ArticlePDF AvailableLiterature Review

Four pillars of heart failure: contemporary pharmacological therapy for heart failure with reduced ejection fraction

Authors:
Open access
1
Straw S, etal. Open Heart 2021;8:e001585. doi:10.1136/openhrt-2021-001585
To cite: Straw S, McGinlay M,
Witte KK. Four pillars of heart
failure: contemporary
pharmacological therapy for
heart failure with reduced
ejection fraction. Open Heart
2021;8:e001585. doi:10.1136/
openhrt-2021-001585
Accepted 11 February 2021
1Leeds Institute of
Cardiovascular and Metabolic
Medicine, University of Leeds,
Leeds, UK
2Cardiorespiratory Clinical
Services Unit, Leeds Teaching
Hospitals NHS Trust, Leeds, UK
Correspondence to
Dr Klaus K Witte; k. k. witte@
leeds. ac. uk
Four pillars of heart failure:
contemporary pharmacological therapy
for heart failure with reduced
ejection fraction
Sam Straw ,1 Melanie McGinlay,2 Klaus K Witte1
Viewpoint
© Author(s) (or their
employer(s)) 2021. Re- use
permitted under CC BY.
Published by BMJ.
INTRODUCTION
The past two decades have heralded dramatic
improvements in outcomes for people living
with heart failure with reduced ejection
fraction (HFrEF).1 The more widespread
implementation of disease modifying phar-
macological therapies,2 supported by land-
mark trials of renin- angiotensin system
inhibitors3 and beta- blockers4 have improved
longevity despite a background of an ageing
and increasingly multimorbid population.
Although the benefits of comprehensive
pharmacological therapies are clear, the
real- world attainment of target doses5 6 and
utilisation of novel agents such as angio-
tensin receptor- neprilysin inhibitors (ARNI)7
remain low. Furthermore, HFrEF remains a
disease associated with significant morbidity
and reduced survival relative to those without
HFrEF, even after taking into account comor-
bidities.8 Recently, trials have demonstrated
improved outcomes in people with HFrEF
receiving sodium- glucose co- transporter
2 inhibitors (SGLT2i).9 10 However, it is
currently unclear how these agents will be
used alongside established therapies. Now
is therefore an opportune moment to pause
and reflect on our current practice, barriers
to further progress and how future guide-
lines might work better for our patients. In
this viewpoint we summarise how our current
linear approach, on a background of increas-
ingly complex pharmacotherapy has the
potential to cause confusion and consequent
delays which could lead to even worse attain-
ment of optimal therapies. On the other
hand, a more parallel approach to the initi-
ation and optimisation of the Four Pillars of
Heart Failure would simplify our approach,
yielding benefits for our patients and health-
care systems.
HOW DID WE GET HERE?
Heart failure guidelines are based around
inhibition of the renin- angiotensin and
sympathetic nervous systems, two funda-
mental pathways which drive the pathophys-
iology of HFrEF using ACE inhibitors (ACEi)
and beta- blockers. In both European2 and
American guidelines11 additional therapies
are recommended for patients who ‘remain
symptomatic’ with persistently impaired left
ventricular (LV) function despite maximally
tolerated doses of ACEi and beta- blockers.
These guidelines differ subtly regarding the
timing of mineralocorticoid receptor antag-
onists (MRA) relative to other therapies but
are otherwise broadly similar, advocating
a linear approach. This attempts to avoid
‘unnecessary’ treatments in patients who
‘respond’ but has several important limita-
tions. First, while guidelines do not stipulate
a time interval between alterations to therapy,
the need for further assessment and re- eval-
uation of LV function inevitably results in
delays initiating additional agents as well as
contributing further follow- up and imaging
costs. In clinical practice it typically takes
many months before patients receive opti-
mised doses of these medications, and many
never do, even where integrated hospital
and community care is available.5 Second,
the barrier of ‘response’ is confusing and
misplaced: does ‘response’ mean asympto-
matic or merely improved? In our experi-
ence, while patients often feel better, they
rarely become asymptomatic (NYHA (New
York Heart Association) class 1),12 an obser-
vation supported by real- world data even
in those receiving ARNI.13 14 Moreover, we
should consider whether a highly subjec-
tive and poorly reproducible assessment is
appropriate to determine our allocation
of life- saving treatments.15 Hence, criteria
Open Heart
2Straw S, etal. Open Heart 2021;8:e001585. doi:10.1136/openhrt-2021-001585
requiring repeat assessment act as a barrier to initiating
additional therapies such as MRA or ARNI,7 which are
regarded as ‘second- line’ due to the hierarchical frame-
work which places greater emphasis on therapies based
on the chronological sequence in which the trials were
performed. There is no logical basis to assume that drug
classes trialled earliest would be the most beneficial, yet
this is what guidelines imply. Therefore, if we are to make
progress, future guidelines must address these limitations
and incorporate the Four Pillars of Heart Failure into a
comprehensive disease modifying programme for all
people living with HFrEF.
LEARNING LESSONS FROM ARNI
The PARADIGM- HF trial showed that a combination
of angiotensin receptor blocker and a neprilysin inhib-
itor (sacubitril- valsartan) was superior to an ACEi in
preventing cardiovascular deaths or hospitalisation
for heart failure (HR 080, 95% CI 0.73 to 0.87) and
reducing all- cause mortality (HR 0.84, 95% CI 0.76 to
0.93).16 Despite overwhelming efficacy which led to the
trial being stopped early, the utilisation of ARNI in the
real- world has been suboptimal, with less than 1% pene-
tration in eligible patients. The reluctance of physicians
to prescribe ARNI may in part be that, unusually, the
recommendations are based on a single trial, studied
in an ‘A+B vs C’ fashion. Furthermore, the comparator
was a submaximal dose of enalapril compounded by
lower blood pressure in those allocated ARNI suggesting
undertreatment in the control arm.16 It has also been
suggested that the trial was additionally biased in favour
of the novel agent due to a double drug run- in period of
unequal times, in which those randomised to ARNI had
already received an ACEi and were therefore pre- selected
(20% of patients were lost during the run- in period).17
Drug doses are related to outcomes in HFrEF18–20 and
the HR for the composite outcome in PARADIGM- HF
between sacubitril- valsartan and enalapril was similar to
the comparison of high and low dosing of lisinopril in
the ATLAS trial.21 However, post- hoc analysis has shown
that the point estimates for the benefit of low dose ARNI
compared with low dose ACEi were identical to the point
estimate of the overall trial,22 and real- world data have
shown clear improvements in outcomes, symptoms and
quality of life compared with standard of care ACEi.23
Another difficulty employing ARNI across the board
includes the wash- out period required following cessation
of ACEi due to risks of angioedema. Although not insur-
mountable, such requirements challenge heart failure
programmes facing reduced face- to- face appointments
due to service redesign and the current pandemic. Hence,
if the benefits of the activity are perceived (whether
correctly or incorrectly) to be minimal, physician inertia
may prevail. To counter this, initiating ARNI at the point
of diagnosis would mitigate the risk of inertia while also
providing a more effective treatment to patients during
the period of highest risk.
