Article

Neurohormonal Blockade in Heart Failure

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Average (ratings)
No ratings
Your rating

Abstract

A key feature of chronic heart failure (HF) is the sustained activation of endogenous neurohormonal systems in response to impaired cardiac pumping and/or filling properties. The clinical use of neurohormonal blockers has revolutionised the care of HF patients over the past three decades. Drug therapy that is active against imbalance in both the autonomic and renin–angiotensin–aldosterone systems consistently reduces morbidity and mortality in chronic HF with reduced left ventricular ejection fraction and in sinus rhythm. This article provides an assessment of the major neurohormonal systems and their therapeutic blockade in patients with chronic HF.

Disclosure:TvL has consulted for Novartis, St Jude Medical and Vifor Pharma, and is a member of the Beta-blockers in Heart Failure (BB-meta-HF) Collaborative Group. DK has received grants from Menarini, non-financial support from Daiichi Sankyo and personal fees from AtriCure, outside the submitted work; and is Chief Investigator for the RAte control Therapy Evaluation in Atrial Fibrillation (RATE-AF) Trial and Steering Committee Lead for the BB-meta-HF Collaborative Group. DA has received speaker honoraria from Novartis, MSD and Vifor Pharma. IH has no conflicts of interest to declare.

Received:

Accepted:

Acknowledgements:Supported by National Health Medical Research Council of Australia Program Grant ID 546272. DK is funded by a National Institute for Health Research (NIHR) Career Development Fellowship (CDF-2015-08-074). The opinions expressed are those of the authors and do not represent the NIHR or the UK Department of Health.

Correspondence Details:Dr Thomas G von Lueder, Department of Cardiology, Oslo University Hospital Ullevål, 0407 Oslo, Norway. E: tomvonoslo@yahoo.com

Copyright Statement:

The copyright in this work belongs to Radcliffe Medical Media. Only articles clearly marked with the CC BY-NC logo are published with the Creative Commons by Attribution Licence. The CC BY-NC option was not available for Radcliffe journals before 1 January 2019. Articles marked ‘Open Access’ but not marked ‘CC BY-NC’ are made freely accessible at the time of publication but are subject to standard copyright law regarding reproduction and distribution. Permission is required for reuse of this content.

Heart Failure (HF) constitutes a major global health problem, evidenced by substantial morbidity and mortality, requiring enormous healthcare-related expenditure. HF is associated with high symptomatic burden, and with a relentless and progressive clinical course towards end-stage disease. A large body of epidemiological data suggests that the prognosis in HF is as poor as in advanced cancer.1 Survival after first hospitalisation for HF is very poor, and less than 50 % of patients are alive after 5 years.2,3 By contrast, cardiac transplantation has very favorable 1- and 10-year survival rates of approximately 90 % and 50 %, respectively, but is restricted to an extremely select group of patients. Medical therapy therefore remains the treatment of choice for most patients with HF. HF is divided clinically according to left ventricular ejection fraction (LVEF) into reduced (<40 %), preserved (>50 %) and the newlyintroduced category of intermediate or “midrange” ejection fraction (40–49 %).4

A key feature of chronic HF is the sustained activation of endogenous neurohormonal systems in response to impaired cardiac pumping and/or filling properties. It is widely believed that neurohormonal systems are essential survival and “injury response” mechanisms that have evolved over thousands of years in order to cope with hostile environments and variable climates.5,6 Neurohormonal systems provide survival benefits through actions such as water and salt conservation or vasoconstriction (for example minimising the impact of haemorrhage). In addition, many neurohormonal systems are essential for normal embryonic development.7,8

While these neurohormonal systems may have compensatory haemodynamic effects in the initial stages of HF, chronic stimulation and dysregulation occurs that exerts profound deleterious actions on a broad range of cardiovascular (CV) tissues. When LVEF is in the midrange or preserved categories, guidelines require additional evidence of elevated natriuretic peptide levels for a diagnosis of HF.4

Based on the above considerations, and following scrutiny of randomised clinical trials (RCTs), pharmacological agents that counteract adverse neurohormonal actions have been introduced into clinical practice over the past three decades. The sympathetic nervous system (SNS) and the renin–angiotensin–aldosterone system (RAAS) are major neurohormonal systems that exert potentially maladaptive actions in HF.9 In patients with HF with reduced ejection fraction (HFrEF) in sinus rhythm, pharmacological blockade of these systems has been shown to markedly reduce mortality and morbidity (see Table 1).4,10–15 As yet, no medical therapy has been shown to improve the prognosis of patients with HF with preserved ejection fraction (HFpEF), despite evidence that both systolic and diastolic dysfunction affect the sympatho–vagal balance.16

This article provides an assessment of the major neurohormonal systems and their therapeutic blockade in patients with chronic HF.

The Sympathetic Nervous System and Pharmacological Blockade

Activation of the SNS increases stroke volume and induces peripheral vasoconstriction in order to maintain arterial perfusion pressure.

Table 1. Important Neurohormonal Systems and their Blockade in Heart Failure

Article image

The interface between the sympathetic nerve fibres and the CV system is formed by the adrenergic receptors. In HF, sustained sympathetic stimulation through elevated catecholamine levels (noradrenaline and others) leads to reductions in cardiac beta-1- adrenergic receptor density and function over time, contributing to disease progression.17–20 Initially thought to be contraindicated in HF, beta-adrenergic receptor antagonists (beta-blockers) represent a cornerstone of the current medical management of HF based on a well-documented reduction in clinical event rates.4,21,22

The beneficial actions of beta-blockers are believed to occur through mechanisms including reduced heart rate and myocardial oxygen demand, to reduce the incidence of arrhythmia and sudden cardiac death, and to provide protection from ischaemia. These adaptations to the pathophysiology of HF and their resultant effects on autonomic and neurohormonal balance translate into tangible patient benefits: in HFrEF with sinus rhythm, beta-blockers lead to a 24 % relative reduction (4 % absolute reduction) in all-cause mortality, and a similar reduction in hospital admissions.15 The beta-blockers with proven survival benefit in HF recommended by the European Society of Cardiology and Heart Failure Association guidelines are bisoprolol, metoprolol, carvedilol and nebivolol.4 While bisoprolol and metoprolol are highly selective for the beta- 1-adrenergic receptor, carvedilol possesses broader substrate specifities, having alpha-adrenergic and proposed pleiotropic and antioxidant properties.23

