You are on page 1of 15

Cardiovascular Research (2023) 00, 1–15 SPOTLIGHT REVIEW

https://doi.org/10.1093/cvr/cvac187

Mechanisms of current therapeutic strategies


for heart failure: more questions than answers?

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


Muhammad Shahzeb Khan1, Izza Shahid2, Stephen J. Greene1,3, Robert J. Mentz1,3,
Adam D. DeVore1,3, and Javed Butler 4*
1
From the Division of Cardiology, Duke University School of Medicine, Durham, NC, USA; 2Division of Cardiovascular Prevention, Houston Methodist Academic Institute, Houston, TX, USA; 3Duke
Clinical Research Institute, Durham, NC, USA; and 4Baylor Scott and White Research Institute, Baylor University Medical Center, 3434 Live Oak St Ste 501, Dallas 75204, TX, USA

Received 13 May 2022; revised 19 October 2022; accepted 21 October 2022; online publish-ahead-of-print 20 December 2022

Abstract Heart failure (HF) is a complex, multifactorial and heterogeneous syndrome with substantial mortality and morbidity. Over the last
few decades, numerous attempts have been made to develop targeted therapies that may attenuate the known pathophysiological
pathways responsible for causing the progression of HF. However, therapies developed with this objective have sometimes failed to
show benefit. The pathophysiological construct of HF with numerous aetiologies suggests that interventions with broad mechan-
isms of action which simultaneously target more than one pathway maybe more effective in improving the outcomes of patients
with HF. Indeed, current therapeutics with clinical benefits in HF have targeted a wider range of intermediate phenotypes. Despite
extensive scientific breakthroughs in HF research recently, questions persist regarding the ideal therapeutic targets which may help
achieve maximum benefit. In this review, we evaluate the mechanism of action of current therapeutic strategies, the pathophysio-
logical pathways they target and highlight remaining knowledge gaps regarding the mode of action of these interventions.
-Keywords
----------------------------------------------------------------------------------------------------------------------------------------------------------------------
HF • HFrEF • pathophysiology • interventions

The neutral or negative results of trials pertaining to interventions dir-


1. Introduction ectly targeting improvement in a cardiac performance led to re-evaluation
Heart failure (HF) is a progressive and complex clinical syndrome charac- of the pathophysiological mechanism responsible for HF. Over time, it was
terized by impaired health-related quality of life, recurrent hospitalizations, hypothesized that a complex neuroendocrine compensatory response
and high mortality.1 HF has a multifactorial pathophysiological construct maybe responsible as the primary pathophysiological construct with differ-
which is responsible for its heterogeneous clinical presentation and ent mechanisms implicated in the two main clinical phenotypes of HF,
course.2 Over the last few decades, numerous attempts have been made namely HF with reduced ejection fraction (HFrEF) and HF with preserved
to develop targeted therapies that may attenuate the pathophysiological ejection fraction (HFpEF).12,13 In HF, long-term reduction of cardiac output
pathways thought to be responsible for causing the progression of HF.3 causes deleterious effects on multiple organ systems, with chronic activa-
However, therapies developed with the objective of altering a single path- tion of the renin–angiotensin–aldosterone system (RAAS) causing further
way have at times yielded little to no clinical benefit in ameliorating adverse impairment of the structure and function of cardiac myocytes. The involve-
events in these patients.4,5 ment of multiple pathophysiologic processes and systems potentially high-
For decades, increasing contractility of the failing heart remained the lights the reason why previous interventions with a specific mechanism of
most prevalent view to improve cardiac performance.6 It was suggested action did not work. Indeed, drugs that seemed counter-intuitive, such as
beta blockers yielded favourable results in improving left ventricular ejec-
that improving cardiac contractility would translate to improvement in cardiac
tion fraction (LVEF), antagonizing cardiac remodelling, and improving sur-
output, thereby curtailing the risk of mortality in HF. However, all clinical trials
vival in patients with HF.14
of positive inotropic drugs failed to show any mortality benefit.4,5 Following
To this end, contemporary trials have shown that interventions often
this, cardiac glycosides also showed no substantial improvement on mortality. initially investigated for purposes other than HF have yielded positive
The neutral or adverse effects on mortality in clinical trials of inotropic agents results in HF.14–16 Current foundational therapy for HF includes angio-
led to an increased focus on vasodilators in reducing afterload as a means to tensin receptor-neprilysin inhibitors, beta blockers, mineralocorticoid
improve cardiac efficiency in HF.7 Similar to positive inotropes, initial clinical receptor antagonists (MRAs) and sodium–glucose co-transporter 2
trials such as Vasodilator Heart Failure Trial (V-HeFT I) showed that short- (SGLT-2) inhibitors.17 However, questions still remain regarding
term improvement in haemodynamics does not necessarily translate to the current therapeutic modalities and the exact pathophysiological
favourable long-term survival outcomes.8 After a series of trials, it was de- mechanisms these drugs alter to achieve clinical benefit in HF. In this re-
duced that in some cases these agents may be doing more harm than good view, we summarize the contemporary understanding of the mechanism(s) of
by increasing the risk of developing worsening HF and mortality.9–11 action of current therapeutic strategies, the pathophysiological pathways

* Corresponding author. Tel: +214 820 2687, E-mail: butlzih@gmail.com.


© The Author(s) 2022. Published by Oxford University Press on behalf of the European Society of Cardiology. All rights reserved. For permissions, please email: journals.permissions@oup.com.
2 M.S. Khan et al.

they target and highlight the knowledge gaps which persist regarding the dobutamine and increased risk of mortality could be inferred from this
mode of action of these drugs. Of note, this review utlises publicly available non-randomized post hoc analysis, alternate methods were suggested to
data, and no new data were generated or analysed in support of this increase cardiac contractility which worked downstream to the β1 recep-
research. tors.21 This involved a string of phosphodiesterase inhibitors (PDEs) such
as amrinone, milrinone, and enoximone which increased intracellular cal-
cium by increasing intracellular cAMP.22–24 Although these conferred a dif-
2. Mechanisms of contemporary ferential mechanism of action to enhance cardiac contractility, they yielded
no clinical benefit in HF. Rather, milrinone therapy was associated with a
pharmacologic therapies in HF: 28% increase in all-cause mortality compared with placebo.20 Although
current evidence and knowledge the majority of positive inotropes tested to date have yielded neutral or

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


negative outcomes, digoxin is the only positive inotrope that has demon-
gaps strated a reduction in HF hospitalizations.
Traditionally, HF is divided into three distinct phenotypes based on the
LVEF. HFrEF is defined as LVEF ≤40%, while HFpEF is designated to pa-
tients showing signs and symptoms of HF with LVEF ≥50%. Patients
4. Digoxin
with LVEF between 41 and 49% are defined as having HF with mildly re- Digoxin is a positive inotrope that exerts its effects by blocking the Na+-K+
duced ejection fraction (HFmrEF). These phenotypes, particularly HFrEF ATPase pump, thereby progressively accumulating Na+ ions inside the sar-
and HFpEF have varying underlying aetiologies and pathophysiology. coplasma.25 This increases the intracellular Ca++ concentration, resulting in
Amongst patients with HFrEF, the overactivation of the neurohormonal a greater quantity of Ca++ release during excitation–contraction coupling
pathway is the most frequently implicated pathophysiological mechanism. which causes a more forceful contraction.26 In addition to positive inotrop-
In contrast, HFpEF has multiple overlapping pathophysiological constructs, ic effects, digoxin is also considered a neurohormonal modulator due to its
such as venous congestion leading to oxidative stress, myocardial stiffness favourable hemodynamic effects.27 Intravenous digoxin reduces cardiac
due to impaired phosphorylation of titin, and altered adiponectin and leptin norepinephrine, with a decline in vascular resistance in patients with
regulation causing epicardial adipose tissue deposition in the surrounding HF.27,28 Furthermore, it enhances the cardiac vagal tone and decreases
myocardium. Although considerable advancement in HF research has de- plasma renin activity.28,29 Although the exact autonomic mechanism of ac-
lineated the underlying mechanisms associated with these phenotypes, tion remains unknown, failure of prior inotropic therapies indicates that
pathophysiological construct of HFpEF remains less well-established. the neurohormonal attenuation of digoxin plausibly translates to the clin-
Consequently, the majority therapeutics have yielded mortality benefits ical benefit observed in reduction of HF hospitalization in large-scale clinical
in patients with HFrEF but not HFpEF, plausibly due to the varying under- trials of this intervention.
lying mechanistic pathways. As observed in the DIG trial (Digitalis Investigation Group), amongst
The activation of multiple pathophysiological pathways in HF requires 8500 chronic HF patients with LVEF ≤45%, digoxin was associated with
pharmacotherapy targeted at inhibiting these systems to achieve opti- a relative risk reduction of 13 and 28% in hospitalization rates for a cardio-
mal clinical benefit. Accordingly, a variety of pharmacological therapies vascular reason or for worsening HF compared to placebo, respectively.30
have been investigated in recent years to inform evidence-based man- However, no difference was observed in the reduction of all-cause mortal-
agement which improves clinical outcomes and health-related quality ity between the two intervention arms. On the contrary, although there
of life in these patients.18 Although a primary focus of HF research was a trend towards reduction in mortality due to worsening HF in digoxin
includes identifying interventions that may alter pathways that are impli- group, an increased risk of cardiovascular death due to other causes was
cated in the adverse progression of HF, to date, a number of interven- also noted in this group (P = 0.04).30 Given the increased risk of toxicity
tions that have yielded substantial clinical benefits, such as the novel observed in prior positive inotropic drug trials, it is plausible that the proar-
SGLT-2 inhibitors, are therapies which were initially devised to counter rhythmic effects of digoxin counterbalanced any benefit on mortality from
a disease process other than HF. This raises concerns that interventions worsening HF. Concerns over digoxin toxicity coupled with better alterna-
which target multiple pathophysiological phenotypes rather than a sin- tives have led to a decline in the utility of this medication in HF.31 Ongoing
gle pathophysiological pathway in HF patients, potentially aid in attenu- trials such as Digitoxin to Improve Outcomes in patients with Advance
ating adverse events in these patients. It therefore remains crucial to Chronic Heart Failure and DECISION (Digoxin Evaluation in Chronic
identify the key pathophysiological mechanisms and pathways which Heart Failure) are anticipated to provide further clarity regarding efficacy,
should be targeted to achieve maximal clinical benefit in these subsets safety and optimum dosage of digoxin in patients with HFrEF.32,33
of patients. Nevertheless, given that digoxin may be used as a viable alternative
amongst patients with severe HF who are unable to tolerate high doses
of other medications, or in patients with symptomatic HFrEF despite
3. Inotropic drugs guideline-directed medical therapy (GDMT), the variable mode of action
of digoxin may translate to a clinical benefit observed in HF.34
For decades, HF research was focused on understanding the decreased
contractility of the left ventricle and the pathways which may be targeted
to mitigate it. Increasing contractility of the myocardium became an em-
phasis to improve cardiac performance. Accordingly, much of the initial re-
5. Beta blockers
search was centred on developing positive inotropic drugs in an effort to Beta blockers are negative inotropes that are used for the management of
increase LVEF, which was postulated to improve long-term survival.4 hypertension and as a first-line therapy for the prevention of angina. Due to
Although increasing the concentration of intracellular calcium or sensitivity their antagonistic effect on β1 receptors in the myocardium, these agents
of sarcomere via stimulation of β1 receptors seemed intuitive, these agents slow the heart rate, thereby reducing the oxygen demand of the myocar-
were only successful in improving short-term surrogate endpoints dium which aids in reducing the duration and frequency of angina.
like hemodynamics, without improving long-term mortality.19,20 On the Consequently, these agents were initially contraindicated in HFrEF due
contrary, these drug trials were often terminated prematurely due to to the transient negative inotropic effect of beta blockade with subsequent
increased risk of adverse events, such as myocardial ischaemia or ventricu- risk of rapid decompensation in patients with acute HF. However, since the
lar arrythmia.5,19,20 early 1990s, multiple landmark clinical trials have demonstrated favourable
Dobutamine was first evaluated in the Flolan International Randomized prognostic impact of specific beta blockers in HFrEF.35–37
Survival Trial, where it showed an increased risk of mortality compared The pharmacologic benefit of beta blockade is exerted via its effects on
with placebo.21 Although only a plausible negative association between multiple pathways (Figure 1). Blocking the B-receptors limits the effect of
Mechanisms of heart failure therapies 3

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


Epi, NE Contractility
β-receptor
β-receptor blocker
blocker

β2 β1
G
Gss
ATP cAMP
Peripheral perfusion
Negative
Ca 2+
PKA inotropy
Ca2+

Ca2+ Activation of SNS

Ca2+
β-blockade

Considered counterintuitive however:

2 Slows HR

Energy expense Vaso- Oxidative Salt/water Cardiac


LVEF constriction stress retention hypertrophy

Figure 1 Mechanism of action of beta blockers.

