You are on page 1of 12

JACC: HEART FAILURE VOL. 7, NO.

5, 2019

ª 2019 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION

PUBLISHED BY ELSEVIER

MINI-FOCUS ISSUE: THE INTERSECTION OF THE KIDNEY AND THE HEART

STATE-OF-THE-ART REVIEW

Medical Management of Heart Failure


With Reduced Ejection Fraction in
Patients With Advanced Renal Disease
Aaron M. Hein, BS,a Julia J. Scialla, MD, MHS,a,b Daniel Edmonston, MD,a,b Lauren B. Cooper, MD, MHS,a,c
Adam D. DeVore, MD, MHS,a,b Robert J. Mentz, MDa,b

JACC: HEART FAILURE CME/MOC/ECME

This article has been selected as the month’s JACC: Heart Failure CME/MOC/ECME Objectives for This Article: Upon completion of this
CME/MOC/ECME activity, available online at http://www.acc.org/jacc- activity, the learner should be able to: 1) discuss the current use of guide-
journals-cme by selecting the JACC Journals CME/MOC/ECME tab. line-directed medical therapy in patients with heart failure with reduced
ejection fraction (HFrEF) and advanced chronic kidney disease (CKD); 2)
Accreditation and Designation Statement select appropriate management options for patients with HFrEF and
advanced CKD; and 3) identify potential adverse effects and monitoring
The American College of Cardiology Foundation (ACCF) is accredited by
considerations in patients with HFrEF and advanced CKD.
the Accreditation Council for Continuing Medical Education (ACCME)
and the European Board for Accreditation in Cardiology (EBAC) to
CME/MOC/ECME Editor Disclosure: Editor-in-Chief Christopher M.
provide continuing medical education for physicians.
O’Connor, MD, has received consultant fees/honoraria from AbbVie, Inc.,
The ACCF designates this Journal-based CME/MOC activity for a maximum Actelion Pharmaceuticals Ltd., Bayer, Bristol-Myers Squibb, Cardiorentis,
of 1 AMAPRA Category 1 Credit or 1 EBAC Credit. Physicians should only claim Merck & Co., Inc., ResMed, and Roche Diagnostics; and has ownership
credit commensurate with the extent of their participation in the activity. interest in Biscardia, LLC.ExecutiveEditorMona Fiuzat,PharmD, has
Successful completion of this CME activity, which includes participation receivedresearch support from ResMed, Gilead, Critical Diagnostics,
in the evaluation component, enables the participant to earn up to 1 Otsuka, and Roche Diagnostics. Tariq Ahmad, MD, MPH, has received a
Medical Knowledge MOC point in the American Board of Internal Medi- travel scholarship from Thoratec. Abhinav Sharma, MD, has received
cine’s (ABIM) Maintenance of Certification (MOC) program. Participants support from Bayer-Canadian Cardiovascular Society, Alberta Innovates
will earn MOC points equivalent to the amount of CME credits claimed Health Solution, Roche Diagnostics, and Takeda. Mitchell Psotka, MD,
for the activity. It is the CME activity provider’s responsibility to submit PhD, and Kishan Parikh, MD, have no relationships relevant to the
participant completion information to ACCME for the purpose of granting contents of this paper to disclose.
ABIM MOC credit.
Author Disclosures: Dr. Scialla has received research support from Na-
Medical Management of Heart Failure With Reduced Ejection Fraction in tional Institute of Diabetes and Digestive and Kidney Diseases grant
Patients With Advanced Renal Disease will be accredited by the European R01DK111952, GlaxoSmithKline, and Sanofi. Dr. Cooper has received
Board for Accreditation in Cardiology (EBAC) for 1 hour of External CME research support from Abbott Laboratories. Dr. DeVore has received
credits. Each participant should claim only those hours of credit that have research support from Amgen, the American Heart Association, the Na-
actually been spent in the educational activity. The Accreditation Council for tional Heart, Lung, and Blood Institute, and Novartis; and is a consultant
Continuing Medical Education (ACCME) and the European Board for for Novartis. Dr. Mentz has received research support from National In-
Accreditation in Cardiology (EBAC) have recognized each other’s accredita- stitutes of Health grants U01HL125511-01A1, U10HL110312, and
tion systems as substantially equivalent. Apply for credit through the post- R01AG045551-01A1; has received funding from Akros, Amgen,
course evaluation. While offering the credits noted above, this program is not AstraZeneca, Bayer, GlaxoSmithKline, Gilead, Luitpold, Medtronic, Merck
intended to provide extensive training or certification in the field. Sharp & Dohme, Novartis, Otsuka, and ResMed; has received honoraria
from Abbott Laboratory, Amgen, AstraZeneca, Bayer, Janssen, Luitpold
Method of Participation and Receipt of CME/MOC/ECME Certificate Pharmaceuticals, Medtronic, Merck Sharp & Dohme, Novartis, and
ResMed; and serves on advisory boards for Amgen, Luitpold, Merck
To obtain credit for JACC: Heart Failure CME/MOC/ECME, you must:
Sharp & Dohme, and Boehringer Ingelheim. All other authors have
1. Be an ACC member or JACC subscriber.
reported that they have no relationships relevant to the contents of this
2. Carefully read the CME/MOC/ECME-designated article available
paper to disclose. Barry Greenberg, MD, served as Guest Editor for this
online and in this issue of the journal.
paper.
3. Answer the post-test questions. A passing score of at least 70% must
be achieved to obtain credit. Medium of Participation: Print (article only); online (article and quiz).
4. Complete a brief evaluation.
CME/MOC/ECME Term of Approval
5. Claim your CME/MOC/ECME credit and receive your certificate
electronically by following the instructions given at the conclusion of Issue date: May 2019
the activity. Expiration date: April 30, 2020

ISSN 2213-1779/$36.00 https://doi.org/10.1016/j.jchf.2019.02.009


Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
372 Hein et al. JACC: HEART FAILURE VOL. 7, NO. 5, 2019

Heart Failure Therapy and Advanced Renal Disease MAY 2019:371–82

Medical Management of Heart Failure With


Reduced Ejection Fraction in Patients With
Advanced Renal Disease
Aaron M. Hein, BS,a Julia J. Scialla, MD, MHS,a,b Daniel Edmonston, MD,a,b Lauren B. Cooper, MD, MHS,a,c
Adam D. DeVore, MD, MHS,a,b Robert J. Mentz, MDa,b

HIGHLIGHTS
 Patients with HF and advanced chronic kidney disease constitute a substantial population
that experiences poor outcomes.
 Evidence for HF therapies in this population is limited by study exclusion and nonstandard
definitions.
 Renin-angiotensin system inhibitors and beta-blockers have yielded some promising
results, whereas mineralocorticoid receptor antagonists should generally be avoided.
 Directed trials and well-powered studies are needed to examine the potential risk-benefit
balance of HF therapies.

ABSTRACT

Large randomized clinical trials (RCT) supporting guidelines for the management of heart failure with reduced ejection
fraction (HFrEF) have typically excluded patients with advanced chronic kidney disease (CKD). Patients with concomitant
advanced CKD and HFrEF experience poor cardiovascular outcomes and mortality relative to either disease in isolation
and have been shown to consistently receive lower rates of HFrEF guideline-directed medical therapy (GDMT).
This review evaluated recent evidence for the use of GDMT in patients with HFrEF and advanced CKD approaching
dialysis from RCTs and observational cohorts. The authors also discuss the limitations and challenges inherent in the
evidence for GDMT in this population, and offer guidance to clinicians for proper clinical use and future research
directions. (J Am Coll Cardiol HF 2019;7:371–82) © 2019 by the American College of Cardiology Foundation.