IMPLEMENTING NOVEL AGENTS INTO HEART FAILURE
PATHWAYS
The efficacy of SGLT2i in addition to standard therapies
for people with HFrEF has been confirmed with consistent
and near identical 25% risk reduction of the primary end
point of cardiovascular death or hospitalisation for heart
failure from both dapagliflozin and empagliflozin.10 24
Both trials also demonstrated a slow of decline in renal
function; EMPEROR- Reduced showed a 50% relative risk
reduction for the composite renal endpoint10 (although
this was non- significant in DAPA- HF).24
It is anticipated that more than four out of five people
with HFrEF in contemporary registries25 will be eligible
based on the inclusion criteria of these trials. The bene-
ficial effects on renal outcomes are particularly attractive
in a disease process typically associated with progressive
decline of kidney function which often prevents the initi-
ation or intensification of renin- angiotensin system inhib-
itors. Furthermore, SGLT2i are safe, with a low incidence
of serious side effects (no patients without diabetes devel-
oped ketoacidosis in DAPA- HF or EMPEROR- Reduced),
a lack of dosing considerations and minimal effects on
blood pressure.
Given the somewhat simpler approach taken in trials of
SGLT2i, the ease of use and clear benefits, it is likely that
uptake among physicians will be enthusiastic, although
it is yet unclear how these agents might fit into our
current practice. There is a risk that SGLT2i become an
additional agent for patients who do not ‘respond’ with
conventional pharmacological therapy, rather than a
fundamental Pillar of Heart Failure. The totality of the
available evidence suggests the benefits of SGLT2i are
consistent across subgroups, including diabetes status,
baseline ARNI and symptoms. SGLT2i must therefore
be regarded as a unique class of medication with a novel
mechanism of action to be used in all eligible patients.
THE POSSIBILITIES OF A COMPREHENSIVE APPROACH
The four drug classes are complementary to each other
and cross- trial comparisons have shown that a compre-
hensive disease modification strategy beyond the treat-
ments which most patients receive (ACEi and beta-
blocker) with ARNI, MRA and SGLT2i are associated with
improved outcomes. A typical patient aged 65 years can
expect to live an additional 5 years if receiving a compre-
hensive strategy with the Four Pillars, compared with
conventional therapy.26
INERTIA: KNOW YOUR ENEMY
Drugs, trial design and side effects aside, the key obstacle
to therapy intensification is physician inertia in patients
who are deemed to have stabilised or ‘responded’ to
treatment. For some with a recent decompensation this
might be appropriate,27 28 but the relevant clinical trials
were carried out in ambulatory patients receiving stable
doses of previous generations of medical therapy, most
3
Straw S, etal. Open Heart 2021;8:e001585. doi:10.1136/openhrt-2021-001585
Viewpoint
of which had class II symptoms.10 16 24 Of particular rele-
vance to SGLT2i, a striking result from DAPA- HF was the
early benefit from dapagliflozin, with a reduction in wors-
ening heart failure events observed within 28 days.24
SIMPLIFY TO PROGRESS
We propose a novel conceptual framework for the imple-
mentation of pharmacological therapies in HFrEF, in
which the Four Pillars of Heart Failure are introduced
in parallel, very early in the patient pathway with subse-
quent optimisation of dosing where required (figure 1).24
The rate of dose increments can be tailored to the patient
and the service. For most patients, low dose ARNI and
SGLT2i could be started simultaneously, followed within
a few days by low dose beta- blocker and MRA, followed
by up- titration. While others have suggested that initia-
tion with beta- blocker alongside SGLT2i might be more
optimal,29 we believe that the exact sequent of initiation
is unimportant, as long as all Four Pillars are introduced
within the first few weeks of diagnosis—although with the
lack of dosing considerations and clear evidence from
randomised trials of an early beneficial effect,27 SGLT2i
are an obvious first choice.
This approach is at odds with current practice and a
fundamental shift away from the linear approach advo-
cated by guidelines, including the recent technology
appraisal of dapagliflozin from the UK.30 Nevertheless, it
remains the case, that very few patients become asymp-
tomatic with even optimal doses of disease- modifying
therapy suggesting that slow up- titration, followed by a
decision to embark on the next step based on symptoms
is folly.
It seems likely that impairment of renal function
following up- titration of ARNI and MRA will become
lesser concerns following the introduction of SGLT2i
into care pathways, however, real- world data are vital to
confirm the safety and feasibility of this approach. Those
caring for people with heart failure must be cognisant
that early increases in creatinine with SGLT2i are tran-
sient and be reassured that in the long- term the less rapid
decline in renal function in patients receiving SGLT2i will
allow more complete renin- angiotensin system blockade.
CONCLUSION
The introduction of SGLT2i to the treatment of HFrEF
is a chance for us to revisit whether current guidelines
for the treatment of HFrEF are fit for purpose. Many
patients have waited for weeks or months before ever
seeing a cardiologist and receiving a diagnosis, and
further delays to treatments have the potential to cause
great harm. Moving forwards, we must recognise heart
failure for what it is, an incurable disease with a mortality
rate similar to many forms of cancer, where any delays
cost lives. Once we have done so we need to implement
pathways that offer rapid initiation and, where required,
up- titration of life- extending therapies.
Twitter Sam Straw @DrSamStraw
Contributors All authors contributed equally.
Funding This work was supported by the British Heart Foundation: CH/13/1/30086.
Competing interests KKW has received speakers’ fees and honoraria from
Medtronic, Cardiac Dimensions, Novartis, Abbott, BMS, Pzer, Bayer and has
received an unconditional research grant from Medtronic.
Patient consent for publication Not required.
Provenance and peer review Not commissioned; externally peer reviewed.
Data availability statement No data are available.
Open access This is an open access article distributed in accordance with the
Creative Commons Attribution 4.0 Unported (CC BY 4.0) license, which permits
others to copy, redistribute, remix, transform and build upon this work for any
purpose, provided the original work is properly cited, a link to the licence is given,
and indication of whether changes were made. See:https:// creativecommons. org/
licenses/ by/ 4. 0/.