increased catecholamine, inhibits renin-release, alters inotropy and chron- While metoprolol and bisoprolol are β1 receptor-selective, carvedilol
otropy, and exerts anti-arrhythmic and anti-ischaemic effects on the myo- exerts its effects by blocking β1, β2 and α1 receptors, which additionally
cardium.38,39 Cardio-selective beta blockers indicated for the management causes vasodilation. The 1996 U.S. Carvedilol Heart Failure Study was
of HFrEF, such as bisoprolol, carvedilol, and metoprolol succinate have all the first trial to show the mortality benefit of this intervention in HF
demonstrated mortality benefits.40 By blocking β1 receptors, these agents (Table 1). Amongst 1094 HF patients with EF <35%, carvedilol was accom-
cause direct antagonism of the cardiotoxic effects of catecholamines and panied by a 38% reduction in the combined risk of hospitalization or death
persistent activation of the sympathetic nervous system. Although initially (24.6% vs. 15.8%, P < 0.001).37 Likewise, in the COPERNICUS study, car-
activated as a compensatory mechanism, persistent activation of RAAS vedilol reduced the risk of mortality by 34% and HF hospitalization by
perpetuates a vicious cycle that leads to increased in cardiac preload, after- 40%.36 Clinical benefits in HF patients have also been seen with metoprolol
load, and induces ventricular remodelling. Blocking the B-receptors is succinate and bisoprolol.47 Metoprolol was evaluated in MERIT-HF trial in
therefore suggested to prevent ventricular remodelling.39 HF patients on baseline angiotensin-converting enzyme (ACE)-inhibitor
Although the use of beta blockers in HF may seem counter-intuitive, this and diuretic therapy.46 This trial was terminated early due to significantly
paradox may exist, in part, due to the inverse relationship between heart lower mortality risk amongst patients taking metoprolol compared with
rate and cardiac contractility in HF.3,41,42 In HF patients, as heart rate increases, placebo (P < 0.001), with fewer deaths due to worsening HF in the meto-
LVEF decreases due to the inability of myocardium to maintain the cardiac out- prolol group.46
put.41 By reducing heart rate, beta blockers increase LVEF and reduce energy Although the suggested benefits of beta blockers are widely stipulated to
expenditure, which is postulated to prevent adverse cardiac remodelling.42–45 be due to anti-sympathomimetic effects, evidence from the MOXCON
4 M.S. Khan et al.

Table 1 Landmark clinical trials of beta blocker therapy in heart failure

Trial Year Type of Adrenergic Patients, N Inclusion Primary outcome Mortality outcome
β-blockers receptor criteria
activity
...............................................................................................................................................................................................
CIBIS-I35 1994 Bisoprolol β1-selective 641 LVEF < 40%, Mortality No significant difference between the
NYHA class two groups
III-V

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


US Carvedilol37 1996 Carvedilol β1-β2-α 1094 LVEF ≤35% Mortality Significant reduction in all-cause
mortality (mortality event rate
3.2% carvedilol vs. 7.8% placebo)
MERIT HF46 1999 Metoprolol β1-selective 3991 LVEF < 40%, Mortality 34% relative risk reduction in all-cause
NYHA class mortality.
II-IV Significant decrease in sudden death
CIBIS II47 1999 Bisoprolol β1-selective 2647 LVEF < 35%, Mortality 34% relative risk reduction in all-cause
NYHA Class III– mortality
IV
COPERNICUS36 2001 Carvedilol β1-β2-α 2289 LVEF < 25% and Mortality, composite of 31% relative risk reduction in all-cause
NYHA Class III– death and mortality
IV hospitalization
SENIORS48 2005 Nebivolol β1-selective 2128 LVEF ≤35% Composite mortality, No significant difference between the
Age ≥70 years cardiovascular two groups
hospitalization

LVEF = left ventricular ejection fraction; NYHA = New York Heart Association.

trial suggests the principal mechanism may likely differ.49 Sustained-release of RAAS served as the key pathophysiological construct of HF, and pro-
preparation of moxonidine (moxonidine SR), a potent central sympathetic moted its progression by causing fluid retention, peripheral arterial vaso-
inhibitor failed to show any benefit in patients with HF.49 On the contrary, constriction, ventricular remodelling, interstitial fibrosis, and myocardial
an excess of early mortality in HF patients taking moxonidine SR compared hypertrophy. By blocking Angiotensin II and interfering with the maladap-
with placebo (5.5% vs. 3.4%; P = 0.012) led to early termination of the tive processes of RAAS in HF, ACEi/ARBs enhance natriuresis, prevent the
MOXCON trial.49 The excess short-term mortality observed in this trial release of aldosterone, lower arterial and venous pressures, decrease pre-
raises concerns regarding generalized sympathetic inhibition in HF, thereby load and afterload, and curtail cardiac remodelling.50,52
suggesting a lack of long-term benefit from inhibition of the vasoconstrictor ACEi and ARBs have shown to reduce the risk of mortality by 20–30% in
effects of SNS stimulation. This likely suggests that although SNS stimula- HF possibly via direct antagonism of RAAS.53–55 The Cooperative North
tion remains pivotal in the pathophysiology of HF, it is plausible that cardio- Scandinavian Enalapril Survival Study (CONSENSUS trial) which evaluated
selective beta blockers achieve optimal counteraction by targeting multiple enalapril in comparison with placebo demonstrated an overall mortality
pathophysiological pathways. risk reduction of 27% in the enalapril group (Table 2).54 This was followed
by the Studies of Left Ventricular Dysfunction trial which similarly observed
an absolute risk reduction of 5% in all-cause mortality in NYHA II
6. Angiotensin-converting enzyme and III HFrEF patients taking enalapril as compared to placebo. Similar
results were observed in landmark ARB trials.55 Candesartan in
inhibitors and angiotensin receptor Heart Failure-Assessment of Reduction in Mortality and Morbidity
(CHARM-alternative) was the first trial to demonstrate mortality and HF
blockers hospitlaization benefit in HFrEF patients treated with candesartan vs. pla-
Angiotensin II is a potent endogenous vasoconstrictor that increases per- cebo. Patients in the candesartan group observed a significantly decreased
ipheral resistance and sympathetic stimulation, thereby elevating blood risk of a composite cardiovascular death and HF hospitalization [0.70
pressure.50 To counter this, ACEis were developed to target the (0.60–0.81)] compared with patients in the placebo group.60 This was con-
angiotensin-converting enzyme (ACE), thereby inhibiting the conversion sistent with results observed in CHARM-added trial, where ARBs were
of Angiotensin I to its physiologically active form, Angiotensin II, while added to background therapy with ACEi and compared with a placebo.59
ARBs were used to block the binding of Angiotensin II to its receptor Although this showed a reduction in the risk of a composite cardiovascular
(Figure 2).51 Accordingly, due to their ability to cause peripheral vasodila- death and HF hospitalization, it was accompanied by increased rates of
tion and reduction of cardiac preload and afterload, ACEi and ARBs worsening renal function and hyperkalemia, thereby serving as a caution
were developed and marketed specifically as anti-hypertensive agents, fo- against concurrent administration of both ACEi and ARBs in HFrEF.
cusing mainly on patients with renin-dependent pathophysiology.52 Given the favourable mortality benefit observed in these trials, it is note-
Initially, ACEi was hypothesized to incur benefit in patients with HF due worthy to acknowledge that ACEi was utilized in addition to usual stan-
to vasodilation and subsequent reduction of blood pressure. However, gi- dards of care in HFrEF such as diuretics, vasodilators, digoxin, and beta
ven that various other more potent vasodilators with a greater reduction in blockers. This likely suggests that multiple mechanisms may be contributing
blood pressure had failed to show any mortality benefit in HF, it is import- to attenuating the progression of HF. As such, several mechanisms have
ant to acknowledge that any benefit observed via ACEi in HF was likely due been proposed in the ACE pathway, including the existence of alternative
to the multimodal mechanism of action of these drugs.50 Indeed, as re- pathways for the conversion of Angiotensin I to Angiotensin II which still
search progressed in HF, it was recognized that deleterious upregulation remain a topic of interest. Although ACEis interfere with RAAS, their effect
Mechanisms of heart failure therapies 5

ACE inhibitor
ARB

Blood pressure Kidney


K+ excretion

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


Renin Aldosterone
ste
Na / H2O retention

Liver Adrenal gland


A

ACE
ACE
CE

Angiotensinogen AT I ATT II
A

Vasoconstriction
ACE 2 NEP / PEP ACE 2 GFR
Urine output Blood pressure

ACE / NEP
Angiotensin (1-9) Angiotensin (1-7)

• Vasodilatation Vasodilatation
• Blood pressure
• Originally suggested
• Anti-prolife
f rative mechanisms for HF

Figure 2 Mechanism of action of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers.

is not directly due to levels of renin in the blood. It is plausible that the ob- (ANP), brain NP (BNP) and c-type NP are secreted from the cardiac myo-
served effects may be due to the upregulation ACE2/Angiotensin 1–7 path- cytes in response to mechanical stress on the heart due to excessive vol-
way of RAAS regulation, which remains less well-established.63 It is ume.66 These NPs alleviate the maladaptive processes triggered due to HF
suggested that ACE2, an endogenous ACE inhibitor converts Ang II into by binding to NP receptor type A and activating the cyclic guanosine mono-
Ang-(1–7), which acts at the Mas receptor to promote vasodilation and so- phosphate (cGMP) cascade, thereby promoting natriuresis, vasodilation,
dium and water excretion, inhibit neurohormonal activation and increase and inhibiting sympathetic outflow which prevents adverse cardiac remod-
nitric oxide production.63–65 These overlapping systemic underpinnings elling.67,68 In an effort to restore fluid homeostasis, promote vasodilation
suggest that targeting multiple pathophysiological mechanisms is likely re- and improve cardiac output, it seemed intuitive to supplement NPs in these
sponsible for the favourable pharmacodynamic profile of ACEi and ARBs. patients. However, clinical trials investigating BNP supplements in HF pa-
tients failed to show any mortality benefit over placebo, thereby shifting
focus to neprilysin inhibitors as a viable alternative.69
7. Angiotensin receptor-neprilysin Neprilysin is responsible for cleaving NP, as well as other vasodilating
inhibitor mediators such as substance P and bradykinin.66,70 Accordingly, inhibition
of neprilysin via neprilysin inhibitors increases endogenous NPs, thereby
To counter the maladaptive neurohormonal changes triggered by the ac- promoting vasodilation and natriuresis.71 However, in addition to increas-
tivation of RAAS in HF, natriuretic peptides (NP) such as atrial NP ing circulating NPs, neprilysin inhibitors also elevate the concentration of
6 M.S. Khan et al.

Table 2 Landmark clinical trials of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers therapy in
heart failure

Trial Year Type of Control Patients, N Follow-up Endpoint Outcome


intervention group (month)
...............................................................................................................................................................................................
ACE inhibitors
CONSENSUS Trial 1987 Enalapril Placebo 253 6 All-cause Mortality 27% reduction
Study Group54

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


SOLVD56 1991 Enalapril Placebo 2569 41.4 Mortality 16% reduction
PEP-CHF57 2006 Perindopril Placebo 850 26.2 Composite all-cause No significant difference
mortality or HHF
HHF
ARBs
Val-HeFT53 2001 Valsartan Placebo 5010 23 All-cause Mortality No significant difference
All-cause Mortality, HF 13% reduction
hospitalization, cardiac
arrest
OPTIMAAL58 2002 Losartan Captopril 5477 32.4 All-cause Mortality No significant difference
CHARM-Added59 2003 Candesartan Placebo 2548 41 All-cause Mortality No significant difference
CV death, HF hospitalization 15% reduction
CHARM-Alternative60 2003 Candesartan Placebo 2028 33.7 CV death, HF hospitalization 30% reduction
CHARM-Preserved61 2003 Candesartan Placebo 699 36.6 CV death or HHF No significant difference
I-PRESERVE62 2008 Irbesartan Placebo 4128 49.5 All-cause mortality or CV No significant difference
hospitalization

ACE = angiotensin-converting enzyme; ARB = angiotensin receptor blocker; HF = heart failure; HHF = hospitalization for heart failure; CV = cardiovascular.