Recent data suggest that the survival benefit of beta-blockers in patients with HFrEF does not extend to those with concomitant atrial fibrillation (AF).21,24 The role of the autonomic nervous system in the (patho)physiology of AF is complex and is related to the modulation of both sympathetic and parasympathetic responses.25 When AF develops in patients with HFrEF, central sympathetic activity is augmented, but the appropriate sympathetic response to exercise is diminished.26,27 These observations raise the possibility that lack of beta-blocker efficacy in AF may be related to differences in autonomic function (and, consequently, the neurohormonal axis), a likelihood supported by the observation that heart rate is associated with mortality in HFrEF with sinus rhythm, but not in HFrEF with AF.28

Blockade of other adrenergic signalling pathways, such as alphaadrenergic receptors, has been ineffective in HF. In patients with hypertension in the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) the alpha receptor-blocker doxazosin doubled the incidence of HF, although overall mortality was similar.29 Some sympatholytics, such as hydralazine and clonidine, have been used in resistant hypertension.30 In African Americans hydralazine has been reported to be of benefit.30 Other centrally-acting sympatholytics have shown signs of harm in HF.31 Non-pharmacological strategies to block the SNS in HF, such as catheter-based renal sympathetic denervation and vagal nerve stimulation, are currently undergoing evaluation in clinical trials.32–34

The Renin–Angiotensin–Aldosterone System and Pharmacological Blockade

The RAAS is a vastly complex neurohormonal system including the protagonist hormones angiotensin-II and aldosterone. Angiotensin- II and aldosterone mediate a range of maladaptive actions upon chronic activation, including renal water and sodium retention, peripheral vasoconstriction leading to hypertension, and cellular effects such as hypertrophy and fibrosis of the heart, kidney and vasculature. The first RAAS blockers were introduced in the late 1980s, with angiotensin-converting enzyme (ACE) inhibitor use being supported by a number of clinical trials in HFrEF that demonstrated substantial reductions in mortality and morbidity.35 Angiotensin receptor blockers (ARBs) are recommended only as an alternative in patients intolerant of an ACE inhibitor.4

Renin is located upstream of ACE in the pathway and constitutes a rate-limiting step in the generation of biologically-active angiotensin-II. Therapeutic inhibition of this first specific step in the cascade using direct renin inhibitors was thought to potentially offer therapeutic advantages over ACE inhibition.36 The recent Aliskiren Trial to Minimize OutcomeS in Patients with HEart failuRE (ATMOSPHERE) trial, however, showed that the addition of aliskiren to enalapril increased adverse events without providing any clinical benefit. In addition, statistical non-inferiority could not be demonstrated for monotherapy with aliskiren as compared with enalapril.37 A number of trials have investigated the potential utility of blocking RAAS at multiple levels – not only in HF but also in other CV diseases – and have failed to demonstrate a consistent benefit for dual-acting RAAS blockade.38–42 It therefore seems that adequate RAAS blockade with a single agent (i.e. the maximum tolerated dose of an ACE inhibitor) ensures adequate blockade of angiotensin-II signalling that cannot be enhanced by the addition of an ARB or a direct renin inhibitor.

Beyond angiotensin-II, the mineralocorticoid hormone aldosterone exerts potent cardiorenal fibrosis and hypertrophy and often escapes RAAS blockade with stand-alone ACE inhibition.43–45 Clinical trials have demonstrated that mineralocorticoid receptor antagonists (MRAs) can improve prognosis in addition to standard therapy with ACE inhibitors and beta-blockers.46,47 MRAs likely promote antifibrotic actions in a broad range of organs such as the heart, kidney, vasculature and lungs, all of which are affected in HF. Despite their class I indication, however, they remain markedly underutilised in daily HF practice, probably due to their real and perceived potential off-target effects on renal function and serum potassium levels.4,48,49 Following encouraging preclinical studies, non-steroidal MRAs are currently being investigated in clinical trials such as the MinerAlocorticoid Receptor antagonist Tolerability Study – Heart Failure (ARTS-HF).14,50–53 These novel compounds appear to induce less hyperkalaemia and less worsening of renal function in HF.

Dual-acting Neprilysin/RAAS Blockers: from Omapatrilat to Sacubitril/Valsartan

The natriuretic peptide (NP) system promotes natriuresis and diuresis and lowers blood pressure. In HF patients, the NP system also counteracts the RAAS and SNS, thereby attenuating the hypertrophy and fibrosis of CV and renal tissues as well as inflammation and neo-angiogenesis.54–56 Hydrolysis by the metallopeptidase neprilysin constitutes the primary breakdown mechanism of NPs; therefore pharmacological targeting of neprilysin has been proposed as a strategy to restore or augment the beneficial actions of NP.57 Singleacting neprilysin inhibitors produce essentially neutral effects in humans, perhaps due to the fact that neprilysin broadly interacts with other vasoactive peptides such as adrenomedullin, bradykinin, endothelin-1, substance P, encephalin and others.58–60 Apart from the membrane-bound fraction of neprilysin, a soluble form exists that is measurable and retains activity in the plasma of patients with HF.61 Better understanding of the molecular mechanisms underlying HF has led to the recognition that in order to exploit the benefits of neprilysin inhibition, RAAS needs to be inhibited concomitantly.62

The vasopeptidase inhibitors were the first class of drugs to inhibit both ACE and neural endopeptidase.63,64 Omapatrilat underwent extensive clinical testing in the treatment of hypertension and HF.65,66 Omapatrilat showed superior antihypertensive effects to stand-alone RAAS blockade in the large Omapatrilat Cardiovascular Treatment versus Enalapril (OCTAVE) trial (n=25,302).67 In the phase-II Inhibition of Metallo Protease by BMS-186716 in a Randomized Exercise and Symptoms Study in Subjects With Heart Failure (IMPRESS) trial, omapatrilat reduced the composite endpoint of all-cause mortality or HF hospitalisation compared to lisinopril.68 In the subsequent phase-III Omapatrilat Versus Enalapril Randomized Trial of Utility in Reducing Events (OVERTURE), however, the primary endpoint was not significantly reduced and the trial failed to meet the pre-specified superiority criterion.69 Important off-target effects, most notably a substantially higher rate of angioedema ascribed to bradykinin accumulation, halted further development of omapatrilat and other vasopeptidase inhibitors.70

