sacubitril/
dase inhibitor that inhibits both ACE (angiotensin-con- valsartan were consistent across study end points includ-
verting enzyme) and NEP, was not shown to be more ing all-cause mortality (HR, 0.84 [95% CI, 0.76–0.93],
effective than ACE inhibition alone in HF patients.30 In P<0.001), cardiovascular mortality (HR, 0.80 [95% CI,
the OVERTURE (Omapatrilat Versus Enalapril Random- 0.71–0.89], P<0.001), and hospitalization for HF (HR,
ized Trial of Utility in Reducing Events), which was con- 0.79 [95% CI, 0.71–0.89], P<0.001). These benefits
ducted in 5770 chronic HF patients, omapatrilat reduced were consistent across a broad array of prespecified
the composite of cardiovascular death or hospitalization subgroups and resulted in an estimated increase in sur-
(hazard ratio [HR], 0.91 [95% CI, 0.84–0.99], P=0.024); vival of 1.4 years for a 55-year-old patient treated with
however, there was no difference between the therapies sacubitril/valsartan compared with enalapril.36 Patients
for the primary end point of death or HF hospitalization receiving sacubitril/valsartan had more symptomatic
(P=0.187).31 Parallel studies with omapatrilat in patients hypotension and nonserious angioedema but less renal
with hypertension demonstrated an increased risk for impairment, hyperkalemia, and cough than the enalapril
serious angioedema, presumably due to increased bra- group. The findings from PARADIGM-HF supported the
dykinin levels secondary to combined ACE inhibition and role of sacubitril/valsartan as superior to ACE inhibition in
NEP inhibition. The clinical development of omapatrilat the management of chronic HFrEF, leading to a class IA
was halted because of safety concerns.32 indication for patients with chronic symptomatic HFrEF
The next therapeutic attempt at NEP inhibition com- (NYHA class II or III) to further reduce morbidity and mor-
bined sacubitril, a prodrug neprilysin inhibitor, with an tality.28,37 Sacubitril/valsartan also has favorable effects
angiotensin receptor blocker. This therapeutic class is on symptom burden in HFrEF. In PARAGIDM-HF, sacubi-
referred to as ARNIs. The rationale for ARNIs was that tril/valsartan treatment led to clinically important benefits

Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158 May 14, 2021   1441


Mann and Felker Translational Advances in HFrEF
Heart Failure Compendium

on health-related quality of life as assessed by the Kan- Mechanism of Action


sas City Cardiomyopathy Questionnaire compared with
Because glucose is a polar molecule, it cannot be trans-
enalapril,38 as well as improvements in functional status
ported across the lipid cell membrane and requires carrier
as estimated by NYHA class. Subsequent randomized
proteins, referred to as glucose transporters, in order, to
clinical trials and additional analyses of completed stud-
enter the cell. Two families of glucose transporters have
ies have added to the overall picture of both the clinical
been identified, including facilitative diffusion GLUTs (glu-
benefits and the safety of sacubitril/valsartan. The PIO-
cose transporters) and SGLTs. There are 2 major SGLT2
NEER-HF (Comparison of Sacubitril/Valsartan Versus
isoforms in human tissues: SGLT1 and SGLT2.43 SGLT1
Enalapril on Effect on NT-proBNP in Patients Stabilized
serves as a high-affinity, low-capacity transporter that is
From an Acute Heart Failure Episode). study established
able to transport glucose against a concentration gradient,
the safety and efficacy of early initiation (in hospital) of
whereas SGLT2 is low affinity, high-capacity transporter.
sacubitril/valsartan in patients hospitalized for HF.39 Quantitative studies of SGLT1 and SGLT2 gene expres-
sion in human tissue have shown that SGLT1 is highly
Effects on LV Remodeling expressed in the small intestine, whereas SGLT2 is very
highly expressed in the kidney.43 In human tissue, SGLT2
Given the atypical sequence of clinical development,
mRNA is expressed ubiquitously (including the heart) and
there was substantial evidence for improvement in clinical
is generally 10- to 100-fold higher than the expression
outcomes with sacubitril/valsartan before understand-
of SGLT1 in the same tissues. One notable exception is
ing whether there were beneficial effects on reverse LV
that SGLT1 mRNA is more highly expressed in the heart
remodeling, which had been observed with prior neuro-
than SGLT2. Relevant to this discussion, SGLT1 mRNA
hormonal therapies that improve outcomes. Subsequent
and protein have been detected in cardiac myocytes,43,44
studies have confirmed the substantial favorable effects
whereas (at the time of the writing) there have been no
on LV remodeling with sacubitril/valsartan. PROVE-HF
comparable studies for SGLT2 in cardiac myocytes. Stud-
(Prospective Study of Biomarkers, Symptom Improve-
ies using Mendelian randomization have estimated that
ment, and Ventricular Remodeling During Sacubitril/
lower SGLT1 expression would be associated with lower
Valsartan Therapy for Heart Failure), a single-arm mul-
rates of obesity, diabetes, HF, and mortality.45
ticenter study, demonstrated significant improvements
In the kidney, SGLT2 is located in the S1 and S2 seg-
in cardiac remodeling with sacubitril/valsartan.40 As
ments of the proximal tubule in the kidneys and accounts
expected, the magnitude of the changes in LV remodeling
Downloaded from http://ahajournals.org by on May 17, 2021

for 90% of glucose reabsorption by the kidney. SGLT1


after initiation of sacubitril/valsartan was related to the
is located in the S3 segment of the proximal tubules and
risk of clinical outcomes, with those patients having more
accounts for the remaining 10% of glucose absorption.
favorable remodeling demonstrating improved progno-
SGLT2 is also responsible for proximal tubular reabsorp-
sis.41 Similar findings on cardiac remodeling have been
tion of sodium, and the passive absorption of chloride that
confirmed in the double-blind EVALUATE-HF (Study of is driven by the resulting electrochemical gradient in the
Effects of Sacubitril/Valsartan vs. Enalapril on Aortic Stiff- proximal tubule lumen. The increased absorption of sodium
ness in Patients With Mild to Moderate HF With Reduced and chloride in the proximal tubule results in lower chloride
Ejection Fraction) trial, which showed decreased systolic concentration delivered to the macula densa, which in turn
and diastolic ventricular volumes, as well as atrial volumes results in dilation of the afferent arteriole and increase glo-
with sacubitril/valsartan compared with enalapril.42 merular filtration through tubuloglomerular feedback, which
preserves renal blood flow and glomerular filtration rate.
Inhibition of SGLT2 results in a 1:1 stoichiometric inhibition
SGLT2 INHIBITORS of sodium and glucose uptake in the proximal tubule of
In contrast to the therapeutic development of ARNIs, the kidney. This leads to contraction of the plasma volume
which leveraged insights provided by the neurohor- and modest lowering of blood pressure, without activa-
monal model, the development of SGLT2i as a therapy tion of the sympathetic nervous system. The contraction
for HF with a reduced ejection was the result of Food of plasma volume may contribute to changes in markers
and Drug Administration policies that required that all of hemoconcentration with SGLT2 inhibitors, including
new antidiabetic drugs should be evaluated in large car- increases in blood urea nitrogen and hematocrit, although
diovascular outcome trials to establish the cardiovascu- the latter may also be on the basis of increased erythropoi-
lar safety profile of new antidiabetic therapies. As will esis. The proximal natriuresis that occurs with SGLT2 inhi-
be discussed below, subsequent studies demonstrated bition results in afferent arteriole vasoconstriction through
a clear benefit for this therapeutic class on important tubuloglomerular feedback, thereby reducing glomerular
cardiovascular and renal outcomes. Although data con- hyperfiltration. Experimental studies showed that SGLT2
tinues to accumulate, it appears that SGLT2is will be inhibitors reduce hyperfiltration and decrease inflammatory
among the most important advances in cardiovascular and fibrotic responses of proximal tubular cells.46 Beyond
therapeutics of recent decades. effects on traditional cardiovascular risk factors, such as