two circulating vasopressors, namely Angiotensin II and Endothelin I.66,72 dosage and changes in BNP.77 This may be attributed to the difference
Consequently, due to opposing effects of promoting vasodilation and vaso- in patient characteristics and plausibly some BNP increase due to neprilysin
constriction simultaneously, this counterbalances any prognostic benefit inhibition being counteracted by a reduction in its production due to off-
neprilysin inhibitors exert when given in isolation. As a result, concomitant loading. In contrast, studies have shown an increase in ANP concentrations
inhibition of neprilysin and RAAS was considered pivotal for the manage- following treatment with ARNi, thereby suggesting that measuring an
ment of hypertension and HF. increase in ANP concentrations following initiation of ARNi may aid
ARNi resulted from the combination of a neprilysin inhibitor and an anti- in identifying a response to sacubitril/valsartan therapy.78 However,
hypertensive drug, sacubitril, and valsartan. While sacubitril inhibits nepri- although neprilysin inhibition contributes as the key mechanism behind
lysin activity, valsartan effectively counteracts activation of the RAAS, a rise in ANP concentration, recent studies suggest that multiple mech-
thereby promoting the dual effect of this therapeutic intervention.73 anistic pathways, such as indirect regulation of microRNAs following
Modulating two essential HF pathways potentially augments the therapeut- ARNi therapy potentially lead to an increase in ANP concentrations
ic benefits observed with this intervention (Figure 3). The Prospective as well.79 Given that neprilysin is responsible for cleaving multiple pep-
Comparison of ARNi with ACEi to Determine Impact on Global tides, the net effect of neprilysin inhibition is more complex. ARNi has
Mortality and Morbidity in Heart Failure (PARADIGM-HF) trial found been observed to increase plasma concentration levels of adrenome-
that ARNi reduced cardiovascular mortality [13.3% vs. 16.5%; HR: 0.80 dullin (ADM) precursor fragment MR-proADM, which is a regulatory
(95% CI: 0.71–0.89)] and hospitalization for HF [12.8% vs. 15.6%; HR: peptide with potent vasodilating, anti-fibrotic, anti-inflammatory and
0.79 (95% CI: 0.71–0.89)] when compared with enalapril in patients with natriuretic properties, in patients with HFrEF.80 Further, as observed
chronic HFrEF (Table 3).74 These findings extend to patients with acute de- in PARADIGM-HF, ARNi also exerts beneficial effects on glycemic
compensated HFrEF. As observed in the PIONEER-HF (Comparison of control by increasing circulating GLP-1 levels.81 A sub-study of this trial
Sacubitril-Valsartan vs. Enalapril on Effect on NT-proBNP in Patients also revealed the anti-fibrotic activity of ARNi, with a significant
Stabilized from an Acute Heart Failure Episode) trial, over a follow-up per- reduction observed in aldosterone, soluble tumorigenicity suppressor,
iod of 8 weeks, sacubitril/valsartan resulted in a greater reduction in matrix metallopeptidase 9 and procollagen type 1 amino-terminal
NT-proBNP and HF hospitalization [8.0% vs. 13.8%; HR: 0.56 (95% CI: propeptide after 8 months of treatment with ARNi compared with en-
0.37–0.84)] when compared with enalapril.75 alapril.82 This, therefore, provides further insights towards various no-
Although ARNi is believed to increase NPs in circulation, conflicting evi- vel mechanism of actions of ARNi which potentially contribute
dence exists regarding BNP levels following the initiation of ARNis.74,77 In towards its therapeutic effect, with questions still persisting regarding
PARADIGM-HF, 19% of study participants demonstrated an increase in the magnitude of therapeutic effect these NPs confer in patients with
BNP. This conflicts with recent pooled analysis of Effects of Sacubitril/ HF.77
Valsartan vs. Enalapril on Aortic Stiffness in Patients with Mild to Further, although neprilysin inhibition enhances the activity of cGMP,
Moderate HF with Reduced Ejection Fraction and PROVE-HF (Effects of which is suggested to reduce myocardial stiffness, no clinical benefit of
Sacubitril/Valsartan Therapy on Biomarkers, Myocardial Remodeling and ARNi in improving survival was observed in patients with HFpEF in the
Outcomes), where initiation of ARNi led to no significant overall change PARAGON-HF trial (Prospective Comparison of ARNi with ARB Global
in BNP levels.77 In addition, no association was observed between ARNi Outcomes in Heart Failure with Preserved Ejection Fraction). However,
Mechanisms of heart failure therapies 7

Adaptive response
Stress on the Maladaptive response
myocardium

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


Secretion of NPs RAAS

Neprilysin
Breakdown NPs

ANP BNP CNP Sacubitril / Ang II


Valsartan

Vasodilation Vasoconstriction

Natriuresis Na / H2O retension


Blood pressure Blood pressure
Sympathetic tone Sympathetic tone

Hypertrophy Hypertrophy

Figure 3 Mechanism of action of angiotensin receptor-neprilysin inhibitor.

a nominal reduction in HF hospitalization supports the use of this thera- locations.83,84 Originally developed as a potent diuretic, the limited scien-
peutic in this subset of patients.76 ARNi, therefore, sustains itself as a tific knowledge regarding aldosterone and its key role in human physiology
GDMT in HF, with likely therapeutic advantages observed due to its con- in 1950s confined the role of MRAs as K-sparing diuretics.85 Accordingly,
comitant inhibition of two key HF pathways. these agents were initially used as anti-hypertensives and diuretics for mo-
bilization of patients with advanced ascites or cirrhosis due to their ability
of blocking the aldosterone receptor, which is an endogenous steroid hor-
8. Mineralocorticoid receptor mone responsible for increased sodium retention and potassium excretion
in the renal distal tubule.86 As described by Funder et al., during this time
antagonists the interaction between aldosterone and mineralocorticoid receptors
Non-selective (e.g. spironolactone) and selective (e.g. eplerenone) MRAs (MR) was summarised as aldosterone being derived mainly by angiotensin,
target the aldosterone receptor in multiple epithelial and non-epithelial aldosterone being the only physiologic ligand of MR, and elevation of blood
8 M.S. Khan et al.

Table 3 Landmark clinical trials of angiotensin receptor-neprilysin inhibitor therapy in heart failure

Trial Year Type of HF Patients, N Follow-up (months) Endpoint Outcome


...............................................................................................................................................................................................
PARADIGM-HF74 2014 HFrEF 8442 27 All-cause mortality; 0.84 (0.76-0.93)
CV death, HF hospitalization; 0.80 (0.73-0.87)
HF hospitalization 0.81 (0.71–0.89)
PIONEER-HF75 2019 HFrEF with acute decompensated HF 881 2 HF Hospitalization 0.56 (0.37–0.84)
PARAGON-HF76 2019 HFpEF 4822 35 All-cause mortality; 0.97 (0.84–1.13)

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


CV death, HF hospitalization; 0.87 (0.75–1.01)
HF hospitalization 0.85 (0.72–1.00)

HF = heart failure; HFrEF = heart failure with reduced ejection fraction; HFpEF = heart failure with preserved ejection fraction; CV = cardiovascular.

Table 4 Landmark clinical trials of mineralocorticoid receptor antagonists in heart failure

Trial Year Type of intervention Patients, N Follow-up (months) Endpoint Outcome


...............................................................................................................................................................................................
RALES90 1999 Spironolactone 1663 24 All-cause mortality; HF hospitalization 0.70 (0.60–0.82);
0.65 (0.54–0.77)
EMPHASIS-HF91 2011 Eplerenone 2737 21 All-cause mortality; 0.76 (0.61–0.94);
CV death, hospitalization 0.63 (0.54–0.74)
TOPCAT92 2014 Spironolactone 3445 39.6 All-cause mortality; HF hospitalization 0.91 (0.77–1.08);
0.83 (0.69–0.99)

HF = heart failure; CV = cardiovascular.

pressure mainly resulting via salt and water retention.85,87 After decades of placebo [HR 0.89; (95% CI: 0.77–1.04)], a significantly lower incidence
research, however, these concepts were later refuted by denoting that of HF hospitalization was observed in the spironolactone group [HR
angiotensin is not the key deriver of aldosterone secretion, aldosterone 0.83; (95% CI: 0.69–0.99)].92 Although these findings raise further ques-
constitutes one of the physiologic ligands of MR and that increase in blood tions regarding the underlying mechanisms which enable the efficacy of
pressure in the setting of aldosterone secretion mainly occurs due to its spironolactone across different LVEFs, various other pathways of aldos-
effects on the vasculature and central nervous system.87,88 As numerous terone activity remain of significant interest. The most noteworthy
studies indicated multiple pathophysiological roles of this hormone, the of these is the role of aldosterone on ACE2/Ang-(1–7)/Mas-R axis
mechanism of action of MRAs expanded from epithelial to non-epithelial pathway, which contributes to the pathogenesis of adverse cardiac
locations. and vascular remodelling.93 Although conclusive evidence remains to
In addition to its initial mechanism of sodium and water retention, aldos- be observed, some studies have shown Ang-(1–7) as a negative modu-
terone is now implicated in promoting pro-inflammatory immune cell phe- lator of aldosterone secretion.94 Therefore, exploring how MRAs may
notypes which potentially result in oxidative stress, cardiovascular target this pathway and the likely clinical benefits it may exert remains
inflammation, endothelial dysfunction, and cardiac fibrosis.89 Treatment an area of interest.
with MRAs can therefore attenuate the aldosterone-mediated
pro-inflammatory state, which perpetuates an increase in collagen synthe-
sis and ventricular remodelling.86 Due to their ability to curtail end-organ
damage, coupled with the natriuretic effects observed, MRAs were given
9. SGLT2 inhibitors
in conjunction with ACEi/ARBs and beta blockers in HFrEF patients to SGLT2 inhibitors were initially developed as antihyperglycemic agents for
amplify the effects of RAAS blockade. Although the clinical relevance of po- the management of Type 2 diabetes mellitus (T2DM). These agents
tential cardiac remodelling after MRAs has not been conclusively evaluated work by blocking the SGLT2 protein located in the proximal convoluted
in humans, accumulated evidence from prior trials demonstrates the mor- tubule of the nephron, which is responsible for reabsorbing up to 90%
tality and morbidity benefits of these agents across the spectrum of HF. of filtered glucose. SGLT2 inhibition, therefore, results in glycosuria with-
The evidence for MRA use was first evaluated in the Randomized out interfering with endogenous insulin.95 Early cardiovascular outcome
Aldactone Evaluation Study (RALES) trial, which was terminated early trials of SGLT2 inhibitors demonstrated a 30–35% lower risk of HF hospi-
due to early mortality benefit of spironolactone amongst 1663 HF patients talization amongst T2DM patients who were at high risk of cardiovascular
with LVEF ≤35% and NYHA Class III or IV symptoms.90 In RALES, the add- disease.96–99 Failure of other, more potent antihyperglycemic therapeutics
ition of spironolactone to standard HF therapy significantly reduced all- to reduce the cardiovascular risk profile of T2DM patients, therefore, sug-
cause mortality (11% absolute reduction), risk of sudden death (RR: gested that SGLT2 inhibitors have a cardioprotective effect independent of
0.71, 95% CI: 0.54–0.95, P = 0.02), and death due to HF (RR: 0.64, 95% their glucose-lowering capability.95
CI: 0.51–0.8, P < 0.001) in the spironolactone group compared with pla- Following cardiovascular risk reduction in T2DM trials, questions arose
cebo (Table 4).90 regarding the efficacy of SGLT2 inhibitors amongst patients with prevalent
More recently, MRAs have also yielded modest results in patients with HF who did not have T2DM. Accumulating evidence from the DAPA-HF
HFpEF. In the TOPCAT (Aldosterone Antagonist Therapy for Heart (Dapagliflozin and Prevention of Adverse outcomes in Heart Failure),
Failure with Preserved Ejection Fraction) trial, although spironolactone EMPEROR-Reduced (Empagliflozin Outcome Trial in Patients with
did not reduce primary outcome consisting of time to cardiac death, resus- Chronic Heart Failure and a Reduced Ejection Fraction) and
citated cardiac arrest, or hospitalization for HF as compared with EMPEROR-Preserved (Empagliflozin in Heart Failure with Preserved
Mechanisms of heart failure therapies 9

Ejection Fraction) trials have demonstrated that the clinical benefits of without any effect on myocardial contractility, blood pressure, intracardiac
SGLT2 inhibitors extend across the HF spectrum, irrespective of diabetes conduction or ventricular repolarization time.115,116 Ivabradine is a heart
status (Table 5).15,16,100 In the DAPA-HF trial, SGLT2 inhibitors reduced rate lowering agent indicated in patients who are in sinus rhythm with a resting
the primary endpoint of worsening heart failure or cardiovascular death heart rate of 70 beats per minute, which selectively inhibits the cardiac pace-
[HR: 0.74 (95% CI: 0.65–0.85); P < 0.001] and cardiovascular mortality maker current channel I(f), which is responsible for regulating heart rate by
[HR: 0.82 (95% CI: 0.69–0.98)] amongst 4744 HFrEF patients controlling the diastolic depolarization in the sinoatrial (SA) node.115,117
compared with placebo.15 Similarly, amongst 3730 HFrEF patients in Blocking of this channel leads to inhibition of sodium–potassium inward cur-
EMPEROR-Reduced trial, SGLT2 inhibitors decreased composite of car- rent, thereby prolonging diastolic depolarization. The slow rate of SA node fir-
diovascular death or hospitalization for HF, [HR 0.75 (95% CI: 0.65–0.86; ing, therefore, leads to a reduction in heart rate without any effect on other
P < 0.001)], with a significant reduction in total hospitalizations for HF in cardiac parameters.115

