Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Apr 1.
Published in final edited form as: Curr Heart Fail Rep. 2017 Apr;14(2):134–145. doi: 10.1007/s11897-017-0327-y

Potential Expanded Indications for Neprilysin Inhibitors

Elizabeth Riddell 1, Justin M Vader 1,*
PMCID: PMC5645149  NIHMSID: NIHMS858914  PMID: 28281174

Abstract

Purpose of review

The goal of this article is to review potential expanded indications for neprilysin inhibitors. This article reviews the rationale and design for ongoing and future trials of sacubitril/valsartan in cardiovascular and non-cardiovascular disease.

Recent findings

Randomized trial data are lacking for use of sacubitril/valsartan in acute heart failure and advanced heart failure. Mechanistic data from animal studies suggest a role for neprilysin inhibition in the treatment of post-myocardial infarction systolic dysfunction and heart failure with preserved ejection fraction. Beyond the cardiovascular system, renal and neurological function may be impacted by neprilysin inhibition. Forthcoming randomized trials will address the clinical impact of sacubitril/valsartan on these conditions.

Summary

Neprolysin inhibition with sacubitril/valsartan offers a new therapeutic strategy with a broad range of potential therapeutic actions. In PARADIGM-HF, the combination of neprolysin and RAAS inhibition was proven to be superior to enalapril for patients with stable NYHA class II–III heart failure and reduced left ventricular ejection fraction. Preliminary data suggests it may also have a role in other cardiovascular and non-cardiovascular disease. Several ongoing and planned studies will determine the extent of its benefit for these other indications.

Keywords: neprilysin inhibitors, cardiovascular disease, sacubitril/valsartan, heart failure

Introduction

Neurohormal pathways in heart failure

The activation of neurohormonal compensatory mechanisms underlies the physiology of heart failure (HF). The contributions of the sympathetic nervous system (SNS) and the renin-angiotensin-aldosterone system (RAAS) to unfavorable changes in renal sodium handling, vascular tone, and cardiomyocyte structure and function were long ago recognized and translated into pharmacotherapies that have been proven in large scale clinical trials to improve survival and morbidity in patients with HF and reduced left ventricular ejection fraction (HFrEF). These agents formed the backbone of modern HF pharmacotherapy: β-blockers, angiotensin converting enzyme inhibitors (ACEI), angiotensin receptor blockers (ARB), and the mineralocorticoid receptor antagonists (MRA). Modulation of other biologic pathways operant in heart failure has failed to demonstrate clinical benefit - endothelin receptor antagonists failed to improved outcomes and were associated with increased adverse events13 while trials of tumor necrosis factor-alpha inhibition were stopped for futility.4 Meanwhile, increased inhibition of the RAAS through multiple or higher-dose drugs failed to demonstrate further mortality improvements, suggesting the need to harness another biological pathway to benefit patients with heart failure.510

The natriurietic peptide system (NPS), also activated in HF, has been an alluring target for drug development - not for inhibition but augmentation. This is due to the natriurietic, diuretic, vasodilatory, and lusitropic properties of these peptides as well as their action to prevent cardiac hypertrophy and fibrosis and to decrease renin release. These peptides include atrial natriurietic peptide (ANP), B-type natriurietic peptide (BNP), and C-type natriurietic peptide (CNP). Synthetic ANP 1113 and synthetic BNP 1416 have been demonstrated to potentiate the above effects, however these molecules are only available in parenteral form and their use has not been demonstrated in large-scale trials to confer a mortality benefit. As an alternative strategy to exogenous administration, targeting the degradation of these molecules presents an opportunity to potentiate their biological effect.

Biological activity of neprilysin and pharmacologic implications

Neprilysin, a predominantly membrane-bound zinc-dependent metalloproteinase distributed broadly throughout the body, is responsible for the breakdown of multiple endogenous vasoactive peptides including bradykinin, natriuretic peptides, and adrenomedullin.1719 Increasing the levels of these peptides through neprilysin inhibition would be expected to counteract the neurohormonal activation and compensatory mechanisms that lead to sodium retention, vasoconstriction, and cardiac remodeling.20,21 Inhibitors of neprilysin were developed in the 1980s (thiorphan)22 and 1990s (sacubitril).23 Early animal studies demonstrated that neprilysin blockade effected a rise in natriuretic peptide levels and natriuriesis, but had inconsistent effects on blood pressure and systemic vascular resistance.17,24,25 Short term use in humans conferred beneficial effects on natriuriesis, diuresis, and hemodynamics, 24,26,27 however longer term use resulted in vasoconstriction and it was subsequently described that neprilysin inhibition also increased the circulating concentration of the vasopressors angiotensin II and endothelin.19,28 Dual potentiation of vasodilatory and vasoconstrictor substances results in a neutralized effect of isolated neprilysin inhibition and thwarts its usefulness in treating heart failure.

The neutralized effect of neprilysin inhibition on vascular tone and sodium handling underscores the need to understand the action of neprilysin on a variety of biological pathways. Neprilysin is involved in the metabolism of a broad array of peptides with various and occasionally contradictory biologic actions (Table 1). In addition to its action on the natriurietic peptides, endothelin, and angiotensin II, neprilysin has a role in the degradation of adrenomedullin and bradykinin, compounds which exert vasodilatory effects. In fact, the enzymatic activity of neprilysin against BNP is relatively less compared with its action on other NPS components, suggesting a more complex biological action of neprilysin inhibitors on the circulation than augmentation of BNP alone.29 The broad enzymatic activity of neprilysin has also led to concerns about implications outside the cardiovascular system. Neprilysin degrades amyloid-β peptide, leading to concerns that its inhibition might contribute to the development of diseases of amyloid-β accumulation such as age-related macular degeneration, cerebral amyloid angiopathy, and Alzheimer disease.30 Neprilysin, through its metabolism of mitogenic peptides may also serve as a check against tumor cell proliferation in prostate,31 breast,32 and other cancers.3335 Clinical ramifications of widespread and prolonged use of neprilysin inhibitors on these non-cardiovascular conditions are unclear.

Table 1.

Substrates of Neprilysin*

Vasoactive Peptides Mitogenesis and Angiogenesis
Adrenomedullin Bombesin-like peptides
Angiotensin I Fibroblast growth
Angiotensin II
Natriurietic peptides (ANP, BNP, CNP, urodilatin) Hypothalamic-Pituitary Axis
Bradykinin Adrenocorticotrophic hormone
Kallidin Gonadotropin-releasing hormone
Endothelin α-melanocyte stimulating hormone
Neurokinin A Oxytocin
Neuropeptide Y
Substance P Digestion and Metabolism
Cholecystokinin
Peptides in Neurologic Processes Gastrin-releasing peptide
Amyloid β Glucagon
Galanin Glucagon-like peptides
Neurotensin Insulin-B chain
Peptide YY
Pain and Inflammation
Calcitonin gene-related peptide
Dynrophin
β endorphin
Enkephalins
Neurokinin A
Vasoactive intestinal peptide
*

Adapted from Campbell, Nature Reviews Cardiology

Combined neprilysin and RAAS inhibition for Heart Failure

Though inhibition of neprilysin alone was not a viable strategy for treating cardiovascular disease, dual inhibition of neprilysin and the RAAS was ultimately explored in large-scale clinical trials. Rodent model data confirmed a greater antihypertensive effect of combined neprilysin inhibition and ACE inhibition36 and cardiac remodeling data from animal models suggested a rationale for this combined therapy in heart failure.3638 The oral agent omipatrilat, a dual inhibitor of neprilysin and ACE was developed for clinical use and in humans demonstrated antihypertensive and NP-augmenting effects.39 Large scale randomized controlled trials of dual neprilysin/ACE inhibition vs. ACE inhibition alone in HF patients followed. In the IMPRESS trial there was a trend towards efficacy of omipatrilat over lisinopril, leading to the OVERTURE trial, in which omapatrilat was superior to enalapril with regard to a secondary outcome of cardiovascular death or hospitalization, but failed to meet the primary endpoint of all-cause mortality or heart failure hospitalization.40,41 Concern was also raised over the increased rate of angioedema in the OVERTURE trial. The subsequent large scale hypertension trial OCTAVE revealed an increased rate and greater severity of angioedema among subjects receiving omipatrilat vs. enalapril, an observation that was more notable among African Americans.42 The failure of omapatrilat to meet the primary endpoints in OVERTURE coupled with concerns over angioedema resulted in an end to its development as a therapy for heart failure.

Despite the failure of omapatrilat to result in an approved pharmacotherapy for heart failure, development of combined neprilysin and RAAS inhibition continued. The action of omapatrilat on both the inhibition of neprilysin-based degradation of bradykinin as well as its inhibition of substance P, which itself breaks down bradykinin, likely accounts for the prohibitive rate of angioedema observed in clinical trials.18 LCZ696, a compound of the neprilysin inhibitor pro-drug sacubitril and the ARB valsartan, emerged as the next, ultimately successful, strategy. This compound demonstrated a favorable hemodynamic profile without cough and angioedema concerns in early phase trials43 and in a phase II study of HF subjects with preserved LVEF it demonstrated more favorable cardiac remodeling and improvements in heart failure status than comparators receiving valsartan.44 Ultimately, it was LCZ696 (sacubitril/valsartan), the first in class angiotensin receptor neprilysin inhibitor (ARNI), that would deliver a trail with a favorable mortality endpoint, leading to FDA approval for use in systolic heart failure.

Current Evidence and Indications for Combined Neprilysin Inhibition and Aldosterone Receptor Blockade

The 2016 ACC/AHA/HFSA Focused Update on New Pharmacological Therapy for Heart Failure includes recommendations for use of sacubitril/valsartan in heart failure patients with reduced ejection fraction (LVEF < 40%).45 Sacubitril/valsartan is recommended in patients with chronic symptomatic HFrEF and NYHA class II or III symptoms (who previously tolerated an ACEI or ARB) to further reduce morbidity and mortality (Class I, Level of Evidence B). Specifically replacement of ACEI or ARB with ARNI is recommended. Current labeling of the drug by the FDA is slightly broader than these guidelines, indicating ARNI for NYHA Class II–IV heart failure. The rationale for these indications is derived from the PARADIGM-HF trial.