ORCID iD
SamStraw http:// orcid. org/ 0000- 0002- 2942- 4574
REFERENCES
1 Cubbon RM, Gale CP, Kearney LC, etal. Changing characteristics
and mode of death associated with chronic heart failure caused by
left ventricular systolic dysfunction: a study across therapeutic eras.
Circ Heart Fail 2011;4:396–403.
2 Ponikowski P, Voors AA, Anker SD, etal. 2016 ESC Guidelines for
the diagnosis and treatment of acute and chronic heart failure: The
Task Force for the diagnosis and treatment of acute and chronic
heart failure of the European Society of Cardiology (ESC)Developed
with the special contribution of the Heart Failure Association (HFA) of
the ESC. Eur Heart J 2016;37:2129–200.
3 SOLVD Investigators, Yusuf S, Pitt B, etal. Effect of enalapril on
survival in patients with reduced left ventricular ejection fractions
and congestive heart failure. N Engl J Med 1991;325:293–302.
4 The cardiac insufciency bisoprolol study II (CIBIS- II): a randomised
trial. Lancet 1999;353:9–13.
5 Greene SJ, Fonarow GC, DeVore AD, etal. Titration of medical
therapy for heart failure with reduced ejection fraction. J Am Coll
Cardiol 2019;73:2365–83.
6 Brunner- La Rocca H- P, Linssen GC, Smeele FJ, etal. Contemporary
Drug Treatment ofChronic Heart Failure With ReducedEjection
Fraction: The CHECK- HF Registry. JACC Heart Fail 2019;7:13–21.
Figure 1 Initiation and optimisation of the Four Pillars
of Heart Failure. All agents are initiated in parallel. This
is followed by up- titration in one, two or three steps, as
required. Additional therapies are then considered as a
nal step. ARNI, angiotensin receptor- neprilysin inhibitors;
BB, beta- blocker; MRA, mineralocorticoid receptor
antagonists;SGLT2i, sodium- glucose co- transporter
2inhibitors.
Open Heart
4Straw S, etal. Open Heart 2021;8:e001585. doi:10.1136/openhrt-2021-001585
7 Greene SJ, Butler J, Albert NM, etal. Medical Therapy for Heart
Failure WithReduced Ejection Fraction: The CHAMP- HF Registry. J
Am Coll Cardiol 2018;72:351–66.
8 Drozd M, Relton SD, Walker AMN, etal. Association of heart failure
and its comorbidities with loss of life expectancy. Heart 2020.
doi:10.1136/heartjnl-2020-317833. [Epub ahead of print: 05 Nov
2020].
9 Colombo G, Casella R, Cazzaniga A, etal. Dapagliozin in patients
with heart failure and reduced ejection fraction. Intern Emerg Med
2020;15:515–7.
10 Packer M, Anker SD, Butler J, etal. Cardiovascular and renal
outcomes with Empagliozin in heart failure. N Engl J Med
2020;383:1413–24.
11 Yancy CW, Jessup M, Bozkurt B, etal. 2017 ACC/AHA/HFSA
focused update of the 2013 ACCF/AHA guideline for the
management of heart failure: a report of the American College of
Cardiology/American heart association Task force on clinical practice
guidelines and the heart failure Society of America. Circulation
2017;136:e137–61.
12 Koshy AO, Gallivan ER, McGinlay M, etal. Prioritizing symptom
management in the treatment of chronic heart failure. ESC Heart Fail
2020;7:2193–207.
13 Polito MV, Silverio A, Rispoli A, etal. Clinical and echocardiographic
benet of Sacubitril/Valsartan in a real- world population with HF with
reduced ejection fraction. Sci Rep 2020;10:6665.
14 Lau CW, Martens P, Lambeets S, etal. Effects of sacubitril/valsartan
on functional status and exercise capacity in real- world patients.
Acta Cardiol 2019;74:405–12.
15 Blacher M, Zimerman A, Engster PHB, etal. Revisiting heart failure
assessment based on objective measures in NYHA functional
classes I and II. Heart 2020. doi:10.1136/heartjnl-2020-317984.
[Epub ahead of print: 23 Dec 2020].
16 McMurray JJV, Packer M, Desai AS, etal. Angiotensin- neprilysin
inhibition versus enalapril in heart failure. N Engl J Med
2014;371:993–1004.
17 Haslam A, Prasad V. Conrmatory trials for drugs Approved on a
single trial. Circ Cardiovasc Qual Outcomes 2019;12:e005494.
18 Witte KK, Drozd M, Walker AMN, etal. Mortality reduction
associated with β-adrenoceptor inhibition in chronic heart failure is
greater in patients with diabetes. Diabetes Care 2018;41:136–42.
19 Straw S, McGinlay M, Relton SD, etal. Effect of disease- modifying
agents and their association with mortality in multi- morbid patients
with heart failure with reduced ejection fraction. ESC Heart Fail
2020;7:3859–70.
20 Bristow MR, Gilbert EM, Abraham WT, etal. Carvedilol produces
dose- related improvements in left ventricular function and survival in
subjects with chronic heart failure. MOCHA Investigators. Circulation
1996;94:2807–16.
21 Packer M, Poole- Wilson PA, Armstrong PW, ATLAS study group.
Comparative effects of low and high doses of the angiotensin-
converting enzyme inhibitor, lisinopril, on morbidity and mortality in
chronic heart failure. Circulation 1999;100:2312–8.
22 Vardeny O, Claggett B, Packer M, etal. Efcacy of sacubitril/
valsartan vs. enalapril at lower than target doses in heart failure with
reduced ejection fraction: the PARADIGM- HF trial. Eur J Heart Fail
2016;18:1228–34.
23 Khariton Y, Fonarow GC, Arnold SV, etal. Association Between
Sacubitril/Valsartan Initiation and Health Status Outcomes
inHeartFailure With Reduced EjectionFraction. JACC Heart Fail
2019;7:933–41.
24 McMurray JJV, Solomon SD, Inzucchi SE, etal. Dapagliozin in
patients with heart failure and reduced ejection fraction. N Engl J
Med 2019;381:1995–2008.
25 Sharma A, Zhao X, Hammill BG, etal. Trends in Noncardiovascular
comorbidities among patients hospitalized for heart failure: insights
from the get with the Guidelines- Heart failure registry. Circ Heart Fail
2018;11:e004646.