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


the empagliflozin group compared with placebo [HR 0.70 (95% CI: 0.58– After ivabradine was reported efficacious, safe and tolerable in clinical
0.85) P < 0.001].16 This clinical benefit extends to patients with HFpEF. trials of patients with stable angina pectoris, it was investigated in patients
As observed in the EMPEROR-Preserved trial, SGLT2 inhibitors proved with systolic dysfunction due to increased evidence of a reduction in heart
to be the first intervention to confer a clinical benefit in patients with rate translating to paradoxical positive inotropism in patients with
HFpEF.100 In this trial of 5988 patients with HFpEF, SGLT2 inhibition HF.116,118,119 In SHIFT trial (Systolic Heart Failure Treatment with the If
with empagliflozin led to a 21% reduction in relative risk in the composite Inhibitor Ivabradine Trial), ivabradine reduced HF hospitalization (HR
of cardiovascular death or hospitalization for HF, which was mainly attrib- 0.74 [95% CI, 0.66–0.83]; P < 0.001) and HF mortality (HR 0.74 [95% CI,
uted to a 29% lower risk of hospitalization for HF with empagliflozin.100 0.58–0.94]; P = 0.01) compared with placebo.120 However, no clinical
Despite the benefits of SGLT2 inhibitors for HF across spectrum of EF, benefit was observed in the reduction of cardiovascular or all-cause mor-
the exact mechanism by which this class of therapy confers a survival ad- tality.120 Despite its direct effect on SA node, beta blockers have demon-
vantage is not well-established. It is suggested that SGLT2 inhibitors simul- strated a greater average reduction in heart rate of up to 12 beats per
taneously target multiple pathways and produce favourable effects on minute (bpm).121 This coupled with the mortality benefits observed in
multiple organ systems thought to be involved in HF (Figure 4).95,102,103 beta blockers led to the recommendation of initiating ivabradine therapy
A major benefit of SGLT2 inhibitors is suggested to be related to the kid- only after maximum optimal dosing of beta blocker has been achieved in
ney, where they inhibit sodium reabsorption in the proximal tubule, there- HFrEF patients with a resting heart rate of ≥70 bpm. This substantiates
by promoting osmotic diuresis which is also speculated as the key that although modulating heart rate serves as one of the pathways which
mechanism of the anti-hypertensive effect observed with this interven- may attenuate adverse HF events, the superiority of beta blockers poten-
tion.104 By lowering blood pressure and antagonizing fluid retention, these tially exists in their ability to modify multiple mechanistic pathways.
agents are suggested to decrease cardiac preload and afterload, which sub-
sequently improves ventricular-arterial coupling.103 It has also been sug-
gested that SGLT2 inhibitors attenuate oxidative stress by decreasing
the underlying systemic inflammation.105 Although the exact mechanism
11. Hydralazine–isosorbide dinitrate
of this in HF remains unknown, it is postulated that these agents potentially Both hydralazine and isosorbide dinitrate (ISDN) have vasodilatory prop-
exert benefit in patients with HFpEF due to their ability to decrease inflam- erties which help in the reduction of preload and afterload. Hydralazine
matory response due to increase macrophagic activity, possible reduction is an arterial vasodilator that reduces peripheral vascular resistance by dir-
of sympathetic nerve activity and inhibition of norepinephrine turn over in ectly relaxing the vascular smooth muscle.122 In isolation, it has been long-
brown adipose tissue and improvement in cardiac energetics by increasing standing treatment of hypertensive crisis and hypertensive disorders in
circulating ketone levels which are utilized by the failing myocardium as an pregnancy due to its potent vasodilation properties.123,124 Although the
energy source, thereby improving cardiac efficiency. However, this evi- exact mechanism of inducing vasodilation remains unknown, it is suggested
dence is largely accumulated by animal data and is yet to be confirmed in that this may be attributed to altering Ca2+ balance in vascular smooth
patients with HF.106–109 muscles or by modulating the effect of compounds released from sympa-
Amongst patients with HF, SGLT2 inhibitors have demonstrated reverse thetic nerve endings.122 In addition, hydralazine is suggested to have anti-
cardiac remodelling in multiple randomized controlled trials and exploratory oxidant properties that help prevent NO degradation.
post hoc analysis.110–112 As observed in EMPA-TROPISM (Empagliflozin in ISDN is a nitric oxide (NO) donor which also aids in the relaxation of
Nondiabetic Patients with Heart Failure and Reduced Ejection Fraction), em- vascular smooth muscle.125 ISDM activates the enzyme soluble guanylyl cy-
pagliflozin in HFrEF patients significantly improved left ventricular volumes, clase in the vascular smooth muscle which subsequently increases cGMP,
mass, systolic function and functional capacity.111 Although only established thereby causing vasodilation and decreases vascular resistance.125 At the
in patients with T2DM, SGLT2 inhibitors augment iron utilization and in- therapeutic level, ISDN is used to treat angina pectoris. The combination
crease erythropoiesis, which is suggested as a marker of reduced metabolic of hydralazine and ISDN maintains a prolonged state of vasodilation and
stress.113 Further, these agents also improve vascular function by attenuating augments the bioavailability of NO, thereby maintaining the vascular
endothelial cell activation via direct vasorelaxation, which results in favour- tone and cellular redox balance.126
able haemodynamics seen with this intervention. The haemodynamic effects of hydralazine and ISDN were first observed
The multimodal mechanism of SGLT2 inhibitors, which simultaneously in the V-HeFT trial, where a 34% risk reduction in mortality was observed
targets multiple pathways further substantiates that therapies with diffuse among patients treated with both hydralazine and ISDN compared with
mechanisms of action confer the highest clinical benefit in curtailing adverse placebo or prazosin.8 The most pronounced effect of this intervention
events of this syndrome. Although SGLT2 inhibitors are now being widely was observed among 1050 Black patients with HFrEF and NYHA Class
investigated in multiple disease processes, it is plausible that many mechan- III–IV symptoms in the A-HeFT trial (The African-American Heart
isms through which they act still remain unknown. Failure Trial).127 This trial was terminated early due to significantly im-
proved survival in hydralazine-ISDN group. At the time of cessation, a high-
er rate of all-cause mortality was observed in the control group (10.2%)
10. Ivabradine compared with the hydralazine-ISDN group (6.2%), with similarly low rates
of HF hospitalization in patients taking hydralazine-ISDN (16.4% vs. 24.4%
Epidemiologic evidence demonstrates a strong correlation between increased in control participants). Survival in patients on hydralazine-ISDN therapy
heart rate and cardiovascular mortality.114 Elevated heart rate increases myo- was therefore increased by 43% (P = 0.02).127 These racial differences
cardial oxygen demand due to shortened length of each cardiac cycle, which are thought to be due to decreased bioavailability of NO in Black patients
reduces diastolic perfusion time.115 Accordingly, ivabradine was first investi- compared with other ethnicities.128 Resultant vasoconstriction in the set-
gated as an antianginal therapeutic due to its ability to reduce heart rate ting of an already failing heart can further contribute to increased cardiac
10 M.S. Khan et al.

Table 5 Landmark clinical trials of sodium–glucose co-transporter 2 inhibitors in heart failure

Trial Year Type of HF Patients, N Follow-up Endpoint Outcome


(months)
...............................................................................................................................................................................................
DAPA-HF15 2019 HFrEF 4744 18.2 All-cause mortality 0.83 (0.71–0.97)
CV death, worsening HF event 0.74 (0.65–0.85)
Worsening HF event 0.70 (0.59–0.83)
EMPEROR-Reduced16 2020 HFrEF 3730 16 CV death, HF hospitalization 0.75 (0.65–0.86)

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


HF hospitalization 0.69 (0.59–0.81)
All-cause mortality 0.92 (0.77–1.10)
SOLOIST-WHF101 2021 HFrEF, HFpEF 1222 9 CV death, HF hospitalization 0.67 (0.52–0.85)
EMPEROR-Preserved100 2021 HFpEF 5988 26.2 CV death, HF hospitalization 0.79 (0.69–0.90)
HF hospitalization 0.71 (0.60–0.83)
All-cause mortality 1.00 (0.87–1.15)
DELIVER 2022 HFmrEF, HFpEF 6263 29.1 HF hospitalization, urgent HF visit 0.79 (0.69–0.91)
HF hospitalization 0.77 (0.67–0.89)
CV death 0.88 (0.74–1.05)

HF = heart failure; HFrEF = heart failure with reduced ejection fraction; HFmrEF = heart failure with mid-range ejection fraction; HFpEF = heart failure with preserved ejection fraction; CV =
cardiovascular.

preload. Given that the neurohormonal system and NO characterize as attenuating cardiac remodelling or improving health-related quality of life
two distinct pathways, conventional HF therapeutics only alter the neuro- in patients with HFpEF.140,141
hormonal overdrive without any alteration of the bioavailability of NO. In The sGC stimulator vericiguat targets a distinct mechanism and repre-
this instance, hydralazine and ISDN is often considered an important ad- sents a novel therapeutic option for patients with HF. Importantly, verici-
junct with optimal HF therapy in Black patients with severe HF. guat targets a pathway (NO-sGC-cGMP) independent of ACEi/ARBs
(RAAS) and beta blockers (sympathetic nervous stimulation), thereby
complementing the currect pharmacotherapy of HFrEF. The clinical bene-
fits of combining vericiguat with other HF drugs to target multiple patho-
12. Soluble guanylate cyclase physiological pathways remains unknown. Additionally, the structural
stimulators optimization of vericiguat, its in-depth interaction with sGC and mechan-
isms by which cGMP regulates downstream pathways to improve HF out-
The NO-soluble guanylate cyclase (sGC)-cGMP pathways have been of comes still requires further research.142
particular interest in the pathophysiology of HF due to the pivotal role
of cGMP in mediating vasodilation and cardiac relaxation.129,130 In HF, de-
ficiency of cGMP due to less bioavailability of NO or sGC perpetuates oxi-
dative stress, which stimulates autophagy, apoptosis, cardiac fibrosis, and
13. Omecamtiv mecarbil
adverse cardiac remodelling.131 To this end, several drugs aimed to modu- Omecamtiv mecarbil is a first-in-class cardiac myosin activator, which is a
late the NO-cGMP pathway via increasing the availability of NO by using novel class of positive inotropes. These agents improve myocardial func-
nitrates, or reducing degradation of cGMP via PDEs have been evaluated tion by selectively binding to cardiac myosin. This increases the number
in HF.132,133 However, the low half-life and high-first pass metabolism of of myosin heads that can bind to the actin filament and initiate a power
NO, and minimal inhibition of PDE in the instance of NO yielded limited stroke at the start of systole without increasing intracellular calcium levels,
clinical applicability of these agents in HF.132,134,135 As a result, sGC ago- thereby augmenting cardiac contractility.143 As a selective cardiac myosin
nists were considered the next best therapeutic target to directly stimulate activator, omecamtiv mecarbil has no effect on the magnitude of intracel-
cGMP production. lular calcium in cardiomyocytes, which were the pivotal drivers of in-
Riociguat was developed as the first sGC stimulator. Although devel- creased incidence of ventricular arrhythmias and mortality in prior
oped for patients with pulmonary hypertension, riociguat demonstrated inotropes. Despite this, omecamtiv mecarbil has yielded modest benefits.
favourable safety profile, improvement in quality of life, and cardiac index In GALACTIC-HF trial (Cardiac Myosin Activation with Omecamtiv
in patients with HFrEF.136 However, its short half-life presented as a limi- Mecarbil in Systolic Heart Failure), HFrEF patients in omecamtiv mecarbil
tation for therapeutic use in HF patients.137 Accordingly, structural opti- group had a relative risk reduction of 8% in a composite of HF events
mization of riociguat was done to develop a therapy tailored for HF. and cardiovascular mortality as compared with placebo.144 However,
This led to the development of vericiguat, which works in synergy with this novel intervention conferred no mortality benefit. It is noteworthy
NO and produces cGMP even under low NO conditions.138 This restores to highlight that although no mortality benefit was observed in the overall
the NO-sGC-cGMP pathway, which is otherwise impaired in HF, thereby population, a possible interaction between the trial group and LVEF at
reducing oxidative stress, improving vascular tone and myocardial dysfunc- baseline was noted. These Phase III trial results are juxtaposed with data
tion, and attenuating adverse ventricular remodelling. The molecular ad- from the Phase II The Chronic Oral Study of Myosin Activation to
vantages of vericiguat also translate to clinical benefits. In the recent Increase Contractility in Heart Failure, where favourable improvement
Vericiguat Global Study in Subjects with Heart Failure with Reduced with omecamtiv mecarbil was noted in stroke volume, systolic ejection
Ejection Fraction, vericiguat reduced the composite primary outcome of time, left ventricular end-systolic and end-diastolic dimensions, and natri-
cardiovascular death or first HF hospitalization [35.5% vs. 38.5%; HR: uretic peptide levels.145 Treatment benefit was also observed in post
0.90 (95% CI: 0.82–0.98)], which was driven by a reduction in HF hospital- hoc analysis of GALACTIC-HF trial, where patients with severe HF (de-
ization [16.4% vs. 17.5%; HR 0.93 (95% CI: 0.81–1.06)] amongst 5050 pa- fined as NYHA symptom Class III–IV, LVEF ≤30%, HF hospitalization in
tients with worsening HFrEF.139 On the contrary, although current the past 6 months and evidence of severe functional impairment) on ome-
evidence remains limited, vericiguat failed to show any benefit in camtiv mecarbil therapy experienced clinically meaningful reduction in
Mechanisms of heart failure therapies 11