The PARADIGM-HF trial compared LCZ696 (sacubitril/valsartan) to enalapril in a prospective, randomized, double-blind, international trial of 9,419 patients with NYHA class II-IV heart failure and reduced left ventricular ejection fraction (≤35%).46 Patients were required to be on 4 weeks of stable medical therapy and have elevated NP levels. Key exclusion criteria included symptomatic hypotension, SBP < 100 mm Hg, serum potassium > 5.2 mmol/L, eGFR < 30 ml/min, or a history of angioedema. Prior to randomization, a single-blind run-in period was required in which patients received enalapril for at least 2 weeks followed by sacubitril/valsartan for a period of 4 to 6 weeks. A total of 9419 subjects entered the run-in and 8442 subjects were randomized. Study drugs were titrated to a goal dose of sacubitril/valsartan 97/103 mg twice daily or enalapril 10 mg twice daily. Most patients enrolled in the study had NYHA class II (70%) or III (24%) heart failure, with < 1% having Class IV HF. The mean LVEF of the study population was 29% and concomitant treatment with guideline-directed heart failure therapies was typical for a heart failure trial population. The trial was concluded early after meeting a pre-specified stopping point for compelling clinical benefit. After a median follow-up of 27 months, subjects taking sacubitril/valsartan had a 20% reduction the combined endpoint of cardiovascular death or HF hospitalization. All-cause mortality was also significantly less among the valsartan/sacubitril group (17% vs. 19.8%).

The impressive results of PARADIGM and the approval of sacubitril/valsartan for clinical use have provided the opportunity to explore the role of neprilysin inhibition, NP potentiation, and the action of ARNI in a variety of cardiovascular and non-cardiovascular disease states. (Table 2). Here we detail the biologic rationale and context for these forthcoming trials.

Table 2.

Ongoing and Forthcoming Trials of Sacubitril/Valsartan

Trial Name NCT/ISDN Population Primary Endpoint Subjects Duration
PIONEER-HF NCT02554890 Recent AHF (HFrEF) Δ NT-proBNP 736 8 weeks
TRANSITION NCT02661217 Inpatient AHF (HFrEF) % taking max dose sac/val at 10 weeks 1000 26 weeks
HFN-LIFE NCT02816736 NYHA Class IV HFrEF Δ NT-proBNP 400 24 weeks
PROVE-HF NCT02887183 NYHA Class II–IV HFrEF Remodeling parameters 830 52 weeks
EVALUATE-HF NCT02874794 NYHA Class I–III with hypertension Δ aortic impedance 432 12 weeks
PARENT NCT02788656 NYHA Class II–III HFrEF Δ mean PA pressure 20 32 weeks
PARADISE-MI NCT02924727 Post-MI LVEF ≤ 40% with risk factors Time to CV death, HF hospitalization, or outpatient HF 4650 156 weeks
PARAGON-HF NCT01920711 HFpEF with elevated NTproBNP, structural heart disease Rate of CV death and total HF hospitalizations 4600 57 months
UK HARP-III ISRCTN 11958993 CKD with an eGFR between 20 and 60 mL/min/1.73m2 Δ GFR 400 12 months
PERSPECTIVE NCT02884206 HFpEF with elevated NTproBNP, structural heart disease Δ Global Cognitive Composite Score 520 36 months
AWAKE-HF NCT02970669 NYHA Class II-IV HFrEF Δ daily actigraphy 136 16 weeks
ENTRESTO-SAS NCT02916160 LVEF ≤ 45% Δ Apnea-Hypopnea Index 100 3 months
Peripheral Arterial Disease Trial NCT02636283 No HF. Caludication with ankle-brachial index ≤ 0.90 Treadmill walk until pain initiated 40 8 weeks
PARASAIL (Post- Approval Study) NCT02690974 NYHA II-III, HFrEF % tolerating max dose 300 12 months
Pediatric trial NCT02678312 Age < 18. NHYHA II- IV, HFrEF Combined Clinical Outcome 360 52 weeks
Japanese trial NCT02468232 Same as PARADIGM Time to CV death or HF Hospitalization 220 40 months
PARABLE NCT02682719 LVEF > 50%, Elevated BNP, LAVI > 28 mL/m2 Δ left atrial volume index 250 18 months
PRIME NCT02687932 LVEF 25–50% with secondary MR (EROA > 0.1 cm2) Δ EROA 118 12 months

Potential Expanded/Future Indications of ARNI use in HFrEF

Acute or Recently Decompensated Heart Failure

In terms of acute heart failure (AHF) , patients treated with sacubitril/valsartan in the PARADIGM trial, experienced reduced readmission, both at 30 and 60 days, for all-cause and HF readmission.47 Further, the benefit of sacubitril/valsartan over enalparil was not attenuated or accentuated by proximity of trial enrollment to most recent prior hospitalization for HF.48 Unfortunately, whether these inferences can be extended to patients with currently or recently decompensated HF is not knowable from the PARADIGM trial, which excluded patients with a current episode of decompensation and patients not taking at least 4 weeks of stable medical therapy with at least 10 mg/day of enalapril or equivalent.

The impact of initiating or up-titrating of any neurohormonal antagonist during an episode of acute heart failure is not well-described. Initiation of beta-blocker prior to discharge during an AHF hospitalization resulted in superior rates of beta-blocker use at 60 days but resulted in no difference in subsequent death or re-hospitalization.49 While analogous prospective data are lacking for ACEI/ARB use at discharge, it is evident from Medicare data that patients discharged on ACEI or ARB are substantially more likely to be maintained on long-term therapy.50 Aggregated data from clinical trials also suggest that ACEI/ARB non-use at discharge is associated with greater risk for post-discharge adverse events.51 Thus, there is interest in the role of early ARNI initiation relative to HF hospitalization.

Two forthcoming trials will address the role of inpatient initiation of ARNI in acute heart failure. PIONEER-HF trial will be an 8 week randomized, double-blind, multicenter study to compare safety and tolerability of initiation of sacubitril/valsartan versus enalapril initiation prior to hospital discharge in patients with HFrEF who have been stabilized following admission for AHF with a primary efficacy endpoint of time-averaged percentage change of NT-proBNP (NCT02554890). Patients will be randomized between 1 and 10 days after hospital presentation provided they meet a definition of stability including SBP ≥100 mmHg for 6 hours prior to randomization with no symptomatic hypotension, no increase in IV diuretic dose or administration of IV vasodilators within 6 hours prior to randomization, and no administration of inotropes for 24 hours prior to randomization. Meanwhile, the TRANSITION trial will be a 1,000 subject international trial similar to IMPACT-HF, randomizing subjects to a strategy of either inpatient initiation of sacubitril/valsartan or outpatient initiation of sacubitril/valsartan from day 1 to 14 post-discharge (NCT02661217). The primary endpoint will be the percentage of subjects receiving maximum dose sacubitril/valsartan at 10 weeks post-randomization.

NYHA Class IV Heart Failure

The effectiveness of sacubtril/valsartan in the patients with the most advanced HF is not clear. When PARADIGM subjects were characterized by use of the MAGGIC risk model, an externally validated predictor of adverse events in HF, a consistent degree of benefit was noted across all quintiles.52 However, while a number of high-risk patients were certainly represented in the trial, PARADIGM included only 60 NYHA Class IV patients, comprising < 1% of the total enrollment. The clinical features that were associated with an inability to complete the run-in period of the PARADIGM trial - an eGFR < 60 ml/m2, lower SBP, and highter NTproBNP–are particularly prevalent in Class IV patients.53 Ultimately, the absolute benefit of ARNI in patients with Class IV heart failure and the relative benefit of ARNI over standard ACEI/ARB use are difficult to ascertain from available data. To this end, the HFN-LIFE trial will prospectively address the comparative effectiveness of sacubitril/valsartan vs. valsartan alone in a randomized, double-blind trial of approximately 400 subjects with NYHA Class IV heart failure (NCT02816736). Subjects will be followed for 24 weeks and the primary study outcome will be an AUC difference in NT-proBNP as assessed at 4, 8, 12, and 24 weeks. Secondary outcomes will include clinical worsening of heart failure and tolerability will provide valuable insight into the practical use of this agent in patients with advanced heart failure.

Structural, hemodynamic, and biochemical evidence of cardiac remodeling

Despite demonstrating improvement in survival and hospitalization, the PARADIGM trial did not include serial echocardiography, thus the degree of ventricular remodeling experienced with ARNI use in the trial is not known. Previous trials of ACEI, ARB, and β-blockers demonstrating mortality improvement have also demonstrated improvement in left ventricular volumes compared to placebo. For ACE inhibitors this favorable remodeling effect extends to patients with asymptomatic LV systolic dysfunction.54 Whether cardiac remodeling undergirds the benefit of ARNI will be assessed in forthcoming trials assessing biomarker changes and ventricular remodeling among patients with NYHA Class II-IV heart failure with reduced LVEF ≤40% (PROVE-HF, NCT02887183), changes in aortic impedance among patients with NYHA Class I-III HF and hypertension (EVALUATE-HF, NCT02874794), changes in functional mitral regurgitation (PRIME, NCT02687932) in patients with LVEF between 25% and 50%, and changes in mean pulmonary artery pressure in patients with LVEF < 35% (PARENT, NCT02788656). Finally, the potential for sacubitril/valsartan to attenuate atrial remodeling in patients with risk for future heart failure will be addressed by the PARABLE study. These trials should provide useful insights into the role of ARNI at several stages in the progression of HF.

Potential Therapeutic Strategies Beyond Systolic Heart Failure

Post Acute Myocardial Infarction

Use of an ACEI or ARB is indicated for all patients with LVEF ≤40% following either ST-segment elevation or non-ST-segment elevation myocardial infarction (Class 1, Level of Evidence A).55,56 Large scale clinical trials demonstrate the mortality benefit of ACEI in this setting.57,58 ARBs are similarly effective, but produce undesired adverse effects when added to ACEI.59,60 Additionally, beta blockers and mineralocorticoid receptor antagonists improve mortality in post-MI patients with reduced LVEF and carry a Class 1, Level of Evidence A recommendation.