26 Vaduganathan M, Claggett BL, Jhund PS, etal. Estimating lifetime
benets of comprehensive disease- modifying pharmacological
therapies in patients with heart failure with reduced ejection fraction:
a comparative analysis of three randomised controlled trials. Lancet
2020;396:121–8.
27 Bhatt DL, Szarek M, Steg PG, etal. Sotagliozin in patients
with diabetes and recent worsening heart failure. N Engl J Med
2021;384:117-128:117–28.
28 Wachter R, Senni M, Belohlavek J, etal. Initiation of sacubitril/
valsartan in haemodynamically stabilised heart failure patients in
hospital or early after discharge: primary results of the randomised
TRANSITION study. Eur J Heart Fail 2019;21:998–1007.
29 McMurray JJV, Packer M. How should we sequence the treatments
for heart failure and a reduced ejection fraction? A redenition of
evidence- based medicine. Circulation 2020;100.
30 National Institute of Health and Care Excelence. Dapagliozin for
treating chronic heart failure with reduced ejection fraction, 2020.
Available: https://www. nice. org. uk/ guidance/ gid- ta10560/ documents/
nal- appraisal- determination- document [Accessed 08 Feb 2021].
... The 2021 European Society of Cardiology (ESC) guidelines for HFrEF emphasize four key pharmacological therapies, often referred to as the "four pillars" of heart failure treatment: angiotensin receptor-neprilysin inhibitors (ARNIs), sodium-glucose cotransporter-2 inhibitors (SGLT2is), beta-blockers, and mineralocorticoid receptor antagonists (MRAs) [116]. ...
... Beta-blockers such as bisoprolol, carvedilol, and metoprolol succinate are essential in heart failure management, as they reduce myocardial oxygen demand, control heart rate, and improve survival rates. The European Society of Cardiology recommends their use as a key strategy in improving long-term outcomes in HFrEF [116]. ...
... MRAs, including spironolactone and eplerenone, antagonize aldosterone, reducing sodium retention, myocardial fibrosis, and endothelial dysfunction. The EMPHASIS-HF trial confirmed their critical role in improving survival, demonstrating that eplerenone significantly reduced mortality and hospitalizations in mildly symptomatic HFrEF patients [114,116]. ...
Article
Full-text available
Congestion is a key clinical feature of heart failure (HF), contributing to hospitalizations, disease progression, and poor outcomes. While traditionally considered a hemodynamic issue, congestion is now recognized as a systemic process affecting multiple organs. Renal dysfunction arises from impaired perfusion and sodium retention, leading to maladaptive left ventricular remodeling. Hepatic congestion contributes to cholestatic liver injury, while metabolic disturbances drive anemia, muscle wasting, and systemic inflammation. Additionally, congestion disrupts the intestinal barrier and immune function, exacerbating HF progression. Given its widespread impact, effective congestion management requires a shift from a cardiovascular-centered approach to a comprehensive, multidisciplinary strategy. Targeted decongestive therapy, metabolic and nutritional optimization, and immune modulation are crucial in mitigating congestion-related organ dysfunction. Early recognition and intervention are essential to slow disease progression, preserve functional capacity, and improve survival. Addressing HF congestion through personalized, evidence-based strategies is vital for optimizing long-term care and advancing treatment paradigms.
... For instance, Simmonds et al. (2020) and Schwinger (2020) have explored the cellular and molecular differences between both phenotypes, while Haydock and Flett (2022) and Murphy et al. (2020) emphasized the clinical diagnosis and prognostic implications of HFrEF. In terms of therapy, Docherty et al. (2022), Straw et al. (2021), and Tromp et al. (2022) presented strong evidence regarding the effectiveness of the four foundational drugs-ARNIs, beta-blockers, MRAs, and SGLT2 inhibitors-in improving outcomes for HFrEF. However, evidence for HFpEF remains inconsistent and limited. ...
... These four medications are recommended for early implementation in heart failure management to reduce mortality, prevent hospitalizations, and potentially support cardiac structural recovery. Drug selection may be influenced by patient comorbidities and financial constraints, but their use should be encouraged whenever possible to achieve better heart failure management outcomesg (Docherty et al., 2022;Straw et al., 2021;Tromp et al., 2022). ...
Article
Heart failure (HF) remains a major clinical challenge, with two main phenotypes: heart failure with reduced ejection fraction (HFrEF) and heart failure with preserved ejection fraction (HFpEF). These conditions exhibit distinct pathophysiology, clinical manifestations, and therapeutic responses, requiring distinct management approaches. The four pillars of HFrEF therapy—renin-angiotensin system (RAS) inhibitors, β-blockers, mineralocorticoid receptor antagonists (MRAs), and sodium-glucose cotransporter-2 (SGLT2) inhibitors—have been shown to reduce mortality and morbidity. In contrast, HFpEF management focuses more on risk factor control and symptom-based therapy, with SGLT2 inhibitors being the only treatment that has shown significant clinical benefit. This literature review aims to evaluate the different roles of the four pillars of therapy in both phenotypes of heart failure and their implications for clinical practice. Although HFrEF treatment has made significant progress with strong clinical trial evidence, HFpEF management still requires further exploration to identify more effective strategies. Therefore, a deeper understanding of each phenotype's pathophysiology and therapeutic response is essential to improve patient outcomes and optimize heart failure management.
... It has been reported that the prognosis of patients with heart failure improves when the left ventricular ejection fraction (LVEF) is improved [1]. Now that evidence is available for angiotensin receptor/neprilysin inhibitors and sodium glucose cotransporter-2 inhibitors (SGLT-2 inhibitors), reports of quadruple medical therapy (also called the Four Pillars [2] or Fantastic Four) have been increasing [3], and multidrug therapy for heart failure has become essential and standard. ...
... In addition, non-HFpEF patients who were undergoing dialysis were not included in this study ( Figure 1). [2] or Fantastic Four) have been increasing [3], and multidrug therapy for heart failure has become essential and standard. ...