1442   May 14, 2021 Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158


Mann and Felker Translational Advances in HFrEF

Heart Failure Compendium


HbA1c and weight, SGLT2 inhibition also reduces plasma ≤40%), 10 mg of empagliflozin daily reduced HF hospi-
uric acid levels by 10% to 15% by increasing uricosuria via talization (HR, 0.75 [95% CI, 0.65–0.86]; P<0.001) com-
exchange of filtered glucose. pared with placebo.54 In this study, however, the reduction
in cardiovascular death with empagliflozin did not reach
statistical significance (HR, 0.92 [95% CI, 0.75–1.12]).
Clinical Trials Quality of life was improved with empagliflozin treatment
Although SGLT2is were developed originally as a glu- compared with placebo, similar to prior results with dapa-
cose-lowering antidiabetic therapy, SGLT2is demon- gliflozin.55 The combined data from the DAPA-HF and
strated significant benefits on cardiovascular mortality, EMPEROR-Reduced trials have been recently summa-
MI, and stroke in cardiovascular outcome trials.47–49 rized in a meta-analysis.56
Somewhat unexpectedly, there was also a consis- Another recent study, the SOLOIST-WHF trial (Effect
tent signal in the trials of HF prevention in the SGLT2i of Sotagliflozin on Cardiovascular Events in Patients With
treatment arm as compared to placebo. The landmark Type 2 Diabetes Post Worsening Heart Failure), focused
EMPA-REG-OUTCOME (BI 10773 [Empagliflozin] Car- on a different population and clinical scenario. This trial
diovascular Outcome Event Trial in Type 2 Diabetes Mel- randomized 1222 patients with diabetes regardless of
litus Patients) study demonstrated a significant reduction EF to either sotagliflozin or placebo.57 Sotagliflozin dif-
in the composite of cardiovascular death, nonfatal MI, or fers from other available SGLT2is in that it inhibits both
stroke (HR in the empagliflozin group, 0.86; [95.02% CI, SGLT1 and SGLT2. Patients were enrolled at the time
0.74–0.99]).49 More striking was the 34% reduction in HF of a discharge from a hospitalization for HF or soon
hospitalization that was consistently observed in patients thereafter. The primary end point was cardiovascular
with and without a prior history of HF (10% of the EMPA- death and total HF hospitalization (including recurrent
REG-OUTCOME population).50 These findings on car- HF hospitalizations). This study showed a dramatic treat-
diovascular outcomes were subsequently replicated for ment effect for sotagliflozin in this population, with an
other SGLT2is, including dapagliflozin (in the DECLARE- HR of 0.67 (95% CI, 0.52–0.85; P<0.001). This was
TIMI58 study [Dapagliflozin Effect on Cardiovascular consistent regardless of EF at study entry, including for
Events–Thrombolysis in Myocardial Infarction-58])48 and patients with EF>50%. Importantly, the study was dis-
canagliflozin (in the CANVAS [Canagliflozin Cardiovas- continued early by the sponsor for financial reasons and
cular Assessment Study] and CREDENCE study [Cana- thus was underpowered to show more definitive effects
gliflozin and Renal Events in Diabetes With Established on cardiovascular mortality, but was broadly consistent
Downloaded from http://ahajournals.org by on May 17, 2021

Nephropathy Clinical Evaluation]).47,51 These findings led with the findings from other HFrEF trials. This is notable
to large programs designed to assess the benefit of these as the first study to show benefit on clinical outcomes
agents in patients across the spectrum of HF patients, in patients with HF with preserved EF using a specific
including those with and without diabetes. drug therapy. An additional notable feature of the long-
The first of these studies to present the results was the term SGLT2i trials has been their impact on slowing the
DAPA-HF (Study to Evaluate the Effect of Dapagliflozin on progression of chronic kidney disease (Figure 3), which
the Incidence of Worsening Heart Failure or Cardiovascu- has been shown with dapagliflozin,59 empagliflozin,60 and
lar Death in Patients With Chronic Heart Failure) study of canagliflozin.51 SGLT2i in HF patients are generally very
dapagliflozin in patients with HFrEF. DAPA-HF random- well tolerated, with very low risk of hypoglycemia, volume
ized 4744 patients with NYHA class II to IV HF and EF depletion, or hypotension similar to placebo.52,54
≤40% to either dapagliflozin or placebo. This study dem-
onstrated a significant reduction in both the combined
end point of cardiovascular death+HF hospitalization Effects on LV Remodeling
(HR, 0.74 [95% CI, 0.65–0.85]; P<0.001) as well as on Four studies have evaluated the effect of SGLT2is on LV
cardiovascular death (HR 0.82 [95% CI, 0.69–0.98]).52 remodeling in HF patients with a reduced EF.61–64 Three
Notably, these findings were completely consistent studies demonstrated a significant decrease in LV volumes
whether or not patients had diabetes at baseline (42% of after a minimum of 3 months of treatment with empa-
DAPA-HF population), showing for the first time important gliflozin as compared to placebo.61,62,64 In contrast, 1 year
clinical benefits in patients without diabetes. Dapagliflozin of treatment with dapagliflozin in diabetic HF patients did
also improved quality of life in this study assessed by the show beneficial effect of LV volumes by cardiac magnetic
Kansas City Cardiomyopathy Questionnaire, making this resonance imaging.63 Given that 100% of the patients
one of the few classes to improve morbidity, mortality, and in the dapagliflozin LV remodeling study had diabetes,
quality of life in HFrEF.53 More recently, the EMPEROR- whereas the number of diabetics varied widely (0%–78%)
Reduced study (Empagliflozin Outcome Trial in Patients in the empagliflozin trials, it is difficult to comment on
with Chronic Heart Failure and a Reduced Ejection Frac- whether the effects of SGLT2is on reverse LV remodeling
tion) with empagliflozin also reported similar beneficial should or should not be viewed as a class effect.
effects in HF patients with reduced EF. In a similar HFrEF The striking results with SGLT2i in patients with HFrEF
population to the DAPA-HF study (NYHA class II–IV, EF represent an exciting example of reverse translation
Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158 May 14, 2021   1443
Mann and Felker Translational Advances in HFrEF
Heart Failure Compendium
Downloaded from http://ahajournals.org by on May 17, 2021

Figure 3. Summary of potential renal protective mechanisms of SGLT2 (sodium-glucose cotransporter 2) inhibitors.
Reproduced from Cherney and Verma.58 CKD indicates chronic kidney disease; and mTORC1, mammalian target pf rapamycin 1. Copyright ©2021.

(bedside to bench), whereby the unexpected results of a neatly into one of the 3 aforementioned conceptual mod-
clinical trial serve as the stimulus to explore novel actions els for HF, rather the pleiotropic mechanisms of action
for a class of drugs that had demonstrated marked of SGLT2is place them at the intersection of metabolic,
effects on clinically meaningful HF outcomes. However, hemodynamic, neurohumoral, and vascular endothelial
what is most provocative about the reverse translation in pathways that impact the heart and the kidney, all of
this instance is that it suggests that there are unknown which are important in the pathogenesis of HF regard-
mechanisms of action for a diabetic drug, that are opera- less of the LVEF. The precise mechanisms underlying the
tive even in the absence of diabetes. Several cardiac and observed cardiorenal benefits of SGLT2is is an area of
extracardiac mechanisms have been proposed, increased active research that is likely to evolve rapidly.
natriuresis and diuresis, renal protective effects, enhanced
cardiac substrate metabolism, improved vascular stiffness,
reduced LV mass, direct inhibitory effects on the cardiac SOLUBLE GUANYLATE CYCLASE
sodium-hydrogen exchanger, decreased inflammation, STIMULATORS
stimulation of cardiac autophagy and mitophagy, reduc-
tion in adipokines, stimulation of EPO (erythropoietin) Mechanism of Action
production, and attenuation or renal afferent sympathetic As noted above, NO is a free radical gas that serves
nervous system activity (Figure 4).65–68 As summarized in as a key signaling molecule in the vasculature and the
Table 3, many of these mechanisms of action do not fit heart. NO is produced by 3 different isoforms of NOS
1444   May 14, 2021 Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158
Mann and Felker Translational Advances in HFrEF

Heart Failure Compendium


Table 3.  Overview of Potential Mechanism of Beneficial
Cardiovascular Effects of SGLT2 Inhibitors