*PCT: Proximal convoluted tubule


**ATP: Adenosine tri-phosphate
*** GLUT: Glucose transporter
Urine PCT cell Blood
Na+
Na/K
ATPase Increased urinary Na+ and glucose excretion
SGLT2 K+

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


GLUT2
Increased
circulating
ketones Reduced
plasma
SGLT2 volume and
inhibition Increased improved
glucagon vs endothelial
insulin ratio function Diuresis
Reduced ATP
consumption in
PCT and
relative hypoxia
in renal cortex
Reduction Reduction
Lypolysis Improved in blood in
glycemic pressure and ventricular
Reversion of afterload preload
control
erythropoietin
Reverse
cardiac
remodelling

Increase in
haematocrit

Increased haematocrit Weight loss Improved ventricular


loading conditions
Demonstrated in patients
with T2DM Mechanism Mechanism
Modest increase observed demonstrated in animal demonstrated in patients
in patients with HF data with HF

Figure 4 Mechanism of action of sodium–glucose co-transporter 2 inhibitors. (Adapted and Modified from Joshi SS, Singh T, Newby DE, Singh J. Sodium–
glucose co-transporter 2 inhibitor therapy: mechanisms of action in heart failure. Heart BMJ Publishing Group Ltd and British Cardiovascular Society; 2021;
heartjnl-2020-318060).

composite of time to first HF event or cardiovascular death compared with optimal prognostic benefit in all patients. As previously mentioned, tar-
patients without severe HF.146 The conflicting results and modest favour- geted therapy is ideal for monogenic disease, where a single pathway
able benefits observed with this intervention substantiates that other neu- might be responsible for adverse reactions to the disease process. In
rohormonal mechanisms not directly targeted by omecamtiv mecarbil in a systemic clinical syndrome like HF, multiple compensatory pathways
this multifactorial disease may be at play and underscores the potential affect more than one organ system. Current therapeutics which have
role of this therapeutic among patients for whom current treatment op- yielded maximum benefits and reduced all-cause mortality in HF pa-
tions are limited, or patients already receiving maximally tolerated or target tients, such as SGLT-2 inhibitors, ARNi, beta blockers, and MRAs
doses of other therapies. have broad mechanisms of action which simultaneously target more
than one pathophysiological pathway. When taken in combination,
these drugs provide incremental benefits with a marked reduction in
14. Future perspectives mortality, recurrent hospitalization, worsening of HF, progression of
HF is a multifactorial syndrome with the integration of multiple systemic kidney disease and enhanced quality of life. Historically, HF is a constant-
pathways. The complexity of this disease process requires a multimodal ly evolving field which despite extensive research has yielded therapies
treatment with a combination of several drugs as the cornerstone for that have not shown benefit in large-scale randomized trials. Failure of
12 M.S. Khan et al.

therapies targeting mechanisms with sound concepts suggests that tar- MECKI Score Research Group. Metabolic exercise test data combined with cardiac and
geting a single mechanism of action may not yield optimal results in kidney indexes, the MECKI score: a multiparametric approach to heart failure prognosis.
Int J Cardiol 2013;167:2710–2718.
multifactorial disease process. Instead, drugs that target multiple path-
3. Ferrari R, Balla C, Fucili A. Heart failure: an historical perspective. Eur Heart J Suppl 2016;18:
ways across different organ systems, such as the heart, kidney, and G3–G10.
endocrine system may aid in attaining maximal clinical benefit in patients 4. Ahmad T, Miller PE, McCullough M, Desai NR, Riello R, Psotka M, Böhm M, Allen LA,
with HF. Teerlink JR, Rosano GMC, Lindenfeld J. Why has positive inotropy failed in chronic heart
Stakeholders should devise a framework that maximizes the potential of failure? Lessons from prior inotrope trials. Eur J Heart Fail 2019;21:1064–1078.
success of novel interventions. Understanding the mechanism of action of 5. Felker GM, O’Connor CM. Inotropic therapy for heart failure: an evidence-based ap-
an intervention in depth serves as the pivotal step. However, HF is a com- proach. Am Heart J 2001;142:393–401.
6. Gault JH, Ross J, Braunwald E. Contractile state of the left ventricle in man: instantaneous
plex disease with several aetiologies. By evaluating the efficacy of novel in-

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


tension-velocity-length relations in patients with and without disease of the left ventricular
terventions based on multiple intermediate phenotypes, conducting
myocardium. Circ Res 1968;22:451–463.
efficacy analyses across multiple different groups and doing a detailed 7. Cohn JN, Franciosa JA. Vasodilator therapy of cardiac failure: (first of two parts). N Engl J
evaluation of quality of life benefit these agents may enable better under- Med 1977; 297:27–31.
standing for future HF drug development. 8. Ghose JC, Chakraborty S, Mondal M, Bhandari B. Effect of vasodilator therapy on mortality
in chronic congestive heart failure. J Assoc Physicians India 1993;41:269–271.
9. Goldstein RE, Boccuzzi SJ, Cruess D, Nattel S, Friday K, Lenkei S, Nattel S, Rubison M.
15. Conclusion Diltiazem increases late-onset congestive heart failure in postinfarction patients with early
reduction in ejection fraction. Circulation 1991;83:52–60.
HF is a multifactorial syndrome with integrated overlapping multiple patho- 10. Franciosa JA, Jordan RA, Wilen MM, Leddy CL. Minoxidil in patients with chronic left heart
physiological pathways. This complexity requires a multimodal treatment failure: contrasting hemodynamic and clinical effects in a controlled trial. Circulation 1984;
with a combination of several drugs as the cornerstone for optimal benefit. 70:63–68.
11. Elkayam U, Amin J, Mehra A, Vasquez J, Weber L, Rahimtoola SH. A prospective, rando-
Very focused and targeted therapy is ideal for monogenic disease where a
mized, double-blind, crossover study to compare the efficacy and safety of chronic nifedi-
single pathway is disrupted and is responsible for the disease process. In a pine therapy with that of isosorbide dinitrate and their combination in the treatment of
systemic clinical syndrome such as HF, multiple pathways are usually in play chronic congestive heart failure. Circulation 1990;82:1954–1961.
affecting more than one organ system, including the myocardium, kidney, 12. Simmonds SJ, Cuijpers I, Heymans S, Jones EAV. Cellular and molecular differences be-
and endocrine. tween HFpEF and HFrEF: a step ahead in an improved pathological understanding. Cells
Current therapeutics which have yielded maximum benefits such as 2020; 9:242.
SGLT-2 inhibitors, ARNi, beta blockers, and MRAs have broad mechanisms 13. Anand IS, Ferrari R, Kalra GS, Wahi PL, Poole-Wilson PA, Harris PC. Edema of cardiac ori-
of action which simultaneously target more than one pathophysiological gin. Studies of body water and sodium, renal function, hemodynamic indexes, and plasma
hormones in untreated congestive cardiac failure. Circulation 1989;80:299–305.
pathway. When taken in combination, these drugs provide incremental
14. Brophy JM, Joseph L, Rouleau JL. Beta-blockers in congestive heart failure. A Bayesian
benefits with a marked reduction in mortality, recurrent hospitalization, meta-analysis. Ann Intern Med 2001;134:550–560.
worsening of HF, progression of end-organ damage, and enhanced quality 15. McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod MN, Martinez FA, Ponikowski
of life. Otherwise, failure of therapies targeting specific mechanisms with P, Sabatine MS, Anand IS, Bělohlávek J, Böhm M, Chiang C-E, Chopra VK, de Boer RA, Desai
sound concept have at large not yielded optimal results in multifactorial dis- AS, Diez M, Drozdz J, Dukát A, Ge J, Howlett JG, Katova T, Kitakaze M, Ljungman CEA,
ease process. Merkely B, Nicolau JC, O’Meara E, Petrie MC, Vinh PN, Schou M, Tereshchenko
S, Verma S, Held C, DeMets DL, Docherty KF, Jhund PS, Bengtsson O, Sjöstrand M,
Conflict of interest: J.B. reports personal consulting fees from Abbott, Langkilde AM; DAPA-HF Trial Committees and Investigators. Dapagliflozin in patients
Adrenomed, Amgen, Applied Therapeutics, Array, AstraZeneca, Bayer, with heart failure and reduced ejection fraction. N Engl J Med 2019;381:1995–2008.
Boehringer Ingelheim, CVRx, G3 Pharma, Impulse Dynamics, Innolife, 16. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, Januzzi J, Verma S, Tsutsui
Janssen, LivaNova, Luitpold, Medtronic, Merck, Novartis, Novo Nordisk, H, Brueckmann M, Jamal W, Kimura K, Schnee J, Zeller C, Cotton D, Bocchi E, Böhm M,
Choi D-J, Chopra V, Chuquiure E, Giannetti N, Janssens S, Zhang J, Gonzalez Juanatey JR,
Relypsa, Sequana Medical, and Vifor Pharma; and payment for lectures,
Kaul S, Brunner-La Rocca H-P, Merkely B, Nicholls SJ, Perrone S, Pina I, Ponikowski P, Sattar
presentations, speakers bureaus, manuscript writing or educational events N, Senni M, Seronde MF, Spinar J, Squire I, Taddei S, Wanner C, Zannad F; EMPEROR-
from AstraZeneca, BI-Lilly, Janssen, and Novartis. S.J.G. has received re- Reduced Trial Investigators. Cardiovascular and renal outcomes with empagliflozin in heart
search support from the American Heart Association, Amgen, failure. N Engl J Med 2020;383:1413–1424.
AstraZeneca, Bristol Myers Squibb, Cytokinetics, Merck, Novartis, Pfizer, 17. McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M, Burri H, Butler J,
and Sanofi; has served on advisory boards for Amgen, AstraZeneca, Celutkiene J, Chioncel O, Cleland JGF, Coats AJS, Crespo-Leiro MG, Farmakis D, Gilard M,
Bristol Myers Squibb, Cytokinetics, and Sanofi; and has served as a consult- Heymans S. 2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart
ant for Amgen, Bayer, Bristol Myers Squibb, Merck, Sanofi and Vifor. R.J.M. failure. Eur Heart J 2021; 42:3599–3726.
18. He Z, Yang L, Nie Y, Wang Y, Wang Y, Niu X, Bai M, Yao Y, Zhang Z. Effects of SGLT-2
has received research support and honoraria from Abbott, American
inhibitors on health-related quality of life and exercise capacity in heart failure patients with
Regent, Amgen, AstraZeneca, Bayer, Boehringer Ingelheim/Eli Lilly, reduced ejection fraction: A systematic review and meta-analysis. Int J Cardiol 2021;345:
Boston Scientific, Cytokinetics, Fast BioMedical, Gilead, Medtronic, 83–88.
Merck, Novartis, Roche, Sanofi, and Vifor. A.D.D. reports research funding 19. Massie B, Bourassa M, DiBianco R, Hess M, Konstam M, Likoff M, Packer M. Long-term oral
through his institution from the American Heart Association, National administration of amrinone for congestive heart failure: lack of efficacy in a multicenter
Heart, Lung, and Blood Institute, and Patient-Centered Outcomes controlled trial. Circulation 1985;71:963–971.
Research Institute. He has also received nonfinancial support from 20. Packer M, Carver JR, Rodeheffer RJ, Ivanhoe RJ, DiBianco R, Zeldis SM, Hendrix GH,
Bommer WJ, Elkayam U, Kukin ML, Mallis GI, Sollano JA, Shannon J, Tandon PK,
Abbott Laboratories for educational activities. The remaining authors
DeMets DL. Effect of oral milrinone on mortality in severe chronic heart failure. N Engl J
have no conflicts of interest to declare. Med 1991;325:1468–1475.
21. O’Connor CM, Gattis WA, Uretsky BF, Adams J, McNulty SE, Grossman SH, McKenna WJ,
Funding Zannad F, Swedberg K, Gheorghiade M, Califf RM. Continuous intravenous dobutamine is
associated with an increased risk of death in patients with advanced heart failure: insights
None declared.
from the flolan international randomized survival trial (FIRST). Am Heart J 1999;138:78–86.
22. Uretsky BF, Generalovich T, Reddy PS, Spangenberg RB, Follansbee WP. The acute hemo-
References dynamic effects of a new agent, MDL 17,043, in the treatment of congestive heart failure.
1. Savarese G, Lund LH. Global public health burden of heart failure. Card Fail Rev 2017;03:7. Circulation 1983;67:823–828.
2. Agostoni P, Corrà U, Cattadori G, Veglia F, La Gioia R, Scardovi AB, Emdin M, Metra M, 23. Maskin CS, Sinoway L, Chadwick B, Sonnenblick EH, Le Jemtel TH. Sustained hemodynamic
Sinagra G, Limongelli G, Raimondo R, Re F, Guazzi M, Belardinelli R, Parati G, Magrì D, and clinical effects of a new cardiotonic agent, WIN 47203, in patients with severe congest-
Fiorentini C, Mezzani A, Salvioni E, Scrutinio D, Ricci R, Bettari L, Di Lenarda A, ive heart failure. Circulation 1983;67:1065–1070.
Pastormerlo LE, Pacileo G, Vaninetti R, Apostolo A, Iorio A, Paolillo S, Palermo 24. Benotti JR, Grossman W, Braunwald E, Davolos DD, Alousi AA. Hemodynamic assessment
P, Contini M, Confalonieri M, Giannuzzi P, Passantino A, Cas LD, Piepoli MF, Passino C; of amrinone. N Engl J Med 1978;299:1373–1377.
Mechanisms of heart failure therapies 13