A logical extension of research efforts into combined neprilysin and RAAS blockade are the angiotensin receptor neprilysin inhibitors (ARNIs).62,71,72 Utilising an ARB rather than ACE inhibitor as the RAAS blocker, ARNIs circumvent the issue of bradykinin accumulation.70 In an experimental angioedema model, vasopeptidase inhibition – but not ARB or neural endopeptidase inhibition or their combination (replicating ARNIs) – induced bradykinin-mediated tracheal plasma extravasation.73 Sacubitril/valsartan, the first-in-class ARNI, has undergone broad clinical testing in HF and hypertension.74,75 The Prospective Comparison of ARNI with ACEI to Determine Impact on Global Mortality and Morbidity in Heart Failure (PARADIGM-HF) trial evaluated sacubitril/valsartan as an alternative to enalapril in patients with HFrEF (i.e. current best therapy based on the Studies Of Left Ventricular Dysfunction (SOLVD) study).76,77 The trial was terminated prematurely due to overwhelming benefit: compared to enalapril, sacubitril/valsartan reduced the risk of the primary composite endpoint of CV mortality or hospitalisation for HF by 20 %. Sacubitril/valsartan was also superior in reducing a number of other pre-specified endpoints, such as time to clinical deterioration and 30-day readmission rates, and was more efficacious regardless of age, LVEF or the presence of AF.78–82 Experimental work suggests that sacubitril/valsartan better protects against angiotensin-II-stimulated myocardial cellular injury, hypertrophy and fibrosis than single-acting RAAS blockade.83,84 Such dual-acting neurohormonal inhibition was also recently reported to offer better renal protection compared to single RAAS blockade.85–87 Based on encouraging results from the phase II Prospective comparison of ARNI with ARB on Management Of heart failUre with preserved ejectioN fracTion (PARAMOUNT-HF) study, sacubitril/valsartan is currently being tested in Efficacy and Safety of LCZ696 Compared to Valsartan, on Morbidity and Mortality in Heart Failure Patients With Preserved Ejection Fraction (PARAGON-HF), a large clinical outcome trial scheduled to enrol 4,300 patients with HFpEF (www.clinicaltrials.gov, NCT01920711).75

Other Neurohormonal Systems with Possible Relevance to HF

Endothelin-1, the major isoform of the endothelin peptide family in the CV system, is an extremely potent vasoconstrictor with additional pro-hypertrophic, pro-fibrotic and mitogenic effects on myocardium and vasculature.88 Endothelin activation in HF disturbs salt and water homeostasis, stimulates the RAAS and SNS, mediates vasoconstriction, and directly contributes to progressive CV and renal dysfunction and remodelling in HF.89,90 Endothelin-1 plasma levels are strongly correlated with mortality and morbidity.91

Fuelled by encouraging experimental and early clinical evidence, several RCTs have explored the putative utility of blocking the endothelin system in acute and chronic HF settings.90,92–96 With the exception of some forms of pulmonary arterial hypertension, the vast majority of large RCTs of endothelin antagonism have failed to show reduced clinical event rates (see Table 1). Unfortunately, some trials (with neutral or negative outcomes presented at scientific meetings) have not been published, or only in abstract form.97–100 In HF, the only current application of endothelin antagonists seems to be to lower pulmonary vascular resistance in high-risk patients on the heart transplant list, although even this indication has been subject of debate.101–103

Among numerous other neurohormones with putative implications in HF pathophysiology are adrenomedullin, bradykinin, serotonin, and urotensin-II.104–107 Their role in HF remains incompletely understood, and no specific pharmacological modulator has advanced into clinical testing. Since several of these neurohormones are substrates of neprilysin, their metabolism could conceivably be altered by neprilysin inhibition.58–60

Remaining Challenges for Neurohormonal Blockade in HF

Concomitant blockade of multiple neurohormonal systems, built on a strong scientific foundation, is the current gold standard of pharmacotherapy in HFrEF. Current treatment recommendations are based on trials that showed clinical benefits for target doses of RAAS and SNS blockers.4 Guideline-adherent treatment is frequently not achieved in practice, however.108 There are various reasons for this, such as the lenient attitude of some caregivers (sometimes termed “therapeutic inertia”) towards patients who appear euvolemic and asymptomatic, and the real or perceived side effects of medical therapy such as hypotension, bradycardia, hyperkalaemia and worsening renal function.109

There is a considerable knowledge gap regarding neurohormonal blockade in various HF entities: renal dysfunction affects at least one in five HF patients and is a major adverse prognostic factor.110 Traditionally these patients have been excluded from RCTs, although there is accumulating evidence for the particular value of neurohormonal blockade in these patients, as discussed above. HF commonly coexists with AF and represents a clinical dilemma.111,112 In patients with HFrEF and AF, the mortality and morbidity benefits of beta-blockers for neurohormonal blockade appear to be absent,15,21 and the data for RAAS antagonists and MRAs are limited.111 Patterns of autonomic activation have not yet been sufficiently studied in patients with concomitant HF and AF, limiting our understanding of the impact of pharmacotherapy.

Some authors have argued that the therapeutic blockade of neurohormonal systems may have been exhausted, and that a ceiling may have been reached. In particular, a discrepancy between promising early-phase and frequently disappointing clinical endpoint trial results of neurohormonal blockade has been noted.113 Recent examples of neurohormonal blockers with promising scientific underpinnings that failed to lower event rates in clinical early-phase or outcome trials include endothelin receptor blockers, adenosine receptor antagonists, tumour necrosis factor antagonists and phosphodiesterase inhibitors.98,114–116 Of note, very recent insights from the PARADIGM-HF study using valsartan/sacubitril support the notion that combination therapy with neurohormonal modulators may be superior to singleacting therapy, even at subtarget doses.117 Such a strategy may better exploit the benefits of abrogating multiple specific maladaptive signalling pathways while circumventing the adverse effects of neurohormonal blocker monotherapy. For instance, renal failure frequently occurs in HF patients, and experimental as well as clinical studies have demonstrated that dual-acting RAAS blockade and neprilysin inhibition offers superior nephroprotection to single-acting therapy.85,86

Finally, no single effective therapy has been identified for patients with HFpEF,118 although this category includes a very heterogeneous population defined by an arbitrary cut-off in LVEF. The limited benefit of neurohormonal blockers in HFpEF may also be explained by older age, more advanced comorbidities and a higher likelihood of death from non-CV causes.119–121 Rates of AF are also higher in patients with HFpEF, leading to additional neurohormonal activation.111,112 In addition, a substantial proportion of patients with HFpEF show evidence of impaired or resolving systolic function.122 The recently-introduced category of HF with midrange ejection fraction has little evidence-base as yet, but will likely increase clinical awareness of these patients.