Conceptual model
of heart failure Mechanism of action
Cardiorenal Stimulation of natriuresis
Stimulation of osmotic diuresis
Decreased tubuloglomerular feedback
Cardiocirculatory Improved systolic and diastolic function
Improved cardiac filling conditions secondary to
reductions in preload and afterload
Inhibition of cardiac fibrosis
Increased cardiac output, increased coronary blood
flow mediated by increased levels of circulating
glucagon
Improved myocardial energetics
Neurohormonal Decreased central nervous system sympathetic
nervous activity
Other Reduction in myocardial CaMKII activity
Increased erythropoietin
Increased circulating proangiogenic progenitor cells
Inhibition of cardiac myocyte Na+/H exchanger
Improved endothelial function
Figure 4. Potential direct myocardial and indirect±systemic Increased mitophagy/autophagy
effects of SGLT2 (sodium-glucose cotransporter 2) inhibitors.
CAMKII indicates calmodulin-dependent protein kinase II; and SGLT2, sodi-
CAMKII indicates calmodulin-dependent protein kinase II; EPO, um-glucose cotransporter 2.
erythropoietin; NHE, sodium/hydrogen exchanger; NLRP3, Adapted from Lam et al, J Am Heart Assoc. 2019;8:e013389.65 Copyright ©2019.
nucleotide-binding oligomerization domain, leucine-rich repeat, and
pyrin domain-containing 3; and SNS, sympathetic nervous system.
Reproduced from Lopaschuk and Verma.68 Copyright ©2020. the identification of sGC as a therapeutic target in HF is
Downloaded from http://ahajournals.org by on May 17, 2021

rooted in biological and physiological insights provided


(NO synthase), all of which are present in the heart, and by the cardiocirculatory and neurohormonal HF models.
include NOS1 (nNOS [neuronal NOS]), NOS2 (iNOS The relatively recent discovery of compounds that acti-
[inducible NOS]), and NOS3 (so-called eNOS [endothe- vate sGC in a NO-independent manner led to the therapeu-
lial-constitutive NOS]). NO exerts its effects, at least in tic development of sGC stimulators and sGC activators.72
part, by binding to the sGC receptor, which consists of a sGC stimulators directly stimulate the reduced form of sGC
larger α-subunit and a smaller heme-binding β-subunit, and enhance the sensitivity of the reduced enzyme to low
which is essential for detecting NO in the cytoplasm (Fig- levels of bioavailable NO. In contrast, sGC activators acti-
ure 2).69 Activation of sGC by NO requires the presence vate the NO-unresponsive heme oxidized enzyme.69 The
of a reduced Fe2+ heme moiety on the β-subunit. Oxida- NO-independent stimulators of sGC have been evaluated
tion of the heme moiety (Fe3+) abolishes endogenous in several different HF trials, as will be discussed below.
NO-induced activation of sGC signaling.70 NO-induced
activation of sGC leads to the production of cGMP, which
Clinical Trials
mediates 3 intracellular effector pathways, including
cGMP-dependent protein kinases I and II, cGMP-gated Vericiguat (BAY 1021189) is a novel oral soluble gua-
ion channels, and cGMP-regulated phosphodiester- nylate cyclase stimulator that has undergone exten-
ases.69 The NO-sGC-cGMP pathway is crucial for the sive clinical testing. Initial studies of vericiguat in HF
control of vascular homeostasis. In healthy subjects, NO included a phase II program composed of 2 studies,
is released continuously by vascular endothelial cells, SOCRATES-Reduced (Soluble Guanylate Cyclase Stim-
thereby promoting vascular smooth muscle cell relaxation ulator in Heart Failure With Reduced Ejection Fraction)
and vasodilation. Endothelium-dependent NO-mediated and SOCRATES-Preserved (Phase IIb Safety and Effi-
dilation of the peripheral vasculature is attenuated in HF cacy Study of Four Dose Regimens of BAY1021189 in
patients, which has been attributed to decreased NO bio- Patients With Heart Failure and Preserved Ejection Frac-
availability secondary to NOS3 uncoupling.71 The loss of tion Suffering From Worsening Chronic Heart Failure).
NO bioavailability impairs endothelium-dependent vaso- SOCRATES-Reduced randomized patients with LVEF
motor regulation and results in peripheral arterial vaso- ≤45% within 4 weeks of a worsening HF event (hospi-
constriction and increased ventricular afterload. Thus, the talization for HF or treatment of worsening HF with IV
identification of impaired NO-sGC-cGMP signaling and diuretics in the ambulatory setting) to either placebo or

Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158 May 14, 2021   1445


Mann and Felker Translational Advances in HFrEF
Heart Failure Compendium

1 of 4 different dosing regimens (from 1.5 mg daily to


10 mg daily).73 The primary end point of this phase 2 trial
EFFECTS ON LV REMODELING
was change in log plasma NT-proBNP (N-terminal pro- In an echocardiographic substudy of the VICTORIA trial,
B-type natriuretic peptide) concentrations from baseline patients were studied at baseline and after 8 months
to 12 weeks. Although the study did not meet its primary of therapy (N=211 in each arm). The primary end point
end point, there was evidence of greater natriuretic pep- was a change in LVEF and LV end-systolic volume index.
tide lowering in the higher dose arms, particularly 10 mg The VICTORIA echocardiographic substudy showed that
daily. The higher dose of 10 mg daily was also associated both LV EF and LV end-systolic volume index significantly
with a reduction in the estimated risk of rehospitalization improved from baseline in both arms through 8 months
of cardiovascular death, although this was not statisti- of treatment. However, treatment with vericiguat had no
cally significant in this phase 2 study (HR, 0.53 [95% additional significant effect on LVEF or LV end-systolic
CI, 0.25–1.16]). In contrast to SOCRATES-Reduced, volume index as compared to placebo.78
the SOCRATES-Preserved study with a similar design in
patients with EF>45% did not show significant improve-
ment on NT-proBNP concentrations.74 CARDIAC MYOSIN ACTIVATORS
The VICTORIA study (VerICiguaT Global Study in Because decreased cardiac output is regarded as a car-
Subjects With Heart Failure With Reduced Ejection Frac- dinal feature of HFrEF, there have been many attempts
tion) randomized 5050 patients with NYHA class II to to develop inotropic agents to improve cardiac output
IV HF and EF≤45% to vericiguat 10 mg daily or pla- by increasing the force of cardiac contraction (cardiocir-
cebo. Patients were selected to be a higher risk cohort, culatory model). However, despite 3 decades of inten-
requiring an HF hospitalization within the prior 6 months, sive efforts, no positive inotrope is currently approved
or outpatient IV diuretic therapy without hospitalization for long-term use in chronic HFrEF.79 Several reasons
within the prior 3 months, as well as elevated natriuretic have been proposed for the negative outcomes of clini-
peptide concentrations. Patients were followed for a cal trials with positive inotropic agents, including patient
median of 10.8 months, and the primary end point was selection, trial design, and trial end points.79 Moreover,
the composite of cardiovascular death or first HF hospi- it should also be recognized that many of the compen-
talization. There was a significant treatment benefit for satory mechanisms that are activated in HFrEF restore
patients randomized to vericiguat with an HR, 0.90 (95% cardiac output to normal or near-normal levels in the
Downloaded from http://ahajournals.org by on May 17, 2021