25. Sagawa T, Sagawa K, Kelly JE, Tsushima RG, Andrew Wasserstrom J. Activation of cardiac Toman J, Böhm M, Anker SD, Thompson SG, Poole-Wilson PA. FASTTRACK
ryanodine receptors by cardiac glycosides. Am J Physiol Hear Circ Physiol 2002;282: Randomized trial to determine the effect of nebivolol on mortality and cardiovascular hos-
H1118–H1126. pital admission in elderly patients with heart failure (SENIORS). Eur Heart J 2005;26:
26. Wasserstrom JA, Aistrup GL. Digitalis: new actions for an old drug. Am J Physiol Hear Circ 215–225.
Physiol 2005;289:H1781–H1793. 49. Cohn JN, Pfeffer MA, Rouleau J, Sharpe N, Swedberg K, Straub M, Wiltse C, Wright TJ.
27. Gheorghiade M, Hall VB, Jacobsen G, Alam M, Rosman H, Goldstein S. Effects of increasing Adverse mortality effect of central sympathetic inhibition with sustained-release moxoni-
maintenance dose of digoxin on left ventricular function and neurohormones in patients dine in patients with heart failure (MOXCON). Eur J Heart Fail 2003;5:659–667.
with chronic heart failure treated with diuretics and angiotensin-converting enzyme inhibi- 50. Bell L, Madri JA. Influence of the angiotensin system on endothelial and smooth muscle cell
tors. Circulation 1995;92:1801–1807. migration. Am J Pathol 1990;137:7–12.
28. Slatton ML, Irani WN, Hall SA, Marcoux LG, Page RL, Grayburn PA, Eichhorn EJ. Does di- 51. Herman LL, Padala SA, Ahmed I, Bashir K. Angiotensin converting enzyme inhibitors (ACEI).
goxin provide additional hemodynamic and autonomic benefit at higher doses in patients StatPearls Publishing; 2021; https://www.ncbi.nlm.nih.gov/books/NBK431051/.

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


with mild to moderate heart failure and normal sinus rhythm? J Am Coll Cardiol 1997;29: 52. Borer JS. Angiotensin-converting enzyme inhibition: a landmark advance in treatment for
1206–1213. cardiovascular diseases. Eur Heart J Suppl 2007;9:E2–E9.
29. Gheorghiade M, Hall V, Lakier JB, Goldstein S. Comparative hemodynamic and neurohor- 53. Hollenberg NK. A randomized trial of the angiotensin-receptor blocker valsartan in chronic
monal effects of intravenous captopril and digoxin and their combinations in patients with heart failure: author’s response. Curr Hypertens Rep 2002;4:412.
severe heart failure. J Am Coll Cardiol 1989;13:134–142. 54. Effects of enalapril on mortality in severe congestive heart failure. N Engl J Med 1987;316:
30. The effect of digoxin on mortality and morbidity in patients with heart failure. N Engl J Med
1429–1435.
1997;336:525–533.
55. Investigators* TS. Effect of enalapril on mortality and the development of heart failure in
31. Konstantinou DiM, Karvounis H, Giannakoulas G. Digoxin in heart failure with a reduced
asymptomatic patients with reduced left ventricular ejection fractions. N Engl J Med
ejection fraction: a risk factor or a risk marker? Cardiol 2016;134:311–319.
1992;327:685–691.
32. EUCTR2018–003789-15-NL. Digoxin Evaluation in Chronic heart failure: Investigational
56. Yusuf S. Effect of enalapril on survival in patients with reduced left ventricular ejection frac-
Study In Outpatients in the Netherlands. https://trialsearch.who.int/Trial2.aspx?TrialID=
tions and congestive heart failure. Ann Intern 1991;115:67.
EUCTR2018-003789-15-NL. 2019. https://www.cochranelibrary.com/central/doi/10.
57. Cleland JGF, Tendera M, Adamus J, Freemantle N, Polonski L, Taylor J. The perindopril in
1002/central/CN-02169107/full (2 October 2022)
elderly people with chronic heart failure (PEP-CHF) study. Eur Heart J 2006;27:2338–2345.
33. Bavendiek U, Berliner D, Dávila LA, Schwab J, Maier L, Philipp SA, Rieth A, Westenfeld R,
58. Dickstein K, Kjekshus J. Effects of losartan and captopril on mortality and morbidity in high-
Piorkowski C, Weber K, Hänselmann A, Oldhafer M, Schallhorn S, von der Leyen H,
risk patients after acute myocardial infarction: the OPTIMAAL randomised trial. Lancet
Schröder C, Veltmann C, Störk S, Böhm M, Koch A, Bauersachs J. Rationale and design
of the DIGIT-HF trial (DIGitoxin to improve ouTcomes in patients with advanced chronic 2002;360:752–760.
heart failure): a randomized, double-blind, placebo-controlled study. Eur J Heart Fail 2019; 59. McMurray JJV, Östergren J, Swedberg K, Granger CB, Held P, Michelson EL, Olofsson B,
21:676–684. Yusuf S, Pfeffer MA. Effects of candesartan in patients with chronic heart failure and re-
34. Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, Deswal A, Drazner duced left-ventricular systolic function taking angiotensin-converting-enzyme inhibitors:
MH, Dunlay SM, Evers LR, Fang JC, Fedson SE, Fonarow GC, Hayek SS, Hernandez AF, the CHARM-added trial. Lancet 2003;362:767–771.
Khazanie P, Kittleson MM, Lee CS, Link MS, Milano CA, Nnacheta LC, Sandhu AT, 60. Granger CB, McMurray JJV, Yusuf S, Held P, Michelson EL, Olofsson B, Östergren J, Pfeffer
Stevenson LW, Vardeny O, Vest AR, Yancy CW, Beckman JA, O’Gara PT, Al-Khatib MA, Swedberg K. Effects of candesartan in patients with chronic heart failure and reduced
SM, Armbruster AL, Writing Committee Members; ACC/AHA Joint Committee left-ventricular systolic function intolerant to angiotensin-converting-enzyme inhibitors:
Members. 2022 AHA/ACC/HFSA guideline for the management of heart failure. J Card the CHARM-alternative trial. Lancet 2003;362:772–776.
Fail 2022;28:e1–e167. 61. Yusuf S, Pfeffer MA, Swedberg K, Granger CB, Held P, McMurray JJV, Michelson EL,
35. Lechat P. A randomized trial of β-blockade in heart failure: the cardiac insufficiency biso- Olofsson B, Östergren J. Effects of candesartan in patients with chronic heart failure and
prolol study (CIBIS). Circulation 1994;90:1765–1773. preserved left-ventricular ejection fraction: the CHARM-preserved trial. Lancet 2003;
36. Fowler MB. Carvedilol prospective randomized cumulative survival (COPERNICUS) trial: 362:777–781.
carvedilol in severe heart failure. Am J Cardiol 2004;93:35–39. 62. Massie BM, Carson PE, McMurray JJ, Komajda M, McKelvie R, Zile MR, Anderson S,
37. Packer M, Bristow MR, Cohn JN, Colucci WS, Fowler MB, Gilbert EM, Shusterman NH. Donovan M, Iverson E, Staiger C, Ptaszynska A. Irbesartan in patients with heart failure
The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. and preserved ejection fraction. N Engl J Med 2008;359:2456–2467.
N Engl J Med 1996;334:1349–1355. 63. Patel VB, Zhong JC, Grant MB, Oudit GY. Role of the ACE2/angiotensin 1–7 axis of the
38. Lefkowitz RJ, Rockman HA, Koch WJ. Catecholamines, cardiac β-adrenergic receptors, and renin-angiotensin system in heart failure. Circ Res 2016;118:1313.
heart failure. Circulation 2000;101:1634–1637. 64. Yousif MHM, Benter IF, Diz DI, Chappell MC. Angiotensin-(1–7)-dependent vasorelaxation
39. Bouzamondo A, Hulot JS, Sanchez P, Cucherat M, Lechat P. Beta-blocker treatment in of the renal artery exhibits unique angiotensin and bradykinin receptor selectivity. Peptides
heart failure. Fundam Clin Pharmacol 2001;15:95–109. 2017;90:10–16.
40. Paolillo S, Dell’Aversana S, Esposito I, Poccia A, Perrone Filardi P. The use of β-blockers in 65. South AM, Shaltout HA, Washburn LK, Hendricks AS, Diz DI, Chappell MC. Fetal program-
patients with heart failure and comorbidities: doubts, certainties and unsolved issues. Eur J ming and the angiotensin-(1-7) axis: a review of the experimental and clinical data. Clin Sci
Intern Med 2021;88:9–14. 2019;133:55–74.
41. Ferrari R. Ivabradine: heart rate and left ventricular function. Cardiol 2014;128:226–230. 66. Mangiafico S, Costello-Boerrigter LC, Andersen IA, Cataliotti A, Burnett JC. Neutral endo-
42. McAlister FA, Wiebe N, Ezekowitz JA, Leung AA, Armstrong PW. Meta-analysis: β-blocker peptidase inhibition and the natriuretic peptide system: an evolving strategy in cardiovas-
dose, heart rate reduction, and death in patients with heart failure. Ann Intern Med 2009; cular therapeutics. Eur Heart J 2013;34:886–893.
150:784–794. 67. Lohmann SM, Vaandrager AB, Smolenski A, Walter U, De Jonge HR. Distinct and specific
43. Fox K, Ford I, Steg PG, Tendera M, Ferrari R, Grancelli H, Freedman B, Eber B,
functions of cGMP-dependent protein kinases. Trends Biochem Sci 1997;22:307–312.
Vanoverschelde JL, Finkov B, Yotov Y, Tardif JC, Hu D, Lau C, Hradec J, Hildebrandt P,
68. Suzuki T, Yamazaki T, Yazaki Y. The role of the natriuretic peptides in the cardiovascular
Eha J, Peuhkurinen K, Danchin N, Meinertz T, Vardas P, Borbola J, Mulcahy D, Maggioni
system. Cardiovasc Res 2001;51:489–494.
A, Erglis A, Jirgensons J, Kalnins U, Laucevicius A, Dickstein K, Ruzyllo W, BEAUTIFUL
69. O’Connor CM, Starling RC, Hernandez AF, Armstrong PW, Dickstein K, Hasselblad V,
Investigators. Ivabradine for patients with stable coronary artery disease and left-
Heizer GM, Komajda M, Massie BM, McMurray JJV, Nieminen MS, Reist CJ, Rouleau JL,
ventricular systolic dysfunction (BEAUTIFUL): a randomised, double-blind, placebo-
Swedberg K, Adams KF, Anker SD, Atar D, Battler A, Botero R, Bohidar NR, Butler J,
controlled trial. Lancet 2008;372:807–816.
Clausell N, Corbalán R, Costanzo MR, Dahlstrom U, Deckelbaum LI, Diaz R, Dunlap
44. Ceconi C, Comini L, Suffredini S, Stillitano F, Bouly M, Cerbai E, Mugelli A, Ferrari R. Heart
ME, Ezekowitz JA, Feldman D, Felker GM, Fonarow GC, Gennevois D, Gottlieb SS, Hill
rate reduction with ivabradine prevents the global phenotype of left ventricular remodel-
ing. Am J Physiol Hear Circ Physiol 2011;300:H366–H373. JA, Hollander JE, Howlett JG, Hudson MP, Kociol RD, Krum H, Laucevicius A, Levy WC,
45. Ferrari R, Fox K. Heart rate reduction in coronary artery disease and heart failure. Nat Rev Méndez GF, Metra M, Mittal S, Oh BH, Pereira NL, Ponikowski P, Tang WH, Tanomsup
Cardiol 2016;13:493–501. S, Teerlink JR, Triposkiadis F, Troughton RW, Voors AA, Whellan DJ, Zannad F, Califf
46. Hjalmarson A, Goldstein S, Fagerberg B, Wedel H, Waagstein F, Kjekshus J, Wikstrand J, RM. Effect of nesiritide in patients with acute decompensated heart failure. N Engl J Med
Westergren G, Thimell M, El Allaf D, Vítovec J, Aldershvile J, Halinen M, Dietz R, 2011;365:32–43.
Neuhaus KL, Jánosi A, Thorgeirsson G, Dunselman P, Gullestad L, Kuch J, Herlitz J, 70. Potter LR. Natriuretic peptide metabolism, clearance and degradation. FEBS J 2011;278:
Rickenbacher P, Ball S, Gottlieb S, Deedwania P, Vandenhoven G, Nováková I, Danker S, 1808–1817.
Lundström M, Meyer-Sabellek W, MERIT-HF Study Group. Effect of metoprolol CR/XL 71. Münzel T, Kurz S, Holtz J, Busse R, Steinhauer H, Just H, Drexler H. Neurohormonal inhib-
in chronic heart failure: metoprolol CR/XL randomised intervention trial in congestive ition and hemodynamic unloading during prolonged inhibition of ANF degradation in pa-
heart failure (MERIT-HF). Lancet 1999;353:2001–2007. tients with severe chronic heart failure. Circulation 1992;86:1089–1098.
47. Dargie HJ, Lechat P. The cardiac insufficiency bisoprolol study II (CIBIS-II): a randomised 72. Birkenfeld AL, Adams F, Schroeder C, Engeli S, Jordan J. Metabolic actions could confound
trial. Lancet 1999;353:9–13. advantageous effects of combined angiotensin II receptor and neprilysin inhibition.
48. Flather MD, Shibata MC, Coats AJS, Van Veldhuisen DJ, Parkhomenko A, Borbola J, Hypertension 2011;57:e4–e5.
Cohen-Solal A, Dumitrascu D, Ferrari R, Lechat P, Soler-Soler J, Tavazzi L, Spinarova L, 73. Menendez JT. The mechanism of action of LCZ696. Card Fail Rev 2016;2:40.
14 M.S. Khan et al.

74. McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, Rouleau JL, Shi VC, Gause-Nilsson IAM, Fredriksson M, Johansson PA, Langkilde A-M, Sabatine MS.
Solomon SD, Swedberg K, Zile MR. Angiotensin–neprilysin inhibition versus enalapril in Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2019;380:
heart failure. N Engl J Med 2014;371:993–1004. 347–357.
75. Morrow DA, Velazquez EJ, Devore AD, Desai AS, Duffy CI, Ambrosy AP, Gurmu Y, 99. Cannon CP, Pratley R, Dagogo-Jack S, Mancuso J, Huyck S, Masiukiewicz U, Charbonnel B,
Mccague K, Rocha R, Braunwald E. Clinical outcomes in patients with acute decompen- Frederich R, Gallo S, Cosentino F, Shih WJ, Gantz I, Terra SG, Cherney DZI, McGuire DK.
sated heart failure randomly assigned to sacubitril/valsartan or enalapril in the Cardiovascular outcomes with ertugliflozin in type 2 diabetes. N Engl J Med 2020;383:
PIONEER-HF trial. Circulation 2019;139:2285–2288. 1425–1435.
76. Solomon SD, McMurray JJV, Anand IS, Ge J, Lam CSP, Maggioni AP, Martinez F, Packer M, 100. Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Böhm M, Brunner–La Rocca H-P,
Pfeffer MA, Pieske B, Redfield MM, Rouleau JL, van Veldhuisen DJ, Zannad F, Zile MR, Desai Choi D-J, Chopra V, Chuquiure-Valenzuela E, Giannetti N, Gomez-Mesa JE, Janssens S,
AS, Claggett B, Jhund PS, Boytsov SA, Comin-Colet J, Cleland J, Düngen H-D, Goncalvesova Januzzi JL, Gonzalez-Juanatey JR, Merkely B, Nicholls SJ, Perrone SV, Piña IL, Ponikowski
E, Katova T, Kerr Saraiva JF, Lelonek M, Merkely B, Senni M, Shah SJ, Zhou J, Rizkala AR, Gong P, Senni M, Sim D, Spinar J, Squire I, Taddei S, Tsutsui H, Verma S, Vinereanu D, Zhang

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


J, Shi VC, Lefkowitz MP; PARAGON-HF Investigators and Committees. Angiotensin–neprilysin J, Carson P, Lam CSP, Marx N, Zeller C, Sattar N, Jamal W, Schnaidt S, Schnee JM,
inhibition in heart failure with preserved ejection fraction. N Engl J Med 2019;381: Brueckmann M, Pocock SJ, Zannad F, Packer M; EMPEROR-Preserved Trial
1609–1620. Investigators. Empagliflozin in heart failure with a preserved ejection fraction. N Engl J
77. Myhre PL, Prescott MF, Murphy SP, Fang JC, Mitchell GF, Ward JH, Claggett B, Desai AS, Med 2021;385:1451–1461.
Solomon SD, Januzzi JL. Early B-type natriuretic peptide change in HFrEF patients treated 101. Bhatt DL, Szarek M, Steg PG, Cannon CP, Leiter LA, McGuire DK, Lewis JB, Riddle MC,
with sacubitril/valsartan: a pooled analysis of EVALUATE-HF and PROVE-HF. JACC Hear Voors AA, Metra M, Lund LH, Komajda M, Testani JM, Wilcox CS, Ponikowski P, Lopes
Fail 2022;10:119–128. RD, Verma S, Lapuerta P, Pitt B. Sotagliflozin in patients with diabetes and recent worsen-
78. Ibrahim NE, McCarthy CP, Shrestha S, Gaggin HK, Mukai R, Szymonifka J, Apple FS, Burnett ing heart failure. N Engl J Med 2021;384:117–128.
JC, Iyer S, Januzzi JL. Effect of neprilysin inhibition on various natriuretic peptide assays. J Am 102. Anker SD, Khan MS, Shahid I, Filippatos G, Coats AJS, Butler J. Sodium–glucose co-
Coll Cardiol 2019;73:1273–1284. transporter 2 inhibitors in heart failure with preserved ejection fraction: reasons for opti-
79. Michel T, Nougué H, Cartailler J, Lefèvre G, Sadoune M, Picard F, Cohen-Solal A, Logeart mism. Eur J Heart Fail 2021;23:1250–1255.
D, Launay J-M, Vodovar N. proANP metabolism provides new insights into sacubitril/val- 103. Lopaschuk GD, Verma S. Mechanisms of cardiovascular benefits of sodium glucose co-
sartan mode of action. Circ Res 2022;130:e44–e57. transporter 2 (SGLT2) inhibitors: a state-of-the-art review. JACC Basic Transl Sci 2020;5:
80. Arfsten H, Goliasch G, Bartko PE, Prausmüller S, Spinka G, Cho A, Novak J, Haslacher H, 632–644.
Strunk G, Struck J, Hülsmann M, Pavo N. Increased concentrations of bioactive adrenome- 104. Sha S, Polidori D, Heise T, Natarajan J, Farrell K, Wang SS, Sica D, Rothenberg P,
dullin subsequently to angiotensin-receptor/neprilysin-inhibitor treatment in chronic sys- Plum-Mörschel L. Effect of the sodium glucose co-transporter 2 inhibitor canagliflozin
tolic heart failure. Br J Clin Pharmacol 2021;87:916–924. on plasma volume in patients with type 2 diabetes mellitus. Diabetes Obes Metab 2014;
81. Seferovic JP, Claggett B, Seidelmann SB, Seely EW, Packer M, Zile MR, Rouleau JL, 16:1087–1095.
Swedberg K, Lefkowitz M, Shi VC, Desai AS, McMurray JJV, Solomon SD. Effect of sacubi- 105. Grubić Rotkvić P, Cigrovski Berković M, Bulj N, Rotkvić L. Minireview: are SGLT2 inhibitors
tril/valsartan versus enalapril on glycaemic control in patients with heart failure and dia- heart savers in diabetes? Heart Fail Rev 2020;25:899–905.
betes: a post-hoc analysis from the PARADIGM-HF trial. Lancet Diabetes Endocrinol 106. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D’Agostino D,
2017;5:333–340. Planavsky N, Lupfer C, Kanneganti TD, Kang S, Horvath TL, Fahmy TM, Crawford PA,
82. Zile MR, O’Meara E, Claggett B, Prescott MF, Solomon SD, Swedberg K, Packer M, Biragyn A, Alnemri E, Dixit VD. The ketone metabolite β-hydroxybutyrate blocks
McMurray JJV, Shi V, Lefkowitz M, Rouleau J. Effects of sacubitril/valsartan on biomarkers NLRP3 inflammasome-mediated inflammatory disease. Nat Med 2015;21:263–269.
of extracellular matrix regulation in patients with HFrEF. J Am Coll Cardiol 2019;73: 107. Chiba Y, Yamada T, Tsukita S, Takahashi K, Munakata Y, Shirai Y, Kodama S, Asai Y,
795–806. Sugisawa T, Uno K, Sawada S, Imai J, Nakamura K, Katagiri H. Dapagliflozin, a sodium-
83. Stier CT, Koenig S, Lee DY, Chawla M, Frishman WH. Aldosterone and aldosterone antag- glucose co-transporter 2 inhibitor, acutely reduces energy expenditure in BAT via neural
onism in cardiovascular disease: focus on eplerenone (inspra). Heart Dis 2003;5:102–118. signals in mice. PLoS One 2016;11:e0150756.
84. Frishman WH, Stier CT. Aldosterone and aldosterone antagonism in systemic hyperten- 108. Al Jobori H, Daniele G, Adams J, Cersosimo E, Triplitt C, DeFronzo RA, Abdul-Ghani M.
sion. Curr Hypertens Rep 2004;6:195–200. Determinants of the increase in ketone concentration during SGLT2 inhibition in NGT,
85. Funder JW. Minireview: aldosterone and mineralocorticoid receptors: past, present, and IFG and T2DM patients. Diabetes Obes Metab 2017;19:809–813.
future. Endocrinology 2010;151:5098–5102. 109. Ferrannini E, Muscelli E, Frascerra S, Baldi S, Mari A, Heise T, Broedl UC, Woerle HJ.
86. Vizzardi E, Regazzoni V, Caretta G, Gavazzoni M, Sciatti E, Bonadei I, Trichaki E, Raddino R, Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic pa-
Metra M. Mineralocorticoid receptor antagonist in heart failure: past, present and future tients. J Clin Invest 2014;124:499–508.
perspectives. IJC Hear Vessel 2014;3:6–14. 110. Lee MMY, Brooksbank KJM, Wetherall K, Mangion K, Roditi G, Campbell RT, Berry C,
87. Epstein M. Aldosterone and mineralocorticoid receptor signaling as determinants of car- Chong V, Coyle L, Docherty KF, Dreisbach JG, Labinjoh C, Lang NN, Lennie V,
diovascular and renal injury: from hans selye to the present. Am J Nephrol 2021;52:209–216. McConnachie A, Murphy CL, Petrie CJ, Petrie JR, Speirits IA, Sourbron S, Welsh P,
88. Funder JW. Reconsidering the roles of the mineralocorticoid receptor. Hypertension 2009; Woodward R, Radjenovic A, Mark PB, McMurray JJV, Jhund PS, Petrie MC, Sattar N.
53:286–290. Effect of empagliflozin on left ventricular volumes in patients with type 2 diabetes, or pre-
89. Bender SB, Demarco VG, Padilla J, Jenkins NT, Habibi J, Garro M, Pulakat L, Aroor AR, Jaffe diabetes, and heart failure with reduced ejection fraction (SUGAR-DM-HF). Circulation
IZ, Sowers JR. Mineralocorticoid receptor antagonism treats obesity-associated cardiac 2021;143:516–525.
diastolic dysfunction. Hypertension 2015;65:1082–1088. 111. Santos-Gallego CG, Vargas-Delgado AP, Requena-Ibanez JA, Garcia-Ropero A, Mancini D,
90. Pitt B, Zannad F, Remme WJ, Cody R, Castaigne A, Perez A, Palensky J, Wittes J. The effect Pinney S, Macaluso F, Sartori S, Roque M, Sabatel-Perez F, Rodriguez-Cordero A, Zafar
of spironolactone on morbidity and mortality in patients with severe heart failure. N Engl J MU, Fergus I, Atallah-Lajam F, Contreras JP, Varley C, Moreno PR, Abascal VM, Lala A,
Med 1999;341:709–717. Tamler R, Sanz J, Fuster V, Badimon JJ. Randomized trial of empagliflozin in nondiabetic pa-
91. Zannad F, McMurray JJV, Krum H, van Veldhuisen DJ, Swedberg K, Shi H, Vincent J, Pocock tients with heart failure and reduced ejection fraction. J Am Coll Cardiol 2021;77:243–255.
SJ, Pitt B. Eplerenone in patients with systolic heart failure and mild symptoms. N Engl J Med 112. Omar M, Jensen J, Ali M, Frederiksen PH, Kistorp C, Videbæk L, Poulsen MK, Tuxen CD,
2011;364:11–21. Möller S, Gustafsson F, Køber L, Schou M, Møller JE. Associations of empagliflozin with left
92. Pitt B, Pfeffer MA, Assmann SF, Boineau R, Anand IS, Claggett B, Clausell N, Desai AS, Diaz ventricular volumes, mass, and function in patients with heart failure and reduced ejection
R, Fleg JL, Gordeev I, Harty B, Heitner JF, Kenwood CT, Lewis EF, O’Meara E, Probstfield JL, fraction: a substudy of the empire HF randomized clinical trial. JAMA Cardiol 2021;6:
Shaburishvili T, Shah SJ, Solomon SD, Sweitzer NK, Yang S, McKinlay SM. Spironolactone 836–840.
for heart failure with preserved ejection fraction. N Engl J Med 2014;370:1383–1392. 113. Sano M, Goto S. Possible mechanism of hematocrit elevation by sodium glucose cotran-
93. Bader M, Alenina N, Andrade-Navarro MA, Santos RA. Mas and its related G protein– sporter 2 inhibitors and associated beneficial renal and cardiovascular effects. Circulation
coupled receptors, Mrgprs. Pharmacol Rev 2014;66:1080–1105. 2019;139:1985–1987.
94. Shefer G, Marcus Y, Knoll E, Dolkart O, Foichtwanger S, Nevo N, Limor R, Stern N. 114. Kannel WB, Kannel C, Paffenbarger RS, Cupples LA. Heart rate and cardiovascular mortal-
Angiotensin 1-7 is a negative modulator of aldosterone secretion in vitro and in vivo. ity: the framingham study. Am Heart J 1987;113:1489–1494.
Hypertension 2016;68:378–384. 115. DiFrancesco D, Camm JA. Heart rate lowering by specific and selective if current inhibition
95. Joshi SS, Singh T, Newby DE, Singh J. Sodium-glucose co-transporter 2 inhibitor therapy: with ivabradine: A new therapeutic perspective in cardiovascular disease. Drugs 2004;64:
mechanisms of action in heart failure. Heart 2021;107:1032–1038. 1757–1765.
96. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, 116. Borer JS, Fox K, Jaillon P, Lerebours G. Antianginal and antiischemic effects of ivabradine, an
Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE. Empagliflozin, cardiovascular outcomes, if inhibitor, in stable angina: a randomized, double-blind, multicentered, placebo-controlled
and mortality in type 2 diabetes. N Engl J Med 2015;373:2117–2128. trial. Circulation 2003;107:817–823.
97. Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, Shaw W, Law G, 117. Nawarskas JJ, Bowman BN, Anderson JR. Ivabradine: a unique and intriguing medication for
Desai M, Matthews DR. Canagliflozin and cardiovascular and renal events in type 2 dia- treating cardiovascular disease. Cardiol Rev 2015;23:201–211.
betes. N Engl J Med 2017;377:644–657. 118. Mulieri LA, Hasenfuss G, Leavitt B, Allen PD, Alpert NR. Altered myocardial force-
98. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Silverman MG, Zelniker TA, frequency relation in human heart failure. Circulation 1992;85:1743–1750.
Kuder JF, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, 119. Ferrari R. The ivabradine journey. Eur Hear Journal Suppl 2015;17:G1–G9.
Mechanisms of heart failure therapies 15