Conclusion

Sustained activation of neurohormonal systems is a hallmark feature of HF. The clinical use of neurohormonal blockers has revolutionised the care of patients over the past four decades. Drug therapy that is active against imbalance in both the autonomic and renin– angiotensin–aldosterone systems consistently reduces morbidity and mortality in chronic HF with reduced LVEF and sinus rhythm. HF is an extraordinarily complex and multi-faceted chronic syndrome, and current knowledge of the interface between the epidemiological, clinical, pathophysiological and molecular features remains limited. Initiation and up-titration of effective neurohormonal therapies remains challenging in patient subcohorts. In addition, optimal medical therapy is frequently not achieved or even attempted despite HF having a similar overall prognosis to cancer. The recent introduction of the novel ARNI drug class attests to superior efficacy of multiple-acting neurohormonal blockade in chronic HF. HFpEF and HF with coexisting AF represent major remaining clinical challenges that appear to be less susceptible to conventional pharmacotherapy. Novel neurohormonal blockers and the refined use of existing therapeutic agents, as well as up-titration to recommended target doses, are needed to reduce adverse clinical events and to improve outcomes in HF.

References

  1. Stewart S, Ekman I, Ekman T, et al. Population impact of heart failure and the most common forms of cancer: a study of 1 162 309 hospital cases in Sweden (1988 to 2004). Circ Cardiovasc Qual Outcomes 2010;3:573–80.
    Crossref | PubMed
  2. Jhund PS, Macintyre K, Simpson CR, et al. Long-term trends in first hospitalization for heart failure and subsequent survival between 1986 and 2003: a population study of 5.1 million people. Circulation 2009;119:515–23.
    Crossref | PubMed
  3. Maggioni AP, Dahlstrom U, Filippatos G, et al.; Heart Failure Association of the European Society of Cardiology. EURObservational Research Programme: regional differences and 1-year follow-up results of the Heart Failure Pilot Survey (ESC-HF Pilot). Eur J Heart Fail 2013;15:808–17.
    Crossref | PubMed
  4. Ponikowski P, Voors AA, Anker SD, et al; Authors/Task Force Members. 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.
    Crossref | PubMed
  5. Tota B, Cerra MC, Gattuso A. Catecholamines, cardiac natriuretic peptides and chromogranin A: evolution and physiopathology of a ‘whip-brake’ system of the endocrine heart. J Exp Biol 2010;213:3081–103.
    Crossref | PubMed
  6. Burnstock G. Evolution of the autonomic innervation of visceral and cardiovascular systems in vertebrates. Pharmacol Rev 1969;21:247–324.
    PubMed
  7. Davisson RL, Kim HS, Krege JH, et al. Complementation of reduced survival, hypotension, and renal abnormalities in angiotensinogen-deficient mice by the human renin and human angiotensinogen genes. J Clin Invest 1997;99:1258–64.
    Crossref | PubMed
  8. Clouthier DE, Hosoda K, Richardson JA, et al. Cranial and cardiac neural crest defects in endothelin-A receptordeficient mice. Development 1998;125:813–24.
    PubMed
  9. Krum H. New and emerging pharmacological strategies in the management of chronic heart failure. Curr Opin Pharmacol. 2001;Apr;1(2):126–33. Review.
    Crossref | PubMed
  10. Goldsmith SR. Interactions between the sympathetic nervous system and the RAAS in heart failure. Curr Heart Fail Rep 2004;1:45–50.
    Crossref | PubMed
  11. von Lueder TG, Krum H. RAAS inhibitors and cardiovascular protection in large scale trials. Cardiovasc Drugs Ther 2013;27:171–9.
    Crossref | PubMed
  12. Parati G, Esler M. The human sympathetic nervous system: its relevance in hypertension and heart failure. Eur Heart J 2012;33:1058–66.
    Crossref | PubMed
  13. Haynes WG, Webb DJ. Endothelin as a regulator of cardiovascular function in health and disease. J Hypertens 1998;16:1081–98.
    Crossref | PubMed
  14. von Lueder TG, Krum H. New medical therapies for heart failure. Nat Rev Cardiol 2015;12:730–40.
    Crossref | PubMed
  15. Kotecha D, Manzano L, Krum H, et al.; Beta-Blockers in Heart Failure Collaborative Group. Effect of age and sex on efficacy and tolerability of beta blockers in patients with heart failure with reduced ejection fraction: individual patient data meta-analysis. BMJ 2016;353:i1855.
    Crossref | PubMed
  16. Arora R, Krummerman A, Vijayaraman P, et al. Heart rate variability and diastolic heart failure. Pacing Clin Electrophysiol 2004;27:299–303.
    Crossref | PubMed
  17. Cohn JN, Levine TB, Olivari MT, et al. Plasma norepinephrine as a guide to prognosis in patients with chronic congestive heart failure. N Engl J Med 1984;311:819–23.
    Crossref | PubMed
  18. Swedberg K, Eneroth P, Kjekshus J, et al. Hormones regulating cardiovascular function in patients with severe congestive heart failure and their relation to mortality. CONSENSUS Trial Study Group. Circulation 1990;82:1730–6.
    Crossref | PubMed
  19. Kaye DM, Lefkovits J, Jennings GL, et al. Adverse consequences of high sympathetic nervous activity in the failing human heart. J Am Coll Cardiol 1995;26:1257–63.
    Crossref | PubMed
  20. Benedict CR, Johnstone DE, Weiner DH, et al. Relation of neurohumoral activation to clinical variables and degree of ventricular dysfunction: a report from the Registry of Studies of Left Ventricular Dysfunction. SOLVD Investigators. J Am Coll Cardiol 1994;23:1410–20.
    Crossref | PubMed