CI, 0.81–1.00).75 Deaths from cardiovascular causes early stages of the disease,80 albeit at the expense of
were nonsignificantly reduced with an HR of 0.93 (95% inappropriate salt and water retention and excessive
CI, 0.81–1.06). Of note, the overall risk profile of patients activation of the SNS and RAAS.
enrolled in VICTORIA was significantly higher than Omecamtiv mecarbil is a first-in-class small-molecule
patients enrolled in many other HFrEF outcome trials, activator of cardiac myosin ATPase that increases the
with a 1-year control group event rate of 34% (compared proportion of myosin heads that are tightly bound to
with 14% in PARADIGM-HF, 16% in DAPA-HF, and actin. Omecamtiv mecarbil increases force generation of
28% for GALACTIC-HF [Global Approach to Lowering the heart by prolonging myocardial contraction. To dif-
Adverse Cardiac Outcomes Through Improving Contrac- ferentiate myosin activators from classic inotropes, they
tility in Heart Failure]), suggesting that the relatively mod- have been referred to as myotropes.81 Whereas inotropic
est relative risk reduction of 10% in the primary end point agents increase cardiac output by increasing intracellular
was associated with a clinically important absolute risk calcium levels in the cell, which can lead to myocardial
reduction, given the high baseline risk of the study popu- ischemia and cardiac arrhythmias, omecamtiv mecarbil
lation.76 Although subgroup analyses generally showed a does not alter intracellular calcium concentrations and
consistent benefit of vericiguat across subgroups, there should presumably have a more favorable safety profile.
was notable heterogeneity with regard to baseline natri- However, given that omecamtiv mecarbil increases sys-
uretic peptide concentrations, with patients with higher tolic ejection time, there is at least theoretical concern
natriuretic peptide levels deriving less benefit.77 The that this agent could adversely affect diastolic coronary
clinical interpretation of this finding remains uncertain, filling and potential myocardial ischemia. Early data in
given the VICTORIA trial focused on higher risk patients patients with known ischemic cardiomyopathy did not
overall but the primary benefit seemed to be in the lower suggest an increase in ischemia with exercise tread-
risk cohorts within the trial. Vericiguat was generally mill testing during OM treatment.82 Clinical trials have
well tolerated, with vericiguat treatment associated with consistently demonstrated a small increase in circulat-
more symptomatic hypotension (9.1% versus 7.9%) and ing cardiac troponin concentrations in patients treated
anemia (7.6% versus 5.7%). Based on the results of the with omecamtiv mecarbil compared with placebo (on
VICTORIA trial, the Food and Drug Administration has the order of 1–4 ng/L), which resolves on discontinua-
approved vericiguat to lower the risk of HF in high-risk tion of omecamtiv mecarbil. Early studies also found that
patients who have symptomatic, chronic HFrEF<45%. supratherapeutic plasma concentrations (>1200 ng/

1446   May 14, 2021 Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158


Mann and Felker Translational Advances in HFrEF

Heart Failure Compendium


mL) were associated with adverse ischemic events. Sub- study population overall, although there were potentially
sequent implementation of a therapeutic drug monitoring important differences noted between those enrolled as
protocol targeting plasma concentrations <300 pg/mL inpatients versus those enrolled as outpatients in the
was successful at avoiding supratherapeutic drug levels effect of omecamtiv mecarbil on quality of life. Although
in subsequent studies.83 Omecamtiv mecarbil prolonged modestly elevated cardiac troponin was observed in
systolic ejection time in a dose-dependent manner and omecamtiv mecarbil treated patients in GALACTIC
thereby improves stroke volume and systolic cardiac consistent with prior studies, there was no increase in
performance in both normal healthy volunteers84 and in myocardial ischemia events, ventricular arrhythmias, or
patients with HFrEF.85 mortality compared with placebo. An additional phase
III trial called METEORIC-HF (Study to Assess the
Effect of Omecamtiv Mecarbil on Exercise Capacity in
Clinical Trials Subjects With Heart Failure) (https://www.clinicaltrials.
The ATOMIC-HF study (Acute Treatment With Omecam- gov; Unique identifier: NCT03759392) and is currently
tiv Mecarbil to Increase Contractility in Acute Heart enrolling focused on the effect of omecamtiv mecarbil
Failure) was a randomized dose-finding clinical trial of on exercise capacity as measured by cardiopulmonary
short-term (48-hour) infusion of omecamtiv mecarbil in exercise testing and actigraphy. The results of the sub-
patients with acute decompensated HF. ATOMIC-HF group analyses of GALACTIC-HF, which suggested that
study showed improvement in systolic ejection time and patients with lower EFs and in normal sinus rhythm were
suggested improvement in dyspnea in the higher dose more likely to have a favorable treatment effect with
group.86 Subsequently in a larger phase 2 clinical trial in omecamtiv mecarbil, suggests that depressed contrac-
HFrEF, the COSMIC study (The Chronic Oral Study of tile function is both necessary and sufficient to explain
Myosin Activation to Increase Contractility in Heart Fail- pathogenesis of HF in patients with more advanced dis-
ure), administration of omecamtiv mecarbil for 20 weeks ease. Multiple ongoing analyses will provide additional
increased LV systolic ejection time and stroke volume, data to clarify the potential role of omecamtiv in HF care.
decreased LV systolic and diastolic volumes suggest-
ing beneficial reverse cardiac remodeling, and reduced
plasma natriuretic peptide concentrations and heart SUMMARY
rate.83 These phase II trial findings were notable in that In the foregoing review, we have discussed the results
Downloaded from http://ahajournals.org by on May 17, 2021

interventions that lower natriuretic peptides and induce of recent HF clinical trials that have employed novel
favorable LV remodeling have generally been shown to therapeutic classes of pharmacological agents. With the
improve HF outcomes in cardiovascular death in larger exception of SGLT2 inhibitors, each of these therapeu-
trials. Subsequently, analyses of these data also sug- tic advances was informed by the insights provided by
gested improvements in health-related quality of life.87 existing conceptual models of HF. For example, ARNIs
The GALACTIC-HF study was a global, phase 3, and soluble guanylate cyclase stimulators attenuate
double-blind, placebo-controlled randomized clinical trial the deleterious effects of SNS and RAAS signaling by
evaluating the efficacy and safety of omecamtiv mecar- upregulating cGMP-mediated signaling pathways that
bil compared with placebo in 8256 patients with symp- directly counteract the deleterious effects of these sig-
tomatic (NYHA class II–IV) HF with an EF ≤35%.88,89 naling pathways (neurohormonal model). The early suc-
Enrolled patients were required to be currently hospital- cess with this therapeutic strategy has given rise to other
ized for HF (inpatients) or had an urgent visit to the emer- approaches to augment endogenous natriuretic peptide
gency department or been hospitalized for HF within 1 and NO-mediated signaling, using phosphodiesterase
year before screening (outpatients). All the patients had 9 inhibitors to delay cGMP degradation.91 Omecamtiv
elevated natriuretic peptides (NT-proBNP) level of ≥400 mecarbil improves the hemodynamic profile of patients
pg/mL (1200 pg/mL for patients in atrial fibrillation) or with advanced heart by directly targeting sarcomere
BNP ≥125 pg/mL (375 pg/mL for patients in atrial fibril- function rather than by altering calcium handling, sug-
lation). GALACTIC demonstrated a significant improve- gesting that small molecules that allosterically modulate
ment in the primary end point, a composite of time to first troponin or myosin might be also be developed in the
HF event or death from cardiovascular causes (HR, 0.92 future (hemodynamic model).92 Although the mecha-
[95% CI, 0.86–0.99]; P=0.03).90 This finding was driven nisms of action of SGLT2is are unknown and represent
primarily by improvement in HF hospitalization events, as an area of active investigation, this therapeutic class of
there was no difference in cardiovascular mortality (HR, drugs has stimulated considerable interest in additional
1.01 [95% CI, 0.92–1.11]). Subgroup analyses sug- strategies to improve cardiac energy production (hemo-
gested that patients with lower EFs and in normal sinus dynamic model) as well as improve renal function (car-
rhythm were more likely to have a favorable treatment diorenal model). The recent observation that SGLT2is
effect with omecamtiv mecarbil. There did not appear may also attenuate excessive SNS signaling (neurohor-
to be a significant improvement in quality of life in the monal model),93 raises the intriguing possibility that a

Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158 May 14, 2021   1447


Mann and Felker Translational Advances in HFrEF
Heart Failure Compendium

single therapeutic class of drug may favorably impact all increase with saline load in patients with dilated cardiomyopathy and mild
heart failure. J Clin Invest. 1991;88:1481–1489. doi: 10.1172/JCI115458
aspects of the pathogenesis of HF. In addition to ongoing 12. Ibebuogu UN, Gladysheva IP, Houng AK, Reed GL. Decompensated
efforts to develop new therapies, how best to combine heart failure is associated with reduced corin levels and decreased cleav-
available therapies in an era of multiple effective drugs age of pro-atrial natriuretic peptide. Circ Heart Fail. 2011;4:114–120. doi:
10.1161/CIRCHEARTFAILURE.109.895581
remains a major focus of clinical research.94 13. Lee CY, Burnett JC Jr. Natriuretic peptides and therapeutic applications.
Heart Fail Rev. 2007;12:131–142. doi: 10.1007/s10741-007-9016-3
14. Kass DA, Baughman KL, Pak PH, Cho PW, Levin HR, Gardner TJ, Halperin
ARTICLE INFORMATION HR, Tsitlik JE, Acker MA. Reverse remodeling from cardiomyoplasty in
human heart failure. External constraint versus active assist. Circulation.
Affiliations 1995;91:2314–2318. doi: 10.1161/01.cir.91.9.2314
Center for Cardiovascular Research, Washington University School of Medicine, 15. Levin HR, Oz MC, Chen JM, Packer M, Rose EA, Burkhoff D. Reversal of
St. Louis, MO (D.L.M.). Duke Clinical Research Institute and Duke University chronic ventricular dilation in patients with end-stage cardiomyopathy by
School of Medicine, Durham, Duke University, NC (G.M.F.). prolonged mechanical unloading. Circulation. 1995;91:2717–2720. doi:
10.1161/01.cir.91.11.2717
Acknowledgments 16. Kramer DG, Trikalinos TA, Kent DM, Antonopoulos GV, Konstam MA,
This study was supported by R01HL107594 (D.L. Mann) and U10 HL110309 Udelson JE. Quantitative evaluation of drug or device effects on ventricular
(D.L. Mann), AN no. 4345132 (D.L. Mann), American Heart Association (AHA) remodeling as predictors of therapeutic effects on mortality in patients with
SFRN (Strategically Focused Network Grant) 30180010 (G.M. Felker), R01 heart failure and reduced ejection fraction: a meta-analytic approach. J Am
HL144928-03 (G.M. Felker), R21 HL152148-01 (G.M. Felker). Coll Cardiol. 2010;56:392–406. doi: 10.1016/j.jacc.2010.05.011
17. Konstam MA, Udelson JE, Anand IS, Cohn JN. Ventricular remodeling in
Disclosures heart failure: a credible surrogate endpoint. J Card Fail. 2003;9:350–353.
D.L. Mann serves on the steering committee for the PARADISE-MI trial (Pro- doi: 10.1054/j.cardfail.2003.09.001
spective ARNI vs ACE Inhibitor Trial to DetermIne Superiority in Reducing 18. Packer M. How should physicians view heart failure? The philosophical and
Heart Failure Events After MI) (Novartis) and the ANTHEM-HFrEF trial (Au- physiological evolution of three conceptual models of the disease. Am J
tonomic Neural Regulation Therapy to Enhance Myocardial Function in Heart Cardiol. 1993;71:3C–11C. doi: 10.1016/0002-9149(93)90081-m
Failure with Reduced Ejection Fraction Study) (LivaNova) and is a member of 19. Mann DL. The evolution of modern theory and therapy for heart failure. Prog
the Scientific Advisory Board for MyoKardia, Tenaya, Cardurion, and Cardior Ped Cardiol. 2014;37:9–12.
and serves as a consultant for Novo Nordisk, G.M. Felker receives research 20. Ellison DH, Felker GM. Diuretic treatment in heart failure. N Engl J Med.
support from, Amgen, Bayer Merck, Cytokinetics, Myokardia; he acts as a 2018;378:684–685. doi: 10.1056/NEJMc1716477
consultant to Novartis, Amgen, BMS (Bristol Meyers Squibb), Cytokinetics, 21. Cohn JN, Archibald DG, Ziesche S, Franciosa JA, Harston WE, Tristani FE,
Medtronic, Cardionomic, Innolife, Boehringer-Ingelheim, American Regent, Dunkman WB, Jacobs W, Francis GS, Flohr KH. Effect of vasodilator ther-
Abbott, Astra-Zeneca, Eidos Therapeutics, Reprieve, and Sequana; and serves apy on mortality in chronic congestive heart failure. Results of a Veterans
on clinical end point committees/data safety monitoring boards for Amgen, Administration Cooperative Study. N Engl J Med. 1986;314:1547–1552.
Merck, Medtronic, EBR Systems, V-Wave, LivaNova, Siemens, and Rocket doi: 10.1056/NEJM198606123142404
Pharma. 22. Packer M, Carver JR, Rodeheffer RJ, Ivanhoe RJ, DiBianco R, Zeldis
SM, Hendrix GH, Bommer WJ, Elkayam U, Kukin ML. Effect of oral mil-
Downloaded from http://ahajournals.org by on May 17, 2021

rinone on mortality in severe chronic heart failure. The PROMISE Study


Research Group. N Engl J Med. 1991;325:1468–1475. doi: 10.1056/
REFERENCES NEJM199111213252103
1. Hartupee J, Mann DL. Neurohormonal activation in heart failure with 23. Metra M, Teerlink JR, Cotter G, Davison BA, Felker GM, Filippatos G,
reduced ejection fraction. Nat Rev Cardiol. 2017;14:30–38. doi: 10.1038/ Greenberg BH, Pang PS, Ponikowski P, Voors AA, et al; RELAX-AHF-2
nrcardio.2016.163 Committees Investigators. Effects of serelaxin in patients with acute heart
2. Mann DL, Barger PM, Burkhoff D. Myocardial recovery: myth, magic or failure. N Engl J Med. 2019;381:716–726. doi: 10.1056/NEJMoa1801291
molecular target? J Amer Coll Cardiol. 2012;60:2465–2472. 24. Teerlink JR, Cotter G, Davison BA, Felker GM, Filippatos G, Greenberg
3. Mann DL, Bristow MR. Mechanisms and models in heart failure: the bio- BH, Ponikowski P, Unemori E, Voors AA, Adams KF Jr, et al; RELAXin
mechanical model and beyond. Circulation. 2005;111:2837–2849. doi: in Acute Heart Failure (RELAX-AHF) Investigators. Serelaxin, recombi-
10.1161/CIRCULATIONAHA.104.500546 nant human relaxin-2, for treatment of acute heart failure (RELAX-AHF):
4. Mann DL. Mechanisms and models in heart failure: a combinatorial approach. a randomised, placebo-controlled trial. Lancet. 2013;381:29–39. doi:
Circulation. 1999;100:999–1008. doi: 10.1161/01.cir.100.9.999 10.1016/S0140-6736(12)61855-8
5. Floras JS, Ponikowski P. The sympathetic/parasympathetic imbalance in 25. Packer M, O’Connor C, McMurray JJV, Wittes J, Abraham WT, Anker SD,
heart failure with reduced ejection fraction. Eur Heart J. 2015;36:1974– Dickstein K, Filippatos G, Holcomb R, Krum H, et al; TRUE-AHF Investiga-
182b. doi: 10.1093/eurheartj/ehv087 tors. Effect of ularitide on cardiovascular mortality in acute heart failure. N
6. van Bilsen M, Patel HC, Bauersachs J, Böhm M, Borggrefe M, Brutsaert D, Engl J Med. 2017;376:1956–1964. doi: 10.1056/NEJMoa1601895
Coats AJS, de Boer RA, de Keulenaer GW, Filippatos GS, et al. The auto- 26. O’Connor CM, Starling RC, Hernandez AF, Armstrong PW, Dickstein K,
nomic nervous system as a therapeutic target in heart failure: a scientific Hasselblad V, Heizer GM, Komajda M, Massie BM, McMurray JJ, et al. Effect
position statement from the Translational Research Committee of the Heart of nesiritide in patients with acute decompensated heart failure. N Engl J
Failure Association of the European Society of Cardiology. Eur J Heart Fail. Med. 2011;365:32–43. doi: 10.1056/NEJMoa1100171
2017;19:1361–1378. doi: 10.1002/ejhf.921 27. Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE Jr, Drazner MH,
7. Schrier RW. Decreased effective blood volume in edematous disorders: Fonarow GC, Geraci SA, Horwich T, Januzzi JL, et al. 2013 ACCF/AHA
what does this mean? J Am Soc Nephrol. 2007;18:2028–2031. doi: guideline for the management of heart failure: a report of the American
10.1681/ASN.2006111302 College of Cardiology Foundation/American Heart Association Task Force
8. Volpe M, Rubattu S, Burnett J Jr. Natriuretic peptides in cardiovascular dis- on Practice Guidelines. Circulation. 2013;128:e240–e327.
eases: current use and perspectives. Eur Heart J. 2014;35:419–425. doi: 28. Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JGF, Coats AJS, Falk
10.1093/eurheartj/eht466 V, González-Juanatey JR, Harjola VP, Jankowska EA, et al; ESC Scientific
9. Buglioni A, Burnett JC Jr. New pharmacological strategies to increase Document Group. 2016 ESC guidelines for the diagnosis and treatment
cGMP. Annu Rev Med. 2016;67:229–243. doi: 10.1146/annurev-med- of acute and chronic heart failure: the task force for the diagnosis and
052914-091923 treatment of acute and chronic heart failure of the European Society of
10. Chen Y, Burnett JC Jr. Biochemistry, therapeutics, and biomarker implica- Cardiology (ESC)Developed with the special contribution of the Heart Fail-
tions of neprilysin in cardiorenal disease. Clin Chem. 2017;63:108–115. doi: ure Association (HFA) of the ESC. Eur Heart J. 2016;37:2129–2200. doi:
10.1373/clinchem.2016.262907 10.1093/eurheartj/ehw128
11. Volpe M, Tritto C, De Luca N, Mele AF, Lembo G, Rubattu S, Romano M, 29. Sucharov CC, Mariner P, Long C, Bristow M, Leinwand L. Yin Yang 1 is
De Campora P, Enea I, Ricciardelli B. Failure of atrial natriuretic factor to increased in human heart failure and represses the activity of the human