120. Swedberg K, Komajda M, Böhm M, Borer JS, Ford I, Dubost-Brama A, Lerebours G, Tavazzi cyclase stimulator BAY 63-2521: an ascending-dose study in healthy male volunteers. J
L. Ivabradine and outcomes in chronic heart failure (SHIFT): A randomised placebo- Clin Pharmacol 2008;48:926–934.
controlled study. Lancet 2010;376:875–885. 138. Mittendorf J, Weigand S, Alonso-Alija C, Bischoff E, Feurer A, Gerisch M, Kern A, Knorr A,
121. Kotecha D, Flather MD, Altman DG, Holmes J, Rosano G, Wikstrand J, Packer M, Coats Lang D, Muenter K, Radtke M, Schirok H, Schlemmer KH, Stahl E, Straub A, Wunder F,
AJS, Manzano L, Böhm M, van Veldhuisen DJ, Andersson B, Wedel H, von Lueder TG, Stasch JP. Discovery of riociguat (BAY 63-2521): a potent, oral stimulator of soluble gua-
Rigby AS, Hjalmarson Å, Kjekshus J, Cleland JGF. Heart rate and rhythm and the benefit nylate cyclase for the treatment of pulmonary hypertension. ChemMedChem 2009;4:
of beta-blockers in patients with heart failure. J Am Coll Cardiol 2017;69:2885–2896. 853–865.
122. Arce C, Segura-Pacheco B, Perez-Cardenas E, Taja-Chayeb L, Candelaria M, 139. Armstrong PW, Pieske B, Anstrom KJ, Ezekowitz J, Hernandez AF, Butler J, Lam CSP,
Dueñnas-Gonzalez A. Hydralazine target: from blood vessels to the epigenome. J Transl Ponikowski P, Voors AA, Jia G, McNulty SE, Patel MJ, Roessig L, Koglin J, O’Connor CM,
Med 2006;4:1–16. VICTORIA Study Group. Vericiguat in Patients with Heart Failure and Reduced Ejection
123. Magee LA, Cham C, Waterman EJ, Ohlsson A, Von Dadelszen P. Hydralazine for treatment Fraction. N Engl J Med 2020;382:1883–1893.

Downloaded from https://academic.oup.com/cardiovascres/advance-article/doi/10.1093/cvr/cvac187/6941155 by guest on 26 January 2023


of severe hypertension in pregnancy: meta-analysis. Br Med J 2003;327:955–960. 140. Pieske B, Maggioni AP, Lam CSP, Pieske-Kraigher E, Filippatos G, Butler J, Ponikowski P,
124. Herting RL, Hunter HL. The physiologic and pharmacologic basis for the clinical treatment Shah SJ, Solomon SD, Scalise AV, Mueller K, Roessig L, Gheorghiade M. Vericiguat in pa-
of hypertension. Med Clin North Am 1967;51:25–37. tients with worsening chronic heart failure and preserved ejection fraction: results of
125. Balasubramanian S, Chowdhury YS. Isosorbide: StatPearls Publishing; 2021. the SOluble guanylate cyclase stimulatoR in heArT failurE patientS with PRESERVED EF
126. Cole RT, Kalogeropoulos AP, Georgiopoulou VV, Gheorghiade M, Quyyumi A, Yancy C, (SOCRATES-PRESERVED) study. Eur Heart J 2017;38:1119–1127.
Butler J. Hydralazine and isosorbide dinitrate in heart failure: historical perspective, me- 141. Armstrong PW, Lam CSP, Anstrom KJ, Ezekowitz J, Hernandez AF, O’Connor CM, Pieske
chanisms, and future directions. . Circulation 2011;123:2414–2422. B, Ponikowski P, Shah SJ, Solomon SD, Voors AA, She L, Vlajnic V, Carvalho F, Bamber L,
127. Taylor AL, Ziesche S, Yancy C, Carson P, D’Agostino R, Ferdinand K, Taylor M, Adams K, Blaustein RO, Roessig L, Butler J. Effect of vericiguat vs placebo on quality of life in patients
Sabolinski M, Worcel M, Cohn JN. Combination of isosorbide dinitrate and hydralazine in
with heart failure and preserved ejection fraction: the VITALITY-HFpEF randomized clin-
blacks with heart failure. N Engl J Med 2004;351:2049–2057.
ical trial. JAMA 2020;324:1512–1521.
128. Ozkor MA, Rahman AM, Murrow JR, Kavtaradze N, Lin J, Manatunga A, Hayek S, Quyyumi
142. Xia J, Hui N, Tian L, Liang C, Zhang J, Liu J, Wang J, Ren X, Xie X, Wang K. Development of
AA. Differences in vascular nitric oxide and endothelium-derived hyperpolarizing factor
vericiguat: the first soluble guanylate cyclase (sGC) stimulator launched for heart failure
bioavailability in blacks and whites. Arterioscler Thromb Vasc Biol 2014;34:1320–1327.
with reduced ejection fraction (HFrEF). Biomed Pharmacother 2022;149:112894.
129. Tsai EJ, Kass DA. Cyclic GMP signaling in cardiovascular pathophysiology and therapeutics.
143. Teerlink JR, Diaz R, Felker GM, McMurray JJV, Metra M, Solomon SD, Legg JC, Büchele G,
Pharmacol Ther 2009;122:216–238.
Varin C, Kurtz CE, Malik FI, Honarpour N. Omecamtiv mecarbil in chronic heart failure
130. Breitenstein S, Roessig L, Sandner P, Lewis KS. Novel sGC stimulators and sGC activators
with reduced ejection fraction: rationale and design of GALACTIC-HF. JACC Heart Fail
for the treatment of heart failure. Handb Exp Pharmacol 2017;243:225–247.
2020;8:329–340.
131. Vettel C, Lämmle S, Ewens S, Cervirgen C, Emons J, Ongherth A, Dewenter M, Lindner D,
144. Teerlink JR, Diaz R, Felker GM, McMurray JJV, Metra M, Solomon SD, Adams KF, Anand I,
Westermann D, Nikolaev VO, Lutz S, Zimmermann WH, El-Armouche A. PDE2-mediated
Arias-Mendoza A, Biering-Sørensen T, Böhm M, Bonderman D, Cleland JGF, Corbalan R,
cAMP hydrolysis accelerates cardiac fibroblast to myofibroblast conversion and is antago-
Crespo-Leiro MG, Dahlström U, Echeverria LE, Fang JC, Filippatos G, Fonseca C,
nized by exogenous activation of cGMP signaling pathways. Am J Physiol Hear Circ Physiol
2014;306:H1246–H1252. Goncalvesova E, Goudev AR, Howlett JG, Lanfear DE, Li J, Lund M, Macdonald P,
132. Tzoumas N, Farrah TE, Dhaun N, Webb DJ. Established and emerging therapeutic uses of Mareev V, Momomura S, O’Meara E, Parkhomenko A, Ponikowski P, Ramires FJA,
PDE type 5 inhibitors in cardiovascular disease. Br J Pharmacol 2020;177:5467–5488. Serpytis P, Sliwa K, Spinar J, Suter TM, Tomcsanyi J, Vandekerckhove H, Vinereanu D,
133. Tfelt-Hansen PC, Tfelt-Hansen J. Nitroglycerin headache and nitroglycerin-induced pri- Voors AA, Yilmaz MB, Zannad F, Sharpsten L, Legg JC, Varin C, Honarpour N, Abbasi
mary headaches from 1846 and onwards: A historical overview and an update. SA, Malik FI, Kurtz CE; GALACTIC-HF Investigators. Cardiac myosin activation with ome-
Headache 2009;49:445–456. camtiv mecarbil in systolic heart failure. N Engl J Med 2021;384:105–116.
134. Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol 145. Teerlink JR, Felker GM, McMurray JJV, Solomon SD, Adams KF, Cleland JGF, Ezekowitz JA,
Rev 2007;87:315–424. Goudev A, Macdonald P, Metra M, Mitrovic V, Ponikowski P, Serpytis P, Spinar J, Tomcsányi
135. Emdin M, Aimo A, Castiglione V, Vergaro G, Georgiopoulos G, Saccaro LF, Lombardi CM, J, Vandekerckhove HJ, Voors AA, Monsalvo ML, Johnston J, Malik FI, Honarpour N. Chronic
Passino C, Cerbai E, Metra M, Senni M. Targeting cyclic guanosine monophosphate to treat oral study of myosin activation to increase contractility in heart failure (COSMIC-HF): a
heart failure: JACC review topic of the week. J Am Coll Cardiol 2020;76:1795–1807. phase 2, pharmacokinetic, randomised, placebo-controlled trial. Lancet 2016;388:
136. Bonderman D, Ghio S, Felix SB, Ghofrani HA, Michelakis E, Mitrovic V, Oudiz RJ, Boateng F, 2895–2903.
Scalise AV, Roessig L, Semigran MJ. Riociguat for patients with pulmonary hypertension 146. Felker GM, Solomon SD, Claggett B, Diaz R, McMurray JJV, Metra M, Anand I, Crespo-Leiro
caused by systolic left ventricular dysfunction: a phase IIb double-blind, randomized, MG, Dahlström U, Goncalvesova E, Howlett JG, Macdonald P, Parkhomenko A, Tomcsányi
placebo-controlled, dose-ranging hemodynamic study. . Circulation 2013;128:502–511. J, Abbasi SA, Heitner SB, Hucko T, Kupfer S, Malik FI, Teerlink JR. Assessment of omecamtiv
137. Frey R, Mück W, Unger S, Artmeier-Brandt U, Weimann G, Wensing G. Single-dose mecarbil for the treatment of patients with severe heart failure: a post hoc analysis of data
pharmacokinetics, pharmacodynamics, tolerability, and safety of the soluble guanylate from the GALACTIC-HF randomized clinical trial. JAMA Cardiol 2022;7:26–34.

You might also like