Article
Full-text available
Background/Objectives: Vericiguat has been shown to reduce cardiovascular mortality and hospitalisation for heart failure in patients with reduced ejection fraction. While Vericiguat is considered one of the standard treatments for heart failure, it is unclear under which conditions Vericiguat would be most effective. With a focus on the prognosis and improved EF of heart failure, we aimed to investigate in which cases Vericiguat is suitable for use in addition to standard cardioprotective drugs. Methods: We prospectively compared echocardiograms taken before and after the administration of Vericiguat in 46 patients with non-dialysis and without heart failure with preserved ejection fraction (non-HFpEF) (left ventricle ejection fraction [LVEF] < 50%) who were able to continue Vericiguat in addition to other standard heart failure drugs (the “Fantastic Four”) for more than 6 months at our hospital. Patients who showed an improvement of 10 points or more in LVEF were defined as improved EF+. Results: LVEF improved significantly from 38 [33–45]% at the time of administration to 46 [35–54.5]% at 6 months (p < 0.001). When comparing patients with and without improved EF, a significant difference was observed in the Hb (OR = 1.66, 95%CI = 1.12–2.83, p = 0.028), early introduction (OR = 12.5, 95%CI = 1.58–149, p = 0.025), and initiation of Vericiguat after the administration of the Fantastic Four (OR = 9.79, 95%CI = 1.71–100.2, p = 0.022). Conclusions: In this study, the early administration of Vericiguat, haemoglobin value, and initiation of Vericiguat after the introduction of the Fantastic Four were identified as independent factors for eligibility in non-dialysis, non-HFpEF patients who were able to continue GDMT treatment for more than 6 months after adding Vericiguat.
... The National Institute for Health and Care Excellence (NICE) guideline NG106 recommends N-terminal pro-B-type natriuretic peptide (NT-proBNP) testing to evaluate suspected HF [73]. For patients with HFrEF, current guidelines advocate quadruple therapy, which includes an RAAS inhibitor (or angiotensin receptor-neprilysin inhibitor), a β-blocker, an MRA and an SGLT2i-collectively termed the "four pillars" of HFrEF management [74,75]. Early initiation of quadruple therapy after discharge, ideally within 30 days, aligns with recent expert consensus recommending rapid sequencing of the four foundational treatments for HFrEF to maximise prognostic benefits [74,76]. ...
Article
Full-text available
Cardiovascular, kidney and metabolic (CKM) conditions are interrelated, significantly contributing to morbidity, mortality and healthcare burden. Despite therapeutic advances, traditional disease-specific approaches often fail to address their complex interplay. Key therapeutic agents—including glucagon-like peptide-1 receptor agonists (GLP-1 RAs), dual GLP-1/glucose-dependent insulinotropic polypeptide RAs, sodium glucose co-transporter inhibitors and the nonsteroidal mineralocorticoid receptor antagonist (MRA) finerenone—offer multi-organ benefits. Emerging therapies, such as triple receptor agonists and second-generation MRAs, target new pathways further expanding treatment options for CKM conditions. A holistic CKM management approach must address and recognise that conditions such as metabolic dysfunction-associated steatotic liver disease, metabolic dysfunction-associated steatohepatitis, obstructive sleep apnoea and obesity are part of the CKM spectrum. Frailty assessment is also important alongside CKM conditions, warranting comprehensive geriatric assessment and deprescribing when appropriate. Multidisciplinary care—including lifestyle interventions, pathway redesign, pharmacological advances and novel technologies—is essential for improving outcomes. As the CKM landscape evolves, future strategies should prioritise early intervention, personalised treatment and addressing unmet needs in high-risk populations. This review advocates for an integrated CKM framework, exploring treatment strategies, emerging therapies and technological innovations. It also examines the role of artificial intelligence and digital health tools in risk stratification, early diagnosis and long-term condition management, alongside ethical and regulatory considerations.
... Research should also focus on the drugs' ability to modulate FD-specific pathways in vivo, alongside the development of novel FD-specific biomarkers to monitor treatment efficacy and enable personalized therapy. Advancing research in this area will expand treatment options available to patients with FD, akin to the 'four-pillar' approach in heart failure [167], cumulative benefits of these widely used drugs may transform care for this complex, multi-system disorder. ...
Article
Full-text available
Fabry disease (FD) is an X-linked lysosomal storage disorder characterized by deficiency of α-galactosidase A (α-GalA), leading to the accumulation of glycosphingolipids and multi-organ dysfunction, particularly affecting the cardiovascular and renal systems. Disease-modifying treatments such as enzyme replacement therapy (ERT) and oral chaperone therapy (OCT) have limited efficacy, particularly in advanced disease, prompting a need for innovative therapeutic approaches targeting underlying molecular mechanisms beyond glycosphingolipid storage alone. Recent insights into the pathophysiology of FD highlights chronic inflammation and mitochondrial, lysosomal, and endothelial dysfunction as key mediators of disease progression. Adjunctive therapies such as sodium-glucose cotransporter-2 (SGLT2) inhibitors, glucagon-like peptide-1 (GLP-1) agonists, and mineralocorticoid receptor antagonists (MRAs) demonstrate significant cardiovascular and renal benefits in conditions including heart failure and chronic kidney disease. These drugs also modulate pathways involved in the pathophysiology of FD, such as autophagy, oxidative stress, and pro-inflammatory cytokine signaling. While theoretical foundations support their utility, dedicated trials are necessary to confirm efficacy in the FD-specific population. This narrative review highlights the importance of expanding therapeutic strategies in FD, advocating for a multi-faceted approach involving evidence-based adjunctive treatments to improve outcomes. Tailored research focusing on diverse FD phenotypes, including females and non-classical variants of disease, will be critical to advancing care and improving outcomes in this complex disorder.
... The primary objective in the new-onset and acute phase of HF is to relieve congestion and manage fluid overload with diuretics. For long-term management of HFrEF, key in vivo 39: 548-558 (2025) medications including MRAs, sodium-glucose cotransporter 2 (SGLT2) inhibitors, beta-blockers and ARNIs are recommended to optimise outcomes in most patients to reduce mortality and hospitalisation (38)(39)(40). These medications should be titrated to the maximum tolerated or recommended doses. ...
Article
Full-text available
Background/Aim Atrial fibrillation (AF) and heart failure (HF) commonly co-occur, significantly increasing morbidity and mortality. Poorly controlled AF can contribute to complications like HF and is associated with conditions, such as stroke and pulmonary embolism (PE). This report involves a man with AF who had persistent respiratory symptoms and left-sided chest pain, initially suspected to be PE, but eventually diagnosed as HF. Case Report A 43-year-old male experienced increasing breathlessness, cough, and fatigue. Initially suspected to have a respiratory infection, his persistent symptoms raised concern for PE. The patient had a history of AF, unsuccessful cardioversion, and long-term non-adherence to beta blockers. Initial assessment revealed persistent respiratory symptoms and elevated levels of C-reactive protein, D-dimer, N-terminal pro-B-type natriuretic peptide, and Troponin T. Chest X-ray showed pulmonary congestion, and echocardiogram confirmed a severely impaired ejection fraction (EF <20%). While the differential diagnosis included community-acquired pneumonia, PE, and HF, the final diagnosis was worsening AF and HF with reduced EF, not PE. Conclusion PE symptoms can overlap with HF, making careful differential diagnosis essential, particularly in AF patients with elevated D-dimer levels, where false positives necessitate caution. This case underscores the importance of thorough differential diagnosis and clinical judgment before ordering tests to avoid misdiagnosis. Long-term non-adherence to beta blockers exacerbated the patient’s symptoms, emphasising the critical role of consistent medication use in managing AF and preventing complications like HF. This case report also highlights the importance of thorough investigations, guideline-based treatments and multidisciplinary care in complex AF-HF cases.