1448   May 14, 2021 Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158


Mann and Felker Translational Advances in HFrEF

Heart Failure Compendium


alpha-myosin heavy chain promoter. J Biol Chem. 2003;278:31233–31239. 47. Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, Shaw
doi: 10.1074/jbc.M301917200 W, Law G, Desai M, Matthews DR; CANVAS Program Collaborative Group.
30. Volpe M, Carnovali M, Mastromarino V. The natriuretic peptides system in Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl
the pathophysiology of heart failure: from molecular basis to treatment. Clin J Med. 2017;377:644–657. doi: 10.1056/NEJMoa1611925
Sci (Lond). 2016;130:57–77. doi: 10.1042/CS20150469 48. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Silverman MG,
31. Packer M, Califf RM, Konstam MA, Krum H, McMurray JJ, Rouleau JL, Zelniker TA, Kuder JF, Murphy SA, et al; DECLARE–TIMI 58 Investigators.
Swedberg K. Comparison of omapatrilat and enalapril in patients with Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med.
chronic heart failure: the Omapatrilat Versus Enalapril Randomized Trial of 2019;380:347–357. doi: 10.1056/NEJMoa1812389
Utility in Reducing Events (OVERTURE). Circulation. 2002;106:920–926. 49. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S,
doi: 10.1161/01.cir.0000029801.86489.50 Mattheus M, Devins T, Johansen OE, Woerle HJ, et al; EMPA-REG
32. Kostis JB, Packer M, Black HR, Schmieder R, Henry D, Levy E. Omapatrilat OUTCOME Investigators. Empagliflozin, cardiovascular outcomes, and
and enalapril in patients with hypertension: the Omapatrilat Cardiovascular mortality in type 2 diabetes. N Engl J Med. 2015;373:2117–2128. doi:
Treatment vs. Enalapril (OCTAVE) trial. Am J Hypertens. 2004;17:103–111. 10.1056/NEJMoa1504720
doi: 10.1016/j.amjhyper.2003.09.014 50. Fitchett D, Zinman B, Wanner C, Lachin JM, Hantel S, Salsali A,
33. Cohn JN, Tognoni G; Valsartan Heart Failure Trial Investigators. A random- Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE; EMPA-REG OUT-
ized trial of the angiotensin-receptor blocker valsartan in chronic heart fail- COME® trial investigators. Heart failure outcomes with empagliflozin in
ure. N Engl J Med. 2001;345:1667–1675. doi: 10.1056/NEJMoa010713 patients with type 2 diabetes at high cardiovascular risk: results of the
34. Ruilope LM, Dukat A, Böhm M, Lacourcière Y, Gong J, Lefkowitz MP. EMPA-REG OUTCOME® trial. Eur Heart J. 2016;37:1526–1534. doi:
Blood-pressure reduction with LCZ696, a novel dual-acting inhibitor of the 10.1093/eurheartj/ehv728
angiotensin II receptor and neprilysin: a randomised, double-blind, placebo- 51. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan
controlled, active comparator study. Lancet. 2010;375:1255–1266. doi: DM, Edwards R, Agarwal R, Bakris G, Bull S, et al; CREDENCE Trial
10.1016/S0140-6736(09)61966-8 Investigators. Canagliflozin and renal outcomes in type 2 diabe-
35. McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, tes and nephropathy. N Engl J Med. 2019;380:2295–2306. doi:
Rouleau JL, Shi VC, Solomon SD, Swedberg K, et al; PARADIGM-HF 10.1056/NEJMoa1811744
Investigators and Committees. Angiotensin-neprilysin inhibition ver- 52. McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod MN, Martinez
sus enalapril in heart failure. N Engl J Med. 2014;371:993–1004. doi: FA, Ponikowski P, Sabatine MS, Anand IS, Bělohlávek J, et al; DAPA-HF Trial
10.1056/NEJMoa1409077 Committees and Investigators. Dapagliflozin in patients with heart failure
36. Claggett B, Packer M, McMurray JJ, Swedberg K, Rouleau J, Zile MR, Jhund
and reduced ejection fraction. N Engl J Med. 2019;381:1995–2008. doi:
P, Lefkowitz M, Shi V, Solomon SD; PARADIGM-HF Investigators. Estimat-
10.1056/NEJMoa1911303
ing the long-term treatment benefits of sacubitril-valsartan. N Engl J Med.
53. Kosiborod MN, Jhund PS, Docherty KF, Diez M, Petrie MC, Verma S, Nicolau
2015;373:2289–2290. doi: 10.1056/NEJMc1509753
JC, Merkely B, Kitakaze M, DeMets DL, et al. Effects of dapagliflozin on
37. Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE Jr, Colvin MM, Drazner
symptoms, function, and quality of life in patients with heart failure and
MH, Filippatos GS, Fonarow GC, Givertz MM, et al. 2017 ACC/AHA/HFSA
reduced ejection fraction: results from the DAPA-HF Trial. Circulation.
focused update of the 2013 ACCF/AHA guideline for the management
2020;141:90–99. doi: 10.1161/CIRCULATIONAHA.119.044138
of heart failure: a report of the American College of Cardiology/Ameri-
54. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, Januzzi J,
can Heart Association Task Force on Clinical Practice Guidelines and the
Verma S, Tsutsui H, Brueckmann M, et al; EMPEROR-Reduced Trial Investi-
Heart Failure Society of America. J Am Coll Cardiol. 2017;70:776–803. doi:
gators. Cardiovascular and renal outcomes with empagliflozin in heart failure.
10.1016/j.jacc.2017.04.025
Downloaded from http://ahajournals.org by on May 17, 2021