P atients with heart failure (HF) experience sig-


nificant morbidity and mortality associated
with chronic kidney disease (CKD). In patients
with HF with reduced ejection fraction (HFrEF),
development of CKD, and end-stage renal disease
due to impaired renal hemodynamics (3,4). Similarly,
patients with impaired renal function are prone to so-
dium and fluid retention and thus are more likely to
the prevalence of CKD greater than stage $G4 develop HF (5). Comorbid CKD is an independent pre-
(Table 1) is approximately 10% (1,2). Pre-existing HF dictor of both short-term and long-term cardiovascu-
predisposes patients to acute kidney injury (AKI), lar outcomes and death in patients with HF, with

From the aDepartment of Medicine, Duke University School of Medicine, Durham, North Carolina; bDuke Clinical Research
Institute, Durham, North Carolina; and the cInova Heart and Vascular Institute, Falls Church, Virginia. Dr. Scialla has received
research support from National Institute of Diabetes and Digestive and Kidney Diseases grant R01DK111952, GlaxoSmithKline, and
Sanofi. Dr. Cooper has received research support from Abbott Laboratories. Dr. DeVore has received research support from Amgen,
the American Heart Association, the National Heart, Lung, and Blood Institute, and Novartis; and is a consultant for Novartis. Dr.
Mentz has received research support from National Institutes of Health grants U01HL125511-01A1, U10HL110312, and
R01AG045551-01A1; has received funding from Akros, Amgen, AstraZeneca, Bayer, GlaxoSmithKline, Gilead, Luitpold, Medtronic,
Merck Sharp & Dohme, Novartis, Otsuka, and ResMed; has received honoraria from Abbott Laboratory, Amgen, AstraZeneca,
Bayer, Janssen, Luitpold Pharmaceuticals, Medtronic, Merck Sharp & Dohme, Novartis, and ResMed; and serves on advisory
boards for Amgen, Luitpold, Merck Sharp & Dohme, and Boehringer Ingelheim. All other authors have reported that they have no
relationships relevant to the contents of this paper to disclose. Barry Greenberg, MD, served as Guest Editor for this paper.

Manuscript received January 27, 2019; accepted February 20, 2019.

Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
JACC: HEART FAILURE VOL. 7, NO. 5, 2019 Hein et al. 373
MAY 2019:371–82 Heart Failure Therapy and Advanced Renal Disease

more advanced renal disease conferring a worse prog- advanced CKD or observational studies with ABBREVIATIONS

nosis (6,7). Patients with HFrEF particularly experi- inherent limitations, as reviewed here. AND ACRONYMS

ence a greater increase in morbidity and mortality Many completed trials, subgroup analyses,
ACE = angiotensin-converting
when advanced CKD is present relative to other HF and observational studies in advanced CKD enzyme
subtypes (6). and HFrEF used serum creatinine (sCr) cutoff
ARB = angiotensin receptor
Despite the high prevalence of concomitant HFrEF values to define levels of renal insufficiency. blocker
and CKD, routine guideline-directed medical therapy Although the use of sCr is a common clinical CKD = chronic kidney disease
(GDMT) of this cohort is often lower than in the approach, sCr alone is an imperfect mea- GDMT = guideline-directed
general HFrEF population (8,9). There remains an surement of kidney function, influenced by medical therapy

unmet need to characterize challenges and recent demographic and patient-specific character- HF = heart failure
therapeutic advances from clinical trials in the HF istics such as age, sex, race, and muscle mass, HFrEF = heart failure with
population, including those with CKD, and to eval- and is used to derive direct estimates of kid- reduced ejection fraction

uate real-world evidence from registries and obser- ney function such as estimated glomerular RCT = randomized controlled

vational cohorts that can inform routine practice. filtration rate (eGFR). With widespread trial

This study evaluated contemporary published adoption of eGFR reporting and increasing aware-
research of pharmacological management of patients ness, recent observational studies and trials largely
with HFrEF and advanced CKD, defined as those with have incorporated eGFR, but challenges remain in
CKD $G4 who are not receiving renal replacement older studies.
therapy. MEDLINE papers published between January Similarly, there is increasing recognition of the
1985 and August 2018 were searched using Medical importance of proteinuria/albuminuria in the evalu-
Subject Headings (MESH) and key words, focusing on ation of kidney dysfunction in patients with HFrEF.
relevant terms for this topic (Online Appendix). We Recent studies have identified that even modest in-
also manually searched reference lists of pertinent creases in proteinuria/albuminuria are independently
reviews and studies to find any relevant citations. All associated with the development of HFrEF and with
citations were screened and evaluated by 1 reviewer morbidity and mortality in those with HFrEF (11–13).
(A.M.H.) to select relevant studies. Unfortunately, many previous studies evaluating
This review discusses the limitations of the current GDMT in patients with advanced CKD, who experi-
evidence and future research directions and offers ence increased proteinuria relative to the general
clinicians guidance regarding the safe use of GDMT in population, do not account for or report data for
this high-risk population. Discussion focuses on proteinuria. Further research should determine the
pharmacotherapy using renin-angiotensin system risk and benefits of GDMT when proteinuria is present
(RAS) inhibitors, angiotensin receptor blockers/ in those with advanced CKD.
neprilysin inhibitors (ARNIs), mineralocorticoid re- Lack of universal outcome definitions represent
ceptor antagonists (MRAs), and beta-blockers and another key limitation. For instance, AKI is a common
briefly considers device therapy in this population.
There is little to no evidence for other HF pharma-
T A B L E 1 Chronic Kidney Disease Categories
cological therapies, such as isosorbide dinitrate and
ivabradine, in advanced CKD, so these therapies were eGFR
eGFR Stages (ml/min per 1.73 m2) Terms
not included in this review. Detailed characteristics
G1 >90 Normal or high
and outcomes for relevant studies are outlined in
G2 60–89 Mildly decreased
Table 2, with studies of note highlighted in the body
G3a 45–59 Mildly to moderately decreased
of the text.
G3b 30–44 Moderately to severely decreased
G4 15–29 Severely decreased
LIMITATIONS OF CURRENT EVIDENCE IN G5 <15 Kidney failure
PATIENTS WITH HFrEF AND ADVANCED CKD Albumin-to-Creatinine Ratio

Albuminuria Albumin Excretion


Many pivotal clinical trials for guideline-directed Categories Rate (mg/24 h) (mg/mmol) (mg/g) Terms

HFrEF therapies excluded patients with severe kid- A1 <30 <3 <30 Normal to mildly increased

ney disease, resulting in limited evidence for A2 30–300 3–30 30–300 Moderately increased
A3 >300 >30 >300 Severely increased
contemporary GDMT in those with advanced CKD and
HFrEF (Central Illustration) (10). Dedicated clinical In the absence of evidence of kidney damage, neither eGFR stage G1 nor G2 fulfill the criteria for CKD. The terms
trials in advanced CKD and HFrEF are uncommon; G2 and A2 are relative to young adult levels. Adapted with permission from the Kidney Disease Improving Global
Outcomes Guidelines (2).
thus, recommendations often rely upon extrapolation eGFR ¼ estimated glomerular filtration rate.
from populations with low numbers or without

Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
374 Hein et al. JACC: HEART FAILURE VOL. 7, NO. 5, 2019

Heart Failure Therapy and Advanced Renal Disease MAY 2019:371–82

T A B L E 2 Outcomes for HF Therapies in Patients with HFrEF and CKD

Follow-Up Exams (n), Population


First Author, Year (Ref. #) Design Time Period and Subgroup Renal Criteria

Renin-angiotensin system inhibitors


Swedberg et al., 1990 (24) PC-RCT* SG analysis 253, 6 months NYHA functional classes IV HFrEF, sCr <3.4 mg/dl; 12% estimated
SG: sCr >1.39 mg/dl with CKD G4
Hillege et al., 2006 (25) PC-RCT SG analysis 2,680, 37.7 median Age >18 yrs, NYHA functional classes sCr <3.0 mg/dl
months II–IV, (LVEF #40%, n ¼ 1,656),
SG: eGFR #45 ml/min per 1.73 m2
Edner et al., 2015 (26) CE-OC 1,204, 1 yr LVEF #39% CKD G4–5 or sCr >2.5 mg/dl
Masoudi et al., 2004 CE-OC 1,258, 1 yr Age $65 yrs, LVEF <40%, after HF sCr >2.5 mg/dl
(Online Ref. 1) hospitalization
Berger et al., 2007 (19) CE-OC 381, 1 yr Age 35–84 yrs, with HF exacerbation CKD G4, n ¼ 238; CKD G5, n ¼ 143
(n ¼ 137 with LVEF <35%)
McAlister et al., 2004 CE-OC 754, 2.5 median yrs Outpatients with HF (57% with CKD $G3; n ¼ 118 with CKD G4–5
(Online Ref. 2) LVEF #35%)
Ahmed et al., 2012 CE-OC 541, 8 yrs Medicare cohort, LVEF #45% eGFR #38.5 ml/min per 1.73 m2
(Online Ref. 3) after HF hospitalization n ¼ 541
Angiotensin receptor blocker/
neprilysin inhibitors
Damman et al., 2018 (31) CER-RCT SG analysis 2,745 NYHA functional classes II–IV, CKD G3–4
LVEF #40%
Solomon et al., 2016 (32) Meta-analysis 14,472 IMPRESS, OVERTURE, -
PARADIGM-HF trial participants
Mineralocorticoid receptor
antagonists
Lu et al., 2016 (41) Meta-analysis 4,935 Adults with CKD CKD G1–5

Inampudi et al., 2014 (42) CE-OC 1,140, 1 yr Hospitalized patients with HF eGFR <45 ml/min per 1.73 m2
and LVEF <45%
Cooper et al., 2017 (43) CE-OC 16,848, 3 yrs Age > 65 yrs, HF hospitalization sCr >2.0 mg/dl or type 2 diabetes
mellitus (mean 1.9 mg/dl)

Beta-blockers
Ghali et al., 2009 (49) PC-RCT SG analysis 493 Age 40-80 yrs, NYHA functional II–IV, CKD $G3b (mean eGFR 36.6 ml/min
LVEF <40% per 1.73 m2)

Erdmann et al., 2001 PC-RCT SG analysis 63, 1.3 yrs NYHA functional classes III–IV, CKD G4–5
(Online Ref. 4) LVEF #35% on ACE inhibitor therapy
Castagno et al., 2010 (50) PC-RCT SG analysis 450, 1.3 yrs NYHA functional classes III–IV, CKD $G3b
LVEF #35% on ACE inhibitor therapy (mean 38.4 ml/min per 1.73 m2)
Badve et al., 2011 (51) Meta-analysis 5,972 Patients with chronic HFrEF CKD $G3

Wali et al., 2011 Meta-analysis 1,116 Patients with systolic left CKD $G3b
(Online Ref. 5) ventricular dysfunction
Nagatomo et al., 2017 (52) CE-RCT 360, 3.8 yrs Age 20–80 yrs, NYHA functional classes Stratification at eGFR 60 ml/min per
II–III, LVEF <40% 1.73 m2
Chang et al., 2013 (9) CE-OC, 2.4 median yrs 668 Age >18 yrs, newly diagnosed HFrEF, CKD $3 or proteinuria, nondialysis;
LVEF <40% 6.3% CKD G4-5
McAlister et al., 2004 CE-OC 419, 2.5 median yrs Outpatients with HF (57% with CKD $G3; n ¼ 118 with CKD G4–5
(Online Ref. 2) LVEF #35%)

The table lists trials and observational studies investigating medical therapies in patients with HFrEF and CKD. Studies not cited in the body of the text are listed in the Online References.
ACE ¼ angiotensin-converting enzyme; ACM ¼ all-cause mortality; ARNI ¼ angiotensin receptor blocker/neprilysin inhibitor; CE-OC ¼ comparative effectiveness observational cohort; CE-RCT ¼
comparative effectiveness randomized controlled trial; CI ¼ confidence interval; eGFR ¼ estimated glomerular filtration rate; G4–5 ¼ stage G4 to G5; HF ¼ heart failure; HR ¼ hazard ratio;
IMPRESS ¼ Inhibition of Metalloprotease by Omapatarilat in Randomized Exercise and Symptoms Study of Heart Failure; LVEF ¼ left ventricular ejection fraction; MACE ¼ major adverse cardiac events;
MRA ¼ mineralocorticoid receptor antagonist; NYHA ¼ New York Heart Association class; OR ¼ odds ratio; OVERTURE ¼ Omapatrilat versus Enalapril Randomized Trial of Utility in Reducing Events;
PARADIGM-HF ¼ Prospective Comparison of ARNI With ACEI to Determine Impact on Global Mortality and Morbidity in Heart Failure; PC-RCT ¼ placebo-controlled randomized controlled trial; RR ¼ risk ratio;
RAS ¼ renin-angiotensin system inhibitor; sCr ¼ serum creatinine; SG ¼ subgroup.

Continued on the next page

feature of decompensated HFrEF, often evaluated as must be sustained or confirmed on repeat testing.
an adverse effect or study outcome. Definitions of the Recent international guidelines in kidney disease
degree of decline, acuity, or timeframe of the change may provide standardized outcome criteria that could
in kidney function vary, and whether the changes reduce this variation in future studies (14).

Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
JACC: HEART FAILURE VOL. 7, NO. 5, 2019 Hein et al. 375
MAY 2019:371–82 Heart Failure Therapy and Advanced Renal Disease

T A B L E 2 Continued

Outcome(s) of Interest
Adjustment Intervention, Comparison Primary Outcome HR/RR (95% CI)

— Enalapril vs. placebo ACM 30% vs. 55%; p ¼ 0.004


— Candesartan vs. placebo Cardiovascular death, unplanned No significant interaction among candesartan, sCr, and primary
readmission, or ACM outcome (p ¼ 0.84)

Propensity matched RAS inhibitor vs. no RAS inhibitor ACM HR ¼ 0.76 (0.67–0.86)
prescription at discharge
Multivariable regression ACE inhibitor vs. no ACE inhibitor ACM RR ¼ 0.65 (0.51–0.80)
prescription at discharge
Risk adjustment models RAS-I vs. no RAS inhibitor prescription 30-day ACM CKD G4: 9.4% vs. 18.5%; p ¼ 0.008
in-hospital CKD G5: 11.9% vs. 22.8%; p ¼ 0.03
Multiple logistic regression ACE inhibitor vs. no ACE inhibitor at 1-yr ACM OR ¼ 0.46 (0.26–0.82)
discharge
Propensity matched RAS inhibitor at target dose vs. no RAS ACM HR ¼ 0.82 (0.70–1.00); reduced ACM for those at target doses
inhibitor prescription at discharge