Article
Purpose To review the key updates in the 2024 KDIGO clinical practice guideline for the evaluation and management of chronic kidney disease (CKD) and highlight the essential role of pharmacists in implementing these recommendations. Summary The updated guideline introduces significant changes in CKD management, including the use of validated equations for estimating glomerular filtration rate (GFR) for drug dosing, with incorporation of serum cystatin C into GFR estimates for specific patient populations, and an emphasis on a comprehensive approach to delay disease progression. The guideline recommends sodium-glucose cotransporter 2 inhibitor (SGLT2i) therapy for kidney disease with proteinuria, with or without diabetes, renin-angiotensin-aldosterone system inhibitors (RAASi) blood pressure control and proteinuria management, and statins to reduce the risk of atherosclerotic cardiovascular disease. New evidence supports the use of finerenone in patients with type 2 diabetes and CKD, and GLP-1 receptor agonists for their kidney-protective effects. The guidelines also emphasize the importance of nephrotoxin stewardship and prevention of acute kidney injury through patient education on sick day medication management. Conclusion Pharmacists play a crucial role in implementing these updated guidelines through comprehensive medication management, nephrotoxin stewardship, drug dosing adjustments, and patient education. Their involvement in interprofessional care teams is essential for optimizing health outcomes in patients with CKD.
Article
Aims Heart failure (HF) following acute myocardial infarction (AMI) is a global health concern, but data on risk factors associated with HF hospitalization post-AMI are limited. Methods and results We analysed data from the Myocardial Ischaemia National Audit Project, including patients admitted with AMI from 1 January 2006 to 31 March 2019. Data linkage with Hospital Episode Statistics Admitted Patient Care and the Office for National Statistics facilitated a longitudinal analysis. High-risk patients were identified using dapagliflozin in patients without diabetes mellitus with acute myocardial infarction (DAPA-MI) and EMPAgliflozin on Hospitalization for Heart Failure and Mortality in Patients With aCuTe Myocardial Infarction (EMPACT-MI) criteria. We assessed clinical outcomes, adherence to European Society of Cardiology quality indicators, and predictors of HF-related hospitalizations. Out of 1 046 480 AMI patients, 9.1% overall, 17.2% in the DAPA-MI cohort, and 16.6% in the EMPACT-MI cohort experienced HF hospitalization within a year post-AMI. High-risk patients, defined by the presence of five risk factors, had nearly one in four hospitalizations with HF at 1-year follow-up. The predicted adjusted incidence rate for heart failure within 1 year almost doubled from 64.5 cases per 1000 person-years [95% confidence interval (CI): 51.1 to 78.0] in 2005, to 118.2 cases per 1000 person-years in 2019 (95% CI: 115.0 to 121.5). Heart failure hospitalization was associated with a three-fold increase in 1-year mortality (hazard ratio 3.01, 95% CI 2.95–3.13). Conclusion One in 10 AMI patients experienced HF hospitalization within the first-year post-AMI, with rising trends in high-risk subgroups. These findings highlight the need for targeted post-AMI care strategies to improve outcomes and address the increasing burden of HF in the modern percutaneous coronary intervention era.
Article
Full-text available
In patients with acute myocardial infarction (MI), heart failure (HF) is one of the most common complications that is associated with a significant burden of mortality and healthcare resources. The clinical benefits of key HF drugs, the so-called “4 pillars” or “fantastic 4”, namely β-blockers, mineralocorticoid receptor antagonists, angiotensin receptor-neprilysin inhibitor, and sodium-glucose cotransporter 2 inhibitors, have been established in patients with HF with reduced ejection fraction, whereas the effects of these drugs are not comprehensively appreciated in patients with acute MI. This review summarizes current evidence on pharmacological and device-based interventions for preventing HF after acute MI.
Article
Full-text available
Objective: Estimating survival can aid care planning, but the use of absolute survival projections can be challenging for patients and clinicians to contextualise. We aimed to define how heart failure and its major comorbidities contribute to loss of actuarially predicted life expectancy. Methods: We conducted an observational cohort study of 1794 adults with stable chronic heart failure and reduced left ventricular ejection fraction, recruited from cardiology outpatient departments of four UK hospitals. Data from an 11-year maximum (5-year median) follow-up period (999 deaths) were used to define how heart failure and its major comorbidities impact on survival, relative to an age-sex matched control UK population, using a relative survival framework. Results: After 10 years, mortality in the reference control population was 29%. In people with heart failure, this increased by an additional 37% (95% CI 34% to 40%), equating to an additional 2.2 years of lost life or a 2.4-fold (2.2-2.5) excess loss of life. This excess was greater in men than women (2.4 years (2.2-2.7) vs 1.6 years (1.2-2.0); p<0.001). In patients without major comorbidity, men still experienced excess loss of life, while women experienced less and were non-significantly different from the reference population (1 year (0.6-1.5) vs 0.4 years (-0.3 to 1); p<0.001). Accrual of comorbidity was associated with substantial increases in excess lost life, particularly for diabetes, chronic kidney and lung disease. Conclusions: Comorbidity accounts for the majority of lost life expectancy in people with heart failure. Women, but not men, without comorbidity experience survival close to reference controls. Keywords: heart failure; heart failure with reduced ejection fraction.