Natriurietic peptide levels rise in the setting of myocardial infarction and are associated with reduced survival.61 The stimulus for NP release appears to be both wall stress and ischemia.62 NPs have potentially favorable effects on the infarcted myocardium, reducing ischemia reperfusion injury, inhibiting neutrophil degranulation, and blunting sympathetic nerve activity.63 In humans with anterior myocardial infarction, infusion of ANP results in a reduction of cardiac sympathetic nerve activity and less LV remodeling64 and infusion of BNP results in improved LVEF and less ventricular dilatation.65 ARNI are a logical consideration for therapy post-MI and indeed animal data show that sacubitril/valsartan attenuates LV dilatation, preserves LV systolic function and mechanics, and reduces myocardial hypertrophy and fibrosis.66 The PARADISE-MI study will test the hypothesis that sacubitril/valsartan is superior to ACEI with regard to the cumulative hazard of CV death, HF hospitalization, or outpatient HF in an international trial of 4,650 subjects with new LV systolic dysfunction (LVEF ≤40% and no prior history of chronic heart failure) following acute myocardial infarction (NCT02924727). In addition to providing data on long-term post-infarct ventricular remodeling with ARNI, the dual effects of potentiating NPs and inhibiting neprilysin should provide insights into post-infarction neutrophil function and the associated consequences on post-infarction myocardial inflammation.

Heart Failure with Preserved LVEF

Patients with heart failure with preserved LV ejection fraction (HFpEF) have a similar, but less severe profile of derangements in neurohormonal activity, exercise capacity, and quality of life compared to patients with heart failure and reduced LV ejection fraction (HFrEF).67 Despite these similarities, clinical trials of RAAS inhibitors and beta blockers have failed to demonstrate statistically significant improvements in survival, while MRAs have shown promise, albeit controversial.68

Modulation of the NPS in patients with HFpEF is appealing, as NP activity appears to adhere to a similar paradigm in HFpEF as in HFrEF. Elevated BNP levels predict adverse clinical outcomes in patients with HFpEF as they do in patients with HFrEF.69 Though BNP levels tend to be lower in patients with HFpEF than in patients with HFrEF, a given BNP level is similarly prognostic.70 In addition to the previously described actions of the NPs in heart failure, the action of the NPS on cardiomyocyte protein-kinase G (PKG) may suggest a particular pathway of benefit. LV biopsy specimens in HFpEF reveal low activity of PKG, a powerful regulator of titin stiffness, and this is associated with an elevation in cardiomyocyte resting passive tension.71 PKG activity is regulated by the availability of cyclic GMP, which is elaborated by guanylate cyclase (GC), occuring in both soluble (sGC) or receptor-bound (rGC) forms. There appear to be separate pools of PKG activity, with nitric oxide synthetase and donors of nitric oxide stimulating sGC and NPs signaling via rGC with separate regulation by phosphodiesterase 9.72 Even as enthusiasm builds for agents that more effectively deliver nitric oxide to hypoxic tissues (eg: inorganic nitrate and inorganic nitrite) or directly stimulate soluble guanylate cyclase (eg: riociguat), the signaling of NPs through the rGC-cGMP-PKA pathway may represent a unique pathway to modulate cardiomyocyte function.73

Phase 2 clinical trial data exist for the use of ARNI in patients with HFpEF. The PARAMOUNT study randomized 308 patients with HFpEF (LVEF ≥45%), hypertension, and elevated NTproBNP > 400 pg/mL to therapy with sacubitril/valsartan or valsartan. Use of sacubitril/valsartan was associated with greater decline in NTproBNP at 12 weeks, greater improvement in left atrial volumes at 36 weeks, no increase in clinical adverse events, and lower levels of high sensitivity troponin.74,75 Moreover, the favorable changes in NYHA class, renal function, left atrial volumes, and NT-proBNP were not correlated with changes in blood pressure, suggesting a more complex mechanism of benefit.76 PARAGON-HF is the subsequent ongoing phase 3 trial of sacubitril/valsartan use in HFpEF (NCT01920711). As the largest HFpEF trial ever conducted, it will enroll 4,600 subjects with NYHA Class II–IV HF with an LVEF ≥45% and compare the rate of CV death and HF hospitalization among subjects treated with sacubitril/valsartan vs. valsartan.

Hypertension

The biologic rationale for blockade of the RAAS and potentiation of the NPS with regard to blood pressure lowering is described in previous sections of this review. At present, three randomized controlled trials of sacubitril/valsartan therapy for hypertension have been reported. A comparison of sacubitril/valsartan vs. valsartan vs. placebo in 1,328 patients with mild-moderate hypertension demonstrated greater lowering of blood pressure in sacubitril/valsartan treated subjects compared to subjects treated with the comparable bioactive dose of valsartan.77 Importantly, no cases of angioedema were reported. Subsequently, Kario et al demonstrated the effectiveness of sacubitril/valsartan compared to placebo in 389 hypertensive Asian subjects in lowering daytime and nighttime blood pressures with no cases of angioedema.78 Finally, the recently-reported PARAMETER study demonstrated that in a group of 454 elderly hypertensive patients with elevated pulse pressure ≥60 mmHg, sacubitril/valsartan was more effective than olmesartan at lowering central aortic blood pressure and 24-hour ambulatory blood pressure at 12 weeks and required fewer add-on antihypertensive therapies over the course of 52 weeks.79 While these data are promising, to date no phase 3 clinical trial of sacubitril/valsartan for the treatment of hypertension is planned.

Potential Non-Cardiac Indications

Renal Disease

In the normal kidney, autoregulation permits the maintenance of GFR across a range of blood pressures, however heart failure, particularly in the setting of therapies that reduce the action of angiotensin II at the glomerulus, is characterized by altered renal function and heightened sensitivity of GFR to reductions in blood pressure and renal perfusion.80 Thus, use of RAAS inhibiting drugs in heart failure may lower GFR. Even as GFR may fall with RAAS blockade in HF, it is evident that compared with placebo ACEI/ARB use provides a clinical benefit for patients with stage III and possibly stage IV CKD81 and that continued ACEI/ARB use even in the face of worsening renal function is beneficial over discontinuation.82 Meanwhile, the NPS, particularly ANP, has been demonstrated to effect an increase in GFR through glomerular afferent arteriolar dilatation and efferent arteriolar constriction in both dog83 and rat models.84 Further, in healthy humans ANP infusion85 and BNP infusion86 have been demonstrated to improve GFR. Through direct effects on the renal vasculature and indirect effects on the RAAS, the NPS appears to be a pathway for treating renal dysfunction and it is tempting to think that dual RAAS inhibition and neprilysin inhibition might permit both improved heart failure outcomes and preserve renal function. What is the evidence for this?

The dual ACEI and neprilysin inhibitor omapatrilat attenuated the progression of renal failure more-so than ACEI in animal models87 and in the IMPRESS study was shown to result in fewer episodes of elevated serum creatinine than ACEI.41 While advancement of this agent was thwarted by angioedema concerns, there is reason to believe the successor ARNI may confer similar favorable effects. In the PARAMOUNT phase 2 trial of subjects with HFpEF and hypertension, treatment with sacubitril/valsartan as compared to valsartan resulted in significantly less decline in eGFR and lower levels of serum creatinine, albeit with no significant difference in cystatin C and a slightly higher urine albumin/creatinine ratio (UACR).88 The observed elevation in UACR was not seen in a study of hypertensive subjects without HFpEF43 and might relate to direct inhibitory effects of natriurietic peptides on glomerular mesangial cell proliferation and contraction.89,90 Finally, in the PARADIGM trial, despite being associated with more symptomatic hypotension, sacubitril/valsartan was associated with fewer episodes of elevated creatinine or serum potassium.46 In aggregate, these data suggest ARNI use should be no less indicated in patients with HF and CKD than ACEI/ARB and may possibly be preferable to these agents in patients with CKD.

Whether the action of ARNI on renal function in non-HF patients is favorable remains to be determined. Considerable data suggest a benefit of ACEI/ARB use on slowing the progression of renal failure among patients with proteinuria and chronic kidney disease, both among diabetics91 and non-diabetics.92 These benefits appear minimal or absent among patients with proteinuria < 500 mg/day.93 Even so, international treatment guidelines recommend the use of ACEI or ARB for the treatment of hypertension in all non-dialysis dependent chronic kidney disease even in settings where there is minimal or no proteinuria.94 It remains an open question whether combined neprilysin and RAAS inhibition may have additive effects, and this will first be studied among patients with CKD and proteinuria. The UK Heart and Renal Protection (HARP)-III is a randomized, controlled trial to be conducted in the UK that will compare the effectiveness of sacubitril/valsartan vs. irbesartan in preserving GFR over 12 months among 360 diabetic and nondiabetic patients with an initial GFR between 20 and 60 ml/m2/1.73m2 and a UACR >=20 mg/mmol (ISRCTN 11958993).

Cognition, Behavior, and Neurologic Disease

As previously described, owing to the broad expression and action of neprilysin, non-cardiovascular concerns have been raised for the use of sacubitril/valsartan. Of particular concern is the possible interaction of neprilysin inhibition and Alzheimer disesase. Neprilysin degrades Aβ peptides and oligomers and in animal models there is an inverse relationship between peripheral expression of neprilysin and brain amyloid burden.95,96 Further, animal models have suggested the possibility of a therapeutic benefit of neprilysin potentiation.97 In humans there are less certain and occasionally conflicting data on this paradigm of neprilysin activity inverse to Alzheimer disease progression.30 However, as neprilysin plays a salutary role in animal models of several amyloid deposition diseases such as age-related macular degeneration, cerebral amyloid angiopathy, and sensorimotor axonal polyneuropathy, concerns remain that ARNI may have long term unfavorable effects on neurologic function.