— Valsartan/sacubitril vs. enalapril Cardiovascular death and HF HR ¼ 0.79 (0.69–0.90)


hospitalization
ARNI vs. single RAS inhibitor therapy Adverse events Increased symptomatic hypotension, but decreased serum
potassium elevation and renal impairment

— MRA vs. non-MRA treatment ACM, MACE, hyperkalemia ACM: RR ¼ 0.78 (0.62–0.97)
MACE: RR ¼ 0.65 (0.50–0.83)
Hyperkalemia: RR ¼ 2.32 (1.83–2.94)
Not significant in patients with HF
Propensity-matched Spironolactone vs. no spironolactone 1-yr all-cause readmission HR ¼ 1.36 (1.13–1.63)
at discharge eGFR <15 ml/min per 1.73 m2: HR ¼ 4.75 (1.84–12.28)
Inverse probability-weighted MRA vs. no MRA prescription at ACM (30-day, 1-yr, 3-yrs) No differences in any ACM;
proportional models discharge 3-yr all-cause readmission: HR ¼ 0.94 (0.89–0.98)
Long-term hyperkalemia readmission: HR ¼ 1.30 (95% CI: 1.11–
1.53)

— Metoprolol succinate vs. placebo ACM, HF mortality, all-cause ACM: HR ¼ 0.41 (95% CI: 0.25–0.68)
hospitalization HF mortality: HR ¼ 0.25 (0.10–0.62)
All-cause hospitalization: HR ¼ 0.61 (0.47–0.79)
— Bisoprolol vs. placebo ACM RR ¼ 0.59 (0.30–1.18)l more permanent treatment
withdrawals in eGFR <30 ml/min per 1.73 m2
— Bisoprolol vs. placebo ACM, composite of ACM or HF HR ¼ 0.71 (0.48–1.05)
hospitalization ACM or HF hospitalization: HR ¼ 0.72 (0.53–0.99)
— Beta-blocker vs. placebo ACM RR ¼ 0.72 (0.64–0.80)
Cardiovascular mortality: RR ¼ 0.66 (0.49–0.89)
— Carvedilol vs. placebo ACM HR ¼ 0.94 (0.72–1.23)
HF mortality: HR ¼ 0.86 (0.61–1.21)
— 2.5 mg vs. 5 mg vs. 20 mg Change in LVEF No difference in LVEF increase between eGFR >60 ml/min per
carvedilol 1.73 m2 and eGFR <60 ml/min per 1.73 m2 for all groups
Nested covariable models New user of beta-blocker vs. ACM HR ¼ 0.75 (0.51–1.12)
no beta-blocker Death or HF hospitalization: HR ¼ 0.67 (0.51–0.88)
Multiple logistic regression Beta-blocker vs. no beta-blocker 1-yr ACM OR ¼ 0.40 (0.23–0.70)
prescription at discharge

Furthermore, worsening renal function suggestive of Importantly, many results from observational
AKI may not reflect true kidney injury in decom- studies are limited by potential confounding, as the
pensated HFrEF or during initiation of RAS inhibitor sickest, most fragile patients, already at increased
therapy (15,16). Conversely, improving measurements risk for poor outcomes, may be less likely to receive
of function during decompensated HFrEF have been GDMT due to concerns for adverse effects. This reality
associated with poor outcomes, reflecting the uncer- may result in attribution of morbidity and mortality
tainty surrounding renal function measurement in benefit to GDMT in an observational cohort that is
this clinical setting (17). truly due to unaccounted factors in participant

Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
376 Hein et al. JACC: HEART FAILURE VOL. 7, NO. 5, 2019

Heart Failure Therapy and Advanced Renal Disease MAY 2019:371–82

baseline status. Circumventing this limitation is enalapril significantly reduced all-cause mortality in
challenging and requires high-quality data sources patients with sCr >1.39 mg/dl compared with placebo
with capture of relevant clinical variables and but did not have a significant effect in those with
rigorous analytic techniques including new-user de- sCr <1.39 mg/dl (24). A post hoc CHARM (Candesartan
signs, propensity scoring, and marginal structural in Heart Failure-Assessment of Reduction in Mortal-
models, among others. ity and Morbidity; NCT00634400) analysis found no
Several medications can cause adverse effects in interaction between renal function and the cardio-
advanced CKD that affect the risk-benefit calculation. vascular benefits of candesartan in those with HF and
For instance, RAS inhibitor therapies may result in or CKD $G3b. Candesartan resulted in a greater
exacerbate hyperkalemia or precipitate AKI in this absolute risk reduction in those with significant
vulnerable population. Due to renal clearance of renal dysfunction who experienced overall worse
many medications used in HFrEF, dose adjustments outcomes (25).
and monitoring may vary greatly based on the degree Only observational data have been reported for the
of renal impairment, as outlined in Figure 1. Concerns population with advanced CKD and HFrEF. In a
for adverse events and polypharmacy often shift the propensity-matched cohort study of patients with
cost-benefit ratio of medication prescription for these HFrEF and CKD $G4, RAS inhibitor therapy was
patients. associated with increased 1-year survival compared
Thus, it is known that those with CKD and HFrEF with nonusers, with efficacy similar to that seen in
experience worse outcomes on average, and unsur- patients without CKD, particularly in those with New
prisingly, they receive lower rates of GDMT. A recent York Heart Association functional class III symptoms
national registry study of patients with end-stage (26). Another cohort study found RAS inhibitor ther-
renal disease found 40% of patients approaching apy during HF hospitalization was associated with
dialysis were receiving an angiotensin-converting decreased 30-day all-cause mortality in patients with
enzyme (ACE) inhibitor or an angiotensin receptor CKD G4 to G5. That study did not independently
blocker (ARB), regardless of HFrEF history, and 60% evaluate patients with HFrEF, however (19). Other
to 75% of those with HFrEF were receiving a beta- studies have demonstrated improved outcomes and a
blocker (9). Other studies in outpatients with potentially higher absolute risk reduction in this
advanced CKD and HF found prescription rates of population using RAS inhibitor therapy (Table 2).
50% to 70%, often 20% lower than their non-CKD Although the aforementioned studies suggest
counterparts (18,19). Studies have shown that GDMT therapeutic benefit for RAS inhibitor therapy in those
improves after initiation of dialysis (19). Given the with HFrEF and advanced CKD, concern for increased
limited life expectancy and comorbid conditions adverse renal outcomes and electrolyte disturbances
often experienced by these patients, in combination are substantial. In a subgroup of patients with un-
with limited clinical trial evidence for efficacy in this specified HF from a population-based study, those
group, clinicians may be dissuaded from initiating with “moderate-to-severe” CKD had a higher risk of
GDMT. an increase of >30% in sCr relative to that in other
groups, and this adverse event conferred an increased
RENIN-ANGIOTENSIN SYSTEM INHIBITORS risk of death in those with comorbid HF (27).
Furthermore, a meta-analysis of patients with
ACE inhibitors and ARBs in large randomized CKD $G3, in which HF history was unspecified, found
controlled trials (RCTs) have decreased cardiovascu- a roughly doubled risk of hyperkalemia with RAS in-
lar events, HF hospitalizations, and mortality, while hibitor monotherapy relative to that of placebo or
improving quality of life in those with HFrEF (20,21). active controls, although RAS inhibitor monotherapy
However, many RCTs that established ACE inhibitors also reduced all-cause mortality (28). However, in an
and ARBs as integral therapies excluded patients with outpatient cohort with HFrEF and CKD G3 to G4,
advanced CKD (10). Although subgroup analyses of chronic RAS inhibitor therapy or up-titration was not
many major trials suggested benefit in those with associated with worsened renal outcomes or hyper-
CKD $G3 similar to or greater than that in the larger kalemia after 1 year, although that study may have
trial (22,23), data for those with advanced CKD are excluded participants with previous adverse effects
scarce. related to RAS inhibitor administration (29).
Of the RAS inhibitor RCTs, only a small proportion Recently, ARNIs have been shown to effectively
of 2 trials included patients with advanced CKD. In a reduce HFrEF hospitalizations and mortality (30).
subgroup analysis of the CONSENSUS (Cooperative However, the tolerability and safety of this new class
North Scandinavian Enalapril Survival Study), of medications in patients with comorbid advanced

Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
JACC: HEART FAILURE VOL. 7, NO. 5, 2019 Hein et al. 377
MAY 2019:371–82 Heart Failure Therapy and Advanced Renal Disease

C ENTR AL I LL U STRA T I ON Limitations in the Evidence for Patients With HFrEF and Advanced CKD

Hein, A.M. et al. J Am Coll Cardiol HF. 2019;7(5):371–82.

Current challenges affecting the paucity of evidence in patients with HFrEF and advanced CKD contributing to reduced utilization of medical therapy in this vulnerable
population. CKD ¼ chronic kidney disease; HFrEF ¼ heart failure with reduced ejection fraction.

CKD is unclear. A PARADIGM (Prospective Compari- excluded patients with CKD G4 to G5, leaving its
son of ARNI with ACEI to Determine Impact on applicability to patients with advanced CKD untested.
Global Mortality and Morbidity in Heart Failure; Regarding adverse events, 1 meta-analysis found
NCT01035255) trial subgroup analysis found patients rates of renal impairment and hyperkalemia were
with CKD G3 receiving sacubitril/valsartan had decreased in patients receiving ARNI therapy relative
decreased rates of cardiovascular mortality and HF to those in patients with HFrEF taking ACE inhibitor
hospitalization compared to those receiving enalapril therapy, although ARNI therapy resulted in increased
in addition to reductions in all-cause mortality, rates of symptomatic hypotension (32).
noting a higher absolute risk reduction than those
without CKD (30,31). Furthermore, the rate of CKD RECOMMENDATIONS AND
progression was reduced in those treated with sacu- FUTURE RESEARCH DIRECTIONS
bitril/valsartan compared with those receiving ena-
lapril, although those patients also experienced an Given the weak evidence base for ACE inhibitor/ARBs
increase in urinary albumin/creatinine ratio. Notably, in this population, current American College of Car-
the increase in albuminuria did not affect the 30-day diology Foundation/American Heart Association
risk of cardiovascular mortality or HF hospitalization (ACCF/AHA) guidelines suggest caution in using RAS
(31). Despite these encouraging findings, the study inhibitor therapy in those with comorbid CKD $G4

Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
378 Hein et al. JACC: HEART FAILURE VOL. 7, NO. 5, 2019

Heart Failure Therapy and Advanced Renal Disease MAY 2019:371–82

F I G U R E 1 Recommended Dosage Adjustments and Monitoring Considerations in Patients With HFrEF and CKD Stages G4 to G5

Dosage adjustments and monitoring adjustments are shown for patients with HFrEF and CKD stages G4 to G5. Adjustments refer to initial dosage with titration as
tolerated based on safety. Dosing adjustments primarily derived from manufacturer labeling and package insert data when possible. †Reduced rate of titration
recommended for captopril. ‡Use with caution. §Twice daily dosing. ACE-Is ¼ angiotensin-converting enzyme inhibitor; ARNI ¼ angiotensin receptor blocker/neprilysin
inhibitor; eGFR ¼ estimated glomerular filtration rate; HFrEF ¼ heart failure with reduced ejection fraction; MRA ¼ mineralocorticoid receptor antagonist.

(33). Kidney Disease: Improving Global Outcomes Although subanalyses of major RCTs showed cardio-
(KDIGO) guidelines recommend specialist supervision vascular benefits in those with mild to moderate CKD
for the use of RAS inhibitor in patients with CKD $G4, and HFrEF (10,38,39), concern for adverse effects,
a reduced starting dose for those with CKD $G3b, notably the heightened risk of hyperkalemia observed
eGFR monitoring, and serum potassium measure- after the completion of the Randomized Aldactone
ment within 1 week of initiation; KDIGO guidelines Evaluation Study (40), resulted in the exclusion of
also advise against routine discontinuation once patients with advanced CKD and HFrEF from major
eGFR reaches <30 ml/min/1.73 m 2 (2,34). However, trials. Although a meta-analysis of 12 studies reported
many of the included studies suggest a survival use of MRA in a general population of patients with
benefit with the use of RAS inhibitor therapy in this CKD $G3 was associated with reduced all-cause
population, and the introduction of therapies to pre- mortality, these mortality benefits were not signifi-
vent hyperkalemia may lessen safety concerns (35). cant in studies incorporating an HF subanalysis (41).
Robust trials with adequate safeguards are needed to Observational studies of MRA use in patients with
confirm the potential benefits in patients with HFrEF HFrEF and advanced CKD have also been scant, but
and advanced CKD. mainly raise concerns about adverse effects without
Current guidelines do not comment on the use of clear evidence of benefit. A propensity-matched
ARNI therapy in those with CKD but suggest the po- analysis of the Alabama Heart Failure registry found
tential for renal insufficiency with this medication MRA prescription at discharge for hospitalized pa-
(33). Although the present evidence is reassuring for tients with HFrEF and CKD $G3b was associated with
ARNI therapy in those with CKD G3, further studies increased readmission rates, as well as the combined
are needed to elucidate whether these beneficial ef- endpoint of 1-year all-cause mortality or readmission;
fects extend to those with comorbid CKD $G4. the risk of 1-year all-cause readmission was greatly
increased in those with CKD G5 not receiving dialysis
MINERALOCORTICOID RECEPTOR (42).
ANTAGONISTS A study incorporating the Get With The Guidelines-
HF registry and Medicare claims found MRA
In major RCTs, MRAs have consistently improved discharge prescription, after adjustment for patient
symptoms while hospitalizations and mortality characteristics and concomitant medication use, was
have been reduced in patients with HFrEF (36,37). not associated with differences in 30-day, 1-year, or

Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
JACC: HEART FAILURE VOL. 7, NO. 5, 2019 Hein et al. 379
MAY 2019:371–82 Heart Failure Therapy and Advanced Renal Disease