  21. Kotecha D, Holmes J, Krum H, et al.; Beta-Blockers in Heart Failure Collaborative Group. Efficacy of β blockers in patients with heart failure plus atrial fibrillation: an individual-patient data meta-analysis. Lancet 2014;384:2235–43.
    Crossref | PubMed
  22. Swedberg K, Hjalmarson A, Waagstein F, et al. Prolongation of survival in congestive cardiomyopathy by beta-receptor blockade. Lancet 1979;1:1374–6.
    Crossref | PubMed
  23. Domanski MJ, Krause-Steinrauf H, Massie BM, et al.; BEST Investigators. A comparative analysis of the results from 4 trials of beta-blocker therapy for heart failure: BEST, CIBISII, MERIT-HF, and COPERNICUS. J Card Fail 2003;9:354–63.
    Crossref | PubMed
  24. Kirchhof P, Benussi S, Kotecha D, et al.; Authors/Task Force Members; Document Reviewers. 2016 ESC Guidelines for the management of atrial fibrillation developed in collaboration with EACTS: The Task Force for the management of atrial fibrillation of the European Society of Cardiology (ESC). Developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. Endorsed by the European Stroke Organisation (ESO). Eur Heart J. 2016 Oct 7;37(38):2893-2962
  25. Shen MJ, Choi EK, Tan AY, et al. Neural mechanisms of atrial arrhythmias. Nat Rev Cardiol 2012;9:30–9.
    Crossref | PubMed
  26. Ikeda T, Murai H, Kaneko S, et al. Augmented singleunit muscle sympathetic nerve activity in heart failure with chronic atrial fibrillation. J Physiol 2012;590:509–18.
    Crossref | PubMed
  27. Gould PA, Esler MD, Kaye DM. Atrial fibrillation is associated with decreased cardiac sympathetic response to isometric exercise in CHF in comparison to sinus rhythm. Pacing Clin Electrophysiol 2008;31:1125–9.
    Crossref | PubMed
  28. Cullington D, Goode KM, Zhang J, et al. Is heart rate important for patients with heart failure in atrial fibrillation? JACC Heart Fail 2014;2:213–20.
    Crossref | PubMed
  29. ALLHAT Collaborative Research Group. Major cardiovascular events in hypertensive patients randomized to doxazosin vs chlorthalidone: the antihypertensive and lipid-lowering treatment to prevent heart attack trial (ALLHAT). JAMA 2000;283:1967–75.
    Crossref | PubMed
  30. McComb MN, Chao JY, Ng TM. Direct vasodilators and sympatholytic agents. J Cardiovasc Pharmacol Ther 2016;21:3–19.
    Crossref | PubMed
  31. Cohn JN, Pfeffer MA, Rouleau J, et al.; MOXCON Investigators. Adverse mortality effect of central sympathetic inhibition with sustained-release moxonidine in patients with heart failure (MOXCON). Eur J Heart Fail 2003;5:659–67.
    Crossref | PubMed
  32. Li M, Zheng C, Sato T, et al. Vagal nerve stimulation markedly improves long-term survival after chronic heart failure in rats. Circulation 2004;109:120–4.
    Crossref | PubMed
  33. Sobotka PA, Krum H, Bohm M, et al. The role of renal denervation in the treatment of heart failure. Curr Cardiol Rep 2012;14:285–92.
    Crossref | PubMed
  34. Gold MR, Van Veldhuisen DJ, Hauptman PJ, et al. Vagus nerve stimulation for the treatment of heart failure: The INOVATEHF Trial. J Am Coll Cardiol 2016;68:149–58.
    Crossref | PubMed
  35. Flather MD, Yusuf S, Køber L, et al. Long-term ACE-inhibitor therapy in patients with heart failure or left-ventricular dysfunction: a systematic overview of data from individual patients. ACE-Inhibitor Myocardial Infarction Collaborative Group. Lancet 2000;355:1575–81.
    Crossref | PubMed
  36. Stanton A. Potential of renin inhibition in cardiovascular disease. J Renin Angiotensin Aldosterone Syst 2003;4:6–10.
    Crossref | PubMed
  37. McMurray JJ, Krum H, Abraham WT, et al.; ATMOSPHERE Committees Investigators. Aliskiren, enalapril, or aliskiren and enalapril in heart failure. N Engl J Med 2016;374:1521–32.
    Crossref | PubMed
  38. McMurray JJ, Ostergren J, Swedberg K, et al.; CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and reduced leftventricular systolic function taking angiotensin-convertingenzyme inhibitors: the CHARM-Added trial. Lancet 2003;362:767–71.
    Crossref | PubMed
  39. Pfeffer MA, McMurray JJ, Velazquez EJ, et al.; Valsartan in Acute Myocardial Infarction Trial Investigators. Valsartan, captopril, or both in myocardial infarction complicated by heart failure, left ventricular dysfunction, or both. N Engl J Med 2003;349:1893–906.
    Crossref | PubMed
  40. Yusuf S, Teo KK, Pogue J, et al. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med 2008;358:1547–59.
    Crossref | PubMed
  41. Parving HH, Brenner BM, McMurray JJ, et al.; ALTITUDE Investigators. Cardiorenal end points in a trial of aliskiren for type 2 diabetes. N Engl J Med 2012;367:2204–13.
    Crossref | PubMed
  42. Gheorghiade M, Bohm M, Greene SJ, et al.; ASTRONAUT Investigators and Coordinators. Effect of aliskiren on postdischarge mortality and heart failure readmissions among patients hospitalized for heart failure: the ASTRONAUT randomized trial. JAMA 2013;309:1125–35.
    Crossref | PubMed
  43. McCurley A, Jaffe IZ. Mineralocorticoid receptors in vascular function and disease. Mol Cell Endocrinol 2012;350:256–65.
    Crossref | PubMed
  44. Funder JW. Aldosterone and mineralocorticoid receptors in the cardiovascular system. Prog Cardiovasc Dis 2010; 52:393–400.
    Crossref | PubMed
  45. Young MJ, Funder JW. Mineralocorticoid receptors and pathophysiological roles for aldosterone in the cardiovascular system. J Hypertens 2002;20:1465–8.
    Crossref | PubMed
  46. Pitt B, Zannad F, Remme WJ, et al. The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators. N Engl J Med 1999;341:709–17.
    Crossref | PubMed
  47. Zannad F, McMurray JJ, Krum H, et al.; EMPHASIS-HF Study Group. Eplerenone in patients with systolic heart failure and mild symptoms. N Engl J Med 2011;364:11–21.