Article
Full-text available
Aims An increasing proportion of patients with heart failure with reduced ejection fraction (HFrEF) have co‐morbidities. The effect of these co‐morbidities on modes of death and the effect of disease‐modifying agents in multi‐morbid patients is unknown. Methods and results We performed a prospective cohort study of ambulatory patients with HFrEF to assess predictors of outcomes. We identified four key co‐morbidities—ischaemic aetiology of heart failure, diabetes mellitus, chronic obstructive pulmonary disease (COPD), and chronic kidney disease (CKD)—that were highly prevalent and associated with an increased risk of all‐cause mortality. We used these data to explore modes of death and the utilization of disease‐modifying agents in patients with and without these co‐morbidities. The cohort included 1789 consecutively recruited patients who had an average age of 69.6 ± 12.5 years, and 1307 (73%) were male. Ischaemic aetiology of heart failure was the most common co‐morbidity, occurring in 1061 (59%) patients; 503 (28%) patients had diabetes mellitus, 283 (16%) had COPD, and 140 (8%) had CKD stage IV/V. During mean follow‐up of 3.8 ± 1.6 years, 737 (41.5%) patients died, classified as progressive heart failure (n = 227, 32%), sudden (n = 112, 16%), and non‐cardiovascular deaths (n = 314, 44%). Multi‐morbid patients were older (P < 0.001), more likely to be male (P < 0.001), and had higher New York Heart Association class (P < 0.001), despite having higher left ventricular (LV) ejection fraction (P = 0.001) and lower LV end‐diastolic diameter (P = 0.001). Multi‐morbid patients were prescribed lower doses of disease‐modifying agents, especially patients with COPD who received lower doses of beta‐adrenoceptor antagonists (2.7 ± 3.0 vs. 4.1 ± 3.4 mg, P < 0.001) and were less likely to be implanted with internal cardioverter defibrillators (7% vs. 13%, P < 0.001). In multivariate analysis, COPD and diabetes mellitus conferred a >2.5‐fold and 1.5‐fold increased risk of sudden death, whilst higher doses of beta‐adrenoceptor antagonists were protective (hazard ratio per milligram 0.92, 95% confidence interval 0.86–0.98, P = 0.009). Each milligram of bisoprolol‐equivalent beta‐adrenoceptor antagonist was associated with 9% (P = 0.001) and 11% (P = 0.023) reduction of sudden deaths in patients with <2 and ≥2 co‐morbidities, respectively. Conclusions Higher doses of beta‐adrenoceptor antagonist are associated with greater protection from sudden death, most evident in multi‐morbid patients. Patients with COPD who appear to be at the highest risk of sudden death are prescribed the lowest doses and less likely to be implanted with implantable cardioverter defibrillators, which might represent a missed opportunity to optimize safe and proven therapies for these patients.
Article
Full-text available
Chronic heart failure (CHF) is a chronic, progressive disease that has detrimental consequences on a patient's quality of life (QoL). In part due to requirements for market access and licensing, the assessment of current and future treatments focuses on reducing mortality and hospitalizations. Few drugs are available principally for their symptomatic effect despite the fact that most patients' symptoms persist or worsen over time and an acceptance that the survival gains of modern therapies are mitigated by poorly controlled symptoms. Additional contributors to the failure to focus on symptoms could be the result of under‐reporting of symptoms by patients and carers and a reliance on insensitive symptomatic categories in which patients frequently remain despite additional therapies. Hence, formal symptom assessment tools, such as questionnaires, can be useful prompts to encourage more fidelity and reproducibility in the assessment of symptoms. This scoping review explores for the first time the assessment options and management of common symptoms in CHF with a focus on patient‐reported outcome tools. The integration of patient‐reported outcomes for symptom assessment into the routine of a CHF clinic could improve the monitoring of disease progression and QoL, especially following changes in treatment or intervention with a targeted symptom approach expected to improve QoL and patient outcomes.
Article
Full-text available
The aim of this study was to evaluate the effects of Sacubitril/Valsartan (S/V) on clinical, laboratory and echocardiographic parameters and outcomes in a real-world population with heart failure with reduced ejection fraction (HFrEF). This was a prospective observational study enrolling patients with HFrEF undergoing treatment with S/V. The primary outcome was the composite of cardiac death and HF rehospitalization at 12 months follow-up; secondary outcomes were all-cause death, cardiac death and the occurrence of rehospitalization for worsening HF. The clinical outcome was compared with a retrospective cohort of 90 HFrEF patients treated with standard medical therapy. The study included 90 patients (66.1 ± 11.7 years) treated with S/V. The adjusted regression analysis showed a significantly lower risk for the primary outcome (HR:0.31; 95%CI, 0.11–0.83; p = 0.019) and for HF rehospitalization (HR:0.27; 95%CI, 0.08–0.94; p = 0.039) in S/V patients as compared to the control group. A significant improvement in NYHA class, left ventricular ejection fraction, left ventricular end systolic volume and systolic pulmonary arterial pressure was observed up to 6 months. S/V did not affect negatively renal function and was associated with a significantly lower dose of furosemide dose prescribed at 6- and 12-month follow-up. In this study, S/V reduced the risk of HF rehospitalization and cardiac death at 1 year in patients with HFrEF. S/V improved NYHA class, echocardiographic parameters and need of furosemide, and preserved renal function.
Article
Objective New York Heart Association (NYHA) functional class plays a central role in heart failure (HF) assessment but might be unreliable in mild presentations. We compared objective measures of HF functional evaluation between patients classified as NYHA I and II in the Re de B rasileira de Estudos em I nsuficiência C ardíaca (ReBIC)-1 Trial. Methods The ReBIC-1 Trial included outpatients with stable HF with reduced ejection fraction. All patients had simultaneous protocol-defined assessment of NYHA class, 6 min walk test (6MWT), N-terminal pro-brain natriuretic peptide (NT-proBNP) levels and patient’s self-perception of dyspnoea using a Visual Analogue Scale (VAS, range 0–100). Results Of 188 included patients with HF, 122 (65%) were classified as NYHA I and 66 (35%) as NYHA II at baseline. Although NYHA class I patients had lower dyspnoea VAS Scores (median 16 (IQR, 4–30) for class I vs 27.5 (11–49) for class II, p=0.001), overlap between classes was substantial (density overlap=60%). A similar profile was observed for NT-proBNP levels (620 pg/mL (248–1333) vs 778 (421–1737), p=0.015; overlap=78%) and for 6MWT distance (400 m (330–466) vs 351 m (286–408), p=0.028; overlap=64%). Among NYHA class I patients, 19%–34% had one marker of HF severity (VAS Score >30 points, 6MWT <300 m or NT-proBNP levels >1000 pg/mL) and 6%–10% had two of them. Temporal change in functional class was not accompanied by variation on dyspnoea VAS (p=0.14). Conclusions Most patients classified as NYHA classes I and II had similar self-perception of their limitation, objective physical capabilities and levels of natriuretic peptides. These results suggest the NYHA classification poorly discriminates patients with mild HF.