In healthy subjects sacubitril/valsartan does not increase CSF levels of the aggregable Aβ isoforms (1–42 and 1–40), but does significantly increase the concentration soluble CSF Aβ 1–38.98 Whether there are clinical consequences to neprilysin inhibition and the described changes in CSF Aβ through ARNI is unclear. A retrospective analysis of PARADIGM revealed no greater rate of dementia-related adverse events in the sacubitril/valsartan arm than the enalapril arm and instead showed that dementia-related AEs were linked to a higher burden of cardiovascular disease and associated risk factors as represented by coronary disease, stroke, atrial fibrillation, higher NT-proBNP levels, and lower eGFR.99 Given the potential for competing effects of Aβ-related disease and modification of the impact of cardiovascular comorbidity on cognition in patients treated with ARNI, ongoing trials will seek to better characterize the effects of these agents on cognition and brain structure and function. The ongoing PARAGON trial includes a Mini Mental State exam and the forthcoming PERSPECTIVE trial will employ a more powerful battery of cognitive tests as well as brain positron emission tomography imaging to the brain using florbetapir-18F to assess changes in amyloid plaque deposition over time (NCT02884206).

Finally, the role of neprilysin and ARNI in disorders of sleep in heart failure is an emerging area of interest. Sleep disordered breathing is common in heart failure, with approximately half of patients affected, the majority of whom have central sleep apnea (CSA).100 Greater elevations in left heart filling pressures are correlated with CSA101 and CSA is associated with reduced survival in heart failure.102 Unfortunately, treatment of HF patients with CSA using adaptive servoventilation was shown to increase mortality, limiting enthusiasm for nocturnal respiratory support in these patients.103 Cardiovascular pharmacotherapies have to date not been show to improve sleep disordered breathing.104 Elevated levels of NPs are associated with CSA in HF, but there are no data to demonstrate that potentiation of NPs has salutary effects on sleep.105,106 That said, in a rat model of sleep deprivation, there is evidence of neprilysin activation in the pituitary gland, potentially implicating neprilysin in sleep regulation.107 Neprilysin or neprilysin-like endopeptidases may also play a role in the regulation of circadian rhythm in Drosphophila models.108 Whether these limited and early observations translate to a role for neprilysin inhibition in the sleep-wake cycle or nocturnal breathing in heart failure is not clear. Currently trials are planned for measuring the impact of sacubitril/valsartan on apnea-hypopnea in patients with sleep apnea (ENTRESTO-SAS, NCT02916160) and daytime activity and nighttime actigraphy in patients with HFrEF treated with sacubitril/valsartan (AWAKE-HF, NCT02970669), potentially illuminating the action of neprilysin inhibition on sleep.

Conclusion

Neprilysin inhibition represents a powerful therapeutic tool in treating chronic heart failure with reduced LVEF and preliminary data suggest a potential role for the use of ARNI in a broader spectrum of cardiovascular and non-cardiovascular disease. Insights from a vast array of clinical trials over the course of the next several years will onstrate whether the promise of combined neprilysin and RAAS inhibition in these disease states will translate to clinical effectiveness.

Footnotes

Compliance with Ethics Guidelines

Conflict of Interest

Elizabeth Riddell and Justin M. Vader declare that they have no conflict of interest