3-year mortality for hospitalized patients with HF included more patients with advanced CKD than in
and comorbid diabetes mellitus type 2 or an sCr RAS inhibitor trials, likely due to decreased safety
>2.0 mg/dl. In contrast, MRA therapy was associated concerns with initiation (10). Multiple subgroup ana-
with modest decreases in 1-year and 3-year all-cause lyses support the benefits of beta-blockers in those
readmissions. Increased risk of short- and long-term with advanced CKD. Analysis of MERIT-HF (Meto-
hyperkalemia readmissions was also observed (43). prolol CR/XL Randomised Intervention Trial in
Congestive Heart Failure) found participants with
RECOMMENDATIONS AND FUTURE
CKD $G3b who received metoprolol had significant
RESEARCH DIRECTIONS
decreases in all-cause mortality, HF mortality, and
all-cause hospitalization (49). One analysis of the
ACCF/AHA guidelines recommend against using MRA
CIBIS-II (Cardiac Insufficiency Bisoprolol Study II)
therapy in men with sCr >2.5 mg/dl and in women
trial likewise found patients with CKD $G3b had
with sCr >2.0 mg/dl due to concern for hyperkalemia,
decreased risk of either all-cause mortality or HF
AKI, and inadequate evidence from RCTs (44). KDIGO
hospitalization, with a lower number needed to treat
guidelines suggest a reduced starting dose for those
due to poor outcomes in this subgroup (50).
with CKD $G3b, eGFR monitoring, and serum potas-
In a large meta-analysis, beta-blocker therapy in
sium measurement within 1 week of initiation but
patients with CKD $G3a reduced all-cause and car-
advise against routine discontinuation once eGFR
diovascular mortality risk but increased the risk of
reaches <30 ml/min/1.73 m 2 (2). The reviewed studies
bradycardia and hypotension; no differences in
suggest that results of MRA therapy in those with
hyperkalemia were noted (51). The effect of increased
HFrEF and advanced CKD raise valid safety concerns
LVEF with carvedilol treatment seen overall after 56
without clear benefit independent of concomitant use
weeks was not blunted for patients with CKD $G3a in
of RAS inhibitor therapy.
the Japanese congestive heart failure trial, which
NOVEL POTASSIUM BINDERS AS randomized patients with HFrEF to receive 3
ADJUNCT THERAPY different doses of carvedilol (52). Each of those
studies suffered from few included participants with
The emergence of novel potassium binders such as CKD G4 to G5, some below 10%, which limits the
patiromer and sodium zirconium cyclosilicate may generalizability to those with advanced CKD.
mitigate the hyperkalemia associated with RAS in- Observational studies involving beta-blocker ther-
hibitor agents, particularly in vulnerable CKD pop- apy in patients with CKD and HFrEF have also been
ulations (35). In a recent trial, patients with CKD G3 to hindered by small proportions of patients with
G4 receiving RAS inhibitors who experienced advanced CKD and have reported mixed results for
elevated serum potassium levels received either mortality benefits. For example, in a retrospective
patiromer or placebo. In the HF subgroup, patiromer study of 668 patients with HFrEF and CKD $G3, beta-
therapy resulted in a significant decrease in serum blocker therapy was not associated with decreased
potassium levels and hyperkalemia recurrence (45). all-cause mortality, but it was associated with a
Patiromer therapy may also decrease the risk of life- decreased risk of the composite outcome of death or
threatening hyperkalemia in those treated with HF hospitalization. There were only 47 patients with
MRAs and recently has been tolerated in CKD patients CKD G4 to G5 in that study, however (53).
up-titrated on spironolactone therapy (46). Adjunc-
tive use of these therapies may help shift the feasi- RECOMMENDATIONS AND
bility of the use of RAS inhibitors in this group. FUTURE RESEARCH DIRECTIONS
However, these therapies also require consideration
of potential adverse events, such as binding of con- Beta-blockers in HFrEF patients have often shown
current oral medications, gastrointestinal upset, hy- beneficial effects on morbidity and mortality in those
pomagnesemia with patiromer, and worsening edema with CKD. ACCF/AHA guidelines support beta-blocker
from an increased sodium load with sodium zirco- therapy in patients with comorbid CKD and HF, and
nium cyclosilicate (35). KDIGO guidelines, while recommending beta-blocker
therapy, suggest dose reduction by 50% for those
BETA-BLOCKERS with CKD G4 to G5 (2,34,44). However, although
many findings from subgroup analyses of RCTs are
Beta-blockers have consistently reduced morbidity promising, there is conflicting observational evidence
and mortality in patients with HFrEF (47,48). Large for efficacy in those with advanced CKD, primarily
RCTs evaluating beta-blocker use in HFrEF often due to a low number of participants included. Careful

Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
380 Hein et al. JACC: HEART FAILURE VOL. 7, NO. 5, 2019

Heart Failure Therapy and Advanced Renal Disease MAY 2019:371–82

monitoring for side effects such as bradyarrhythmia CONCLUSIONS


and hypotension is suggested with beta-blocker
initiation in this population. The lack of high-quality evidence for GDMT in pa-
tients with HFrEF and advanced CKD leaves little
IMPLANTABLE DEVICE THERAPY
guidance for the management of this vulnerable
population. GDMT in this population can yield
Device therapy in patients with HFrEF and advanced
dangerous side effects, but overall poor outcomes
CKD is another area of continued controversy, as
may result in a greater absolute therapeutic benefit.
multiple trials have shown benefit in patient pop-
Although RAS inhibitor therapy in patients with
ulations with HFrEF, but application to the advanced
HFrEF and advanced CKD has generally shown posi-
CKD population is less clear. The risk of sudden car-
tive outcomes, the role of MRAs is less clear. ARNI
diac death increases with worsening kidney
therapy requires thorough evaluation in CKD but may
dysfunction (54), suggesting patients with advanced
promise decreased safety concerns compared with
CKD may benefit from device therapy with an
ACE inhibitor/ARB therapy. Potassium binding ther-
implantable cardioverter-defibrillator (ICD). Howev-
apies may prove beneficial as adjunctive therapy to
er, a meta-analysis of 3 primary prevention ICD RCTs
RAS inhibitor therapies in patients with CKD experi-
found no survival benefit from ICD therapy in pa-
encing hyperkalemia. Beta-blockers, although often
tients with CKD $G3 (55). Also, a secondary analysis
beneficial in populations with comorbid CKD-HFrEF,
of the Multicenter Automatic Defibrillator Implanta-
require careful monitoring for side effects, particu-
tion Trial-II found ICD therapy was not associated
larly in patients nearing dialysis. Finally, consider-
with decreased mortality in patients with eGFR <35
2 ation of benefit from device therapy in this
ml/min per 1.73 m (54). Another meta-analysis of 4
population must weigh the elevated risks of device
primary prevention ICD trials found the therapeutic
placement, life expectancy, and comorbidity burden.
benefit was attenuated with increasing comorbidity
Clearly, adequately powered clinical trials with care-
burden (56). Although 1 study showed eGFR
ful adverse event monitoring and follow-up are
improvement with cardiac resynchronization therapy
required to evaluate the benefits of these therapies.
in those with CKD G3 (57), no studies specifically
evaluated the benefit of cardiac resynchronization
therapy in advanced CKD (58). In this population, ADDRESS FOR CORRESPONDENCE: Dr. Robert J.
factors such as life expectancy and periprocedural Mentz, Duke Clinical Research Institute, 2301 Erwin
risk must be balanced when considering the potential Road, P.O. Box 17969, Durham, North Carolina 27715.
benefit of device therapy. E-mail: robert.mentz@duke.edu.