N Engl J Med. 2020;383:1413–1424. doi: 10.1056/NEJMoa2022190


38. Lewis EF, Claggett BL, McMurray JJV, Packer M, Lefkowitz MP, Rouleau
55. Butler J, Anker SD, Filippatos G, Khan MS, Ferreira JP, Pocock SJ, Giannetti
JL, Liu J, Shi VC, Zile MR, Desai AS, et al. Health-related quality of life
N, Januzzi JL, Piña IL, Lam CSP, et al. Empagliflozin and health-related
outcomes in PARADIGM-HF. Circ Heart Fail. 2017;10:e003430. doi:
quality of life outcomes in patients with heart failure with reduced ejection
10.1161/CIRCHEARTFAILURE.116.003430
fraction: the EMPEROR-Reduced trial. Eur Heart J. 2021;42:1203–1212.
39. Velazquez EJ, Morrow DA, DeVore AD, Duffy CI, Ambrosy AP, McCague
doi: 10.1093/eurheartj/ehaa1007
K, Rocha R, Braunwald E; PIONEER-HF Investigators. Angiotensin-
neprilysin inhibition in acute decompensated heart failure. N Engl J Med. 56. Zannad F, Ferreira JP, Pocock SJ, Anker SD, Butler J, Filippatos G,
2019;380:539–548. doi: 10.1056/NEJMoa1812851 Brueckmann M, Ofstad AP, Pfarr E, Jamal W, et al. SGLT2 inhibitors in
40. Januzzi JL Jr, Prescott MF, Butler J, Felker GM, Maisel AS, McCague K, patients with heart failure with reduced ejection fraction: a meta-analysis of
Camacho A, Pina IL, Rocha RA, Shah AM, et al. Association of change in the EMPEROR-Reduced and DAPA-HF trials. Lancet. 2020;396:819–829.
N-terminal pro-B-type natriuretic peptide following initiation of sacubitril- doi: 10.1016/S0140-6736(20)31824-9
valsartan treatment with cardiac structure and function in patients with heart 57. Bhatt DL, Szarek M, Steg PG, Cannon CP, Leiter LA, McGuire DK, Lewis
failure with reduced ejection fraction. JAMA. 2019;322:1–11. JB, Riddle MC, Voors AA, Metra M, et al; SOLOIST-WHF Trial Investigators.
41. Januzzi JL Jr, Camacho A, Piña IL, Rocha R, Williamson KM, Maisel Sotagliflozin in patients with diabetes and recent worsening heart failure. N
AS, Felker GM, Prescott MF, Butler J, Solomon SD; PROVE-HF Inves- Engl J Med. 2021;384:117–128. doi: 10.1056/NEJMoa2030183
tigators. Reverse cardiac remodeling and outcome after initiation of 58. Cherney DZI, Verma S. DAPA-CKD: the beginning of a new era in renal
sacubitril/valsartan. Circ Heart Fail. 2020;13:e006946. doi: 10.1161/ protection. JACC Basic Transl Sci. 2021;6:74–77.
CIRCHEARTFAILURE.119.006946 59. Jhund PS, Solomon SD, Docherty KF, Heerspink HJL, Anand IS, Böhm
42. Desai AS, Solomon SD, Shah AM, Claggett BL, Fang JC, Izzo J, McCague M, Chopra V, de Boer RA, Desai AS, Ge J, et al. Efficacy of dapagliflozin
K, Abbas CA, Rocha R, Mitchell GF; EVALUATE-HF Investigators. Effect on renal function and outcomes in patients with heart failure with reduced
of sacubitril-valsartan vs enalapril on aortic stiffness in patients with heart ejection fraction: results of DAPA-HF. Circulation. 2021;143:298–309. doi:
failure and reduced ejection fraction: a randomized clinical trial. JAMA. 10.1161/CIRCULATIONAHA.120.050391
2019;322:1077–1084. 60. Zannad F, Ferreira JP, Pocock SJ, Zeller C, Anker SD, Butler J, Filippatos
43. Zhou L, Cryan EV, D’Andrea MR, Belkowski S, Conway BR, Demarest KT. G, Hauske SJ, Brueckmann M, Pfarr E, et al. Cardiac and kidney benefits
Human cardiomyocytes express high level of Na+/glucose cotransporter 1 of empagliflozin in heart failure across the spectrum of kidney function:
(SGLT1). J Cell Biochem. 2003;90:339–346. doi: 10.1002/jcb.10631 insights from EMPEROR-Reduced. Circulation. 2021;143:310–321. doi:
44. Banerjee SK, McGaffin KR, Pastor-Soler NM, Ahmad F. SGLT1 is a novel 10.1161/CIRCULATIONAHA.120.051685
cardiac glucose transporter that is perturbed in disease states. Cardiovasc 61. Omar M, Jensen J, Ali M, Frederiksen PH, Kistorp C, Videbæk L, Poulsen
Res. 2009;84:111–118. doi: 10.1093/cvr/cvp190 MK, Tuxen CD, Möller S, Gustafsson F, et al. Associations of empagliflozin
45. Seidelmann SB, Feofanova E, Yu B, Franceschini N, Claggett B, Kuokkanen with left ventricular volumes, mass, and function in patients with heart
M, Puolijoki H, Ebeling T, Perola M, Salomaa V, et al. Genetic variants in failure and reduced ejection fraction: a substudy of the Empire HF Ran-
SGLT1, glucose tolerance, and cardiometabolic risk. J Am Coll Cardiol. domized Clinical Trial [published online January 6, 2021]. JAMA Cardiol.
2018;72:1763–1773. doi: 10.1016/j.jacc.2018.07.061 2021:e206827. doi: 10.1001/jamacardio.2020.6827
46. Cherney DZ, Odutayo A, Aronson R, Ezekowitz J, Parker JD. Sodium glu- 62. Lee MMY, Brooksbank KJM, Wetherall K, Mangion K, Roditi G, Campbell
cose cotransporter-2 inhibition and cardiorenal protection. J Amer Coll Car- RT, Berry C, Chong V, Coyle L, Docherty KF, et al. Effect of empagliflozin on
diol. 2019;74:2511–2524. left ventricular volumes in patients with type 2 diabetes, or prediabetes, and

Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158 May 14, 2021   1449


Mann and Felker Translational Advances in HFrEF
Heart Failure Compendium

heart failure with reduced ejection fraction (SUGAR-DM-HF). Circulation. 80. Carlsson M, Andersson R, Bloch KM, Steding-Ehrenborg K, Mosén H,
2021;143:516–525. doi: 10.1161/CIRCULATIONAHA.120.052186 Stahlberg F, Ekmehag B, Arheden H. Cardiac output and cardiac index mea-
63. Singh JSS, Mordi IR, Vickneson K, Fathi A, Donnan PT, Mohan M, Choy sured with cardiovascular magnetic resonance in healthy subjects, elite ath-
AMJ, Gandy S, George J, Khan F, et al. Dapagliflozin versus placebo on left ven- letes and patients with congestive heart failure. J Cardiovasc Magn Reson.
tricular remodeling in patients with diabetes and heart failure: the REFORM 2012;14:51. doi: 10.1186/1532-429X-14-51
Trial. Diabetes Care. 2020;43:1356–1359. doi: 10.2337/dc19-2187 81. Psotka MA, Gottlieb SS, Francis GS, Allen LA, Teerlink JR, Adams KF Jr,
64. Santos-Gallego CG, Vargas-Delgado AP, Requena-Ibanez JA, Garcia-Ropero Rosano GMC, Lancellotti P. Cardiac calcitropes, myotropes, and mitotropes:
A, Mancini D, Pinney S, Macaluso F, Sartori S, Roque M, Sabatel-Perez F, JACC review topic of the week. J Am Coll Cardiol. 2019;73:2345–2353.
et al; EMPA-TROPISM (ATRU-4) Investigators. Randomized trial of empa- doi: 10.1016/j.jacc.2019.02.051
gliflozin in nondiabetic patients with heart failure and reduced ejection frac- 82. Greenberg BH, Chou W, Saikali KG, Escandón R, Lee JH, Chen MM,
tion. J Am Coll Cardiol. 2021;77:243–255. doi: 10.1016/j.jacc.2020.11.008 Treshkur T, Megreladze I, Wasserman SM, Eisenberg P, et al. Safety and
65. Lam CSP, Chandramouli C, Ahooja V, Verma S. SGLT-2 inhibitors in heart tolerability of omecamtiv mecarbil during exercise in patients with isch-
failure: current management, unmet needs, and therapeutic prospects. J Am emic cardiomyopathy and angina. JACC Heart Fail. 2015;3:22–29. doi:
Heart Assoc. 2019;8:e013389. doi: 10.1161/JAHA.119.013389 10.1016/j.jchf.2014.07.009
66. Verma S, Rawat S, Ho KL, Wagg CS, Zhang L, Teoh H, Dyck JE, Uddin GM, 83. Teerlink JR, Felker GM, McMurray JJ, Solomon SD, Adams KF Jr, Cleland
Oudit GY, Mayoux E, et al. Empagliflozin increases cardiac energy produc- JG, Ezekowitz JA, Goudev A, Macdonald P, Metra M, et al; COSMIC-HF
tion in diabetes. JACC Basic Transl Sci. 2018;3:575–587. Investigators. Chronic oral study of myosin activation to increase con-
67. Herat LY, Magno AL, Rudnicka C, Hricova J, Carnagarin R, Ward NC, tractility in heart failure (COSMIC-HF): a phase 2, pharmacokinetic, ran-
Arcambal A, Kiuchi MG, Head GA, Schlaich MP, et al. SGLT2 inhibitor- domised, placebo-controlled trial. Lancet. 2016;388:2895–2903. doi:
induced sympathoinhibition: a novel mechanism for cardiorenal protection. 10.1016/S0140-6736(16)32049-9
JACC Basic Transl Sci. 2020;5:169–179. doi: 10.1016/j.jacbts.2019.11.007 84. Teerlink JR, Clarke CP, Saikali KG, Lee JH, Chen MM, Escandon RD, Elliott
68. Lopaschuk GD, Verma S. Mechanisms of cardiovascular benefits of sodium L, Bee R, Habibzadeh MR, Goldman JH, et al. Dose-dependent augmenta-
glucose co-transporter 2 (SGLT2) inhibitors: a state-of-the-art review. JACC tion of cardiac systolic function with the selective cardiac myosin activator,
Basic Transl Sci. 2020;5:632–644. doi: 10.1016/j.jacbts.2020.02.004 omecamtiv mecarbil: a first-in-man study. Lancet. 2011;378:667–675. doi:
69. Evgenov OV, Pacher P, Schmidt PM, Haskó G, Schmidt HH, Stasch JP. NO- 10.1016/S0140-6736(11)61219-1
independent stimulators and activators of soluble guanylate cyclase: discov- 85. Cleland JG, Teerlink JR, Senior R, Nifontov EM, Mc Murray JJ, Lang
ery and therapeutic potential. Nat Rev Drug Discov. 2006;5:755–768. doi: CC, Tsyrlin VA, Greenberg BH, Mayet J, Francis DP, et al. The effects
10.1038/nrd2038 of the cardiac myosin activator, omecamtiv mecarbil, on cardiac func-
70. Foerster J, Harteneck C, Malkewitz J, Schultz G, Koesling D. A functional tion in systolic heart failure: a double-blind, placebo-controlled, cross-
heme-binding site of soluble guanylyl cyclase requires intact N-termini of over, dose-ranging phase 2 trial. Lancet. 2011;378:676–683. doi:
alpha 1 and beta 1 subunits. Eur J Biochem. 1996;240:380–386. doi: 10.1016/S0140-6736(11)61126-4
10.1111/j.1432-1033.1996.0380h.x 86. Teerlink JR, Felker GM, McMurray JJV, Ponikowski P, Metra M, Filippatos
71. Farah C, Michel LYM, Balligand JL. Nitric oxide signalling in cardio- GS, Ezekowitz JA, Dickstein K, Cleland JGF, Kim JB, et al; ATOMIC-AHF
vascular health and disease. Nat Rev Cardiol. 2018;15:292–316. doi: Investigators. Acute treatment with omecamtiv mecarbil to increase con-
10.1038/nrcardio.2017.224 tractility in acute heart failure: the ATOMIC-AHF Study. J Am Coll Cardiol.
72. Boerrigter G, Burnett JC Jr. Soluble guanylate cyclase: not a dull enzyme. 2016;67:1444–1455. doi: 10.1016/j.jacc.2016.01.031
Circulation. 2009;119:2752–2754. doi: 10.1161/CIRCULATIONAHA. 87. Felker GM, Solomon SD, McMurray JJV, Cleland JGF, Abbasi SA, Malik
109.860288 FI, Zhang H, Globe G, Teerlink JR; COSMIC-HF Investigators. Effects of
Downloaded from http://ahajournals.org by on May 17, 2021