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

References

  • 1.Teerlink JR, Bourge RC, Cleland JGF, Jondeau G, Krum H, Metra M, Connor CMO, Parker JD, Lewsey J, Frey A, Rainisio M. Effects of Tezosentan on Symptoms. Heart Fail. 2009;298:2009–2019. [Google Scholar]
  • 2.Prasad SK, Dargie HJ, Smith GC, Barlow MM, Grothues F, Groenning BA, Cleland JGF, Pennell DJ. Comparison of the dual receptor endothelin antagonist enrasentan with enalapril in asymptomatic left ventricular systolic dysfunction: a cardiovascular magnetic resonance study. Heart [Internet] 2006;92:798–803. doi: 10.1136/hrt.2004.049734. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16339819%5Cnhttp://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=PMC1860639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Anand PI, McMurray PJ, Cohn PJN, Konstam PMA, Notter T, Quitzau K, Ruschitzka F, L??scher PTF. 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;364:347–354. doi: 10.1016/S0140-6736(04)16723-8. [DOI] [PubMed] [Google Scholar]
  • 4.Mann DL, McMurray JJV, Packer M, Swedberg K, Borer JS, Colucci WS, Djian J, Drexler H, Feldman A, Kober L, Krum H, Liu P, Nieminen M, Tavazzi L, Van Veldhuisen DJ, Waldenstrom A, Warren M, Westheim A, Zannad F, Fleming T. Targeted Anticytokine Therapy in Patients with Chronic Heart Failure: Results of the Randomized Etanercept Worldwide Evaluation (RENEWAL) Circulation. 2004;109:1594–1602. doi: 10.1161/01.CIR.0000124490.27666.B2. [DOI] [PubMed] [Google Scholar]
  • 5.McMurray JJV, Krum H, Abraham WT, Dickstein K, Køber LV, Desai AS, Solomon SD, Greenlaw N, Ali MA, Chiang Y, Shao Q, Tarnesby G, Massie BM ATMOSPHERE Committees Investigators. Aliskiren, Enalapril, or Aliskiren and Enalapril in Heart Failure. N Engl J Med [Internet] 2016;374:1521–32. doi: 10.1056/NEJMoa1514859. Available from: http://www.nejm.org/doi/10.1056/NEJMoa1514859%5Cnhttp://www.ncbi.nlm.nih.gov/pubmed/27043774. [DOI] [PubMed] [Google Scholar]
  • 6.Inhibitor AE, Packer M, Poole-wilson PA, Armstrong PW, Cleland JGF, Horowitz JD, Massie BM, Ryde L, Thygesen K. Clinical Investigation and Reports Comparative Effects of Low and High Doses of the. Circulation. 2015;100:2312–2318. doi: 10.1161/01.cir.100.23.2312. [DOI] [PubMed] [Google Scholar]
  • 7.Konstam MA, Neaton JD, Dickstein K, Drexler H, Komajda M, Martinez FA, Riegger GA, Malbecq W, Smith RD, Guptha S, Poole-Wilson PA. Effects of high-dose versus low-dose losartan on clinical outcomes in patients with heart failure (HEAAL study): a randomised, double-blind trial. Lancet [Internet] 2009;374:1840–1848. doi: 10.1016/S0140-6736(09)61913-9. Available from: http://dx.doi.org/10.1016/S0140-6736(09)61913-9. [DOI] [PubMed] [Google Scholar]
  • 8.Mcmurray JJV, Ostergren J, Swedberg K, Granger CB. Effects of candesartan in patients with chronic heart failure and reduced lef. Lancet. 2003;362:767–771. doi: 10.1016/S0140-6736(03)14283-3. [DOI] [PubMed] [Google Scholar]
  • 9.Cohn JN, Tognoni G Investigators VHFT. A Randomized Trial of the Angiotensin-Receptor Blocker Valsartan in Chronic Heart Failure. N Engl J Med. 2001;345:1667–1675. doi: 10.1056/NEJMoa010713. [DOI] [PubMed] [Google Scholar]
  • 10.Gheorghiade M, Böhm M, Greene SJ, Fonarow GC, Lewis EF, Zannad F, Solomon SD, Baschiera F, Botha J, Hua Ta, Gimpelewicz CR, Jaumont X, Lesogor A, Maggioni AP. Effect of aliskiren on postdischarge mortality and heart failure readmissions among patients hospitalized for heart failure: the ASTRONAUT randomized trial. JAMA [Internet] 2013;309:1125–35. doi: 10.1001/jama.2013.1954. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23478743. [DOI] [PubMed] [Google Scholar]
  • 11.Hata N, Seino Y, Tsutamoto T, Hiramitsu S, Kaneko N, Yoshikawa T, Yokoyama H, Tanaka K, Mizuno K, Nejima J, Kinoshita M. Effects of carperitide on the long-term prognosis of patients with acute decompensated chronic heart failure: the PROTECT multicenter randomized controlled study. Circ J [Internet] 2008;72:1787–93. doi: 10.1253/circj.cj-08-0130. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18812677. [DOI] [PubMed] [Google Scholar]
  • 12.Nomura F, Kurobe N, Mori Y, Hikita A, Kawai M, Suwa M, Okutani Y. Multicenter prospective investigation on efficacy and safety of carperitide as a first-line drug for acute heart failure syndrome with preserved blood pressure: COMPASS: Carperitide Effects Observed Through Monitoring Dyspnea in Acute Decompensated Heart. Circ J. 2008;72:1777–1786. doi: 10.1253/circj.cj-07-0760. [DOI] [PubMed] [Google Scholar]
  • 13.Matsue Y, Kagiyama N, Yoshida K, Kume T, Okura H, Suzuki M, Matsumura A, Yoshida K, Hashimoto Y. Carperitide Is Associated With Increased In-Hospital Mortality in Acute Heart Failure: A Propensity Score-Matched Analysis. J Card Fail [Internet] 2015;21:859–864. doi: 10.1016/j.cardfail.2015.05.007. Available from: http://dx.doi.org/10.1016/j.cardfail.2015.05.007. [DOI] [PubMed] [Google Scholar]
  • 14.O’Connor CM, Starling RC, Hernandez AF, Armstrong PW, Dickstein K, Hasselblad V, Heizer GM, Komajda M, Massie BM, McMurray JJV, Nieminen MS, Reist CJ, Rouleau JL, Swedberg K, Adams KF, Anker SD, Atar D, Battler A, Botero R, Bohidar NR, Butler J, Clausell N, Corbalán R, Costanzo MR, Dahlstrom U, Deckelbaum LI, Diaz R, Dunlap ME, Ezekowitz JA, Feldman D, Felker GM, Fonarow GC, Gennevois D, Gottlieb SS, Hill JA, Hollander JE, Howlett JG, Hudson MP, Kociol RD, Krum H, Laucevicius A, Levy WC, Méndez GF, Metra M, Mittal S, Oh B-H, Pereira NL, Ponikowski P, Tang WHW, Tanomsup S, Teerlink JR, Triposkiadis F, Troughton RW, Voors AA, Whellan DJ, Zannad F, Califf RM. Effect of Nesiritide in Patients with Acute Decompensated Heart Failure. N Engl J Med [Internet] 2011;365:32–43. doi: 10.1056/NEJMoa1100171. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21732835%5Cnhttp://www.nejm.org/doi/abs/10.1056/NEJMoa1100171. [DOI] [PubMed] [Google Scholar]
  • 15.Colucci WS, Elkayam U, Horton DP, Abraham WT, Bourge RC, Johnson AD, Wagoner LE, Givertz MM, Liang C-S, Neibaur M, Haught WH, LeJemtel TH, Group NS. Intravenous Nesiritide, a natriurietic peptide, in the treatment of decompensated congestive heart failure. N Engl J Med. 2000;343:246–53. doi: 10.1056/NEJM200007273430403. [DOI] [PubMed] [Google Scholar]
  • 16.Investigators V. Intravenous Nesiritide vs Nitroglycerin for Treatment of Decompensated Congestive Heart Failure: A randomized controlled trial. JAMA J Am Med Assoc. 2015;287:1531–77. doi: 10.1001/jama.287.12.1531. [DOI] [PubMed] [Google Scholar]
  • 17.Rademaker MT, Charles CJ, Espiner EA, Nicholls MG, Richards AM, Kosoglou T. Neutral Endopeptidase Inhibition: Augmented Atrial and Brain Natriuretic Peptide, Haemodynamic and Natriuretic Responses in Ovine Heart Failure. Clin Sci [Internet] 1996;91:283 LP-291. doi: 10.1042/cs0910283. Available from: http://www.clinsci.org/content/91/3/283.abstract. [DOI] [PubMed] [Google Scholar]
  • 18.Cruden NLM, Fox KAA, Ludlam CA, Johnston NR, Newby DE. Neutral Endopeptidase Inhibition Augments Vascular Actions of Bradykinin in Patients Treated With Angiotensin-Converting Enzyme Inhibition. Hypertension [Internet] 2004;44:913 LP-918. doi: 10.1161/01.HYP.0000146483.78994.56. Available from: http://hyper.ahajournals.org/content/44/6/913.abstract. [DOI] [PubMed] [Google Scholar]
  • 19.McDowell G, Coutie W, Shaw C, Buchanan KD, Struthers AD, Nicholls DP. The effect of the neutral endopeptidase inhibitor drug, candoxatril, on circulating levels of two of the most potent vasoactive peptides. Br J Clin Pharmacol [Internet] 1997;43:329–332. doi: 10.1046/j.1365-2125.1997.00545.x. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2042739/ [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Maric C, Zheng W, Walther T. Interactions between angiotensin ll and atrial natriuretic peptide in renomedullary interstitial cells: the role of neutral endopeptidase. Nephron Physiol. 2006;103:p149–56. doi: 10.1159/000092457. [DOI] [PubMed] [Google Scholar]
  • 21.Kuhn M. Molecular physiology of natriuretic peptide signalling. Basic Res Cardiol. 2004;99:76–82. doi: 10.1007/s00395-004-0460-0. [DOI] [PubMed] [Google Scholar]
  • 22.Roques BP, Fournie-Zaluski MC, Soroca E, Lecomte JM, Malfroy B, Llorens C, Schwartz J-C. The enkephalinase inhibitor thiorphan shows antinociceptive activity in mice. Nature [Internet] 1980;288:286–288. doi: 10.1038/288286a0. Available from: http://dx.doi.org/10.1038/288286a0. [DOI] [PubMed] [Google Scholar]
  • 23.Ksander GM, Ghai RD, deJesus R, Diefenbacher C, Yuan A, Berry C, Sakane Y, Trapani A. Dicarboxylic Acid Dipeptide Neutral Endopeptidase Inhibitors. J Med Chem [Internet] 1995;38:1689–1700. doi: 10.1021/jm00010a014. Available from: http://dx.doi.org/10.1021/jm00010a014. [DOI] [PubMed] [Google Scholar]
  • 24.Cavero PG, Margulies KB, Winaver J, Seymour AA, Delaney NG, Burnett JC., Jr Cardiorenal actions of neutral endopeptidase inhibition in experimental congestive heart failure. Circulation. 1990;82:196–201. doi: 10.1161/01.cir.82.1.196. [DOI] [PubMed] [Google Scholar]
  • 25.Seymour AA, Asaad MM, Lanoce VM, Langenbacher KM, Fennell SA, Rogers WL. Systemic hemodynamics, renal function and hormonal levels during inhibition of neutral endopeptidase 3.4.24.11 and angiotensin-converting enzyme in conscious dogs with pacing-induced heart failure. J Pharmacol Exp Ther [Internet] 1993;266:872 LP-883. Available from: http://jpet.aspetjournals.org/content/266/2/872.abstract. [PubMed] [Google Scholar]