REFERENCES

1. McAlister FA, Ezekowitz J, Tarantini L, et al. failure: the atherosclerosis risk in communities 10. Damman K, Tang WH, Felker GM, et al. Current
Renal dysfunction in patients with heart failure (ARIC) study. J Am Soc Nephrol 2007;18:1307–15. evidence on treatment of patients with chronic
with preserved versus reduced ejection fraction: systolic heart failure and renal insufficiency:
6. Lofman I, Szummer K, Dahlstrom U, Jernberg T,
impact of the new Chronic Kidney Disease- practical considerations from published data. J Am
Lund LH. Associations with and prognostic impact
Epidemiology Collaboration Group formula. Circ Coll Cardiol 2014;63:853–71.
of chronic kidney disease in heart failure with
Heart Fail 2012;5:309–14.
preserved, mid-range, and reduced ejection frac- 11. de Boer RA, Nayor M, deFilippi CR, et al. As-
2. Kidney disease: improving global outcomes tion. Eur J Heart Fail 2017;19:1606–14. sociation of cardiovascular biomarkers with inci-
(KDIGO) CKD work group. KDIGO 2012 clinical dent heart failure with preserved and reduced
7. Damman K, Valente MA, Voors AA,
practice guideline for the evaluation and man- ejection fraction. JAMA Cardiol 2018;3:215–24.
O’Connor CM, van Veldhuisen DJ, Hillege HL.
agement of chronic kidney disease. Kidney Int
Renal impairment, worsening renal function, and 12. Nayor M, Larson MG, Wang N, et al. The as-
2013;3: Suppl:1–150.
outcome in patients with heart failure: an updated sociation of chronic kidney disease and micro-
3. Rahman M, Xie D, Feldman HI, et al. Association meta-analysis. Eur Heart J 2014;35:455–69. albuminuria with heart failure with preserved vs.
between chronic kidney disease progression and reduced ejection fraction. Eur J Heart Fail 2017;19:
8. Heywood JT, Fonarow GC, Yancy CW, et al.
cardiovascular disease: results from the CRIC 615–23.
Influence of renal function on the use of guideline-
Study. Am J Nephrol 2014;40:399–407.
recommended therapies for patients with heart
13. Anand IS, Bishu K, Rector TS, Ishani A,
4. Bosselmann H, Gislason G, Gustafsson F, et al. failure. Am J Cardiol 2010;105:1140–6. Kuskowski MA, Cohn JN. Proteinuria, chronic
Incidence and predictors of end-stage renal dis- kidney disease, and the effect of an angiotensin
9. Chang TI, Zheng Y, Montez-Rath ME,
ease in outpatients with systolic heart failure. Circ
Winkelmayer WC. Antihypertensive medication use receptor blocker in addition to an angiotensin-
Heart Fail 2013;6:1124–31.
in older patients transitioning from chronic kidney converting enzyme inhibitor in patients with
5. Kottgen A, Russell SD, Loehr LR, et al. Reduced disease to end-stage renal disease on dialysis. Clin moderate to severe heart failure. Circulation
kidney function as a risk factor for incident heart J Am Soc Nephrol 2016;11:1401–12. 2009;120:1577–84.

Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
JACC: HEART FAILURE VOL. 7, NO. 5, 2019 Hein et al. 381
MAY 2019:371–82 Heart Failure Therapy and Advanced Renal Disease

14. Baigent C, Herrington WG, Coresh J, et al. 27. Schmidt M, Mansfield KE, Bhaskaran K, et al. Randomized Aldactone Evaluation Study. N Engl J
Challenges in conducting clinical trials in Serum creatinine elevation after renin-angiotensin Med 2004;351:543–51.
nephrology: conclusions from a Kidney Disease- system blockade and long term cardiorenal risks:
41. Lu R, Zhang Y, Zhu X, et al. Effects of miner-
Improving Global Outcomes (KDIGO) contro- cohort study. BMJ 2017;356.
alocorticoid receptor antagonists on left ventric-
versies conference. Kidney Int 2017;92:297–305.
28. Xie X, Liu Y, Perkovic V, et al. Renin-angio- ular mass in chronic kidney disease patients: a
15. Bakris GL, Weir MR. Angiotensin-converting tensin system inhibitors and kidney and cardio- systematic review and meta-analysis. Int Urol
enzyme inhibitor-associated elevations in serum vascular outcomes in patients with CKD: A Nephrol 2016;48:1499–509.
creatinine: is this a cause for concern? Arch Intern Bayesian network meta-analysis of randomized
42. Inampudi C, Parvataneni S, Morgan CJ, et al.
Med 2000;160:685–93. clinical trials. Am J Kidney Dis 2016;67:728–41.
Spironolactone use and higher hospital read-
16. Fudim M, Loungani R, Doerfler SM, et al. 29. Frohlich H, Nelges C, Tager T, et al. Long-term mission for Medicare beneficiaries with heart fail-
Worsening renal function during decongestion changes of renal function in relation to ace in- ure, left ventricular ejection fraction <45%, and
among patients hospitalized for heart failure: hibitor/angiotensin receptor blocker dosing in estimated glomerular filtration rate <45 ml/min/
findings from the Evaluation Study of Congestive patients with heart failure and chronic kidney 1.73 m(2). Am J Cardiol 2014;114:79–82.
Heart Failure and Pulmonary Artery Catheteriza- disease. Am Heart J 2016;178:28–36.
43. Cooper LB, Lippmann SJ, Greiner MA, et al.
tion Effectiveness (ESCAPE) trial. Am Heart J 30. McMurray JJ, Packer M, Desai AS, et al. Use of mineralocorticoid receptor antagonists in
2018;204:163–73. Angiotensin-neprilysin inhibition versus enalapril patients with heart failure and comorbid diabetes