    Crossref | PubMed
  48. Albert NM, Yancy CW, Liang L, et al. Use of aldosterone antagonists in heart failure. JAMA 2009;302:1658–65.
    Crossref | PubMed
  49. Rossignol P, Dobre D, McMurray JJ, et al. Incidence, determinants, and prognostic significance of hyperkalemia and worsening renal function in patients with heart failure receiving the mineralocorticoid receptor antagonist eplerenone or placebo in addition to optimal medical therapy: results from the Eplerenone in Mild Patients Hospitalization and Survival Study in Heart Failure (EMPHASIS-HF). Circ Heart Fail 2014;7:51–8.
    Crossref | PubMed
  50. Barfacker L, Kuhl A, Hillisch A, et al. Discovery of BAY 94-8862: a nonsteroidal antagonist of the mineralocorticoid receptor for the treatment of cardiorenal diseases. ChemMedChem 2012;7:1385–403.
    Crossref | PubMed
  51. Kolkhof P, Delbeck M, Kretschmer A, et al. Finerenone, a novel selective nonsteroidal mineralocorticoid receptor antagonist protects from rat cardiorenal injury. J Cardiovasc Pharmacol 2014;64:69–78.
    Crossref | PubMed
  52. Pitt B, Kober L, Ponikowski P, et al. Safety and tolerability of the novel non-steroidal mineralocorticoid receptor antagonist BAY 94-8862 in patients with chronic heart failure and mild or moderate chronic kidney disease: a randomized, double-blind trial. Eur Heart J 2013;34:2453–63.
    Crossref | PubMed
  53. Pitt B, Anker SD, Bohm M, et al. Rationale and design of MinerAlocorticoid Receptor antagonist Tolerability Study- Heart Failure (ARTS-HF): a randomized study of finerenone vs. eplerenone in patients who have worsening chronic heart failure with diabetes and/or chronic kidney disease. Eur J Heart Fail 2015;17:224–32.
    Crossref | PubMed
  54. Rubattu S, Bigatti G, Evangelista A, et al. Association of atrial natriuretic peptide and type a natriuretic peptide receptor gene polymorphisms with left ventricular mass in human essential hypertension. J Am Coll Cardiol 2006;48:499–505.
    Crossref | PubMed
  55. Rubattu S, Sciarretta S, Valenti V, et al. Natriuretic peptides: an update on bioactivity, potential therapeutic use, and implication in cardiovascular diseases. Am J Hypertens 2008;21:733–41.
    Crossref | PubMed
  56. Kuhn M, Volker K, Schwarz K, et al. The natriuretic peptide/guanylyl cyclase--a system functions as a stressresponsive regulator of angiogenesis in mice. J Clin Invest 2009;119:2019–30.
    Crossref | PubMed
  57. Munzel T, Kurz S, Holtz J, et al. Neurohormonal inhibition and hemodynamic unloading during prolonged inhibition of ANF degradation in patients with severe chronic heart failure. Circulation 1992;86:1089–98.
    Crossref | PubMed
  58. von Lueder TG, Atar D, Krum H. Current role of neprilysin inhibitors in hypertension and heart failure. Pharmacol Ther 2014;144:41–9.
    Crossref | PubMed
  59. Dalzell JR, Seed A, Berry C, et al. Effects of neutral endopeptidase (neprilysin) inhibition on the response to other vasoactive peptides in small human resistance arteries: studies with thiorphan and omapatrilat. Cardiovasc Ther 2014;32:13–8.
    Crossref | PubMed
  60. Pham I, Gonzalez W, el Amrani AI, et al. Effects of converting enzyme inhibitor and neutral endopeptidase inhibitor on blood pressure and renal function in experimental hypertension. J Pharmacol Exp Ther 1993;265:1339–47.
    PubMed
  61. Bayes-Genis A, Prickett TC, Richards AM, et al. Soluble neprilysin retains catalytic activity in heart failure. J Heart Lung Transplant 2016;35:684–5.
    Crossref | PubMed
  62. Mangiafico S, Costello-Boerrigter LC, Andersen IA, et al. Neutral endopeptidase inhibition and the natriuretic peptide system: an evolving strategy in cardiovascular therapeutics. Eur Heart J 2013;34:886–93c.
    Crossref | PubMed
  63. Burnett JC, Jr. Vasopeptidase inhibition: a new concept in blood pressure management. J Hypertens Suppl 1999;17:S37– 43.
    PubMed
  64. Armstrong PW, Lorell BH, Nissen S, et al. Omapatrilat. Circulation 2002;106:e9011-2.
    PubMed
  65. Coats AJ. Omapatrilat--the story of Overture and Octave. Int J Cardiol 2002;86:1–4.
    Crossref | PubMed
  66. Coats AJ. Omapatrilat--the ups and downs of an exciting but complicated new drug. Int J Cardiol 2000;74:1–3.
    Crossref | PubMed
  67. Kostis JB, Packer M, Black HR, et al. Omapatrilat and enalapril in patients with hypertension: the Omapatrilat Cardiovascular Treatment vs. Enalapril (OCTAVE) trial. Am J Hypertens 2004; 17:103–11.
    Crossref | PubMed
  68. Rouleau JL, Pfeffer MA, Stewart DJ, et al. Comparison of vasopeptidase inhibitor, omapatrilat, and lisinopril on exercise tolerance and morbidity in patients with heart failure: IMPRESS randomised trial. Lancet 2000;356:615-20.
    Crossref | PubMed
  69. Packer M, Califf RM, Konstam MA, et al. Comparison of omapatrilat and enalapril in patients with chronic heart failure: the Omapatrilat Versus Enalapril Randomized Trial of Utility in Reducing Events (OVERTURE). Circulation 2002;106:920-6.
    Crossref | PubMed
  70. Messerli FH, Nussberger J. Vasopeptidase inhibition and angio-oedema. Lancet 2000;356:608-9.
    Crossref | PubMed
  71. von Lueder TG, Sangaralingham SJ, Wang BH, et al. Reninangiotensin blockade combined with natriuretic Peptide system augmentation: novel therapeutic concepts to combat heart failure. Circ Heart Fail 2013;6:594–605.
    Crossref | PubMed
  72. McMurray JJ. Neprilysin inhibition to treat heart failure: a tale of science, serendipity, and second chances. Eur J Heart Fail 2015;17:242–7.
    Crossref | PubMed