  • 26.Münzel T, Kurz S, Holtz J, Busse R, Steinhauer H, Just H, Drexler H. Neurohormonal inhibition and hemodynamic unloading during prolonged inhibition of ANF degradation in patients with severe chronic heart failure. Circulation. 1992;86:1089–1098. doi: 10.1161/01.cir.86.4.1089. [DOI] [PubMed] [Google Scholar]
  • 27.Northridge D, Alabaster C, Connell JC, Dilly S, Lever A, Jardine A, Barclay P, Dargie H, Findlay I, Samuels GR. EFFECTS OF UK 69–578: A NOVEL ATRIOPEPTIDASE INHIBITOR. Lancet [Internet] 1989;334:591–593. doi: 10.1016/s0140-6736(89)90714-9. Available from: file://www.sciencedirect.com/science/article/pii/S0140673689907149. [DOI] [PubMed] [Google Scholar]
  • 28.Ferro CJ, Spratt JC, Haynes WG, Webb DJ. Inhibition of Neutral Endopeptidase Causes Vasoconstriction of Human Resistance Vessels In Vivo. Circulation [Internet] 1998;97:2323–2330. doi: 10.1161/01.cir.97.23.2323. Available from: http://circ.ahajournals.org/content/97/23/2323.short%5Cnhttp://circ.ahajournals.org/cgi/doi/10.1161/01.CIR.97.23.2323. [DOI] [PubMed] [Google Scholar]
  • 29.Semenov AG, Katrukha AG. Different Susceptibility of B-Type Natriuretic Peptide (BNP) and BNP Precursor (proBNP) to Cleavage by Neprilysin: The N-Terminal Part Does Matter. Clin Chem [Internet] 2016;62:617 LP-622. doi: 10.1373/clinchem.2016.254524. Available from: http://clinchem.aaccjnls.org/content/62/4/617.abstract. [DOI] [PubMed] [Google Scholar]
  • 30.Campbell DJ. Long-term neprilysin inhibition — implications for ARNIs. Nat Rev Cardiol [Internet] 2016:1–16. doi: 10.1038/nrcardio.2016.200. Available from: http://www.nature.com/doifinder/10.1038/nrcardio.2016.200. [DOI] [PubMed]
  • 31.Shen R, Sumitomo M, Dai J, Harris A, Kaminetzky D, Gao M, Burnstein KL, Nanus DM. Androgen-induced growth inhibition of androgen receptor expressing androgen-independent prostate cancer cells is mediated by increased levels of neutral endopeptidase. Endocrinology. 2000;141:1699–1704. doi: 10.1210/endo.141.5.7463. [DOI] [PubMed] [Google Scholar]
  • 32.Stephen HM, Khoury RJ, Majmudar PR, Blaylock T, Hawkins K, Salama MS, Cosminsky B, Utreja NK. Epigenetic suppression of neprilysin regulates breast cancer invasion. Oncogensis [Internet] 2016;5:e207–10. doi: 10.1038/oncsis.2016.16. Available from: http://dx.doi.org/10.1038/oncsis.2016.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Meng F, DeMorrow S, Venter J, Frampton G, Han Y, Francis H, Standeford H, Avila S, McDaniel K, McMillin M, Afroze S, Guerrier M, Quezada M, Ray D, Kennedy L, Hargrove L, Glaser S, Alpini G. Overexpression of membrane metalloendopeptidase inhibits substance P stimulation of cholangiocarcinoma growth. Am J Physiol - Gastrointest Liver Physiol [Internet] 2014;306:G759 LP-G768. doi: 10.1152/ajpgi.00018.2014. Available from: http://ajpgi.physiology.org/content/306/9/G759.abstract. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Terauchi M, Kajiyama H, Shibata K, Ino K, Mizutani S, Kikkawa F. Anti-Progressive Effect of Neutral Endopeptidase 24.11 (NEP/CD10) on Cervical Carcinoma in vitro and in vivo. Oncology [Internet] 2005;69:52–62. doi: 10.1159/000087476. Available from: http://www.karger.com/DOI/10.1159/000087476. [DOI] [PubMed] [Google Scholar]
  • 35.Kajiyama H, Shibata K, Terauchi M, Morita T, Ino K, Mizutani S, Kikkawa F. Neutral Endopeptidase 24.11/CD10 Suppresses Progressive Potential in Ovarian Carcinoma <em>In vitro</em> and <em>In vivo</em>. Clin Cancer Res [Internet] 2005;11:1798 LP-1808. doi: 10.1158/1078-0432.CCR-04-2395. Available from: http://clincancerres.aacrjournals.org/content/11/5/1798.abstract. [DOI] [PubMed] [Google Scholar]
  • 36.Seymour AA, Swerdel JN, Abboa-Offei B. Antihypertensive activity during inhibition of neutral endopeptidase and angiotensin converting enzyme. J Cardiovasc Pharmacol. 1991;17:456–465. doi: 10.1097/00005344-199103000-00015. [DOI] [PubMed] [Google Scholar]
  • 37.Rademaker MT, Charles CJ, Espiner EA, Nicholls MG, Richards AM, Kosoglou T. Combined neutral endopeptidase and angiotensin-converting enzyme inhibition in heart failure: role of natriuretic peptides and angiotensin II. J Cardiovasc Pharmacol. 1998;31:116–125. doi: 10.1097/00005344-199801000-00017. [DOI] [PubMed] [Google Scholar]
  • 38.Trippodo NC, Fox M, Monticello TM, Panchal BC, Asaad MM. Vasopeptidase inhibition with omapatrilat improves cardiac geometry and survival in cardiomyopathic hamsters more than does ACE inhibition with captopril. J Cardiovasc Pharmacol. 1999;34:782–790. doi: 10.1097/00005344-199912000-00003. [DOI] [PubMed] [Google Scholar]
  • 39.Campese VM, Lasseter KC, Ferrario CM, Smith WB, Ruddy MC, Grim CE, Smith RD, Vargas R, Habashy MF, Vesterqvist O, Delaney CL, Liao WC. Omapatrilat versus lisinopril: efficacy and neurohormonal profile in salt-sensitive hypertensive patients. Hypertension. 2001;38:1342–1348. doi: 10.1161/hy1201.096569. [DOI] [PubMed] [Google Scholar]
  • 40.Packer M, Califf RM, Konstam MA, Krum H, McMurray JJ, Rouleau J-L, Swedberg K. 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;106:920–926. doi: 10.1161/01.cir.0000029801.86489.50. [DOI] [PubMed] [Google Scholar]
  • 41.Rouleau JL, Pfeffer MA, Stewart DJ, Isaac D, Sestier F, Kerut EK, Porter CB, Proulx G, Qian C, Block AJ. Comparison of vasopeptidase inhibitor, omapatrilat, and lisinopril on exercise tolerance and morbidity in patients with heart failure: IMPRESS randomised trial. Lancet. 2000;356:615–620. doi: 10.1016/s0140-6736(00)02602-7. [DOI] [PubMed] [Google Scholar]
  • 42.Kostis JB, Packer M, Black HR, Schmieder R, Henry D, Levy E. Omapatrilat and enalapril in patients with hypertension: the Omapatrilat Cardiovascular Treatment vs. Enalapril (OCTAVE) trial. Am J Hypertens. 2004;17:103–111. doi: 10.1016/j.amjhyper.2003.09.014. [DOI] [PubMed] [Google Scholar]
  • 43.Ruilope LM, Dukat A, Bhm M, Lacourcire Y, Gong J, Lefkowitz MP. Blood-pressure reduction with LCZ696, a novel dual-acting inhibitor of the angiotensin II receptor and neprilysin: a randomised, double-blind, placebo-controlled, active comparator study. Lancet. 2010;375:1255–1266. doi: 10.1016/S0140-6736(09)61966-8. [DOI] [PubMed] [Google Scholar]
  • 44.Jhund PS, Claggett B, Packer M, Zile MR, Voors AA, Pieske B, Lefkowitz M, Shi V, Bransford T, McMurray JJV, Solomon SD. Independence of the blood pressure lowering effect and efficacy of the angiotensin receptor neprilysin inhibitor, LCZ696, in patients with heart failure with preserved ejection fraction: An analysis of the PARAMOUNT trial. Eur J Heart Fail. 2014;16:671–677. doi: 10.1002/ejhf.76. [DOI] [PubMed] [Google Scholar]
  • 45.Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DEJ, Colvin MM, Drazner MH, Filippatos G, Fonarow GC, Givertz MM, Hollenberg SM, Lindenfeld J, Masoudi FA, McBride PE, Peterson PN, Stevenson LW, Westlake C. 2016 ACC/AHA/HFSA Focused Update on New Pharmacological Therapy for Heart Failure: An 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. J Am Coll Cardiol. 2016;68:1476–1488. doi: 10.1016/j.jacc.2016.05.011. [DOI] [PubMed] [Google Scholar]
  • 46.McMurray JJV, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, Rouleau JL, Shi VC, Solomon SD, Swedberg K, Zile MR. Angiotensin Neprilysin Inhibition versus Enalapril in Heart Failure. N Engl J Med [Internet] 2014;371:993–1004. doi: 10.1056/NEJMoa1409077. Available from: http://dx.doi.org/10.1056/NEJMoa1409077. [DOI] [PubMed] [Google Scholar]
  • 47.Desai AS, Claggett BL, Packer M, Zile MR, Rouleau JL, Swedberg K, Shi V, Lefkowitz M, Starling R, Teerlink J, McMurray JJV, Solomon SD. Influence of Sacubitril/Valsartan (LCZ696) on 30-Day Readmission After Heart Failure Hospitalization. J Am Coll Cardiol. 2016;68:241–248. doi: 10.1016/j.jacc.2016.04.047. [DOI] [PubMed] [Google Scholar]
  • 48.Solomon SD, Claggett B, Packer M, Desai A, Zile MR, Swedberg K, Rouleau J, Shi V, Lefkowitz M, McMurray JJV. Efficacy of Sacubitril/Valsartan Relative to a Prior Decompensation: The PARADIGM-HF Trial. JACC Heart Fail. 2016;4:816–822. doi: 10.1016/j.jchf.2016.05.002. [DOI] [PubMed] [Google Scholar]
  • 49.Gattis WA, O’Connor CM, Gallup DS, Hasselblad V, Gheorghiade M. Predischarge initiation of carvedilol in patients hospitalized for decompensated heart failure: results of the Initiation Management Predischarge: Process for Assessment of Carvedilol Therapy in Heart Failure (IMPACT-HF) trial. J Am Coll Cardiol. 2004;43:1534–1541. doi: 10.1016/j.jacc.2003.12.040. [DOI] [PubMed] [Google Scholar]
  • 50.Butler J, Arbogast PG, Daugherty J, Jain MK, Ray WA, Griffin MR. Outpatient utilization of angiotensin-converting enzyme inhibitors among heart failure patients after hospital discharge. J Am Coll Cardiol. 2004;43:2036–2043. doi: 10.1016/j.jacc.2004.01.041. [DOI] [PubMed] [Google Scholar]
  • 51.Vader JM, LaRue SJ, Stevens SR, Mentz RJ, DeVore AD, Lala A, Groarke JD, AbouEzzeddine OF, Dunlay SM, Grodin JL, Davila-Roman VG, de Las Fuentes L. Timing and Causes of Readmission After Acute Heart Failure Hospitalization-Insights From the Heart Failure Network Trials. J Card Fail. 2016;22:875–883. doi: 10.1016/j.cardfail.2016.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Simpson J, Jhund PS, Silva Cardoso J, Martinez F, Mosterd A, Ramires F, Rizkala AR, Senni M, Squire I, Gong J, Lefkowitz MP, Shi VC, Desai AS, Rouleau JL, Swedberg K, Zile MR, McMurray JJV, Packer M, Solomon SD. Comparing LCZ696 with enalapril according to baseline risk using the MAGGIC and EMPHASIS-HF risk scores: an analysis of mortality and morbidity in PARADIGM-HF. J Am Coll Cardiol. 2015;66:2059–2071. doi: 10.1016/j.jacc.2015.08.878. [DOI] [PubMed] [Google Scholar]