17. Testani JM, McCauley BD, Chen J, Coca SG, in heart failure. N Engl J Med 2014;371:993–1004. mellitus or chronic kidney disease. J Am Heart
Assoc 2017 2017 Dec 23;6(12). pii:e006540.
Cappola TP, Kimmel SE. Clinical characteristics and 31. Damman K, Gori M, Claggett B, et al. Renal
outcomes of patients with improvement in renal effects and associated outcomes during 44. Yancy CW, Jessup M, Bozkurt B, et al. 2013
function during the treatment of decompensated angiotensin-neprilysin inhibition in heart failure. ACCF/AHA guideline for the management of heart
heart failure. J Card Fail 2011;17:993–1000. J Am Coll Cardiol HF 2018;6:489–98. failure: a report of the American College of Car-
diology Foundation/American Heart Association
18. Dounaevskaia V, Yan AT, Charytan D, et al. The 32. Solomon SD, Claggett B, McMurray JJ,
Task Force on Practice Guidelines. J Am Coll Car-
management of left ventricular systolic dysfunc- Hernandez AF, Fonarow GC. Combined neprilysin
diol 2013;62:e147–239.
tion in patients with advanced chronic kidney and renin-angiotensin system inhibition in heart
disease. J Nephrol 2011;24:41–9. failure with reduced ejection fraction: a meta- 45. Pitt B, Bakris GL, Bushinsky DA, et al. Effect of
analysis. Eur J Heart Fail 2016;18:1238–43. patiromer on reducing serum potassium and pre-
19. Berger AK, Duval S, Manske C, et al. Angio-
venting recurrent hyperkalaemia in patients with
tensin-converting enzyme inhibitors and angio- 33. Yancy CW, Jessup M, Bozkurt B, et al. 2016
heart failure and chronic kidney disease on RAAS
tensin receptor blockers in patients with ACC/AHA/HFSA focused update on new pharma-
inhibitors. Eur J Heart Fail 2015;17:1057–65.
congestive heart failure and chronic kidney dis- cological therapy for heart failure: an update of
ease. Am Heart J 2007;153:1064–73. the 2013 ACCF/AHA guideline for the manage- 46. Pitt B, Bushinsky DA, Kitzman DW, et al.
ment of heart failure: a report of the American Evaluation of an individualized dose titration
20. Consensus Trial Study Group. Effects of ena- College of Cardiology/American Heart Association regimen of patiromer to prevent hyperkalaemia in
lapril on mortality in severe congestive heart Task Force on Clinical Practice Guidelines and the patients with heart failure and chronic kidney
failure. Results of the Cooperative North Scandi-
Heart Failure Society of America. J Am Coll Cardiol disease. ESC Heart Fail 2018;5:257–66.
navian Enalapril Survival Study (CONSENSUS).
2016;134:e282–93.
N Engl J Med 1987;316:1429–35. 47. Packer M, Bristow MR, Cohn JN, et al. The
34. Herzog CA, Asinger RW, Berger AK, et al. effect of carvedilol on morbidity and mortality in
21. Pfeffer MA, Swedberg K, Granger CB, et al. Cardiovascular disease in chronic kidney disease. A patients with chronic heart failure. U.S. Carvedilol
Effects of candesartan on mortality and morbidity clinical update from Kidney Disease: Improving Heart Failure Study Group. N Engl J Med 1996;
in patients with chronic heart failure: the CHARM- Global Outcomes (KDIGO). Kidney Int 2011;80: 334:1349–55.
Overall programme. Lancet 2003;362:759–66. 572–86.
48. MERIT-HF Study Group. Effect of metoprolol
22. Bowling CB, Sanders PW, Allman RM, et al. 35. Packham DK, Kosiborod M. Potential new CR/XL in chronic heart failure: Metoprolol CR/XL
Effects of enalapril in systolic heart failure pa- agents for the management of hyperkalemia. Am J Randomised Intervention Trial in Congestive Heart
tients with and without chronic kidney disease: Cardiovasc Drugs 2016;16:19–31. Failure (MERIT-HF). Lancet 1999;353:2001–7.
insights from the SOLVD treatment trial. Int J
36. Zannad F, McMurray JJ, Krum H, et al. Epler- 49. Ghali JK, Wikstrand J, Van Veldhuisen DJ,
Cardiol 2013;167:151–6.
enone in patients with systolic heart failure and et al. The influence of renal function on clinical
23. Ahmed A, Love TE, Sui X, Rich MW. Effects of mild symptoms. N Engl J Med 2011;364:11–21. outcome and response to beta-blockade in systolic
angiotensin-converting enzyme inhibitors in sys- heart failure: insights from Metoprolol CR/XL
37. Pitt B, Zannad F, Remme WJ, et al. The effect
tolic heart failure patients with chronic kidney Randomized Intervention Trial in Chronic HF
of spironolactone on morbidity and mortality in
disease: a propensity score analysis. J Card Fail (MERIT-HF). J Card Fail 2009;15:310–8.
patients with severe heart failure. Randomized
2006;12:499–506.
Aldactone Evaluation Study Investigators. N Engl J 50. Castagno D, Jhund PS, McMurray JJ, et al.
24. Swedberg K, Eneroth P, Kjekshus J, Snapinn S. Med 1999;341:709–17. Improved survival with bisoprolol in patients with
Effects of enalapril and neuroendocrine activation heart failure and renal impairment: an analysis of
38. Vardeny O, Wu DH, Desai A, et al. Influence of
on prognosis in severe congestive heart failure the cardiac insufficiency bisoprolol study II (CIBIS-
baseline and worsening renal function on efficacy
(follow-up of the CONSENSUS trial). CONSENSUS II) trial. Eur J Heart Fail 2010;12:607–16.
of spironolactone in patients with severe heart
trial study group. Am J Cardiol 1990;66: Suppl D: failure: insights from RALES (Randomized Aldac- 51. Badve SV, Roberts MA, Hawley CM, et al. Effects
40–5. tone Evaluation Study). J Am Coll Cardiol 2012; of beta-adrenergic antagonists in patients with
25. Hillege HL, Nitsch D, Pfeffer MA, et al. Renal 60:2082–9. chronic kidney disease: a systematic review and
function as a predictor of outcome in a broad meta-analysis. J Am Coll Cardiol 2011;58:1152–61.
39. Eschalier R, McMurray JJ, Swedberg K, et al.
spectrum of patients with heart failure. Circulation Safety and efficacy of eplerenone in patients at 52. Nagatomo Y, Yoshikawa T, Okamoto H,
2006;113:671–8. high risk for hyperkalemia and/or worsening renal Kitabatake A, Hori M, Investigators JC. Anemia is
function: analyses of the EMPHASIS-HF study associated with blunted response to beta-blocker
26. Edner M, Benson L, Dahlstrom U, Lund LH.
subgroups (Eplerenone in Mild Patients Hospital- therapy using carvedilol—insights from Japanese
Association between renin-angiotensin system
antagonist use and mortality in heart failure with ization And SurvIval Study in Heart Failure). J Am Chronic Heart Failure (J-CHF) study. Circ J 2018;
severe renal insufficiency: a prospective pro- Coll Cardiol 2013;62:1585–93. 82:691–8.

pensity score-matched cohort study. Eur Heart J 40. Juurlink DN, Mamdani MM, Lee DS, et al. 53. Chang TI, Yang J, Freeman JV, Hlatky MA,
2015;36:2318–26. Rates of hyperkalemia after publication of the Go AS. Effectiveness of beta-blockers in heart

Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.
382 Hein et al. JACC: HEART FAILURE VOL. 7, NO. 5, 2019

Heart Failure Therapy and Advanced Renal Disease MAY 2019:371–82

failure with left ventricular systolic dysfunction 56. Steinberg BA, Al-Khatib SM, Edwards R, et al.
and chronic kidney disease. J Card Fail 2013;19: Outcomes of implantable cardioverter- KEY WORDS chronic kidney disease,
176–82. defibrillator use in patients with comorbidities: heart failure management, heart failure with
results from a combined analysis of 4 randomized reduced ejection fraction
54. Goldenberg I, Moss AJ, McNitt S, et al. Re-
clinical trials. J Am Coll Cardiol HF 2014;2:623–9.
lations among renal function, risk of sudden car-
A PPE NDI X For sample MEDLINE searches
diac death, and benefit of the implanted cardiac 57. Boerrigter G, Costello-Boerrigter LC,
and supplemental references, please see the
defibrillator in patients with ischemic left ven- Abraham WT, et al. Cardiac resynchronization
online version of this paper.
tricular dysfunction. Am J Cardiol 2006;98: therapy improves renal function in human heart
485–90. failure with reduced glomerular filtration rate.
J Card Fail 2008;14:539–46. Go to http://www.acc.org/
55. Pun PH, Al-Khatib SM, Han JY, et al.
jacc-journals-cme to take
Implantable cardioverter-defibrillators for primary 58. Cannizzaro LA, Piccini JP, Patel UD,
the CME/MOC/ECME
prevention of sudden cardiac death in CKD: a Hernandez AF. Device therapy in heart failure pa-
quiz for this article.
meta-analysis of patient-level data from 3 ran- tients with chronic kidney disease. J Am Coll Car-
domized trials. Am J Kidney Dis 2014;64:32–9. diol 2011;58:889–96.

Descargado para Anonymous User (n/a) en Univ Del Rosario de ClinicalKey.es por Elsevier en mayo 08, 2019.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2019. Elsevier Inc. Todos los derechos reservados.

You might also like