  73. Hegde LG, Yu C, Renner T, et al. Concomitant angiotensin AT1 receptor antagonism and neprilysin inhibition produces omapatrilat-like antihypertensive effects without promoting tracheal plasma extravasation in the rat. J Cardiovasc Pharmacol 2011;57:495–504.
    Crossref | PubMed
  74. Ruilope LM, Dukat A, Bohm M, et al. Blood-pressure reduction with LCZ696, a novel dual-acting inhibitor of the angiotensin II receptor and neprilysin: a randomised, doubleblind, placebo-controlled, active comparator study. Lancet 2010;375:1255–66.
    Crossref | PubMed
  75. Solomon SD, Zile M, Pieske B, et al.; Prospective comparison of ARNI with ARB on Management Of heart failUre with preserved ejectioN fracTion (PARAMOUNT) Investigators. The angiotensin receptor neprilysin inhibitor LCZ696 in heart failure with preserved ejection fraction: a phase 2 doubleblind randomised controlled trial. Lancet 2012;380:1387–95.
    Crossref | PubMed
  76. McMurray JJ, Packer M, Desai AS, et al.; PARADIGM-HF Investigators and Committees. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med 2014;371:993–1004.
    Crossref | PubMed
  77. The SOLVD Investigators. Effect of enalapril on mortality and the development of heart failure in asymptomatic patients with reduced left ventricular ejection fractions. N Engl J Med 1992;327:685–91.
    Crossref | PubMed
  78. McMurray J, Packer M, Desai A, et al. A putative placebo analysis of the effects of LCZ696 on clinical outcomes in heart failure. Eur Heart J 2015;36:434–9.
    Crossref | PubMed
  79. Kristensen SL, Preiss D, Jhund PS, et al.; PARADIGM-HF Investigators and Committees. Risk Related to Pre-Diabetes Mellitus and Diabetes Mellitus in Heart Failure With Reduced Ejection Fraction: Insights From Prospective Comparison of ARNI With ACEI to Determine Impact on Global Mortality and Morbidity in Heart Failure Trial. Circ Heart Fail 2016;9:e002560.
    Crossref | PubMed
  80. Desai AS, Claggett BL, Packer M, et al.; PARADIGM-HF Investigators. Influence of sacubitril/valsartan (LCZ696) on 30-Day readmission after heart failure hospitalization. J Am Coll Cardiol 2016;68:241–8.
    Crossref | PubMed
  81. Solomon SD, Claggett B, Desai AS, et al. Influence of ejection fraction on outcomes and efficacy of sacubitril/ valsartan (LCZ696) in heart failure with reduced ejection rraction: The Prospective Comparison of ARNI with ACEI to Determine Impact on Global Mortality and Morbidity in Heart Failure (PARADIGM-HF) trial. Circ Heart Fail 2016;9:e002744.
    Crossref | PubMed
  82. Packer M, McMurray JJ, Desai AS, et al.; PARADIGM-HF Investigators and Coordinators. Angiotensin receptor neprilysin inhibition compared with enalapril on the risk of clinical progression in surviving patients with heart failure. Circulation 2015;131:54–61.
    Crossref | PubMed
  83. Jhund PS, Claggett BL, Voors AA, et al. Elevation in highsensitivity troponin T in heart failure and preserved ejection fraction and influence of treatment with the angiotensin receptor neprilysin inhibitor LCZ696. Circ Heart Fail 2014;7: 953–9.
    Crossref | PubMed
  84. von Lueder TG, Wang BH, Kompa AR, et al. Angiotensin receptor neprilysin inhibitor LCZ696 attenuates cardiac remodeling and dysfunction after myocardial infarction by reducing cardiac fibrosis and hypertrophy. Circ Heart Fail 2015;8:71–8.
    Crossref | PubMed
  85. Bodey F, Hopper I, Krum H. Neprilysin inhibitors preserve renal function in heart failure. Int J Cardiol 2015;179: 329–30.
    Crossref | PubMed
  86. Wang BH, von Lueder TG, Kompa AR, et al. Combined angiotensin receptor blockade and neprilysin inhibition attenuates angiotensin-II mediated renal cellular collagen synthesis. Int J Cardiol 2015;186:104–5.
    Crossref | PubMed
  87. Voors AA, Gori M, Liu LC, et al.; PARAMOUNT Investigators. Renal effects of the angiotensin receptor neprilysin inhibitor LCZ696 in patients with heart failure and preserved ejection fraction. Eur J Heart Fail 2015;17:510–7.
    Crossref | PubMed
  88. Yanagisawa M, Kurihara H, Kimura S, et al. A novel peptide vasoconstrictor, endothelin, is produced by vascular endothelium and modulates smooth muscle Ca2+ channels. J Hypertens Suppl 1988;6:S188–91.
    Crossref | PubMed
  89. Davenport AP, Hyndman KA, Dhaun N, et al. Endothelin. Pharmacol Rev 2016;68:357–418.
    Crossref | PubMed
  90. Kiowski W, Sutsch G, Hunziker P, et al. Evidence for endothelin-1-mediated vasoconstriction in severe chronic heart failure. Lancet 1995;346:732–6.
    Crossref | PubMed
  91. Lerman A, Kubo SH, Tschumperlin LK, et al. Plasma endothelin concentrations in humans with end-stage heart failure and after heart transplantation. J Am Coll Cardiol 1992;20:849–53.
    Crossref | PubMed
  92. Sakai S, Miyauchi T, Kobayashi M, et al. Inhibition of myocardial endothelin pathway improves longterm survival in heart failure. Nature 1996;384:353–5.
    Crossref | PubMed
  93. Fraccarollo D, Hu K, Galuppo P, et al. Chronic endothelin receptor blockade attenuates progressive ventricular dilation and improves cardiac function in rats with myocardial infarction: possible involvement of myocardial endothelin system in ventricular remodeling. Circulation 1997;96:3963-73.
    Crossref | PubMed
  94. Mulder P, Richard V, Derumeaux G, et al. Role of endogenous endothelin in chronic heart failure: effect of long- term treatment with an endothelin antagonist on survival, hemodynamics, and cardiac remodeling. Circulation 1997;96:1976–82.
    Crossref | PubMed