  • 53.Solomon SD, Claggett B, Desai AS, Packer M, Zile M, Swedberg K, Rouleau JL, Shi VC, Starling RC, Kozan O, Dukat A, Lefkowitz MP, McMurray JJV. Influence of Ejection Fraction on Outcomes and Efficacy of Sacubitril/Valsartan (LCZ696) in Heart Failure with Reduced Ejection Fraction: 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. doi: 10.1161/CIRCHEARTFAILURE.115.002744. [DOI] [PubMed] [Google Scholar]
  • 54.Konstam MA, Kronenberg MW, Rousseau MF, Udelson JE, Melin J, Stewart D, Dolan N, Edens TR, Ahn S, Kinan D. Effects of the angiotensin converting enzyme inhibitor enalapril on the long-term progression of left ventricular dilatation in patients with asymptomatic systolic dysfunction. SOLVD (Studies of Left Ventricular Dysfunction) Investigators. Circulation. 1993;88:2277–2283. doi: 10.1161/01.cir.88.5.2277. [DOI] [PubMed] [Google Scholar]
  • 55.O'Gara PT, Kushner FG, Ascheim DD, Casey DE, Chung MK, de Lemos JA, Ettinger SM, Fang JC, Fesmire FM, Franklin BA, Granger CB, Krumholz HM, Linderbaum JA, Morrow DA, Newby LK, Ornato JP, Ou N, Radford MJ, Tamis-Holland JE, Tommaso CL, Tracy CM, Woo YJ, Zhao DX. 2013 ACCF/AHA Guideline for the Management of ST-Elevation Myocardial Infarction. J Am Coll Cardiol [Internet] 2013;61:e78 LP-e140. doi: 10.1016/j.jacc.2012.11.019. Available from: http://www.onlinejacc.org/content/61/4/e78.abstract. [DOI] [PubMed] [Google Scholar]
  • 56.Amsterdam EA, Wenger NK, Brindis RG, Casey DE, Ganiats TG, Holmes DR, Jaffe AS, Jneid H, Kelly RF, Kontos MC, Levine GN, Liebson PR, Mukherjee D, Peterson ED, Sabatine MS, Smalling RW, Zieman SJ. 2014 AHA/ACC Guideline for the Management of Patients With Non-ST-Elevation Acute Coronary Syndromes. Circulation [Internet] 2014 doi: 10.1016/j.jacc.2014.09.017. Available from: http://circ.ahajournals.org/content/early/2014/09/22/CIR.0000000000000134.abstract. [DOI] [PubMed]
  • 57.Kober L, Torp-Pedersen C, Carlsen JE, Bagger H, Eliasen P, Lyngborg K, Videbaek J, Cole DS, Auclert L, Pauly NC. A clinical trial of the angiotensin-converting-enzyme inhibitor trandolapril in patients with left ventricular dysfunction after myocardial infarction. Trandolapril Cardiac Evaluation (TRACE) Study Group. N Engl J Med. 1995;333:1670–1676. doi: 10.1056/NEJM199512213332503. [DOI] [PubMed] [Google Scholar]
  • 58.Pfeffer MA, Braunwald E, Moyé LA, Basta L, Brown EJ, Cuddy TE, Davis BR, Geltman EM, Goldman S, Flaker GC, Klein M, Lamas GA, Packer M, Rouleau J, Rouleau JL, Rutherford J, Wertheimer JH, Hawkins CM. Effect of Captopril on Mortality and Morbidity in Patients with Left Ventricular Dysfunction after Myocardial Infarction. N Engl J Med [Internet] 1992;327:669–677. doi: 10.1056/NEJM199209033271001. Available from: http://dx.doi.org/10.1056/NEJM199209033271001. [DOI] [PubMed] [Google Scholar]
  • 59.Pfeffer MA, McMurray JJV, Velazquez EJ, Rouleau J-L, Kober L, Maggioni AP, Solomon SD, Swedberg K, Van de Werf F, White H, Leimberger JD, Henis M, Edwards S, Zelenkofske S, Sellers MA, Califf RM. Valsartan, captopril, or both in myocardial infarction complicated by heart failure, left ventricular dysfunction, or both. N Engl J Med. 2003;349:1893–1906. doi: 10.1056/NEJMoa032292. [DOI] [PubMed] [Google Scholar]
  • 60.Yusuf S, Teo KK, Pogue J, Dyal L, Copland I, Schumacher H, Dagenais G, Sleight P, Anderson C. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med. 2008;358:1547–1559. doi: 10.1056/NEJMoa0801317. [DOI] [PubMed] [Google Scholar]
  • 61.de Lemos JA, Morrow DA. Brain Natriuretic Peptide Measurement in Acute Coronary Syndromes. Circulation [Internet] 2002;106:2868 LP-2870. doi: 10.1161/01.cir.0000042763.07757.c0. Available from: http://circ.ahajournals.org/content/106/23/2868.abstract. [DOI] [PubMed] [Google Scholar]
  • 62.Möllmann H, Nef HM, Kostin S, Dragu A, Maack C, Weber M, Troidl C, Rolf A, Elsässer A, Böhm M, Brantner R, Hamm CW, Holubarsch CJF. Ischemia triggers BNP expression in the human myocardium independent from mechanical stress. Int J Cardiol [Internet] 2017;143:289–297. doi: 10.1016/j.ijcard.2009.03.012. Available from: http://dx.doi.org/10.1016/j.ijcard.2009.03.012. [DOI] [PubMed] [Google Scholar]
  • 63.Nishikimi T, Maeda N, Matsuoka H. The role of natriuretic peptides in cardioprotection. Cardiovasc Res [Internet] 2006;69:318–328. doi: 10.1016/j.cardiores.2005.10.001. Available from: http://dx.doi.org/10.1016/j.cardiores.2005.10.001. [DOI] [PubMed] [Google Scholar]
  • 64.Kasama S, Toyama T, Hatori T, Sumino H, Kumakura H, Takayama Y, Ichikawa S, Suzuki T, Kurabayashi M. Effects of intravenous atrial natriuretic peptide on cardiac sympathetic nerve activity and left ventricular remodeling in patients with first anterior acute myocardial infarction. J Am Coll Cardiol. 2007;49:667–674. doi: 10.1016/j.jacc.2006.09.048. [DOI] [PubMed] [Google Scholar]
  • 65.Chen HH, Martin FL, Gibbons RJ, Schirger JA, Wright RS, Schears RM, Redfield MM, Simari RD, Lerman A, Cataliotti A, Burnett JCJ. Low-dose nesiritide in human anterior myocardial infarction suppresses aldosterone and preserves ventricular function and structure: a proof of concept study. Heart. 2009;95:1315–1319. doi: 10.1136/hrt.2008.153916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.von Lueder TG, Wang BH, Kompa AR, Huang L, Webb R, Jordaan P, Atar D, Krum H. Angiotensin Receptor Neprilysin Inhibitor LCZ696 Attenuates Cardiac Remodeling and Dysfunction After Myocardial Infarction by Reducing Cardiac Fibrosis and Hypertrophy. Circ Hear Fail [Internet] 2015;8:71–78. doi: 10.1161/CIRCHEARTFAILURE.114.001785. Available from: http://circheartfailure.ahajournals.org/content/8/1/71.abstract. [DOI] [PubMed] [Google Scholar]
  • 67.Kitzman DW, Little WC, Brubaker PH, Anderson RT, Hundley WG, Marburger CT, Brosnihan B, Morgan TM, Stewart KP. Pathophysiological characterization of isolated diastolic heart failure in comparison to systolic heart failure. JAMA. 2002;288:2144–2150. doi: 10.1001/jama.288.17.2144. [DOI] [PubMed] [Google Scholar]
  • 68.Pitt B, Pfeffer MA, Assmann SF, Boineau R, Anand IS, Claggett B, Clausell N, Desai AS, Diaz R, Fleg JL, Gordeev I, Harty B, Heitner JF, Kenwood CT, Lewis EF, O’Meara E, Probstfield JL, Shaburishvili T, Shah SJ, Solomon SD, Sweitzer NK, Yang S, McKinlay SM. Spironolactone for heart failure with preserved ejection fraction. N Engl J Med. 2014;370:1383–1392. doi: 10.1056/NEJMoa1313731. [DOI] [PubMed] [Google Scholar]
  • 69.Grewal J, McKelvie RS, Persson H, Tait P, Carlsson J, Swedberg K, Ostergren J, Lonn E. Usefulness of N-terminal pro-brain natriuretic Peptide and brain natriuretic peptide to predict cardiovascular outcomes in patients with heart failure and preserved left ventricular ejection fraction. Am J Cardiol. 2008;102:733–737. doi: 10.1016/j.amjcard.2008.04.048. [DOI] [PubMed] [Google Scholar]
  • 70.van Veldhuisen DJ, Linssen GCM, Jaarsma T, van Gilst WH, Hoes AW, Tijssen JGP, Paulus WJ, Voors AA, Hillege HL. B-Type Natriuretic Peptide and Prognosis in Heart Failure Patients With Preserved and Reduced Ejection Fraction. J Am Coll Cardiol [Internet] 2013;61:1498 LP-1506. doi: 10.1016/j.jacc.2012.12.044. Available from: http://www.onlinejacc.org/content/61/14/1498.abstract. [DOI] [PubMed] [Google Scholar]
  • 71.van Heerebeek L, Hamdani N, Falcao-Pires I, Leite-Moreira AF, Begieneman MPV, Bronzwaer JGF, van der Velden J, Stienen GJM, Laarman GJ, Somsen A, Verheugt FWA, Niessen HWM, Paulus WJ. Low myocardial protein kinase G activity in heart failure with preserved ejection fraction. Circulation. 2012;126:830–839. doi: 10.1161/CIRCULATIONAHA.111.076075. [DOI] [PubMed] [Google Scholar]
  • 72.Lee DI, Zhu G, Sasaki T, Cho G-S, Hamdani N, Holewinski R, Jo S-H, Danner T, Zhang M, Rainer PP, Bedja D, Kirk JA, Ranek MJ, Dostmann WR, Kwon C, Margulies KB, Van Eyk JE, Paulus WJ, Takimoto E, Kass DA. Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease. Nature. 2015;519:472–476. doi: 10.1038/nature14332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Shah SJ, Kitzman DW, Borlaug BA, van Heerebeek L, Zile MR, Kass DA, Paulus WJ. Phenotype-Specific Treatment of Heart Failure With Preserved Ejection Fraction: A Multiorgan Roadmap. Circulation. 2016;134:73–90. doi: 10.1161/CIRCULATIONAHA.116.021884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Jhund PS, Claggett BL, Voors AA, Zile MR, Packer M, Pieske BM, Kraigher-Krainer E, Shah AM, Prescott MF, Shi V, Lefkowitz M, McMurray JJV, Solomon SD. Elevation in high-sensitivity troponin T in heart failure and preserved ejection fraction and influence of treatment with the angiotensin receptor neprilysin inhibitor LCZ696. Circ Heart Fail. 2014;7:953–959. doi: 10.1161/CIRCHEARTFAILURE.114.001427. [DOI] [PubMed] [Google Scholar]
  • 75.Solomon SD, Zile M, Pieske B, Voors A, Shah A, Kraigher-Krainer E, Shi V, Bransford T, Takeuchi M, Gong J, Lefkowitz M, Packer M, McMurray JJV. The angiotensin receptor neprilysin inhibitor LCZ696 in heart failure with preserved ejection fraction: a phase 2 double-blind randomised controlled trial. Lancet. 2012;380:1387–1395. doi: 10.1016/S0140-6736(12)61227-6. [DOI] [PubMed] [Google Scholar]
  • 76.Jhund PS, Claggett B, Packer M, Zile MR, Voors AA, Pieske B, Lefkowitz M, Shi V, Bransford T, McMurray JJV, Solomon SD. Independence of the blood pressure lowering effect and efficacy of the angiotensin receptor neprilysin inhibitor, LCZ696, in patients with heart failure with preserved ejection fraction: an analysis of the PARAMOUNT trial. Eur J Heart Fail. 2014;16:671–677. doi: 10.1002/ejhf.76. [DOI] [PubMed] [Google Scholar]