73. Gheorghiade M, Greene SJ, Butler J, Filippatos G, Lam CS, Maggioni AP, omecamtiv mecarbil on symptoms and health-related quality of life in patients
Ponikowski P, Shah SJ, Solomon SD, Kraigher-Krainer E, et al; SOCRATES- with chronic heart failure: results from the COSMIC-HF Study. Circ Heart
REDUCED Investigators and Coordinators. Effect of vericiguat, a soluble Fail. 2020;13:e007814. doi: 10.1161/CIRCHEARTFAILURE.120.007814
guanylate cyclase stimulator, on natriuretic peptide levels in patients 88. Teerlink JR, Diaz R, Felker GM, McMurray JJV, Metra M, Solomon SD, Legg
with worsening chronic heart failure and reduced ejection fraction: the JC, Büchele G, Varin C, Kurtz CE, et al. Omecamtiv mecarbil in chronic
SOCRATES-REDUCED Randomized Trial. JAMA. 2015;314:2251–2262. heart failure with reduced ejection fraction: rationale and design of GALAC-
doi: 10.1001/jama.2015.15734 TIC-HF. JACC Heart Fail. 2020;8:329–340. doi: 10.1016/j.jchf.2019.12.001
74. Pieske B, Maggioni AP, Lam CSP, Pieske-Kraigher E, Filippatos G, Butler J, 89. Teerlink JR, Diaz R, Felker GM, McMurray JJV, Metra M, Solomon SD, Adams
Ponikowski P, Shah SJ, Solomon SD, Scalise AV, et al. Vericiguat in patients KF, Anand I, Arias-Mendoza A, Biering-Sorensen T, et al; GALACTIC-HF
with worsening chronic heart failure and preserved ejection fraction: results Investigators. Omecamtiv mecarbil in chronic heart failure with reduced
of the SOluble guanylate Cyclase stimulatoR in heArT failurE patientS ejection fraction, GALACTIC-HF: baseline characteristics and comparison
with PRESERVED EF (SOCRATES-PRESERVED) study. Eur Heart J. with contemporary clinical trials. Eur J Heart Fail. 2020;22:2160–2171. doi:
2017;38:1119–1127. doi: 10.1093/eurheartj/ehw593 10.1002/ejhf.2015
75. Armstrong PW, Pieske B, Anstrom KJ, Ezekowitz J, Hernandez AF, Butler 90. Teerlink JR, Diaz R, Felker GM, McMurray JJV, Metra M, Solomon
J, Lam CSP, Ponikowski P, Voors AA, Jia G, et al; VICTORIA Study Group. SD, Adams KF, Anand I, Arias-Mendoza A, Biering-Sørensen T, et al;
Vericiguat in patients with heart failure and reduced ejection fraction. N Engl GALACTIC-HF Investigators. Cardiac myosin activation with omecamtiv
J Med. 2020;382:1883–1893. doi: 10.1056/NEJMoa1915928 mecarbil in systolic heart failure. N Engl J Med. 2021;384:105–116. doi:
76. Butler J, Anstrom KJ, Armstrong PW. Comparing the benefit of novel 10.1056/NEJMoa2025797
therapies across clinical trials: insights from the VICTORIA Trial. Circulation. 91. Richards DA, Aronovitz MJ, Liu P, Martin GL, Tam K, Pande S, Karas
2020;142:717–719. doi: 10.1161/CIRCULATIONAHA.120.047086 RH, Bloomfield DM, Mendelsohn ME, Blanton RM. CRD-733, a novel
77. Ezekowitz JA, O’Connor CM, Troughton RW, Alemayehu WG, Westerhout PDE9 (Phosphodiesterase 9) inhibitor, reverses pressure overload-
CM, Voors AA, Butler J, Lam CSP, Ponikowski P, Emdin M, et al. N-terminal induced heart failure. Circ Heart Fail. 2021;14:e007300. doi: 10.1161/
pro-B-type natriuretic peptide and clinical outcomes: vericiguat heart failure CIRCHEARTFAILURE.120.007300
with reduced ejection fraction study. JACC Heart Fail. 2020;8:931–939. doi: 92. Alsulami K, Marston S. Small molecules acting on myofilaments as treat-
10.1016/j.jchf.2020.08.008 ments for heart and skeletal muscle diseases. Int J Mol Sci. 2020;21:E9599.
78. Stiles S. Discordant VICTORIA Results Deepen Mystery of Vericiguat in doi: 10.3390/ijms21249599
Low-EF Heart Failure. 2020. Accessed March 21, 2021. https://www.med- 93. Verma S. Are the cardiorenal benefits of SGLT2 inhibitors due to inhibition
scape.com/viewarticle/931976 of the sympathetic nervous system? JACC Basic Transl Sci. 2020;5:180–
79. Ahmad T, Miller PE, McCullough M, Desai NR, Riello R, Psotka M, Böhm M, 182. doi: 10.1016/j.jacbts.2020.01.011
Allen LA, Teerlink JR, Rosano GMC, et al. Why has positive inotropy failed 94. Bhatt AS, Abraham WT, Lindenfeld J, Bristow M, Carson PE, Felker GM,
in chronic heart failure? Lessons from prior inotrope trials. Eur J Heart Fail. Fonarow GC, Greene SJ, Psotka MA, Solomon SD, et al. Treatment of HF in
2019;21:1064–1078. doi: 10.1002/ejhf.1557 an Era of multiple therapies. JACC Heart Fail. 2021;9:1–12.

1450   May 14, 2021 Circulation Research. 2021;128:1435–1450. DOI: 10.1161/CIRCRESAHA.121.318158

You might also like