  95. Sutsch G, Kiowski W, Yan XW, et al. Short-term oral endothelin-receptor antagonist therapy in conventionally treated patients with symptomatic severe chronic heart failure. Circulation 1998;98:2262–8.
    Crossref | PubMed
  96. Love MP, Haynes WG, Gray GA, et al. Vasodilator effects of endothelin-converting enzyme inhibition and endothelin ETA receptor blockade in chronic heart failure patients treated with ACE inhibitors. Circulation 1996;94:2131–7.
    Crossref | PubMed
  97. Kelland NF, Webb DJ. Clinical trials of endothelin antagonists in heart failure: publication is good for the public health. Heart 2007;93:2–4.
    Crossref | PubMed
  98. Kelland NF, Webb DJ. Clinical trials of endothelin antagonists in heart failure: a question of dose? Exp Biol Med (Maywood) 2006;231:696–9.
    PubMed
  99. Teerlink JR. Endothelins: pathophysiology and treatment implications in chronic heart failure. Curr Heart Fail Res 2005;2:191–7.
    Crossref | PubMed
  100. Gottlieb SS. The impact of finally publishing a negative study: new conclusions about endothelin antagonists. J Card Fail 2005;11:21–2.
    Crossref | PubMed
  101. Hefke T, Zittermann A, Fuchs U, et al. Bosentan effects on hemodynamics and clinical outcome in heart failure patients with pulmonary hypertension awaiting cardiac transplantation. Thorac Cardiovasc Surg 2012;60:26–34.
    Crossref | PubMed
  102. Padeletti M, Caputo M, Zaca V, et al. Effect of bosentan on pulmonary hypertension secondary to systolic heart failure. Pharmacology 2013;92:281–5.
    Crossref | PubMed
  103. Perez-Villa F, Farrero M, Cardona M, et al. Bosentan in heart transplantation candidates with severe pulmonary hypertension: efficacy, safety and outcome after transplantation. Clin Transplant 2013;27:25–31.
    Crossref | PubMed
  104. Nagaya N, Satoh T, Nishikimi T, et al. Hemodynamic, renal, and hormonal effects of adrenomedullin infusion in patients with congestive heart failure. Circulation 2000;101:498–503.
    Crossref | PubMed
  105. Ames RS, Sarau HM, Chambers JK, et al. Human urotensin- II is a potent vasoconstrictor and agonist for the orphan receptor GPR14. Nature 1999;401:282–6.
    Crossref | PubMed
  106. Douglas SA, Tayara L, Ohlstein EH, et al. Congestive heart failure and expression of myocardial urotensin II. Lancet 2002;359:1990–7.
    Crossref | PubMed
  107. Jougasaki M, Wei CM, McKinley LJ, et al. Elevation of circulating and ventricular adrenomedullin in human congestive heart failure. Circulation 1995;92:286–9.
    Crossref | PubMed
  108. Chin KL, Skiba M, Tonkin A, et al. The treatment gap in patients with chronic systolic heart failure: a systematic review of evidence-based prescribing in practice. Heart Fail Rev 2016;21:675–97.
    Crossref | PubMed
  109. Packer M. Heart failure’s dark secret: Does anyone really care about optimal medical therapy? Circulation 2016; 134:629–31.
    Crossref | PubMed
  110. Cohen-Solal A, Kotecha D, van Veldhuisen DJ, et al.; SENIORS Investigators. Efficacy and safety of nebivolol in elderly heart failure patients with impaired renal function: insights from the SENIORS trial. Eur J Heart Fail 2009;11:872–80.
    Crossref | PubMed
  111. Kotecha D, Piccini JP. Atrial fibrillation in heart failure: what should we do? Eur Heart J 2015;36:3250–7.
    Crossref | PubMed
  112. Kotecha D, Lam CS, Van Veldhuisen DJ, et al. Vicious twins - heart failure with preserved ejection fraction and atrial fibrillation. J Am Coll Cardiol 2016:68:2217–28.
    Crossref | PubMed
  113. Gheorghiade M, Larson CJ, Shah SJ, et al. Developing new treatments for heart failure: focus on the heart. Circ Heart Fail 2016;9:e002727.
    Crossref | PubMed
  114. Mann DL, McMurray JJ, Packer M, et al. Targeted anticytokine therapy in patients with chronic heart failure: results of the Randomized Etanercept Worldwide Evaluation (RENEWAL). Circulation. 2004;109:1594–602.
    Crossref | PubMed
  115. Anand I, McMurray J, Cohn JN, et al.; EARTH Investigators. Long-term effects of darusentan on left-ventricular remodelling and clinical outcomes in the Endothelin: A Receptor Antagonist Trial in Heart Failure (EARTH): randomised, double-blind, placebo-controlled trial. Lancet 2004;364:347–54.
    Crossref | PubMed
  116. Borlaug BA, Lewis GD, McNulty SE, et al. Effects of sildenafil on ventricular and vascular function in heart failure with preserved ejection fraction. Circ Heart Fail 2015;8:533–41.
    Crossref | PubMed
  117. Packer M. Kicking the tyres of a heart failure trial: physician response to the approval of sacubitril/valsartan in the USA. Eur J Heart Fail 2016;18:1211–9.
    Crossref | PubMed
  118. Campbell RT, Jhund PS, Castagno D, et al. What have we learned about patients with heart failure and preserved ejection fraction from DIG-PEF, CHARM-preserved, and I-PRESERVE? J Am Coll Cardiol 2012;60:2349–56.
    Crossref | PubMed
  119. Mentz RJ, Kelly JP, von Lueder TG, et al. Noncardiac Comorbidities in heart failure with reduced versus preserved ejection fraction. J Am Coll Cardiol 2014;64:2281–93.
    Crossref | PubMed
  120. Chamberlain AM, St Sauver JL, Gerber Y, et al. Multimorbidity in heart failure: a community perspective. Am J Med 2015;128:38–45.
    Crossref | PubMed
  121. Gerber Y, Weston SA, Redfield MM, et al. A contemporary appraisal of the heart failure epidemic in Olmsted County, Minnesota, 2000 to 2010. JAMA Intern Med 2015;175:996–1004.
    Crossref | PubMed
  122. Shah AM, Claggett B, Sweitzer NK, et al. Prognostic importance of impaired systolic function in heart failure with preserved ejection fraction and the impact of spironolactone. Circulation 2015;132:402–14.
    Crossref | PubMed