  • 77.Ruilope LM, Dukat A, Bohm M, Lacourciere Y, Gong J, Lefkowitz MP. Blood-pressure reduction with LCZ696, a novel dual-acting inhibitor of the angiotensin II receptor and neprilysin: a randomised, double-blind, placebo-controlled, active comparator study. Lancet. 2010;375:1255–1266. doi: 10.1016/S0140-6736(09)61966-8. [DOI] [PubMed] [Google Scholar]
  • 78.Kario K, Sun N, Chiang F-T, Supasyndh O, Baek SH, Inubushi-Molessa A, Zhang Y, Gotou H, Lefkowitz M, Zhang J. Efficacy and safety of LCZ696, a first-in-class angiotensin receptor neprilysin inhibitor, in Asian patients with hypertension: a randomized, double-blind, placebo-controlled study. Hypertension. 2014;63:698–705. doi: 10.1161/HYPERTENSIONAHA.113.02002. [DOI] [PubMed] [Google Scholar]
  • 79.Williams B, Cockcroft JR, Kario K, Zappe DH, Brunel PC, Wang Q, Guo W. Effects of Sacubitril/Valsartan Versus Olmesartan on Central Hemodynamics in the Elderly With Systolic Hypertension: The PARAMETER Study. Hypertension. 2017:69. doi: 10.1161/HYPERTENSIONAHA.116.08556. [DOI] [PubMed] [Google Scholar]
  • 80.Palmer BF. Renal Dysfunction Complicating the Treatment of Hypertension. N Engl J Med [Internet] 2002;347:1256–1261. doi: 10.1056/NEJMra020676. Available from: http://dx.doi.org/10.1056/NEJMra020676. [DOI] [PubMed] [Google Scholar]
  • 81.Damman K, Tang WHW, Felker GM, Lassus J, Zannad F, Krum H, McMurray JJV. Current evidence on treatment of patients with chronic systolic heart failure and renal insufficiency: practical considerations from published data. J Am Coll Cardiol. 2014;63:853–871. doi: 10.1016/j.jacc.2013.11.031. [DOI] [PubMed] [Google Scholar]
  • 82.Clark H, Krum H, Hopper I. Worsening renal function during renin-angiotensin-aldosterone system inhibitor initiation and long-term outcomes in patients with left ventricular systolic dysfunction. Eur J Heart Fail. 2014;16:41–48. doi: 10.1002/ejhf.13. [DOI] [PubMed] [Google Scholar]
  • 83.Ohishi K, Hishida A, Honda N. Direct vasodilatory action of atrial natriuretic factor on canine glomerular afferent arterioles. Am J Physiol. 1988;255:F415–20. doi: 10.1152/ajprenal.1988.255.3.F415. [DOI] [PubMed] [Google Scholar]
  • 84.Marin-Grez M, Fleming JT, Steinhausen M. Atrial natriuretic peptide causes pre-glomerular vasodilatation and post-glomerular vasoconstriction in rat kidney. Nature. 1986;324:473–476. doi: 10.1038/324473a0. [DOI] [PubMed] [Google Scholar]
  • 85.Pham I, Sediame S, Maistre G, Roudot-Thoraval F, Chabrier PE, Carayon A, Adnot S. Renal and vascular effects of C-type and atrial natriuretic peptides in humans. Am J Physiol. 1997;273:R1457–64. doi: 10.1152/ajpregu.1997.273.4.R1457. [DOI] [PubMed] [Google Scholar]
  • 86.Jensen KT, Carstens J, Pedersen EB. Effect of BNP on renal hemodynamics, tubular function and vasoactive hormones in humans. Am J Physiol. 1998;274:F63–72. doi: 10.1152/ajprenal.1998.274.1.F63. [DOI] [PubMed] [Google Scholar]
  • 87.Taal MW, Nenov VD, Wong W, Satyal SR, Sakharova O, Choi JH, Troy JL, Brenner BM. Vasopeptidase inhibition affords greater renoprotection than angiotensin-converting enzyme inhibition alone. J Am Soc Nephrol. 2001;12:2051–2059. doi: 10.1681/ASN.V12102051. [DOI] [PubMed] [Google Scholar]
  • 88.Voors AA, Gori M, Liu LCY, Claggett B, Zile MR, Pieske B, McMurray JJV, Packer M, Shi V, Lefkowitz MP, Solomon SD. Renal effects of the angiotensin receptor neprilysin inhibitor LCZ696 in patients with heart failure and preserved ejection fraction. Eur J Heart Fail. 2015;17:510–517. doi: 10.1002/ejhf.232. [DOI] [PubMed] [Google Scholar]
  • 89.Appel RG, Wang J, Simonson MS, Dunn MJ. A mechanism by which atrial natriuretic factor mediates its glomerular actions. Am J Physiol. 1986;251:F1036–42. doi: 10.1152/ajprenal.1986.251.6.F1036. [DOI] [PubMed] [Google Scholar]
  • 90.Canaan-Kuhl S, Ostendorf T, Zander K, Koch KM, Floege J. C-type natriuretic peptide inhibits mesangial cell proliferation and matrix accumulation in vivo. Kidney Int. 1998;53:1143–1151. doi: 10.1046/j.1523-1755.1998.00895.x. [DOI] [PubMed] [Google Scholar]
  • 91.Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, Remuzzi G, Snapinn SM, Zhang Z, Shahinfar S. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med. 2001;345:861–869. doi: 10.1056/NEJMoa011161. [DOI] [PubMed] [Google Scholar]
  • 92.Ruggenenti P, Perna A, Gherardi G, Garini G, Zoccali C, Salvadori M, Scolari F, Schena FP, Remuzzi G. Renoprotective properties of ACE-inhibition in non-diabetic nephropathies with non-nephrotic proteinuria. Lancet (London, England) 1999;354:359–364. doi: 10.1016/S0140-6736(98)10363-X. [DOI] [PubMed] [Google Scholar]
  • 93.Kent DM, Jafar TH, Hayward RA, Tighiouart H, Landa M, de Jong P, de Zeeuw D, Remuzzi G, Kamper A-L, Levey AS. Progression risk, urinary protein excretion, and treatment effects of angiotensin-converting enzyme inhibitors in nondiabetic kidney disease. J Am Soc Nephrol. 2007;18:1959–1965. doi: 10.1681/ASN.2006101081. [DOI] [PubMed] [Google Scholar]
  • 94.KDIGO Clinical Practice Guideline for the Management of Blood Pressure in Chronic Kidney Disease KDIGO Clinical Practice Guideline for the Management of Blood Pressure in Chronic Kidney Disease. Kidney Int Suppl. 2012;2:405–414. [Google Scholar]
  • 95.Guan H, Liu Y, Daily A, Police S, Kim M-H, Oddo S, LaFerla FM, Pauly JR, Murphy MP, Hersh LB. Peripherally expressed neprilysin reduces brain amyloid burden: a novel approach for treating Alzheimer’s disease. J Neurosci Res. 2009;87:1462–1473. doi: 10.1002/jnr.21944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Kanemitsu H, Tomiyama T, Mori H. Human neprilysin is capable of degrading amyloid beta peptide not only in the monomeric form but also the pathological oligomeric form. Neurosci Lett. 2003;350:113–116. doi: 10.1016/s0304-3940(03)00898-x. [DOI] [PubMed] [Google Scholar]
  • 97.Park MH, Lee JK, Choi S, Ahn J, Jin HK, Park J-S, Bae J. Recombinant soluble neprilysin reduces amyloid-beta accumulation and improves memory impairment in Alzheimer’s disease mice. Brain Res. 2013;1529:113–124. doi: 10.1016/j.brainres.2013.05.045. [DOI] [PubMed] [Google Scholar]
  • 98.Langenickel TH, Tsubouchi C, Ayalasomayajula S, Pal P, Valentin M-A, Hinder M, Jhee S, Gevorkyan H, Rajman I. The effect of LCZ696 (sacubitril/valsartan) on amyloid-beta concentrations in cerebrospinal fluid in healthy subjects. Br J Clin Pharmacol. 2016;81:878–890. doi: 10.1111/bcp.12861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Cannon JA, Shen L, Jhund PS, Kristensen SL, Kober L, Chen F, Gong J, Lefkowitz MP, Rouleau JL, Shi VC, Swedberg K, Zile MR, Solomon SD, Packer M, McMurray JJV. Dementia-related adverse events in PARADIGM-HF and other trials in heart failure with reduced ejection fraction. Eur J Heart Fail. 2017;19:129–137. doi: 10.1002/ejhf.687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Javaheri S, Parker TJ, Liming JD, Corbett WS, Nishiyama H, Wexler L, Roselle GA. Sleep apnea in 81 ambulatory male patients with stable heart failure. Types and their prevalences, consequences, and presentations. Circulation. 1998;97:2154–2159. doi: 10.1161/01.cir.97.21.2154. [DOI] [PubMed] [Google Scholar]
  • 101.Solin P, Bergin P, Richardson M, Kaye DM, Walters EH, Naughton MT. Influence of pulmonary capillary wedge pressure on central apnea in heart failure. Circulation. 1999;99:1574–1579. doi: 10.1161/01.cir.99.12.1574. [DOI] [PubMed] [Google Scholar]
  • 102.Lanfranchi PA, Braghiroli A, Bosimini E, Mazzuero G, Colombo R, Donner CF, Giannuzzi P. Prognostic value of nocturnal Cheyne-Stokes respiration in chronic heart failure. Circulation. 1999;99:1435–1440. doi: 10.1161/01.cir.99.11.1435. [DOI] [PubMed] [Google Scholar]
  • 103.Cowie MR, Woehrle H, Wegscheider K, Angermann C, d’Ortho M-P, Erdmann E, Levy P, Simonds AK, Somers VK, Zannad F, Teschler H. Adaptive Servo-Ventilation for Central Sleep Apnea in Systolic Heart Failure. N Engl J Med. 2015;373:1095–1105. doi: 10.1056/NEJMoa1506459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kraiczi H, Hedner J, Peker Y, Grote L. Comparison of atenolol, amlodipine, enalapril, hydrochlorothiazide, and losartan for antihypertensive treatment in patients with obstructive sleep apnea. Am J Respir Crit Care Med. 2000;161:1423–1428. doi: 10.1164/ajrccm.161.5.9909024. [DOI] [PubMed] [Google Scholar]
  • 105.Calvin AD, Somers VK, van der Walt C, Scott CG, Olson LJ. Relation of natriuretic peptide concentrations to central sleep apnea in patients with heart failure. Chest. 2011;140:1517–1523. doi: 10.1378/chest.10-2472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Carmona-Bernal C, Quintana-Gallego E, Villa-Gil M, Sanchez-Armengol A, Martinez-Martinez A, Capote F. Brain natriuretic peptide in patients with congestive heart failure and central sleep apnea. Chest. 2005;127:1667–1673. doi: 10.1378/chest.127.5.1667. [DOI] [PubMed] [Google Scholar]
  • 107.Ogawa T, Kiryu-Seo S, Tanaka M, Konishi H, Iwata N, Saido T, Watanabe Y, Kiyama H. Altered expression of neprilysin family members in the pituitary gland of sleep-disturbed rats, an animal model of severe fatigue. J Neurochem. 2005;95:1156–1166. doi: 10.1111/j.1471-4159.2005.03436.x. [DOI] [PubMed] [Google Scholar]
  • 108.Isaac RE, Johnson EC, Audsley N, Shirras AD. Metabolic inactivation of the circadian transmitter, pigment dispersing factor (PDF), by neprilysin-like peptidases in Drosophila. J Exp Biol. 2007;210:4465–4470. doi: 10.1242/jeb.012088. [DOI] [PubMed] [Google Scholar]

RESOURCES