Article
Background Sodium–glucose cotransporter 2 (SGLT2) inhibitors reduce the risk of hospitalization for heart failure or death from cardiovascular causes among patients with stable heart failure. However, the safety and efficacy of SGLT2 inhibitors when initiated soon after an episode of decompensated heart failure are unknown. Methods We performed a multicenter, double-blind trial in which patients with type 2 diabetes mellitus who were recently hospitalized for worsening heart failure were randomly assigned to receive sotagliflozin or placebo. The primary end point was the total number of deaths from cardiovascular causes and hospitalizations and urgent visits for heart failure (first and subsequent events). The trial ended early because of loss of funding from the sponsor. Results A total of 1222 patients underwent randomization (608 to the sotagliflozin group and 614 to the placebo group) and were followed for a median of 9.0 months; the first dose of sotagliflozin or placebo was administered before discharge in 48.8% and a median of 2 days after discharge in 51.2%. Among these patients, 600 primary end-point events occurred (245 in the sotagliflozin group and 355 in the placebo group). The rate (the number of events per 100 patient-years) of primary end-point events was lower in the sotagliflozin group than in the placebo group (51.0 vs. 76.3; hazard ratio, 0.67; 95% confidence interval [CI], 0.52 to 0.85; P<0.001). The rate of death from cardiovascular causes was 10.6 in the sotagliflozin group and 12.5 in the placebo group (hazard ratio, 0.84; 95% CI, 0.58 to 1.22); the rate of death from any cause was 13.5 in the sotagliflozin group and 16.3 in the placebo group (hazard ratio, 0.82; 95% CI, 0.59 to 1.14). Diarrhea was more common with sotagliflozin than with placebo (6.1% vs. 3.4%), as was severe hypoglycemia (1.5% vs. 0.3%). The percentage of patients with hypotension was similar in the sotagliflozin group and the placebo group (6.0% and 4.6%, respectively), as was the percentage with acute kidney injury (4.1% and 4.4%, respectively). The benefits of sotagliflozin were consistent in the prespecified subgroups of patients stratified according to the timing of the first dose. Conclusions In patients with diabetes and recent worsening heart failure, sotagliflozin therapy, initiated before or shortly after discharge, resulted in a significantly lower total number of deaths from cardiovascular causes and hospitalizations and urgent visits for heart failure than placebo. (Funded by Sanofi and Lexicon Pharmaceuticals; SOLOIST-WHF ClinicalTrials.gov number, NCT03521934.)
Article
Background: Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce the risk of hospitalization for heart failure in patients regardless of the presence or absence of diabetes. More evidence is needed regarding the effects of these drugs in patients across the broad spectrum of heart failure, including those with a markedly reduced ejection fraction. Methods: In this double-blind trial, we randomly assigned 3730 patients with class II, III, or IV heart failure and an ejection fraction of 40% or less to receive empagliflozin (10 mg once daily) or placebo, in addition to recommended therapy. The primary outcome was a composite of cardiovascular death or hospitalization for worsening heart failure. Results: During a median of 16 months, a primary outcome event occurred in 361 of 1863 patients (19.4%) in the empagliflozin group and in 462 of 1867 patients (24.7%) in the placebo group (hazard ratio for cardiovascular death or hospitalization for heart failure, 0.75; 95% confidence interval [CI], 0.65 to 0.86; P<0.001). The effect of empagliflozin on the primary outcome was consistent in patients regardless of the presence or absence of diabetes. The total number of hospitalizations for heart failure was lower in the empagliflozin group than in the placebo group (hazard ratio, 0.70; 95% CI, 0.58 to 0.85; P<0.001). The annual rate of decline in the estimated glomerular filtration rate was slower in the empagliflozin group than in the placebo group (-0.55 vs. -2.28 ml per minute per 1.73 m2 of body-surface area per year, P<0.001), and empagliflozin-treated patients had a lower risk of serious renal outcomes. Uncomplicated genital tract infection was reported more frequently with empagliflozin. Conclusions: Among patients receiving recommended therapy for heart failure, those in the empagliflozin group had a lower risk of cardiovascular death or hospitalization for heart failure than those in the placebo group, regardless of the presence or absence of diabetes. (Funded by Boehringer Ingelheim and Eli Lilly; EMPEROR-Reduced ClinicalTrials.gov number, NCT03057977.).
Article
Background Three drug classes (mineralocorticoid receptor antagonists [MRAs], angiotensin receptor–neprilysin inhibitors [ARNIs], and sodium/glucose cotransporter 2 [SGLT2] inhibitors) reduce mortality in patients with heart failure with reduced ejection fraction (HFrEF) beyond conventional therapy consisting of angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs) and β blockers. Each class was previously studied with different background therapies and the expected treatment benefits with their combined use are not known. Here, we used data from three previously reported randomised controlled trials to estimate lifetime gains in event-free survival and overall survival with comprehensive therapy versus conventional therapy in patients with chronic HFrEF. Methods In this cross-trial analysis, we estimated treatment effects of comprehensive disease-modifying pharmacological therapy (ARNI, β blocker, MRA, and SGLT2 inhibitor) versus conventional therapy (ACE inhibitor or ARB and β blocker) in patients with chronic HFrEF by making indirect comparisons of three pivotal trials, EMPHASIS-HF (n=2737), PARADIGM-HF (n=8399), and DAPA-HF (n=4744). Our primary endpoint was a composite of cardiovascular death or first hospital admission for heart failure; we also assessed these endpoints individually and assessed all-cause mortality. Assuming these relative treatment effects are consistent over time, we then projected incremental long-term gains in event-free survival and overall survival with comprehensive disease-modifying therapy in the control group of the EMPHASIS-HF trial (ACE inhibitor or ARB and β blocker). Findings The hazard ratio (HR) for the imputed aggregate treatment effects of comprehensive disease-modifying therapy versus conventional therapy on the primary endpoint of cardiovascular death or hospital admission for heart failure was 0·38 (95% CI 0·30–0·47). HRs were also favourable for cardiovascular death alone (HR 0·50 [95% CI 0·37–0·67]), hospital admission for heart failure alone (0·32 [0·24–0·43]), and all-cause mortality (0·53 [0·40–0·70]). Treatment with comprehensive disease-modifying pharmacological therapy was estimated to afford 2·7 additional years (for an 80-year-old) to 8·3 additional years (for a 55-year-old) free from cardiovascular death or first hospital admission for heart failure and 1·4 additional years (for an 80-year-old) to 6·3 additional years (for a 55-year-old) of survival compared with conventional therapy. Interpretation Among patients with HFrEF, the anticipated aggregate treatment effects of early comprehensive disease-modifying pharmacological therapy are substantial and support the combination use of an ARNI, β blocker, MRA, and SGLT2 inhibitor as a new therapeutic standard. Funding None.